ABSTRACTCancer immunotherapy uses the body’s immune system to combat cancer, marking a significant advancement in treatment. This review traces its evolution from the late 19th century to its current status. It began with William Coley’s pioneering work using bacterial toxins to stimulate the immune system against cancer cells, establishing the foundational concept of immunotherapy. In the mid-20th century, cytokine therapies like interferons and interleukins emerged, demonstrating that altering the immune response could reduce tumors and highlighting the complex interplay between cancer and the immune system. The discovery of immune checkpoints, regulatory pathways that prevent autoimmunity but are exploited by cancer cells to evade detection, was a pivotal development. Another major breakthrough is CAR-T cell therapy, which involves modifying a patient’s T cells to target cancer-specific antigens. This personalized treatment has shown remarkable success in certain blood cancers. Additionally, cancer vaccines aim to trigger immune responses against tumor-specific or associated antigens, and while challenging, ongoing research is improving their efficacy. The historical progression of cancer immunotherapy, from Coley’s toxins to modern innovations like checkpoint inhibitors and CAR-T cell therapy, underscores its transformative impact on cancer treatment. As research delves deeper into the immune system’s complexities, immunotherapy is poised to become even more crucial in oncology, offering renewed hope to patients globally.
INTRODUCTIONScientists have been honored with more than seventeen Nobel Prizes for their contributions to immunology and associated fields [1]. Cancer immunotherapy represents a groundbreaking approach in the field of oncology, revolutionizing how we combat various forms of cancer [2–4]. This innovative treatment method revolves around leveraging the body’s own immune system, our most potent defense mechanism, to combat malignant cells [5]. Unlike conventional cancer treatments like chemotherapy and radiation therapy, which can be accompanied by severe side effects due to their non-specific targeting, immunotherapy offers a more targeted and nuanced strategy [6].
At its core, cancer immunotherapy aims to activate or enhance the body’s immune response against cancer cells [2,7]. This is achieved through a variety of mechanisms, including the administration of immune checkpoint inhibitors, adoptive cell transfer, cytokine therapy, and therapeutic cancer vaccines, among others [8–10]. These approaches are designed to either stimulate the immune system or remove barriers that prevent it from recognizing and attacking cancer cells effectively [8]. Each approach targets different aspects of the immune response to enhance its effectiveness against cancer cells.
High-dose interleukin (IL)-2 was the initial documented immunotherapy to demonstrate effectiveness in producing lasting and broad responses in patients with advanced melanoma and kidney cancer [11,12]. One of the key features of cancer immunotherapy is its ability to induce long-lasting responses, potentially leading to sustained remission or even cure in some cases [13,14]. Moreover, immunotherapy can offer a more favorable side effect profile compared to traditional treatments, as it predominantly targets cancer cells while sparing healthy tissues [15].
However, it’s essential to acknowledge that cancer immunotherapy is still an evolving field with many challenges including efficacy, response rate, resistance, side effects, resistance; and its efficacy and response rate can vary depending on factors such as the type and stage of cancer, as well as individual patient characteristics [16,17]. Ongoing research and clinical trials continue to refine our understanding of immunotherapy’s mechanisms of action and identify novel strategies to improve outcomes for patients across diverse cancer types.
THE FOUNDATION OF CANCER IMMUNOTHERAPYHistorical perspectives to current scenarioCancer has existed for millennia, predating humanity itself, and has been a significant challenge since our earliest days [18–20]. Key milestones in cancer understanding and management over time are highlighted in Table 1 [18,21–83]. Early insights into tumor immunity stem from ancient observations that hinted at the immune system’s role in cancer. For instance, spontaneous tumor regression, sometimes observed following infection and high fever, was noted as far back as ancient Egypt, 3,000 years ago [84–86]. These early observations, although anecdotal, laid the groundwork for later scientific inquiry into the interaction between the immune system and cancer.
A crucial figure in ancient cancer theory was the Greek physician Galen, who lived around 2,000 years ago [87]. He speculated on the role of inflammation in cancer, observing that tumors often arose in regions of previous injury or inflammation. His hypothesis that chronic inflammation could lead to cancer growth, though limited by the medical knowledge of his era, laid an early foundation for exploring the links between the immune system and cancer [85,87,88]. Galen’s term “oncos” for tumors persists in modern terminology, and his theory of “black bile” causing incurable cancers, while “yellow bile” indicated curable tumors, reflects an early attempt to categorize cancer based on perceived pathology [25].
By the 19th century, scientific understanding began to evolve. In 1828, French surgeon Jean Nicholas Marjolin noted the development of squamous cell carcinoma in burn scars, introducing the idea that chronic irritation could lead to cancer [33,36,89]. English surgeon Caesar Hawkins corroborated this observation in 1833 by noting cases of skin cancer arising near old burn wounds [35,36]. This era’s most significant contribution came from Rudolf Virchow, who in the late 19th century identified leukocytes within tumor tissues. He proposed the groundbreaking theory that tumors could originate from areas of chronic inflammation, advancing the link between inflammation and cancer significantly [39,40,90].
The idea that infections could influence cancer regression was further explored by German physicians Fehleisen and Busch in the 19th century. Both independently observed tumor regressions in cancer patients following erysipelas (a Streptococcus pyogenes infection) [91–94]. While initial attempts to harness this for systematic cancer treatment had mixed success, Fehleisen succeeded in isolating the bacterial strain responsible for both the infection and tumor reduction, marking a milestone in immunotherapy [94–96]. This work would later inspire William Bradley Coley’s famous experiments with bacterial toxins, laying the foundation for modern cancer immunotherapy in the late 19th and early 20th centuries [3,43,85].
In the early 20th century, Paul Ehrlich introduced the concept of “immune surveillance,” suggesting that the immune system continuously monitors the body for abnormal cells, including cancerous ones [97–99]. This hypothesis marked a major shift in understanding the immune system’s role in cancer prevention. Although experimental tools at the time were insufficient to verify the theory, Ehrlich’s ideas laid the groundwork for future research on how immune dysfunction could contribute to cancer progression. Later, Lewis Thomas proposed that the immune system detects newly developing tumors through the recognition of tumor-specific neoantigens expressed on tumor cells, mirroring the rejection process observed in tissue grafts. This mechanism aids in maintaining tissue balance in complex multicellular organisms [100]. The initial clear demonstration of the immune system’s specific capability to elicit a response occurred in 1943. Gross achieved this by immunizing C3H mice, bred through continuous sibling mating for over 20 years, against a sarcoma via intradermal injection. Subsequent research has explored and confirmed these aspects, demonstrating the active role of the immune system in identifying and eliminating cancerous cells through various immune mechanisms, including cytotoxic T cells, natural killer (NK) cells, and other components of the immune response [101,102].
The mid-20th century saw a deeper understanding of immune mechanisms. Moritz Wilhelm Hugo Ribbert, a pathologist from Zurich, suggested that chronic inflammation and mechanical irritation could lead to epithelial and connective tissue proliferation, predisposing tissues to cancer [45,91]. Research by George Klein later expanded this understanding, revealing that the immune system could distinguish and eliminate tumor cells. Klein’s work on major histocompatibility complex (MHC) antigens and immune responses to tumors played a pivotal role in tumor immunology and provided key insights into how tumors evade immune detection [103–106]. This era also saw the development of transplantable tumor models, which allowed scientists to study immune responses to cancer systematically, laying the foundation for modern immunotherapy [107–115].
In the latter part of the 20th century, cytokine therapy emerged as a promising cancer treatment. Alick Isaacs and Jean Lindenmann’s discovery of interferons in 1957 and subsequent discoveries of IL-1 and IL-2 marked major milestones [116–122]. Interferon-based therapies, particularly interferon-α, became the first cytokine therapy approved by the Food and Drug Administration (FDA) for treating hairy cell leukemia in 1986, opening the door to cytokine-based cancer treatments [123–125]. Similarly, IL-2 therapy gained approval for treating metastatic renal cell carcinoma and melanoma in the 1990s, further solidifying the role of cytokines in cancer immunotherapy [126].
In the mid-to-late 20th century, groundbreaking advancements in biotechnology emerged with the advent of hybridoma technology, pioneered by Georges Köhler and César Milstein. For their revolutionary contributions, they received the Nobel Prize in 1984 [127–129]. This technique facilitated the production of monoclonal antibodies, marking a significant milestone in biomedical research and therapeutic development [130]. The origins of antibody understanding, however, trace back to Behring and Shibasaburo Kitasato’s work in 1890, where they identified “antitoxins” in blood, now understood as antibodies. These antibodies neutralized bacterial toxins, such as those causing diphtheria and tetanus, laying the foundation for our understanding of antibody-mediated immunity [131,132].
Subsequent research in the 1930s by Michael Heidelberger and Oswald Avery further elucidated the chemical nature of antibodies and their antigen-specific binding capacity, providing key insights into the molecular basis of the immune response [133,134]. Astrid Fagraeus’s work in the late 1940s demonstrated that plasma cells produce antibodies, solidifying our understanding of how the adaptive immune system responds to antigens [135]. In the 1960s, Sir Gustav Nossal and Joshua Lederberg provided strong evidence for the clonal selection theory, initially proposed by Frank Macfarlane Burnet and David W. Talmage. Their experiments demonstrated that each B cell clone produces antibodies of a single specificity, underpinning the principles of monoclonal antibody technology [136,137].
Monoclonal antibodies, characterized by their specificity to a single epitope, became a focal point of cancer research for their potential in targeted therapy. The approval of rituximab, a monoclonal antibody targeting the CD20 antigen for lymphoma treatment in 1997, marked a watershed moment in medical history [138–140]. Today, monoclonal antibodies are integral to precision medicine, offering targeted therapeutic options across a range of malignancies.
This milestone paved the way for the exploration of novel antibody-based treatments for various malignancies and spurred further research and development efforts, leading to the discovery and approval of additional monoclonal antibody therapies for different types of cancer [140]. Hybridoma technology and monoclonal antibodies continue to play a crucial role in modern medicine, offering targeted and personalized treatment options for patients battling cancer and other diseases [128,141].
In the 1990s, the discovery of immune checkpoints like cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) transformed the landscape of cancer immunotherapy. James P. Allison and Tasuku Honjo’s groundbreaking research led to the development of immune checkpoint inhibitors, which block the inhibitory signals that prevent the immune system from attacking cancer cells [142–147]. Drugs like ipilimumab and pembrolizumab have since revolutionized cancer treatment, particularly for previously untreatable malignancies.
In the 2010s, adoptive cell transfer therapies, including tumor-infiltrating lymphocyte and chimeric antigen receptor (CAR) T-cell therapies, brought even more innovative approaches to immunotherapy [148,149]. CAR-T cell therapy, in particular, demonstrated remarkable success in treating hematologic cancers by genetically engineering T-cells to target specific cancer antigens [70,150,151]. The first CAR-T therapy approval in 2017 marked a pivotal moment in cancer treatment, offering a new avenue for targeting and eliminating cancer cells with precision [152–157].
Throughout the history of cancer immunology, the evolving understanding of the immune system’s role in cancer progression and treatment has paved the way for the development of increasingly sophisticated therapies, transforming the field of oncology and offering new hope for patients worldwide. Preclinical, and early clinical studies have shown promising results for both CAR-M and CAR-NK cells [157–163].
Over the last five decades, there has been a concentrated effort to develop cancer vaccines, yielding only modest achievements [164,165]. Yet, recent progress in genetics, molecular biology, biochemistry, and immunology has reignited enthusiasm for these immunotherapies, leading to the emergence of promising candidates for cancer, since the 2000s the development of cancer vaccines, encompassing both therapeutic and preventive approaches, has represented a significant frontier in cancer immunotherapy [165,166]. Furthermore, ongoing efforts are focused on combining cancer vaccines with other immunotherapies, such as checkpoint inhibitors, to enhance anti-tumor immune responses and improve treatment outcomes [167,168]. While significant challenges remain, including issues related to vaccine efficacy, safety, and patient selection, the development of cancer vaccines holds great promise for the future of cancer treatment and prevention [169]. Continued research and clinical trials are critical for realizing the full potential of cancer vaccines in improving patient outcomes and reducing the global burden of cancer.
Milestones in immunotherapy research
Fig. 1 presents the chronological progression of significant discoveries and breakthroughs in immunotherapy. It began in 1891 with Coley’s toxin, an early attempt at using bacterial toxins to treat cancer. In 1957, Burnet’s immunosurveillance theory was proposed, suggesting that the immune system plays a role in recognizing and destroying cancer cells. In 1975, hybridoma technology was developed, enabling the mass production of monoclonal antibodies, revolutionizing targeted cancer therapies.
The timeline continues with the discovery of crucial immune checkpoints: CTLA-4 in 1995, which led to the development of immune checkpoint inhibitors, and PD-1 in 1992, another key checkpoint protein that plays a role in preventing the immune system from attacking normal cells. These discoveries paved the way for immunotherapy drugs like pembrolizumab, which received FDA approval in 2014 for the treatment of melanoma, marking a major milestone in immune checkpoint blockade therapy.
In 2012, the Nobel Prize for the discovery of CRISPR was awarded, revolutionizing gene editing technology with applications in cancer research. This was followed by the FDA approval of CAR-T cell therapy in 2017, a breakthrough in personalized cancer immunotherapy targeting blood cancers. The timeline concludes with the FDA approval of Opdualag in 2022, a novel combination of immune checkpoint inhibitors (nivolumab and relatlimab), signifying continued advancements in immunotherapy for treating melanoma.
TUMOR IMMUNOLOGY: UNRAVELING THE IMMUNE RESPONSE TO CANCERDiscovery of tumor immunogenicityThe quest to understand tumor immunogenicity in the context of cancer therapy has been a profound and multifaceted journey, intertwining decades of scientific research, clinical observations, and technological progress [170]. This narrative unfolds against a backdrop of perseverance, skepticism, and ultimately, groundbreaking discoveries that have revolutionized the field of oncology [171]. Tumor immunogenicity is determined by the tumor cells themselves and is also influenced by factors within the tumor microenvironment (TME), including the activity of antigen-presenting cells like dendritic cells (DCs) [172]. The primary factors that determine tumor immunogenicity are the presence of tumor-specific antigens and the efficiency of antigen processing and presentation [173].
The foundation of understanding tumor immunogenicity in cancer therapy can be traced back to the pioneering efforts of Dr. William B. Coley (1862–1936), a bone sarcoma surgeon, in the late 19th century, around 1891 [43]. Dr. Coley’s empirical observations of some cancer patients experiencing spontaneous tumor regression following erysipelas, a streptococcal skin infection, served as the catalyst for the recognition of the immune system’s potential in combating cancer. He hypothesized that the immune response triggered by the infection might also be attacking the cancer cells [174,175]. Coley extensively reviewed medical records, case reports, and literature available to him in the late nineteenth century. He found numerous instances of potentially incurable cancers undergoing spontaneous remission following acute bacterial infections. This provided further evidence supporting the potential role of the immune system in fighting cancer. Building upon his observations and understanding, Coley began experimenting at Memorial Sloan-Kettering Cancer Center in New York, by injecting a mixture of heat-killed streptococcal bacteria and another organism, now known as Serratia marcescens (Coley’s toxin), directly into tumors (mostly bone and soft-tissue sarcomas), achieving a cure rate of over 10% [43,174–176]. By doing so, he aimed to stimulate the immune system’s response against cancer cells. This approach laid the foundation for what can be considered the first immune-based treatment for cancer. In 1891, Coley made his initial endeavor to utilize the immune system for treating bone cancer [43,86]. Coley’s experiments encountered skepticism in his era because of the unclear understanding of their mechanisms of action and concerns regarding the potential risks of infection. Significant criticisms emerged within the scientific community because many physicians did not believe his results, particularly highlighted in the Journal of the American Medical Association (JAMA) in 1894, where a statement was issued condemning the use of his toxins. The part of statement issued in JAMA read as “There is no longer much question of the entire failure of the toxin injections, as a cure for sarcomata and malignant growths. During the last 6 months the alleged remedy has been faithfully tried by many surgeons, but so far not a single well-authenticated case of recovery has been reported” [43,177]. Additionally, the FDA reclassified “Coley’s toxins” in 1963 as an investigational drug due to the absence of safety and efficacy data, despite more than 70 years of application and numerous published studies [43]. Despite facing initial skepticism, Coley’s work laid a cornerstone for future investigations into the intricate interplay between the immune system and malignant cells. Several years after his death in 1962, his toxins underwent reevaluation in a controlled trial, revealing their ability to induce anti-tumor effects [43,178]. Presently Coley’s principles, demonstrating that certain cancers respond positively to an augmented immune response has been validated and oncolytic virus therapies have been developed, employing genetically modified viruses to infect tumor cells, thus inducing a pro-inflammatory environment to bolster systemic anti-tumor immunity [179]. With vigorous research in the area of cancer immunotherapy, William B. Coley rightfully deserves and often regarded as the “Father of Immunotherapy” [43,180].
The mid-20th century ushered in a transformative period for scientific research, as investigators began to explore the intricate mechanisms underlying the immune system’s responses to tumors. During the 1950s and 1960s, pioneering work by scientists like Lewis Thomas and others brought significant advancements in the understanding of immune surveillance. This phenomenon describes the immune system’s role as a vigilant guardian, constantly monitoring and capable of recognizing and eliminating abnormal cells, including cancerous ones [98,181]. This groundbreaking concept laid the foundation for the modern understanding of tumor immunogenicity, emphasizing the innate capacity of the immune system to distinguish between self and non-self-antigens, including those presented by tumors [182].
In 1971, Professor Lloyd J. Old, a trailblazer in cancer immuno-oncology, observed that “there exists a distinct quality in cancer cells, setting them apart from normal cells, which the body’s immune system is capable of recognizing. He suggested that this unique attribute could be pivotal in understanding and harnessing the immune system’s ability to identify and combat cancer cells [183]. The subsequent decades witnessed a flurry of discoveries that further elucidated the complexities of tumor immunogenicity. Notably, the identification of tumor antigens in experimental animal models by researchers such as George Klein in the 1970s was an important scientific discovery [103,182]. George Klein played a key role in identifying tumor-associated antigens and tumor-specific antigens, contributing to our understanding that these antigens can be expressed by cancer cells at higher concentrations compared to normal cells [103]. However, research has since shown that the absolute specificity of these antigens might have limitations, and our understanding of their expression and immune response continues to evolve [184]. This pivotal insight paved the way for subsequent efforts to delineate tumor antigens in human cancers, laying the groundwork for the development of personalized immunotherapies tailored to individual patients’ tumor profiles. Advancements in molecular biology in the latter half of the 20th century catalyzed a paradigm shift in our understanding of tumor immunogenicity [185]. The discovery of oncogenes and tumor suppressor genes, unraveled the genetic underpinnings of cancer, unveiling a landscape marked by genetic alterations that drive tumorigenesis and shape the immunogenicity of tumors [186–188]. Importantly, mutations in oncogenes and tumor suppressor genes were found to give rise to neoantigens—novel antigens derived from mutated proteins expressed by cancer cells. These neoantigens, recognized as foreign by the immune system, emerged as promising targets for immunotherapeutic interventions aimed at eliciting potent anti-tumor immune responses [189–191].
The emergence of high-throughput genomic technologies in the 21st century has revolutionized our capacity to analyze the immune landscape of tumors with unparalleled accuracy [192–195]. Large-scale genomic projects, such as the Cancer Genome Atlas (TCGA), undertook the ambitious goal of cataloging the genomic alterations driving cancer across various tumor types [196,197]. Through comprehensive genomic analyses, TCGA and similar projects unveiled the heterogeneity of tumor antigens, including neoantigens, and provided invaluable insights into the molecular underpinnings of cancer immunogenicity [190,198]. These efforts laid the groundwork for the development of novel immunotherapeutic strategies aimed at exploiting the immunogenic potential of tumors for therapeutic benefit [199,200].
A significant milestone in the effort to leverage tumor immunogenicity for cancer therapy was the discovery of immune checkpoints—a class of inhibitory receptors that regulate immune responses and ensure immune homeostasis [8,147]. Through the advancement of therapies that inhibit specific checkpoints, such as ipilimumab (anti-CTLA-4), pembrolizumab and nivolumab (anti-PD-1), and atezolizumab and durvalumab (anti-PD-L1), scientists have harnessed the potential of the immune system, enabling it to generate robust anti-tumor immune reactions [147].
Advances in technologies such as next-generation sequencing have made it possible to identify neoantigens—antigens that arise from mutations specific to tumors [190,201,202]. This has opened the door to personalized cancer immunotherapy strategies, such as cancer vaccines and adoptive cell therapies, which are designed to target neoantigens specific to each patient’s unique tumor [190].
Insights into tumor immunosurveillanceThe notion that the immune system has the ability to recognize and destroy cancer cells was initially proposed by Paul Ehrlich in 1909 [99]. However, more precise concepts of tumor immunosurveillance developed from the pivotal observations made by Macfarlane Burnet and Lewis Thomas in the mid-20th century [40,110]. Building upon the earlier work of Paul Ehrlich and others, Burnet proposed the idea that the immune system plays a critical role in recognizing and eliminating cancerous cells, akin to its function in combating infectious pathogens [110,203]. Burnet expressed that “In large long-lived animals, like most of the warm-blooded vertebrates, inheritable genetic changes must be common in somatic cells and a proportion of these changes will represent a step toward malignancy. It is an evolutionary necessity that there should be some mechanism for eliminating or inactivating such potentially dangerous mutant cells and it is postulated that this mechanism is of immunological character” [110]. He further hypothesized that neoantigens specific to tumor cells could trigger a potent immune response capable of eradicating emerging cancers [110,203]. This notion was further refined by Lewis Thomas, who introduced the term “immunosurveillance” to describe the immune system’s vigilant surveillance of the body for aberrant cells, including those with tumorigenic potential [111].
Although it initially faced skepticism, the concept of tumor immunosurveillance gained acceptance with the development of experimental models that showed the immune system’s capacity to identify and destroy tumors [114]. Studies in animal models, such as the observation of increased tumor incidence in immunocompromised mice and vice versa, provided compelling evidence for the existence of immunosurveillance mechanisms capable of suppressing tumor development [98,204,205]. Furthermore, clinical observations, such as the increased incidence of certain cancers in immunocompromised individuals, lent support to the notion of immune-mediated tumor suppression [206].
Tumor immunosurveillance involves a complex interaction between the immune system and cancer cells, driven by a wide range of cellular and molecular mechanisms [192]. Central to this process is the recognition of tumor-specific antigens by immune effector cells, including cytotoxic T lymphocytes (CTLs; CD8+ T cells) and NK cells, leading to the elimination of cancerous cells [114,204,207,208]. This recognition is facilitated by the presentation of tumor antigens by MHC molecules on the surface of antigen-presenting cells, such as DCs [209]. The innate immune system serves as the first line of defense against tumors, detecting and eliminating transformed cells through a variety of mechanisms [210]. NK cells, endowed with the ability to recognize and kill aberrant cells lacking self MHC class I molecules or abnormal/altered MHC class I expression, play a pivotal role in tumor immunosurveillance [211,212]. Additionally, macrophages and DCs contribute to the elimination of cancer cells through phagocytosis and antigen presentation, respectively, initiating adaptive immune responses against tumors [213,214]. The adaptive immune system provides a more specialized and potent response to cancer cells, especially mediated by antigen-specific T lymphocytes [3,215]. CTLs recognize and eliminate cancerous cells expressing tumor-specific antigens in the context of MHC class I molecules, while helper T cells orchestrate the immune response by secreting cytokines and activating other immune cells [216,217]. While B lymphocytes have the capacity to generate antibodies directed at tumor antigens, the significance of humoral immune responses in combating cancer is controversial and a subject of debate [218].
Beyond cellular immunity, soluble factors like cytokines and chemokines are vital to tumor immunosurveillance [219]. Interferons, ILs, and tumor necrosis factor-alpha directly combat tumors by triggering apoptosis, suppressing cell proliferation, and boosting immune cell activity. Chemokines, on the other hand, manage the movement of immune cells to the tumor site, aiding in their infiltration and the subsequent elimination of the tumor [220].
Immuno-evasion mechanisms employed by cancer cellsImmuno-evasion mechanisms employed by cancer cells represent a complex array of strategies meticulously orchestrated to evade the host immune system’s surveillance and thwart immune-mediated destruction [115]. These mechanisms reflect the remarkable adaptability of cancer cells in evading recognition and eradication by the immune system, thereby promoting tumor growth, progression, and metastasis [115,221].
Disruption of antigen presentation1) MHC class I downregulationOne fundamental aspect of cancer immune-evasion involves the manipulation of antigen presentation pathways [222]. The presentation of tumor antigens by MHC molecules is essential for the initiation of effective anti-tumor immune responses [222,223]. However, cancer cells often exploit various mechanisms to subvert antigen presentation, thereby evading detection by CTLs and other immune effector cells [115]. This may entail downregulation or loss of MHC-I molecules on the surface of cancer cells, thereby impairing the recognition of tumor antigens by CTLs (CD8+ T cells), by becoming less stimulatory or even invisible to CTLs (CD8+ T cells), without impairing their ability to grow and metastasize [3,223,224]. Consequently, the significance of modulating MHC-I in evading the immune system in cancer has garnered attention in recent years. Various studies have highlighted that the loss or reduction of MHC-I serves as a primary mechanism for cancer immune evasion, hindering the display of tumor-associated antigens on the cell surface. This, in turn, diminishes the cytotoxicity of CD8+ T cells (CTLs) and compromises the effectiveness of the adaptive immune response [223,225,226].
2) Antigen processing pathway impairmentAdditionally, cancer cells may disrupt antigen processing and presentation pathways, leading to inefficient presentation of tumor-derived antigens to immune cells [223,226]. This disruption can occur at multiple levels of the antigen processing machinery. The proteasome is responsible for degrading proteins into peptide fragments, which are then transported into the endoplasmic reticulum to be loaded onto MHC class I molecules. Cancer cells can alter proteasome function, reducing the production of antigenic peptides that are essential for immune recognition [227,228]. Transporter associated with antigen processing (TAP) proteins transport peptide fragments from the cytosol into the endoplasmic reticulum, where they are loaded onto MHC class I molecules. Cancer cells can downregulate or inhibit TAP, preventing the transfer of these peptides and thereby reducing the number of antigens presented on the cell surface [229]. Cancer cells may manipulate enzymes involved in the generation of peptide epitopes, such as endoplasmic reticulum aminopeptidase 1. By modifying the activity of these enzymes, cancer cells can produce non-immunogenic peptides or degrade antigenic peptides, thus evading immune detection [223,226].
Modulation of immune checkpoint pathways1) PD-1/PD-L1 axisAnother pivotal immune-evasion mechanism employed by cancer cells involves the modulation of immune checkpoint pathways [230]. Immune checkpoints serve as crucial regulators of immune responses, maintaining self-tolerance and preventing excessive immune activation [231,232]. However, cancer cells exploit these checkpoints to evade immune surveillance and suppress anti-tumor immune responses in several ways [231,232]. Cancer cells can increase the expression of ligands for immune checkpoint receptors on their surface [231]. For example, they often upregulate the expression of PD-L1. When PD-L1 binds to its receptor, PD-1 on T cells, it transmits an inhibitory signal that reduces the activity of T cells, thereby preventing them from attacking the tumor [233, 234].
2) CTLA-4 pathwayThe CTLA-4 pathway is another critical immune checkpoint exploited by tumors. CTLA-4 competes with CD28 for binding to B7 molecules (CD80/CD86) on antigen-presenting cells. When CTLA-4 binds to B7, it inhibits T cell activation and proliferation [235,236]. Tumor cells can promote the expression of CTLA-4 on regulatory T cells (Tregs) and effector T cells within the TME, dampening the immune response against the tumor [237,238]. Tumor cells can also influence the TME to promote the expression of immune checkpoint receptors such as PD-1, CTLA-4 (CTL-associated protein 4), and others on T cells and other immune cells. This induction can occur through the secretion of immunosuppressive cytokines, such as IL-10 and transforming growth factor-beta (TGF-β), which create an environment conducive to immune evasion [3,239].
TME-mediated immunosuppression1) Recruitment of immunosuppressive cellsAdditionally, tumors establish an immunosuppressive microenvironment (immunosuppressive TME), which serves to shield tumor cells from immune detection [240]. Cancer cells actively attract immunosuppressive cells to the TME, thereby further promoting immune evasion [241]. T-regs, myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs) are among the immune cell populations recruited by cancer cells to promote immune tolerance and dampen anti-tumor immune responses [242–244].
2) Cytokine secretionT-regs suppress immune responses through the secretion of immunosuppressive cytokines such as IL-10 and TGF-β, and by competition for growth factors by Treg cells leading to cytokine (like IL-2) deprivation for T-cells, thereby inhibiting effector T cell function [245,246]. MDSCs exert immunosuppressive effects by inhibiting T cell activation and proliferation, promoting angiogenesis, and inducing immune tolerance. They achieve this by upregulating the expression of negative immune checkpoint molecules like PD-L1, VISTA, GAL-9, and CD-155 to induce T-cell anergy, depriving essential amino acids crucial for T cell metabolism and function, secreting reactive nitrogen and oxygen species to impair T cell function, and hampering T cell trafficking in hosts with tumors [247–251]. TAMs, on the other hand, exhibit a dual role in the TME, M2-like TAMs contribute to tumor progression by suppressing anti-tumor immunity [243,252]. By recruiting these immunosuppressive cell populations, cancer cells create an immune-permissive niche that facilitates tumor immune evasion and survival.
NK cell evasion1) Downregulation of NK cell activating ligandsMoreover, cancer cells employ various mechanisms to evade innate immune surveillance, particularly by resisting natural NK cell-mediated cytotoxicity [253,254]. NK cells play a critical role in tumor surveillance and elimination by recognizing and killing abnormal or stressed cells, including cancer cells [253,255]. However, cancer cells evade NK cell recognition and killing through various mechanisms, including downregulation of NK cell-activating ligands (e.g., natural killer group 2 member D [NKG2D] ligands), recruiting suppressive immune cells that dampen NK cell activity, and altering their surface architecture/expression of surface molecules to make them less accessible to NK cells [253,256,257].
2) Induction of immunosuppressive factorsAnother significant mechanism of cancer immune evasion involves, cancer cells producing and secreting immunosuppressive cytokines and signaling molecules [115]. These cytokines, such as IL-10, TGF-β, and vascular endothelial growth factor, suppress the immune system by inhibiting effector T cell function, promoting Treg expansion, and inducing immune tolerance [258]. Additionally, cancer cells can activate signaling pathways like the Janus kinase/signal transducer and activator of transcription pathway or the nuclear factor-kappa B pathway, which regulate immune responses and facilitate immune evasion [259–261]. By secreting immunosuppressive cytokines and activating signaling pathways, cancer cells create an immunosuppressive microenvironment that facilitates tumor immune escape and progression.
Immunosuppressive exosomes and metabolic reprogramming1) Exosome mediated suppressionFurthermore, tumor-derived exosomes represent another mechanism by which cancer cells evade immune surveillance and promote immune tolerance [262–264]. Exosomes are small extracellular vesicles also released by cancer cells that contain a cargo of proteins, lipids, and nucleic acids which can modulate immune responses [265]. Cancer-derived exosomes can transfer immunosuppressive molecules such as TGF-c, IL-10, and PD-L1 to immune cells, thereby inhibiting their function and promoting immune evasion. Additionally, tumor-derived exosomes can induce apoptosis or dysfunction in immune cells, impairing their ability to mount an effective anti-tumor immune response [266]. Exosomes containing TGF-β have the potential to diminish the expression of NKG2D on CD8 T cells, consequently hindering cell activation [267]. Newer research has also discovered elevated levels of exosomal PD-L1, possessing a similar structure to cellular PD-L1. It has been observed that exosomal PD-L1 engages with PD-1 receptors on CD8 T cells, leading to the deactivation of these T cells and facilitating immune evasion by tumor cells [268]. Exosomes carrying UL16-binding proteins and MHC class I chain-related protein A have the capability to suppress the NKG2D signaling pathway, which plays a crucial role in the cytotoxic function of T cells [265,269]. Galectin-9 found in exosomes derived from nasopharyngeal cancer cells can trigger the apoptosis of numerous Epstein–Barr virus-specific CD4 T cells and suppress the activity of T helper 1 (Th1) cells [270]. Research has revealed that in individuals with nasopharyngeal cancer, circulating exosomes contain a significant amount of miR-24-3p. This miRNA hinders the differentiation of Th1 and Th17 cells by suppressing fibroblast growth factor-11 leading to immunosuppression [271]. By exploiting exosomal-mediated immune modulation, cancer cells create an immune-suppressive microenvironment that facilitates tumor immune escape and progression.
2) Metabolic reprogrammingMoreover, cancer cells undergo metabolic reprogramming to sustain their rapid proliferation and survival within the TME, thereby creating an immunosuppressive metabolic landscape that facilitates tumor immune evasion [272–275]. This metabolic adaptation entails heightened glucose absorption and glycolysis, resulting in lactate buildup and acidosis. Additionally, it involves the alteration of lipid, amino acid, nucleotide, and mitochondrial biogenesis within tumor cells amidst the TME [274,276]. Such changes can hinder immune cell activity and foster immune tolerance. Additionally, cancer cells may exploit metabolic intermediates such as adenosine, arginine, kynurenine, prostaglandin E2, and norepinephrine and epinephrine to suppress immune responses and promote immune evasion [276–281].
FUTURE TRENDS IN CANCER IMMUNOTHERAPYCancer immunotherapy has seen transformative developments over the past few decades, culminating in remarkable successes in treating various malignancies. As we look to the future, several trends and emerging technologies hold promise for further advancing this field and improving patient outcomes.
Checkpoint inhibitors and combination therapiesThe success of checkpoint inhibitors, such as ipilimumab (yervoy) (anti-CTLA-4), pembrolizumab/cemiplimab (Libtayo)/nivolumab (anti-PD-1), and atezolizumab/avelumab (bavencio)/durvalumab (imfinzi) (anti-PD-L1), has revolutionized cancer treatment. These therapies work by blocking inhibitory pathways that prevent T-cells from attacking cancer cells [147,282]. In future, combining these inhibitors with other treatments, including additional immunotherapies, targeted therapies, and conventional treatments like chemotherapy and radiation, can have promising results. Combining therapies can lead to synergistic effects, enhancing the overall anti-tumor response and potentially overcoming resistance mechanisms.
CAR-T cell therapyCAR-T cell therapy represents a significant leap in personalized cancer treatment. By genetically modifying a patient’s T-cells to express receptors specific to cancer antigens, CAR-T cell therapy has shown substantial success in hematologic cancers like leukemia and lymphoma [283,284]. Future research aims to extend this therapy to solid tumors, improve its safety and efficacy, and reduce side effects such as cytokine release syndrome. Advances in gene editing technologies, including CRISPR/Cas9, are expected to further refine and enhance CAR therapies. CAR-M and CAR-NK cells are promising candidates [285–288].
Adoptive T-cell therapyAdoptive T-cell therapy (ACT) involves expanding and reinfusing a patient’s own T-cells that have been activated to fight cancer. This approach has shown promise in treating various cancers and is continually being refined [289,290]. Efforts in positive directions led to, lifileucel, receiving US FDA approval on February 15, 2024, as the first cellular therapy for solid cancer [82]. Future developments in ACT include enhancing the specificity and persistence of T-cells, optimizing the expansion process, and combining ACT with other immunotherapies to boost effectiveness.
Gene editing and cellular reprogrammingGene editing technologies, particularly CRISPR/Cas9, have opened new avenues for cancer immunotherapy [291]. These tools enable precise modifications to immune cells, enhancing their ability to target and destroy cancer cells. Additionally, cellular reprogramming techniques, such as converting fibroblasts into DCs, are being explored to create more effective cancer vaccines and therapies [292]. These innovations have the potential to generate personalized treatments, according to individual patients’ tumor profiles.
Immune system memory and long-term remissionImmune system’s ability to remember cancer antigens offers the potential for long-term remission. Research is focused on understanding how to stimulate and sustain immune memory responses to prevent cancer recurrence [293,294]. Vaccines and therapies that enhance immune memory are being developed to provide lasting protection against cancer [295].
TME and microbiomeThe TME, including the surrounding stroma, immune cells, and vasculature, plays a crucial role in cancer progression and response to therapy [240]. Modulating the TME to enhance immunotherapy effectiveness is a key area of research. Additionally, the microbiome—the community of microorganisms in the body—has been found to influence cancer treatment outcomes [296]. Table 2 depicts the various immunotherapy clinical trials in phase 1 [297–310]. Manipulating the microbiome through approaches like fecal microbiota transplantation could improve responses to immunotherapy [311].
Biomarker identification and personalized medicineIdentifying biomarkers that predict responses to immunotherapy is essential for personalized cancer treatment [312]. Researchers are actively investigating additional biomarkers to enhance patient selection and optimize therapeutic outcomes [313]. Tumors with high mutational burden or specific genetic features, such as microsatellite instability, are known to respond better to certain immunotherapies [314,315]. Personalized approaches based on these biomarkers can enhance treatment efficacy and reduce adverse effects.
Overcoming resistance and relapseDespite the success of immunotherapy, some patients develop resistance or relapse after treatment [316,317]. Understanding the mechanisms of resistance is important for developing strategies to overcome it [318]. This includes targeting multiple immune checkpoints, enhancing antigen presentation, and preventing the development of escape mutations [318–320]. New therapeutic combinations and novel agents are being explored to address resistance and improve long-term outcomes.
CONCLUSIONThe future of cancer immunotherapy is filled with promise. By leveraging the power of the immune system and harnessing cutting-edge technologies, researchers and clinicians are poised to make significant strides in the fight against cancer. These advancements offer hope for improved treatments and better outcomes for patients worldwide.
REFERENCES1. Stone MJ. Monoclonal antibodies in the prehybridoma era: a brief historical perspective and personal reminiscence. Clin Lymphoma 2001;2:148-54.
2. Zhang Y, Zhang Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol 2020;17:807-21.
3. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 2020;20:651-68.
4. Want MY, Bashir Z, Najar RA. T cell based immunotherapy for cancer: approaches and strategies. Vaccines (Basel) 2023;11:835.
6. Sahu M, Suryawanshi H. Immunotherapy: the future of cancer treatment. J Oral Maxillofac Pathol 2021;25:371.
7. Mukherjee AG, Wanjari UR, Namachivayam A, Murali R, Prabakaran DS, Ganesan R, et al. Role of immune cells and receptors in cancer treatment: an immunotherapeutic approach. Vaccines (Basel) 2022;10:1493.
8. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov 2019;18:175-96.
9. Kciuk M, Yahya EB, Mohamed Ibrahim Mohamed M, Rashid S, Iqbal MO, Kontek R, et al. Recent advances in molecular mechanisms of cancer immunotherapy. Cancers (Basel) 2023;15:2721.
10. Wang DR, Wu XL, Sun YL. Therapeutic targets and biomarkers of tumor immunotherapy: response versus non-response. Signal Transduct Target Ther 2022;7:331.
11. Lotze MT, Frana LW, Sharrow SO, Robb RJ, Rosenberg SA. In vivo administration of purified human interleukin 2. I. Half-life and immunologic effects of the Jurkat cell line-derived interleukin 2. J Immunol 1985;134:157-66.
12. Lotze MT, Custer MC, Sharrow SO, Rubin LA, Nelson DL, Rosenberg SA. In vivo administration of purified human interleukin-2 to patients with cancer: development of interleukin-2 receptor positive cells and circulating soluble interleukin-2 receptors following interleukin-2 administration. Cancer Res 1987;47:2188-95.
13. Koury J, Lucero M, Cato C, Chang L, Geiger J, Henry D, et al. Immunotherapies: exploiting the immune system for cancer treatment. J Immunol Res 2018;2018:9585614.
14. D’Errico G, Machado HL, Sainz B Jr. A current perspective on cancer immune therapy: step-by-step approach to constructing the magic bullet. Clin Transl Med 2017;6:3.
15. Ventola CL. Cancer immunotherapy, Part 2: efficacy, safety, and other clinical considerations. P T 2017;42:452-63.
16. Peterson C, Denlinger N, Yang Y. Recent advances and challenges in cancer immunotherapy. Cancers (Basel) 2022;14:3972.
18. Hajdu SI. A note from history: landmarks in history of cancer, part 1. Cancer 2011;117:1097-102.
19. Mitrus I, Bryndza E, Sochanik A, Szala S. Evolving models of tumor origin and progression. Tumour Biol 2012;33:911-7.
20. Binder M, Roberts C, Spencer N, Antoine D, Cartwright C. On the antiquity of cancer: evidence for metastatic carcinoma in a young man from ancient Nubia (c. 1200 BC). PLoS One 2014;9:e90924.
21. Kapur BN. The fruits of long endeavors: 200 years of oncology. Med J Armed Forces India 2014;70:95-7.
22. Brucher BL, Lyman G, van Hillegersberg R, Pollock RE, Lordick F, Yang HK, et al. Imagine a world without cancer. BMC Cancer 2014;14:186.
23. Spencer WG. Celsus De Medicina Volume 3:Harvard University Press;1935;
24. Novick M, Gard DA, Hardy SB, Spira M. Burn scar carcinoma: a review and analysis of 46 cases. J Trauma 1977;17:809-17.
26. Dalfardi B, Mahmoudi Nezhad GS. Insights into Avicenna’s contributions to the science of surgery. World J Surg 2014;38:2175-9.
27. Laios K. Wilhelm Fabricius von Hilden (1560–1634): the pioneer of German surgery. Surg Innov 2018;25:301-3.
28. Fabricius H. Observationum et curationum chirurgicarum centuriae: Cent II. IA Huguetan; 1641.
29. Ventura HO. Giovanni Battista Morgagni and the foundation of modern medicine. Clin Cardiol 2000;23:792-4.
31. Brown JR, Thornton JL. Percivall Pott (1714–1788) and chimney sweepers’ cancer of the scrotum. Br J Ind Med 1957;14:68-70.
33. Marjolin J. Ulcer. Adelon NP, editor. Dictionnaire de medicine. Bechet; 1828. p. 31-50.
34. Bazalinski D, Przybek-Mita J, Baranska B, Wiech P. Marjolin’s ulcer in chronic wounds: review of available literature. Contemp Oncol (Pozn) 2017;21:197-202.
35. Hawkins C. On warty tumors in cicatrices. Lond Med Gaz 1833;13:481-2.
36. Piotrowski I, Kulcenty K, Suchorska W. Interplay between inflammation and cancer. Rep Pract Oncol Radiother 2020;25:422-7.
37. Capp JP. Cancer stem cells: from historical roots to a new perspective. J Oncol 2019;2019:5189232.
41. Plesca M, Bordea C, El Houcheimi B, Ichim E, Blidaru A. Evolution of radical mastectomy for breast cancer. J Med Life 2016;9:183-6.
42. Paget S. The distribution of secondary growths in cancer of the breast. Cancer Metastasis Rev 1989;8:98-101.
43. McCarthy EF. The toxins of William B. Coley and the treatment of bone and soft-tissue sarcomas. Iowa Orthop J 2006;26:154-8.
44. Boveri T. Concerning the origin of malignant tumours by Theodor Boveri: translated and annotated by Henry Harris. J Cell Sci 2008;121:Suppl 1. 1-84.
47. Fujiki H. Gist of Dr. Katsusaburo Yamagiwa’s papers entitled “Experimental study on the pathogenesis of epithelial tumors” (I to VI reports). Cancer Sci 2014;105:143-9.
48. Stolt CM, Klein G, Jansson AT. An analysis of a wrong Nobel Prize-Johannes Fibiger, 1926: a study in the Nobel archives. Adv Cancer Res 2004;92:1-12.
49. Tan SY, Tatsumura Y. George Papanicolaou (1883–1962): discoverer of the Pap smear. Singapore Med J 2015;56:586-7.
50. Swailes AL, Hossler CE, Kesterson JP. Pathway to the Papanicolaou smear: the development of cervical cytology in twentieth-century America and implications in the present day. Gynecol Oncol 2019;154:3-7.
51. National Cancer Institute. National Cancer Act of 1937 [Internet]. National Cancer Institute;c2024. [cited 2024 Jul 28]. Available from: https://www.cancer.gov/about-nci/overview/history/national-cancer-act-1937.
52. Desai K, McManus JM, Sharifi N. Hormonal therapy for prostate cancer. Endocr Rev 2021;42:354-73.
53. Proctor RN. The history of the discovery of the cigarette-lung cancer link: evidentiary traditions, corporate denial, global toll. Tob Control 2012;21:87-91.
54. Hecht SS, Szabo E. Fifty years of tobacco carcinogenesis research: from mechanisms to early detection and prevention of lung cancer. Cancer Prev Res (Phila) 2014;7:1-8.
55. John Hopkins Medicine. The Immortal Life of Henrietta Lacks [Internet]. John Hopkins Medicine;c2024. [cited 2024 Jul 28]. Available from: https://www.hopkinsmedicine.org/henrietta-lacks/immortal-life-of-henrietta-lacks.
56. Nowell PC, Hungerford DA. A minute chromosome in human chronic granulocytic leukemia. Harper PS, editor. Landmarks in medical genetics. Oxford University Press; 2004. p. 103.
58. Rowley JD. Letter: a new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature 1973;243:290-3.
59. Bister K. Discovery of oncogenes: the advent of molecular cancer research. Proc Natl Acad Sci U S A 2015;112:15259-60.
60. Jordan VC. Tamoxifen as the first targeted long-term adjuvant therapy for breast cancer. Endocr Relat Cancer 2014;21:R235-46.
61. Baumann M, von Knebel Doeberitz M. Harald zur Hausen, virologist who linked viruses to cancer (1936–2023). Nature 2023;619:93.
63. Hurst JH. Pioneering geneticist Mary-Claire King receives the 2014 Lasker~Koshland Special Achievement Award in Medical Science. J Clin Invest 2014;124:4148-51.
64. Boyiadzis M, Foon KA. Approved monoclonal antibodies for cancer therapy. Expert Opin Biol Ther 2008;8:1151-8.
65. Iqbal N, Iqbal N. Imatinib: a breakthrough of targeted therapy in cancer. Chemother Res Pract 2014;2014:357027.
66. Sharma R, Sharma CL. Quadrivalent human papillomavirus recombinant vaccine: the first vaccine for cervical cancers. J Cancer Res Ther 2007;3:92-5.
67. Vaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB. Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers (Basel) 2020;12:738.
68. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012;337:816-21.
69. Raedler LA. Keytruda (pembrolizumab): first PD-1 inhibitor approved for previously treated unresectable or metastatic melanoma. Am Health Drug Benefits 2015;8(Spec Feature):96-100.
70. Mitra A, Barua A, Huang L, Ganguly S, Feng Q, He B. From bench to bedside: the history and progress of CAR T cell therapy. Front Immunol 2023;14:1188049.
71. Guo ZS. The 2018 Nobel Prize in medicine goes to cancer immunotherapy (editorial for BMC cancer). BMC Cancer 2018;18:1086.
72. Marcus L, Donoghue M, Aungst S, Myers CE, Helms WS, Shen G, et al. FDA approval summary: entrectinib for the treatment of NTRK gene fusion solid tumors. Clin Cancer Res 2021;27:928-32.
73. Foundation Medicine. FDA approves Foundation Medicine’s FoundationOne®Liquid CDx, a comprehensive pan-tumor liquid biopsy test with multiple companion diagnostic indications for patients with advanced cancer. foundation medicine [Internet]. Foundation Medicine;c2020. [cited 2024 Jul 28]. Available from: https://www.foundationmedicine.com/press-releases/fda-approves-foundation-medicine%27s-foundationone%C2%AEliquid-cdx%2C-a-comprehensive-pan-tumor-liquid-biopsy-test-with-multiple-companion-diagnostic-indications-for-patients-with-advanced-cancer.
74. Passaro A, Al Bakir M, Hamilton EG, Diehn M, Andre F, Roy-Chowdhuri S, et al. Cancer biomarkers: emerging trends and clinical implications for personalized treatment. Cell 2024;187:1617-35.
75. Nakajima EC, Drezner N, Li X, Mishra-Kalyani PS, Liu Y, Zhao H, et al. FDA approval summary: sotorasib for KRAS G12C-mutated metastatic NSCLC. Clin Cancer Res 2022;28:1482-6.
76. US Food and Drug Administration (FDA). FDA approves idecabtagene vicleucel for multiple myeloma. U.S. FDA;c2021. [cited 2024 Jul 28]. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-idecabtagene-vicleucel-multiple-myeloma.
77. US Food and Drug Administration (FDA). FDA approves dostarlimab-gxly with chemotherapy for endometrial cancer. U.S. FDA;c2023. [cited 2024 Jul 28]. Available from: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-dostarlimab-gxly-chemotherapy-endometrial-cancer.
78. de Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell 2023;41:374-403.
79. Baghy K, Ladanyi A, Reszegi A, Kovalszky I. Insights into the tumor microenvironment-components, functions and therapeutics. Int J Mol Sci 2023;24:17536.
80. Shegekar T, Vodithala S, Juganavar A. The emerging role of liquid biopsies in revolutionising cancer diagnosis and therapy. Cureus 2023;15:e43650.
81. Connal S, Cameron JM, Sala A, Brennan PM, Palmer DS, Palmer JD, et al. Liquid biopsies: the future of cancer early detection. J Transl Med 2023;21:118.
83. Newswire PR. WestGene’s mRNA therapeutic cancer vaccine receives FDA approval [Internet]. PR Newswire;c2024. [cited 2024 Jul 10]. Available from: https://tinyurl.com/2acwcea3.
84. Decker WK, da Silva RF, Sanabria MH, Angelo LS, Guimaraes F, Burt BM, et al. Cancer immunotherapy: historical perspective of a clinical revolution and emerging preclinical animal models. Front Immunol 2017;8:829.
85. Dobosz P, Dzieciatkowski T. The intriguing history of cancer immunotherapy. Front Immunol 2019;10:2965.
86. Kleef R, Dieter Hager E. Fever, pyrogens and cancer. Madame Curie Bioscience Database [Internet]. Landes Bioscience;c2013. [cited 2024 Jul 28]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK6084/.
87. Institut Pasteur. Viruses and human cancers [Internet]. Institut Pasteur;c2019. [cited 2024 Jul 28]. Available from: https://www.pasteur.fr/fr/international/cours-internationaux/cours-internationaux-2019/viruses-and-human-cancers.
89. Pavlovic S, Wiley E, Guzman G, Morris D, Braniecki M. Marjolin ulcer: an overlooked entity. Int Wound J 2011;8:419-24.
90. Virchow R. Cellular pathology as based upon physiological and pathological histology. J.B. Lippincott; 1863.
91. Ribbert MWH. Geschwulstlehere fur Arzte und Studierende. Cohen; 1904;
93. Busch W. Aus der Sitzung der medicinischen Section vom 13 November 1867. Berlin Klin Wochenschr 1868;5:137.
94. Fehleisen F. Ueber die Züchtung der Erysipelkokken auf künstlichem Nährboden und ihre Übertragbarkeit auf den Menschen. Dtsch Med Wochenschr 1882;8:553-4.
95. Oiseth SJ, Aziz MS. Cancer immunotherapy: a brief review of the history, possibilities, and challenges ahead. J Cancer Metastasis Treat 2017;3:250-61.
98. Ribatti D. The concept of immune surveillance against tumors: the first theories. Oncotarget 2017;8:7175-80.
99. Ehrlich P. Ueber den jetzigen Stand der Karzinomforschung. Ned Tijdschr Geneeskd 1909;5:273-90.
100. Thomas L. Discussion. Lawrence HS, editor. Cellular and humoral aspects of the hypersensitive states. Hoeber-Harper; 1959. p. 529-32.
101. Raskov H, Orhan A, Christensen JP, Gogenur I. Cytotoxic CD8+ T cells in cancer and cancer immunotherapy. Br J Cancer 2021;124:359-67.
102. Yoon SR, Kim TD, Choi I. Understanding of molecular mechanisms in natural killer cell therapy. Exp Mol Med 2015;47:e141.
104. Kaminski JM, Summers JB, Ward MB, Huber MR, Minev B. Immunotherapy and prostate cancer. Cancer Treat Rev 2003;29:199-209.
106. Srivastava PK. George Klein (1925–2016) a prescient, luminous voice. Cancer Immunol Res 2017;5:272.
108. Kim R, Emi M, Tanabe K. Cancer immunoediting from immune surveillance to immune escape. Immunology 2007;121:1-14.
109. Olson B, Li Y, Lin Y, Liu ET, Patnaik A. Mouse models for cancer immunotherapy research. Cancer Discov 2018;8:1358-65.
112. Budhu S, Wolchok J, Merghoub T. The importance of animal models in tumor immunity and immunotherapy. Curr Opin Genet Dev 2014;24:46-51.
113. Chulpanova DS, Kitaeva KV, Rutland CS, Rizvanov AA, Solovyeva VV. Mouse tumor models for advanced cancer immunotherapy. Int J Mol Sci 2020;21:4118.
114. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 2002;3:991-8.
115. Kim SK, Cho SW. The evasion mechanisms of cancer immunity and drug intervention in the tumor microenvironment. Front Pharmacol 2022;13:868695.
116. Ponomarev AV, Shubina IZ. Insights into mechanisms of tumor and immune system interaction: association with wound healing. Front Oncol 2019;9:1115.
117. Conlon KC, Miljkovic MD, Waldmann TA. Cytokines in the treatment of cancer. J Interferon Cytokine Res 2019;39:6-21.
118. Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol 2022;19:237-53.
119. Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci 1957;147:258-67.
120. Isaacs A, Lindenmann J, Valentine RC. Virus interference. II. Some properties of interferon. Proc R Soc Lond B Biol Sci 1957;147:268-73.
121. Dinarello CA, Goldin NP, Wolff SM. Demonstration and characterization of two distinct human leukocytic pyrogens. J Exp Med 1974;139:1369-81.
122. Morgan DA, Ruscetti FW, Gallo R. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science 1976;193:1007-8.
123. Lan T, Chen L, Wei X. Inflammatory cytokines in cancer: comprehensive understanding and clinical progress in gene therapy. Cells 2021;10:100.
124. Qiu Y, Su M, Liu L, Tang Y, Pan Y, Sun J. Clinical application of cytokines in cancer immunotherapy. Drug Des Devel Ther 2021;15:2269-87.
125. Waldmann TA. Cytokines in cancer immunotherapy. Cold Spring Harb Perspect Biol 2018;10:a028472.
126. Alva A, Daniels GA, Wong MK, Kaufman HL, Morse MA, McDermott DF, et al. Contemporary experience with high-dose interleukin-2 therapy and impact on survival in patients with metastatic melanoma and metastatic renal cell carcinoma. Cancer Immunol Immunother 2016;65:1533-44.
127. Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975;256:495-7.
128. Parray HA, Shukla S, Samal S, Shrivastava T, Ahmed S, Sharma C, et al. Hybridoma technology a versatile method for isolation of monoclonal antibodies, its applicability across species, limitations, advancement and future perspectives. Int Immunopharmacol 2020;85:106639.
129. Zhang C. Hybridoma technology for the generation of monoclonal antibodies. Methods Mol Biol 2012;901:117-35.
130. Holzlohner P, Hanack K. Generation of murine monoclonal antibodies by hybridoma technology. J Vis Exp 2017;(119):54832.
131. Kaufmann SH. Remembering Emil von Behring: from tetanus treatment to antibody cooperation with phagocytes. mBio 2017;8:e00117-17.
135. Fagraeus A. Plasma cellular reaction and its relation to the formation of antibodies in vitro. Nature 1947;159:499.
138. Mohammed R, Milne A, Kayani K, Ojha U. How the discovery of rituximab impacted the treatment of B-cell non-Hodgkin’s lymphomas. J Blood Med 2019;10:71-84.
139. Casan JM, Wong J, Northcott MJ, Opat S. Anti-CD20 monoclonal antibodies: reviewing a revolution. Hum Vaccin Immunother 2018;14:2820-41.
140. Lu RM, Hwang YC, Liu IJ, Lee CC, Tsai HZ, Li HJ, et al. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci 2020;27:1.
141. Quinteros DA, Bermudez JM, Ravetti S, Cid A, Allemandi DA, Palma SD. Therapeutic use of monoclonal antibodies: general aspects and challenges for drug delivery. Andronescu E, Grumezescu AM, editors. Nanostructures for drug delivery. Elsevier; 2017. p. 807-33.
142. Huang PW, Chang JW. Immune checkpoint inhibitors win the 2018 Nobel Prize. Biomed J 2019;42:299-306.
143. Zappasodi R, Merghoub T, Wolchok JD. Emerging concepts for immune checkpoint blockade-based combination therapies. Cancer Cell 2018;33:581-98.
144. Topalian SL, Weiner GJ, Pardoll DM. Cancer immunotherapy comes of age. J Clin Oncol 2011;29:4828-36.
145. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 2015;27:450-61.
146. Wojtukiewicz MZ, Rek MM, Karpowicz K, Gorska M, Politynska B, Wojtukiewicz AM, et al. Inhibitors of immune checkpoints-PD-1, PD-L1, CTLA-4: new opportunities for cancer patients and a new challenge for internists and general practitioners. Cancer Metastasis Rev 2021;40:949-82.
147. Shiravand Y, Khodadadi F, Kashani SM, Hosseini-Fard SR, Hosseini S, Sadeghirad H, et al. Immune checkpoint inhibitors in cancer therapy. Curr Oncol 2022;29:3044-60.
148. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer 2008;8:299-308.
149. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma: a preliminary report. N Engl J Med 1988;319:1676-80.
150. Assi R, Salman H. Harnessing the potential of chimeric antigen receptor T-cell therapy for the treatment of T-cell malignancies: a dare or double dare? Cells 2022;11:3971.
151. Zhang X, Zhu L, Zhang H, Chen S, Xiao Y. CAR-T cell therapy in hematological malignancies: current opportunities and challenges. Front Immunol 2022;13:927153.
152. Keshavarz A, Salehi A, Khosravi S, Shariati Y, Nasrabadi N, Kahrizi MS, et al. Recent findings on chimeric antigen receptor (CAR)-engineered immune cell therapy in solid tumors and hematological malignancies. Stem Cell Res Ther 2022;13:482.
153. Wang S, Yang Y, Ma P, Zha Y, Zhang J, Lei A, et al. CAR-macrophage: an extensive immune enhancer to fight cancer. EBioMedicine 2022;76:103873.
154. Hadiloo K, Tahmasebi S, Esmaeilzadeh A. CAR-NKT cell therapy: a new promising paradigm of cancer immunotherapy. Cancer Cell Int 2023;23:86.
155. Shin MH, Oh E, Kim Y, Nam DH, Jeon SY, Yu JH, et al. Recent advances in CAR-based solid tumor immunotherapy. Cells 2023;12:1606.
156. Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, Basar R, et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N Engl J Med 2020;382:545-53.
157. Li H, Song W, Li Z, Zhang M. Preclinical and clinical studies of CAR-NK-cell therapies for malignancies. Front Immunol 2022;13:992232.
158. Liu M, Liu J, Liang Z, Dai K, Gan J, Wang Q, et al. CAR-macrophages and CAR-T cells synergistically kill tumor cells in vitro. Cells 2022;11:3692.
159. Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, Zeeman M, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol 2020;38:947-53.
160. Morrissey MA, Williamson AP, Steinbach AM, Roberts EW, Kern N, Headley MB, et al. Chimeric antigen receptors that trigger phagocytosis. Elife 2018;7:e36688.
161. Reiss KA, Yuan Y, Ueno NT, Johnson ML, Gill S, Dees EC, et al. A phase 1, first-in-human (FIH) study of the anti-HER2 CAR macrophage CT-0508 in subjects with HER2 overexpressing solid tumors. J Clin Oncol 2022;40(16 suppl):2533.
162. Ma R, Lu T, Li Z, Teng KY, Mansour AG, Yu M, et al. An oncolytic virus expressing IL15/IL15Rα combined with off-the-shelf EGFR-CAR NK cells targets glioblastoma. Cancer Res 2021;81:3635-48.
163. Marin D, Li Y, Basar R, Rafei H, Daher M, Dou J, et al. Safety, efficacy and determinants of response of allogeneic CD19-specific CAR-NK cells in CD19+ B cell tumors: a phase 1/2 trial. Nat Med 2024;30:772-84.
164. Donninger H, Li C, Eaton JW, Yaddanapudi K. Cancer vaccines: promising therapeutics or an unattainable dream. Vaccines (Basel) 2021;9:668.
165. Tay BQ, Wright Q, Ladwa R, Perry C, Leggatt G, Simpson F, et al. Evolution of cancer vaccines-challenges, achievements, and future directions. Vaccines (Basel) 2021;9:535.
166. Liu J, Fu M, Wang M, Wan D, Wei Y, Wei X. Cancer vaccines as promising immuno-therapeutics: platforms and current progress. J Hematol Oncol 2022;15:28.
167. Kim CG, Sang YB, Lee JH, Chon HJ. Combining cancer vaccines with immunotherapy: establishing a new immunological approach. Int J Mol Sci 2021;22:8035.
168. Mougel A, Terme M, Tanchot C. Therapeutic cancer vaccine and combinations with antiangiogenic therapies and immune checkpoint blockade. Front Immunol 2019;10:467.
169. Kaczmarek M, Poznanska J, Fechner F, Michalska N, Paszkowska S, Napierala A, et al. Cancer vaccine therapeutics: limitations and effectiveness: a literature review. Cells 2023;12:2159.
170. Wang S, He Z, Wang X, Li H, Liu XS. Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. Elife 2019;8:e49020.
171. Lee D, Huntoon K, Wang Y, Jiang W, Kim BY. Harnessing innate immunity using biomaterials for cancer immunotherapy. Adv Mater 2021;33:e2007576.
172. Mellman I, Steinman RM. Dendritic cells: specialized and regulated antigen processing machines. Cell 2001;106:255-8.
173. Blankenstein T, Coulie PG, Gilboa E, Jaffee EM. The determinants of tumour immunogenicity. Nat Rev Cancer 2012;12:307-13.
174. Coley WB. The treatment of malignant tumors by repeated inoculations of erysipelas: with a report of ten original cases. Am J Med Sci 1893;105:487-511.
175. Coley WB. The treatment of inoperable sarcoma by bacterial toxins (the mixed toxins of the Streptococcus erysipelas and the Bacillus prodigiosus). Proc R Soc Med 1910;3(Surg Sect):1-48.
176. Wiemann B, Starnes CO. Coley’s toxins, tumor necrosis factor and cancer research: a historical perspective. Pharmacol Ther 1994;64:529-64.
178. Johnston BJ, Novales ET. Clinical effect of Coley’s toxin. II. A seven-year study. Cancer Chemother Rep 1962;21:43-68.
179. Zimmermannova O, Caiado I, Ferreira AG, Pereira CF. Cell fate reprogramming in the era of cancer immunotherapy. Front Immunol 2021;12:714822.
180. Carlson RD, Flickinger JC Jr, Snook AE. Talkin’ toxins: from Coley’s to modern cancer immunotherapy. Toxins (Basel) 2020;12:241.
182. Kunimasa K, Goto T. Immunosurveillance and immunoediting of lung cancer: current perspectives and challenges. Int J Mol Sci 2020;21:597.
184. Srinivasan R, Wolchok JD. Tumor antigens for cancer immunotherapy: therapeutic potential of xenogeneic DNA vaccines. J Transl Med 2004;2:12.
185. Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell 2018;175:313-26.
186. Lyu J, Li JJ, Su J, Peng F, Chen YE, Ge X, et al. DORGE: discovery of oncogenes and tumor suppressor genes using genetic and epigenetic features. Sci Adv 2020;6:eaba6784.
187. Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P. Molecular biology of the cell. 4th ed. Garland Science; 2002.
188. Chakravarthi BV, Nepal S, Varambally S. Genomic and epigenomic alterations in cancer. Am J Pathol 2016;186:1724-35.
189. Fang X, Guo Z, Liang J, Wen J, Liu Y, Guan X, et al. Neoantigens and their potential applications in tumor immunotherapy. Oncol Lett 2022;23:88.
190. Xie N, Shen G, Gao W, Huang Z, Huang C, Fu L. Neoantigens: promising targets for cancer therapy. Signal Transduct Target Ther 2023;8:9.
191. Wirth TC, Kuhnel F. Neoantigen targeting-dawn of a new era in cancer immunotherapy? Front Immunol 2017;8:1848.
192. Ginsburg O, Ashton-Prolla P, Cantor A, Mariosa D, Brennan P. The role of genomics in global cancer prevention. Nat Rev Clin Oncol 2021;18:116-28.
193. Dai X, Shen L. Advances and trends in omics technology development. Front Med (Lausanne) 2022;9:911861.
194. Restrepo JC, Duenas D, Corredor Z, Liscano Y. Advances in genomic data and biomarkers: revolutionizing NSCLC diagnosis and treatment. Cancers (Basel) 2023;15:3474.
195. Burak GI, Ozge S, Cem M, Gulgun B, Zeynep DY, Atil B. The emerging clinical relevance of genomic profiling in neuroendocrine tumours. BMC Cancer 2021;21:234.
196. Tomczak K, Czerwinska P, Wiznerowicz M. The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Contemp Oncol (Pozn) 2015;19(1A):A68-77.
197. India Project Team of the International Cancer Genome Consortium. Mutational landscape of gingivo-buccal oral squamous cell carcinoma reveals new recurrently-mutated genes and molecular subgroups. Nat Commun 2013;4:2873.
198. Brown SD, Warren RL, Gibb EA, Martin SD, Spinelli JJ, Nelson BH, et al. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res 2014;24:743-50.
200. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189-99.
201. Kast F, Klein C, Umana P, Gros A, Gasser S. Advances in identification and selection of personalized neoantigen/T-cell pairs for autologous adoptive T cell therapies. Oncoimmunology 2021;10:1869389.
202. De Mattos-Arruda L, Vazquez M, Finotello F, Lepore R, Porta E, Hundal J, et al. Neoantigen prediction and computational perspectives towards clinical benefit: recommendations from the ESMO Precision Medicine Working Group. Ann Oncol 2020;31:978-90.
203. Burnet M. Cancer: a biological approach. III. Viruses associated with neoplastic conditions. IV. Practical applications. Br Med J 1957;1:841-7.
205. Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, et al. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001;410:1107-11.
206. Ilham S, Willis C, Kim K, Chung KC, Wood BM, Tan MS, et al. Cancer incidence in immunocompromised patients: a single-center cohort study. BMC Cancer 2023;23:33.
207. Smyth MJ, Thia KY, Street SE, MacGregor D, Godfrey DI, Trapani JA. Perforin-mediated cytotoxicity is critical for surveillance of spontaneous lymphoma. J Exp Med 2000;192:755-60.
208. Escors D. Tumour immunogenicity, antigen presentation and immunological barriers in cancer immunotherapy. New J Sci 2014;2014:734515.
209. Nouri-Shirazi M, Banchereau J, Bell D, Burkeholder S, Kraus ET, Davoust J, et al. Dendritic cells capture killed tumor cells and present their antigens to elicit tumor-specific immune responses. J Immunol 2000;165:3797-803.
210. Pandya PH, Murray ME, Pollok KE, Renbarger JL. The immune system in cancer pathogenesis: potential therapeutic approaches. J Immunol Res 2016;2016:4273943.
211. Sun JC, Lanier LL. Natural killer cells remember: an evolutionary bridge between innate and adaptive immunity? Eur J Immunol 2009;39:2059-64.
213. Del Prete A, Salvi V, Soriani A, Laffranchi M, Sozio F, Bosisio D, et al. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell Mol Immunol 2023;20:432-47.
214. DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol 2019;19:369-82.
215. Halliday GM, Patel A, Hunt MJ, Tefany FJ, Barnetson RS. Spontaneous regression of human melanoma/nonmelanoma skin cancer: association with infiltrating CD4+ T cells. World J Surg 1995;19:352-8.
216. Sykulev Y. Factors contributing to the potency of CD8+ T cells. Trends Immunol 2023;44:693-700.
217. Schirrmacher V, Schild HJ, Guckel B, von Hoegen P. Tumour-specific CTL response requiring interactions of four different cell types and recognition of MHC class I and class II restricted tumour antigens. Immunol Cell Biol 1993;71(Pt 4):311-26.
218. Yuen GJ, Demissie E, Pillai S. B lymphocytes and cancer: a love-hate relationship. Trends Cancer 2016;2:747-57.
219. Matsuo K, Yoshie O, Nakayama T. Multifaceted roles of chemokines and chemokine receptors in tumor immunity. Cancers (Basel) 2021;13:6132.
220. Nishikawa H, Koyama S. Mechanisms of regulatory T cell infiltration in tumors: implications for innovative immune precision therapies. J Immunother Cancer 2021;9:e002591.
221. Kallingal A, Olszewski M, Maciejewska N, Brankiewicz W, Baginski M. Cancer immune escape: the role of antigen presentation machinery. J Cancer Res Clin Oncol 2023;149:8131-41.
222. Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res 2015;21:687-92.
223. Dhatchinamoorthy K, Colbert JD, Rock KL. Cancer immune evasion through loss of MHC class I antigen presentation. Front Immunol 2021;12:636568.
224. Cornel AM, Mimpen IL, Nierkens S. MHC class I downregulation in cancer: underlying mechanisms and potential targets for cancer immunotherapy. Cancers (Basel) 2020;12:1760.
225. Wu X, Li T, Jiang R, Yang X, Guo H, Yang R. Targeting MHC-I molecules for cancer: function, mechanism, and therapeutic prospects. Mol Cancer 2023;22:194.
226. Mpakali A, Stratikos E. The role of antigen processing and presentation in cancer and the efficacy of immune checkpoint inhibitor immunotherapy. Cancers (Basel) 2021;13:134.
227. Vigneron N, Van den Eynde BJ. Proteasome subtypes and regulators in the processing of antigenic peptides presented by class I molecules of the major histocompatibility complex. Biomolecules 2014;4:994-1025.
228. Sijts EJ, Kloetzel PM. The role of the proteasome in the generation of MHC class I ligands and immune responses. Cell Mol Life Sci 2011;68:1491-502.
229. Ritz U, Seliger B. The transporter associated with antigen processing (TAP): structural integrity, expression, function, and its clinical relevance. Mol Med 2001;7:149-58.
230. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252-64.
231. Singh S, Singh N, Baranwal M, Sharma S, Devi SS, Kumar S. Understanding immune checkpoints and PD-1/PD-L1-mediated immune resistance towards tumour immunotherapy. 3 Biotech 2023;13:411.
232. Meng L, Wu H, Wu J, Ding P, He J, Sang M, et al. Mechanisms of immune checkpoint inhibitors: insights into the regulation of circular RNAS involved in cancer hallmarks. Cell Death Dis 2024;15:3.
233. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res 2020;10:727-42.
234. Gutic B, Bozanovic T, Mandic A, Dugalic S, Todorovic J, Stanisavljevic D, et al. Programmed cell death-1 and its ligands: current knowledge and possibilities in immunotherapy. Clinics (Sao Paulo) 2023;78:100177.
235. Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol 2016;39:98-106.
236. Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood 2018;131:58-67.
237. Sobhani N, Tardiel-Cyril DR, Davtyan A, Generali D, Roudi R, Li Y. CTLA-4 in regulatory T cells for cancer immunotherapy. Cancers (Basel) 2021;13:1440.
238. Watanabe T, Ishino T, Ueda Y, Nagasaki J, Sadahira T, Dansako H, et al. Activated CTLA-4-independent immunosuppression of Treg cells disturbs CTLA-4 blockade-mediated antitumor immunity. Cancer Sci 2023;114:1859-70.
239. Ahuja S, Zaheer S. Multifaceted TGF-β signaling, a master regulator: from bench-to-bedside, intricacies, and complexities. Cell Biol Int 2024;48:87-127.
240. Aden D, Zaheer S, Ahluwalia H, Ranga S. Cancer-associated fibroblasts: is it a key to an intricate lock of tumorigenesis? Cell Biol Int 2023;47:859-93.
241. Tie Y, Tang F, Wei YQ, Wei XW. Immunosuppressive cells in cancer: mechanisms and potential therapeutic targets. J Hematol Oncol 2022;15:61.
242. Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ. The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 2013;138:105-15.
243. Malik S, Sureka N, Ahuja S, Aden D, Zaheer S, Zaheer S. Tumor-associated macrophages: a sentinel of innate immune system in tumor microenvironment gone haywire. Cell Biol Int 2024;48:1406-49.
244. Pan Y, Yu Y, Wang X, Zhang T. Tumor-associated macrophages in tumor immunity. Front Immunol 2020;11:583084.
245. Sojka DK, Huang YH, Fowell DJ. Mechanisms of regulatory T-cell suppression: a diverse arsenal for a moving target. Immunology 2008;124:13-22.
246. Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med 2014;211:781-90.
247. Li K, Shi H, Zhang B, Ou X, Ma Q, Chen Y, et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther 2021;6:362.
248. Yang Y, Li C, Liu T, Dai X, Bazhin AV. Myeloid-derived suppressor cells in tumors: from mechanisms to antigen specificity and microenvironmental regulation. Front Immunol 2020;11:1371.
249. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol 2021;21:485-98.
250. Ma T, Renz BW, Ilmer M, Koch D, Yang Y, Werner J, et al. Myeloid-derived suppressor cells in solid tumors. Cells 2022;11:310.
251. Krishnamoorthy M, Gerhardt L, Maleki Vareki S. Immunosuppressive effects of myeloid-derived suppressor cells in cancer and immunotherapy. Cells 2021;10:1170.
252. Yang Q, Guo N, Zhou Y, Chen J, Wei Q, Han M. The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy. Acta Pharm Sin B 2020;10:2156-70.
253. Meza Guzman LG, Keating N, Nicholson SE. Natural killer cells: tumor surveillance and signaling. Cancers (Basel) 2020;12:952.
254. Sordo-Bahamonde C, Lorenzo-Herrero S, Payer AR, Gonzalez S, Lopez-Soto A. Mechanisms of apoptosis resistance to NK cell-mediated cytotoxicity in cancer. Int J Mol Sci 2020;21:3726.
255. Sharma P, Kumar P, Sharma R. Natural killer cells: their role in tumour immunosurveillance. J Clin Diagn Res 2017;11:BE01-5.
256. Cantoni C, Wurzer H, Thomas C, Vitale M. Escape of tumor cells from the NK cell cytotoxic activity. J Leukoc Biol 2020;108:1339-60.
257. Jones AB, Rocco A, Lamb LS, Friedman GK, Hjelmeland AB. Regulation of NKG2D stress ligands and its relevance in cancer progression. Cancers (Basel) 2022;14:2339.
258. McRitchie BR, Akkaya B. Exhaust the exhausters: targeting regulatory T cells in the tumor microenvironment. Front Immunol 2022;13:940052.
259. Hu X, Li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther 2021;6:402.
260. Thomas SJ, Snowden JA, Zeidler MP, Danson SJ. The role of JAK/STAT signalling in the pathogenesis, prognosis and treatment of solid tumours. Br J Cancer 2015;113:365-71.
261. Bapat AS, O’Connor CH, Schwertfeger KL. Targeting the NF-κB pathway enhances responsiveness of mammary tumors to JAK inhibitors. Sci Rep 2023;13:5349.
262. Yu S, Liu C, Su K, Wang J, Liu Y, Zhang L, et al. Tumor exosomes inhibit differentiation of bone marrow dendritic cells. J Immunol 2007;178:6867-75.
263. Valenti R, Huber V, Filipazzi P, Pilla L, Sovena G, Villa A, et al. Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-beta-mediated suppressive activity on T lymphocytes. Cancer Res 2006;66:9290-8.
264. Tai YL, Chen KC, Hsieh JT, Shen TL. Exosomes in cancer development and clinical applications. Cancer Sci 2018;109:2364-74.
265. Xie QH, Zheng JQ, Ding JY, Wu YF, Liu L, Yu ZL, et al. Exosome-mediated immunosuppression in tumor microenvironments. Cells 2022;11:1946.
266. Han Q, Zhao H, Jiang Y, Yin C, Zhang J. HCC-derived exosomes: critical player and target for cancer immune escape. Cells 2019;8:558.
267. Lazarova M, Steinle A. Impairment of NKG2D-mediated tumor immunity by TGF-β. Front Immunol 2019;10:2689.
268. Ye L, Zhu Z, Chen X, Zhang H, Huang J, Gu S, et al. The importance of exosomal PD-L1 in cancer progression and its potential as a therapeutic target. Cells 2021;10:3247.
269. Ashiru O, Boutet P, Fernandez-Messina L, Aguera-Gonzalez S, Skepper JN, Vales-Gomez M, et al. Natural killer cell cytotoxicity is suppressed by exposure to the human NKG2D ligand MICA* 008 that is shed by tumor cells in exosomes. Cancer Res 2010;70:481-9.
270. Klibi J, Niki T, Riedel A, Pioche-Durieu C, Souquere S, Rubinstein E, et al. Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood 2009;113:1957-66.
271. Ye SB, Zhang H, Cai TT, Liu YN, Ni JJ, He J, et al. Exosomal miR-24-3p impedes T-cell function by targeting FGF11 and serves as a potential prognostic biomarker for nasopharyngeal carcinoma. J Pathol 2016;240:329-40.
272. Ahuja S, Sureka N, Zaheer S. Unraveling the intricacies of cancer-associated fibroblasts: a comprehensive review on metabolic reprogramming and tumor microenvironment crosstalk. APMIS 2024;132:906-27.
273. Xu Y, He L, Fu Q, Hu J. Metabolic reprogramming in the tumor microenvironment with immunocytes and immune checkpoints. Front Oncol 2021;11:759015.
274. Navarro C, Ortega A, Santeliz R, Garrido B, Chacin M, Galban N, et al. Metabolic reprogramming in cancer cells: emerging molecular mechanisms and novel therapeutic approaches. Pharmaceutics 2022;14:1303.
275. Chen X, Ma Z, Yi Z, Wu E, Shang Z, Tuo B, et al. The effects of metabolism on the immune microenvironment in colorectal cancer. Cell Death Discov 2024;10:118.
276. Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, et al. The cancer metabolic reprogramming and immune response. Mol Cancer 2021;20:28.
277. Phillips MM, Sheaff MT, Szlosarek PW. Targeting arginine-dependent cancers with arginine-degrading enzymes: opportunities and challenges. Cancer Res Treat 2013;45:251-62.
278. Gouasmi R, Ferraro-Peyret C, Nancey S, Coste I, Renno T, Chaveroux C, et al. The kynurenine pathway and cancer: why keep it simple when you can make it complicated. Cancers (Basel) 2022;14:2793.
279. Duffin R, O’Connor RA, Crittenden S, Forster T, Yu C, Zheng X, et al. Prostaglandin E2 constrains systemic inflammation through an innate lymphoid cell-IL-22 axis. Science 2016;351:1333-8.
280. Lu W, Yu W, He J, Liu W, Yang J, Lin X, et al. Reprogramming immunosuppressive myeloid cells facilitates immunotherapy for colorectal cancer. EMBO Mol Med 2021;13:e12798.
281. Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer 2007;7:95-106.
282. Okobi TJ, Uhomoibhi TO, Akahara DE, Odoma VA, Sanusi IA, Okobi OE, et al. Immune checkpoint inhibitors as a treatment option for bladder cancer: current evidence. Cureus 2023;15:e40031.
283. Cappell KM, Kochenderfer JN. Long-term outcomes following CAR-T cell therapy: what we know so far. Nat Rev Clin Oncol 2023;20:359-71.
284. Pegram HJ, Smith EL, Rafiq S, Brentjens RJ. CAR therapy for hematological cancers: can success seen in the treatment of B-cell acute lymphoblastic leukemia be applied to other hematological malignancies? Immunotherapy 2015;7:545-61.
285. Chen Y, Zhu X, Liu H, Wang C, Chen Y, Wang H, et al. The application of HER2 and CD47 CAR-macrophage in ovarian cancer. J Transl Med 2023;21:654.
286. Hadiloo K, Taremi S, Heidari M, Esmaeilzadeh A. The CAR macrophage cells, a novel generation of chimeric antigen-based approach against solid tumors. Biomark Res 2023;11:103.
287. Li T, Niu M, Zhang W, Qin S, Zhou J, Yi M. CAR-NK cells for cancer immunotherapy: recent advances and future directions. Front Immunol 2024;15:1361194.
288. Zhong Y, Liu J. Emerging roles of CAR-NK cell therapies in tumor immunotherapy: current status and future directions. Cell Death Discov 2024;10:318.
289. Fang KK, Lee JB, Zhang L. Adoptive cell therapy for T-cell malignancies. Cancers (Basel) 2022;15:94.
290. Du S, Yan J, Xue Y, Zhong Y, Dong Y. Adoptive cell therapy for cancer treatment. Exploration (Beijing) 2023;3:20210058.
291. Ding S, Liu J, Han X, Tang M. CRISPR/Cas9-mediated genome editing in cancer therapy. Int J Mol Sci 2023;24:16325.
292. Perez CR, De Palma M. Engineering dendritic cell vaccines to improve cancer immunotherapy. Nat Commun 2019;10:5408.
293. Alvero AB, Fox A, Madina BR, Krady MM, Gogoi R, Chehade H, et al. Immune modulation of innate and adaptive responses restores immune surveillance and establishes antitumor immunologic memory. Cancer Immunol Res 2024;12:261-74.
294. Hiam-Galvez KJ, Allen BM, Spitzer MH. Systemic immunity in cancer. Nat Rev Cancer 2021;21:345-59.
295. Le I, Dhandayuthapani S, Chacon J, Eiring AM, Gadad SS. Harnessing the immune system with cancer vaccines: from prevention to therapeutics. Vaccines (Basel) 2022;10:816.
296. Bhatt AP, Redinbo MR, Bultman SJ. The role of the microbiome in cancer development and therapy. CA Cancer J Clin 2017;67:326-44.
297. Tawbi HA, Schadendorf D, Lipson EJ, Ascierto PA, Matamala L, Castillo Gutierrez E, et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N Engl J Med 2022;386:24-34.
298. Tian T, Li Z. Targeting Tim-3 in cancer with resistance to PD-1/PD-L1 blockade. Front Oncol 2021;11:731175.
299. Li X, Zhao L, Li W, Gao P, Zhang N. HER2-targeting CAR-T cells show highly efficient anti-tumor activity against glioblastoma both in vitro and in vivo. Genes Immun 2024;25:201-8.
300. Beatty GL, O’Hara MH, Lacey SF, Torigian DA, Nazimuddin F, Chen F, et al. Activity of mesothelin-specific chimeric antigen receptor T cells against pancreatic carcinoma metastases in a phase 1 trial. Gastroenterology 2018;155:29-32.
301. Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol 2015;33:2780-8.
302. Ziogas DC, Martinos A, Petsiou DP, Anastasopoulou A, Gogas H. Beyond immunotherapy: seizing the momentum of oncolytic viruses in the ideal platform of skin cancers. Cancers (Basel) 2022;14:2873.
303. Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017;547:217-21.
304. Le DT, Wang-Gillam A, Picozzi V, Greten TF, Crocenzi T, Springett G, et al. Safety and survival with GVAX pancreas prime and Listeria Monocytogenes-expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J Clin Oncol 2015;33:1325-33.
305. Chen YL, Cui Y, Liu X, Liu G, Dong X, Tang L, et al. A bispecific antibody targeting HER2 and PD-L1 inhibits tumor growth with superior efficacy. J Biol Chem 2021;297:101420.
306. Glud EN, Rasmussen M, Zhang Y, Mandrup OA, Salachan PV, Borre M, et al. Identification of a high-risk immunogenic prostate cancer patient subset as candidates for T-cell engager immunotherapy and the introduction of a novel albumin-fused anti-CD3 × anti-PSMA bispecific design. Br J Cancer 2022;127:2186-97.
307. Goff SL, Dudley ME, Citrin DE, Somerville RP, Wunderlich JR, Danforth DN, et al. Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J Clin Oncol 2016;34:2389-97.
308. Stevanovic S, Helman SR, Wunderlich JR, Langhan MM, Doran SL, Kwong ML, et al. A phase II study of tumor-infiltrating lymphocyte therapy for human Papillomavirus-associated epithelial cancers. Clin Cancer Res 2019;25:1486-93.
309. Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster E, et al. CRISPR-engineered T cells in patients with refractory cancer. Science 2020;367:eaba7365..
310. Chamberlain CA, Bennett EP, Kverneland AH, Svane IM, Donia M, Met O. Highly efficient PD-1-targeted CRISPR-Cas9 for tumor-infiltrating lymphocyte-based adoptive T cell therapy. Mol Ther Oncolytics 2022;24:417-28.
311. Zhao LY, Mei JX, Yu G, Lei L, Zhang WH, Liu K, et al. Role of the gut microbiota in anticancer therapy: from molecular mechanisms to clinical applications. Signal Transduct Target Ther 2023;8:201.
312. Sankar K, Ye JC, Li Z, Zheng L, Song W, Hu-Lieskovan S. The role of biomarkers in personalized immunotherapy. Biomark Res 2022;10:32.
313. Qin Y, Huo M, Liu X, Li SC. Biomarkers and computational models for predicting efficacy to tumor ICI immunotherapy. Front Immunol 2024;15:1368749.
314. Palmeri M, Mehnert J, Silk AW, Jabbour SK, Ganesan S, Popli P, et al. Real-world application of tumor mutational burden-high (TMB-high) and microsatellite instability (MSI) confirms their utility as immunotherapy biomarkers. ESMO Open 2022;7:100336.
315. Schrock AB, Ouyang C, Sandhu J, Sokol E, Jin D, Ross JS, et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann Oncol 2019;30:1096-103.
316. Borcoman E, Nandikolla A, Long G, Goel S, Le Tourneau C. Patterns of response and progression to immunotherapy. Am Soc Clin Oncol Educ Book 2018;38:169-78.
317. Bai R, Chen N, Li L, Du N, Bai L, Lv Z, et al. Mechanisms of cancer resistance to immunotherapy. Front Oncol 2020;10:1290.
318. Pathak R, Pharaon RR, Mohanty A, Villaflor VM, Salgia R, Massarelli E. Acquired resistance to PD-1/PD-L1 blockade in lung cancer: mechanisms and patterns of failure. Cancers (Basel) 2020;12:3851.
Table 1
FDA, Food and Drug Administration; CML, chronic myeloid leukemia; HPV, human papillomavirus; CAR, chimeric antigen receptor; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; TIL, tumor-infiltrating lymphocyte; mRNA, messenger RNA; IND, investigational new drug. Table 2
LAG-3, lymphocyte-activation gene 3; TIM-3, T-cell immunoglobulin and mucin-domain containing-3; TIGIT, T-cell immunoreceptor with Ig and ITIM domains; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; CAR, chimeric antigen receptor; HER2, human epidermal growth factor receptor 2; T-VEC, talimogene laherparepvec; HNSCC, head and neck squamous cell carcinoma; mRNA, messenger RNA; NSCLC, non-small cell lung cancer; BiTEs, bispecific T-cell engagers; PSMA, prostate-specific membrane antigen; TIL, tumor-infiltrating lymphocyte. |
|