Fact-checked by Grok 2 weeks ago

Cell-mediated immunity

Cell-mediated immunity is a critical branch of the adaptive immune system that protects against intracellular pathogens, such as viruses and certain bacteria, by deploying specialized T lymphocytes to recognize and eliminate infected or abnormal cells. This form of immunity contrasts with humoral immunity, which relies on antibodies produced by B cells to target extracellular threats, as cell-mediated responses focus on direct cellular destruction and coordination of immune activities without antibody involvement. It plays an essential role in controlling infections, preventing cancer progression, and contributing to vaccine efficacy by generating long-lasting memory T cells that enable rapid responses upon re-exposure to antigens. The process begins with antigen-presenting cells (APCs), such as dendritic cells and macrophages, capturing and processing pathogens to display peptide antigens on (MHC) molecules. Naive T cells in lymphoid organs encounter these antigen-MHC complexes: CD4+ helper T cells bind to on APCs and differentiate into subsets like Th1 cells that secrete cytokines (e.g., IFN-γ) to activate macrophages, while CD8+ cytotoxic T cells bind to and proliferate into effectors that induce in infected cells via perforin and granzymes. This activation requires co-stimulatory signals and leads to clonal expansion, ensuring a targeted and amplified response. Cell-mediated immunity is vital for host defense against pathogens that evade humoral mechanisms, such as those residing inside cells, and is implicated in autoimmune diseases when dysregulated. In development, eliciting robust T cell responses enhances protection against chronic infections like or , often through adjuvants that stimulate APCs and promote memory formation. Its study has advanced , including the use of cytokine-induced killer cells for , highlighting its broader therapeutic potential.

Introduction

Definition and Scope

Cell-mediated immunity (CMI) refers to an adaptive mediated primarily by T lymphocytes that targets intracellular pathogens, virus-infected cells, tumor cells, and foreign tissues, operating independently of antibodies through direct cell-cell contact or the secretion of cytokines. This branch of immunity involves the specific recognition of antigenic peptides presented on the surface of infected or abnormal cells via (MHC) molecules, which activates naive T cells to undergo clonal expansion and differentiate into effector cells capable of inducing targeted or modulating other immune components. The scope of CMI extends to both adaptive mechanisms, centered on antigen-specific T cell responses that provide immunological memory for enhanced protection upon re-exposure, and innate contributions from cells like natural killer (NK) cells that offer rapid, non-specific surveillance against intracellular threats. While CMI emphasizes adaptive processes for precision and durability, it contrasts with humoral immunity by focusing on intracellular rather than extracellular challenges, such as free-floating bacteria or toxins. Evolutionarily, CMI emerged as a hallmark of adaptive immunity in jawed vertebrates, enabling sophisticated defense against diverse pathogens that evade humoral mechanisms by hiding within host cells. CMI is essential for controlling viral infections through the elimination of infected host cells, managing certain fungal diseases by activating macrophages to engulf and destroy intracellular fungi, and driving via recognition of allogeneic MHC molecules on grafted tissues. In its core process, professional antigen-presenting cells process antigens and display fragments bound to (for CD8+ cytotoxic T cells) or class II (for CD4+ helper T cells) molecules, initiating T cell activation that culminates in effector functions like perforin-mediated or cytokine-driven inflammation to resolve the threat.

Distinction from Humoral Immunity

Humoral immunity refers to the arm of the mediated by B lymphocytes, which produce and secrete antibodies to target extracellular pathogens such as and their toxins. These antibodies circulate in bodily fluids, neutralizing threats by binding to antigens, facilitating through opsonization, or activating the to lyse pathogens. In contrast, cell-mediated immunity (CMI) primarily involves T lymphocytes that directly interact with infected or abnormal cells through cell-cell contact, such as via cytotoxic mechanisms that induce target cell . While relies on soluble mediators to address extracellular invaders, CMI targets intracellular pathogens like viruses that reside within host cells, using (MHC) molecules to present antigenic peptides for T cell recognition. Specifically, molecules display intracellular peptides to + cytotoxic T cells, and molecules present extracellularly derived peptides to + helper T cells, enabling precise immune surveillance absent in humoral responses that recognize free antigens. CMI and exhibit complementary roles in orchestrating a comprehensive ; for instance, + helper T cells in CMI indirectly activate B cells to enhance production and class switching. This synergy ensures that CMI clears intracellular infections while humoral components neutralize extracellular threats, together providing robust protection against diverse pathogens. Representative examples illustrate these distinctions: CMI facilitates viral clearance by cytotoxic T cells eliminating virus-infected host cells, whereas neutralizes bacterial toxins through binding, preventing their harmful effects.

Historical Background

Early Discoveries

The earliest observations of cell-mediated immune phenomena emerged in the late through studies on . In 1890, described the tuberculin reaction, a delayed hypersensitivity response elicited by injecting a filtrate from cultures into previously infected individuals; this reaction, peaking 24-48 hours after injection, indicated a specific immune distinct from immediate antibody-mediated responses. By the early , researchers began recognizing that immunity to certain pathogens, particularly intracellular ones like the tubercle bacillus, could not be fully explained by humoral factors alone. This led to recognition of the importance of cellular mechanisms in resistance, emphasizing the role of host cells in containing without reliance on soluble antibodies. In the 1940s, experimental evidence solidified the cellular basis of these responses. and Merrill W. Chase demonstrated in guinea pigs that delayed-type to simple chemical compounds could be passively transferred using viable peritoneal cells from sensitized donors, but not cell-free , highlighting a non-antibody mechanism. Concurrently, Peter Medawar's studies on in rabbits during burn treatments revealed that allograft rejection involved an adaptive mediated by host cells, as rejected grafts showed lymphocytic infiltration rather than humoral involvement. These findings laid the experimental foundations through animal models, where immunity to or contact sensitizers was transferable via leukocytes or lymphoid cells, but not , underscoring the distinct cellular arm of adaptive immunity.

Key Milestones and Contributors

In the early 1960s, the foundational understanding of cell-mediated immunity advanced significantly with the discovery of the 's critical role in immune function. Australian immunologist Jacques F. A. P. demonstrated in 1961 that in newborn mice led to profound , particularly in cell-mediated responses, revealing that thymus-derived lymphocytes—later termed T cells—were essential for such immunity. This work built on earlier observations but pinpointed the as the site of T cell maturation, shifting focus from humoral to cellular mechanisms. By the late 1960s, the distinction between T cells and B cells emerged through experiments showing that T cells mediated graft rejection and delayed-type , while B cells drove production; key studies by and colleagues in 1968-1969 used reconstitution assays in thymectomized mice to delineate these lineages. The 1970s and 1980s brought molecular insights into T cell recognition and activation. In 1974, Rolf M. Zinkernagel and Peter C. Doherty identified MHC restriction, showing that cytotoxic T cells recognize viral antigens only when presented by self-major histocompatibility complex (MHC) molecules on infected cells, a discovery that earned them the 1996 Nobel Prize in Physiology or Medicine. This principle explained the specificity of cell-mediated responses and influenced vaccine design. Concurrently, the role of cytokines in T cell proliferation was elucidated; in 1976, Robert C. Gallo and Francis W. Ruscetti discovered interleukin-2 (IL-2), a T cell growth factor secreted by activated T cells that sustains their expansion during immune responses. By the mid-1980s, T cell receptor (TCR) genes were cloned—first in mice by Hedrick et al. in 1984, followed by human TCR by Yanagi et al.—revealing the molecular basis for antigen specificity in cell-mediated immunity. Co-stimulation mechanisms were also uncovered, with CD28 identified as a key receptor providing the second signal for T cell activation; monoclonal antibody studies in 1986 by Martin et al. demonstrated that CD28 engagement enhanced T cell proliferation and cytokine production. From the late 1980s onward, these insights fueled therapeutic innovations in cell-mediated immunity. Tim R. Mosmann and Robert L. Coffman proposed in 1986 the Th1/Th2 paradigm, classifying CD4+ T helper cells into subsets based on distinct profiles—Th1 cells promoting cell-mediated responses via IFN-γ, and Th2 cells favoring —providing a framework for understanding immune polarization. In 1989, Zelig Eshhar and colleagues conceptualized chimeric antigen receptors (CARs), T cells with synthetic receptors to bypass and directly target tumor antigens, laying the groundwork for CAR-T cell therapies. Post-2010, CRISPR-Cas9 revolutionized T cell for enhanced cell-mediated therapies; seminal work by Schumann et al. in 2015 achieved precise multiplex in human T cells, enabling knockouts of inhibitory genes like PD-1 to boost antitumor activity, while later applications created universal allogeneic CAR-T cells resistant to rejection. These efforts culminated in clinical breakthroughs, including the U.S. Food and Drug Administration's approval of the first CAR-T therapy, (Kymriah), in August 2017 for certain blood cancers, marking the first using modified patient T cells. Additionally, the 2018 in Physiology or Medicine was awarded to and for their discoveries of cancer therapy by inhibition of negative immune regulation, which unleashes T cell-mediated attacks on tumors via checkpoint inhibitors targeting CTLA-4 and PD-1. These milestones, driven by pioneers like , Zinkernagel, Doherty, and Mosmann, transformed cell-mediated immunity from a descriptive concept to a manipulable system for .

Cellular Components

T Lymphocytes

T lymphocytes, also known as T cells, originate from hematopoietic stem cells in the , where common lymphoid progenitors differentiate into early T cell precursors that subsequently migrate to the for further maturation. This migration ensures that T cells develop in a specialized microenvironment conducive to their antigen-specific receptor diversity and self-tolerance. T cells are broadly classified into several subtypes based on their function and surface markers, with CD4+ helper T cells playing a central role in coordinating immune responses by activating other immune cells through secretion, CD8+ cytotoxic T cells directly eliminating infected or malignant cells via perforin and granzymes, and regulatory T cells (Tregs) maintaining immune by suppressing excessive responses to prevent . CD4+ T cells typically recognize antigens presented by (MHC) class II molecules, while CD8+ T cells interact with , enabling precise targeting in cell-mediated immunity. Key surface markers define T cell identity and function: the T cell receptor (TCR) complex, composed of αβ or γδ chains, specifically recognizes peptide-MHC complexes on antigen-presenting cells; the associated CD3 complex transduces activation signals into the cell; and co-receptors CD4 or CD8 enhance TCR-MHC binding affinity and stabilize interactions during immune surveillance. Naive T cells, which have not yet encountered their specific antigen, exhibit long lifespans and continuously recirculate through blood and secondary lymphoid organs such as lymph nodes to patrol for pathogens, whereas memory T cells, generated after initial activation, persist for years or even decades to provide rapid and robust responses upon re-exposure, contributing to long-term adaptive immunity. In human peripheral blood, T cells constitute approximately 60-70% of circulating lymphocytes, underscoring their prominence in the adaptive immune system. Unlike innate effectors such as natural killer cells, T cells require antigen-specific priming for full functionality.

Natural Killer Cells and Other Innate Effectors

Natural killer (NK) cells are large granular lymphocytes that constitute approximately 5–15% of circulating lymphocytes in humans. These innate immune effectors provide rapid responses without prior sensitization, distinguishing them from adaptive T cells that require antigen-specific recognition. NK cells recognize target cells through the "missing-self" hypothesis, where inhibitory receptors such as killer-cell immunoglobulin-like receptors (KIRs) detect the absence of class I (MHC-I) molecules on healthy cells, preventing activation. Upon encountering stressed or infected cells lacking sufficient MHC-I, activating receptors trigger NK cell responses, including (ADCC) via and release such as interferon-gamma (IFN-γ). In cell-mediated immunity, NK cells serve as early defenders against viral infections and tumors by directly lysing infected or malignant cells and secreting IFN-γ to modulate the immune environment. This production enhances adaptive cell-mediated responses by activating macrophages and promoting T . Unlike adaptive immune cells, NK cells do not undergo clonal expansion or form classical immunological memory. However, evidence since the 2010s indicates that certain NK cell subsets can exhibit adaptive-like memory, such as enhanced responses to infection through epigenetic modifications. Other innate effectors supporting cell-mediated immunity include γδ T cells, which act in a semi-innate manner by recognizing stress-induced ligands via their T cell receptors without MHC restriction. Macrophages contribute as phagocytic cells that eliminate pathogens and present antigens to bridge innate and adaptive phases. Dendritic cells similarly function as antigen-presenting cells, capturing and processing antigens to initiate T cell responses while providing innate surveillance.

Development and Activation

T Cell Maturation in the Thymus

T cell progenitors, derived from hematopoietic cells in the , enter the through blood vessels at the corticomedullary junction, guided by such as CCL19, CCL21, and CCL25 via receptors CCR7 and CCR9. These early thymic progenitors (ETPs) initially lack expression of and co-receptors, marking the double-negative (DN) stage, which is subdivided into DN1 to DN4 phases based on and CD25 expression. During the DN stages, thymocytes proliferate and undergo T cell receptor (TCR) β-chain gene rearrangement, followed by pre-TCR signaling that promotes survival and progression to the double-positive (DP) stage. In the DP phase, thymocytes express both and and migrate to the thymic , where they complete TCR α-chain rearrangement to form the full αβ TCR. Positive selection occurs primarily in the among DP thymocytes, mediated by cortical thymic epithelial cells (cTECs) presenting self-peptides on (MHC) molecules. Thymocytes with TCRs that bind weakly to self-MHC survive and receive survival signals, leading to lineage commitment: those recognizing differentiate into + single-positive (SP) cells, while those recognizing become + SP cells; approximately 10% of DP thymocytes succeed in this process. Post-positive selection, SP thymocytes migrate to the thymic medulla for negative selection, where strong TCR affinity for self-peptide-MHC complexes triggers apoptosis to eliminate potentially autoreactive clones. This medullary deletion is facilitated by medullary thymic epithelial cells (mTECs) and dendritic cells presenting a broad array of self-antigens, including tissue-restricted antigens (TRAs) promiscuously expressed under the control of the autoimmune regulator (AIRE) transcription factor. AIRE enables mTECs to express over 1,000 TRAs, ensuring central tolerance by deleting high-affinity self-reactive T cells and promoting regulatory T cell development. The surviving SP thymocytes, now mature naive CD4+ or CD8+ T cells, complete maturation in the medulla over several days before exiting the via efferent lymphatics or blood vessels to seed peripheral lymphoid organs. Only about 1-5% of entering progenitors emerge as naive T cells, reflecting the stringent dual selection. begins around , characterized by progressive replacement of thymic with , leading to a 3% annual decline in thymic cellularity until age 35-45, followed by a slower 1% rate thereafter. This age-related reduces output of new naive T cells, impairing the diversity of the T cell repertoire and contributing to diminished efficiency of cell-mediated immunity in older individuals.

Antigen-Specific Activation Processes

Antigen-specific activation of naive T cells occurs primarily in secondary lymphoid organs, such as lymph nodes and the , where antigen-presenting cells, particularly dendritic cells, migrate to present processed antigens to circulating T lymphocytes. Dendritic cells capture and process antigens from peripheral tissues, then transport them to lymphoid tissues via lymphatic vessels, forming a structured environment that facilitates high-affinity interactions between T cells and antigen-major complex (MHC) complexes. This process initiates the adaptive by converting naive T cells, which have undergone thymic selection to recognize self-MHC, into antigen-specific effectors capable of targeting infected or abnormal cells.00267-8) The activation of naive T cells requires the coordinated delivery of three distinct signals to ensure productive immunity while preventing inappropriate responses. Signal 1 involves the binding of the (TCR) to antigens presented by MHC molecules on dendritic cells, triggering initial intracellular signaling cascades such as of zeta-chain associated 70 (ZAP-70) and activation of downstream pathways like nuclear factor of activated T cells (NFAT).00267-8) Signal 2 provides , typically through the interaction of on T cells with B7-1 () or B7-2 () ligands on antigen-presenting cells, which amplifies TCR signaling, promotes expression of anti-apoptotic proteins like , and sustains production.00267-8) Signal 3 consists of signaling, such as interleukin-12 (IL-12) for type 1 responses or IL-4 for type 2, which directs T cell differentiation and survival through activation of transcription factors like signal transducer and activator of transcription () proteins.00267-8) Upon receiving all three signals, activated T cells undergo clonal expansion, a rapid proliferation phase that amplifies antigen-specific clones from rare naive precursors to billions of effectors. This expansion is driven by an autocrine loop involving IL-2, where activated T cells produce and secrete IL-2, which binds to IL-2 receptors (primarily the high-affinity heterotrimeric form including CD25) on the same cells, promoting cell cycle progression via cyclin-dependent kinases and preventing apoptosis. During this phase, both CD4+ and CD8+ T cells proliferate extensively, with CD8+ T cells differentiating into cytotoxic T lymphocytes (CTLs) that express perforin and granzymes for target cell lysis, while naive CD4+ T cells polarize into subsets such as Th1 (promoted by IL-12 and T-bet transcription factor for interferon-gamma production), Th2 (driven by IL-4 and GATA3 for IL-4/IL-5/IL-13), Th17 (induced by transforming growth factor-beta and IL-6 via RORγt), or regulatory T cells (Tregs, fostered by IL-2 and Foxp3 for immunosuppression). A portion of these expanded cells differentiates into long-lived central memory T cells that recirculate through lymphoid tissues, providing rapid secondary responses upon re-encountering the same antigen. If activation signals are incomplete—particularly the absence of (signal 2)—T cells enter a state of anergy, a reversible hyporesponsive condition characterized by impaired IL-2 production, proliferation defects, and epigenetic changes like at promoter regions, which collectively prevent by tolerizing self-reactive clones. This anergic state can be induced by without B7 ligands, as demonstrated in models where T cells fail to expand or produce cytokines upon subsequent full , underscoring the role of co-stimulation in gating peripheral tolerance.80284-8)

Mechanisms of Effector Function

Cytotoxic Activity

Cytotoxic activity represents a core effector function in cell-mediated immunity, primarily executed by + cytotoxic T lymphocytes (CTLs) and natural killer () cells to eliminate infected or malignant target cells through direct induction of . These effectors deploy specialized lytic machinery upon of aberrant cells, ensuring precise destruction without widespread tissue damage. Target recognition by CTLs occurs via the (TCR) binding to antigenic peptides presented on class I (MHC-I) molecules on the surface of infected or transformed cells. In contrast, NK cells identify targets through a balance of activating and inhibitory receptors; they preferentially attack cells lacking MHC-I expression (the "missing self" hypothesis) or those coated with antibodies via Fc receptors, facilitating (ADCC). Stressed cells upregulate ligands for NK activating receptors, such as , further enhancing recognition. The primary mechanism of cytotoxicity involves the perforin/granzyme pathway, where effector cells release cytotoxic granules containing perforin and granzymes upon target engagement. Perforin polymerizes to form pores in the target cell membrane, enabling granzymes—serine proteases—to enter the cytosol and trigger caspase-dependent and -independent apoptosis pathways, leading to DNA fragmentation and cell death. A complementary pathway utilizes Fas ligand (FasL) expressed on the effector cell surface, which binds to Fas (CD95) receptors on the target, recruiting adaptor proteins like FADD and activating caspase-8 to initiate extrinsic apoptosis. CTLs and NK cells exhibit serial killing capacity, allowing a single effector to sequentially eliminate multiple targets by detaching from one dying and engaging another, often within minutes, thereby amplifying their impact during infections or tumors. This process is facilitated by spatiotemporal uncoupling of lytic granule release from sustained TCR or receptor signaling. To prevent to bystander healthy cells, cytotoxic activity is tightly regulated by inhibitory signals; for instance, cells converge lytic granules directionally at the , minimizing granule spillover, while both CTLs and cells express inhibitory receptors (e.g., KIRs on cells or PD-1 on CTLs) that dampen upon engagement with self-MHC or healthy ligands.

Cytokine-Mediated Responses

In cell-mediated immunity, cytokines serve as soluble messengers secreted primarily by activated T lymphocytes and other effectors, coordinating immune responses by amplifying , recruiting cells, and modulating effector functions without direct cell contact. Among the key cytokines, interferon-gamma (IFN-γ), produced mainly by Th1 cells and natural killer cells, plays a central role in activating macrophages to enhance their phagocytic and microbicidal activities, thereby promoting the classical pathway of cell-mediated immunity against intracellular pathogens. Tumor necrosis factor-alpha (TNF-α), released by macrophages and T cells, induces and in infected or transformed cells, further supporting the containment of infections. Interleukin-2 (IL-2), secreted predominantly by activated + T cells, drives the proliferation and survival of T lymphocytes and natural killer cells, sustaining the expansion of antigen-specific clones during immune responses. Helper T cells, particularly the Th1 subset, orchestrate cytokine-mediated responses by secreting IFN-γ, which not only reinforces macrophage activation but also influences interactions with innate immune cells like dendritic cells and neutrophils to amplify clearance.00118-7) While Th1 cells provide limited direct help to B cells compared to Th2 subsets, their cytokines such as IFN-γ can indirectly support humoral responses by enhancing and class-switching to IgG subclasses effective against intracellular threats. The polarization of naive CD4+ T cells into Th1 effectors is dictated by the local cytokine milieu; for instance, interleukin-12 (IL-12) produced by dendritic cells and macrophages during infection promotes Th1 through STAT4 signaling, ensuring a toward cell-mediated over humoral immunity.30128-1) Cytokine-mediated responses extend to systemic effects, where pro-inflammatory signals like TNF-α and IL-6 trigger the acute phase response in the liver, leading to the production of and other opsonins that aid in opsonization and clearance. These cytokines also contribute to fever induction by acting on the to elevate body temperature, creating an inhospitable environment for while enhancing immune motility. In chronic infections, IFN-γ and TNF-α are essential for formation, organizing macrophages and lymphocytes into structured aggregates that isolate and contain persistent intracellular microbes such as . Post-2020 research on severe has illuminated the risks of dysregulation in cell-mediated immunity, where hyperproduction of IFN-γ, TNF-α, and IL-6 culminates in storms, leading to widespread and damage that underscores the need for balanced effector responses.

Specific Pathways

Type 1 Immunity

Type 1 immunity represents a critical arm of cell-mediated immunity characterized by the differentiation and activation of T helper 1 (Th1) cells, which drive interferon-gamma (IFN-γ) production in response to interleukin-12 (IL-12) signaling, ultimately activating macrophages to eliminate intracellular pathogens. This pathway is initiated when antigen-presenting cells, such as dendritic cells, detect microbial signals and secrete IL-12, which binds to the IL-12 receptor on naïve + T cells, triggering signal transducer and activator of transcription 4 (STAT4) and promoting Th1 . The resulting Th1 cells then produce high levels of IFN-γ, a potent activator of classical M1 macrophages that enhances their phagocytic and microbicidal activities through upregulation of inducible (iNOS) and production. This IFN-γ/IL-12 axis is essential for coordinating innate and adaptive responses against infections confined within host cells, distinguishing it from by emphasizing cellular and over production. Central to Th1 differentiation is the T-bet (encoded by Tbx21), which acts as a master regulator by directly binding to the Ifng promoter to drive IFN-γ expression and suppress alternative T helper lineages. T-bet expression is induced early in response to IL-12 and IFN-γ signals, forming a feed-forward loop that reinforces Th1 commitment through and coordination with other factors like STAT4 and RUNX3. This lineage-specific program ensures robust Th1 responses tailored to intracellular threats, with T-bet-deficient models exhibiting impaired IFN-γ production and increased susceptibility to infections. Type 1 immunity primarily targets viruses and intracellular bacteria such as , where Th1-derived IFN-γ activates to restrict pathogen replication within . For instance, in mycobacterial infections, Th1 cells promote the production of opsonizing IgG isotypes like IgG1 and IgG3, which facilitate and complement activation without shifting toward Th2-dominated responses. In viral contexts, such as or LCMV infections, Th1 responses enhance cytotoxic T lymphocyte function and control of viral spread. Recent advancements in design have leveraged Th1 pathways to improve efficacy; for example, BCG revaccination in latently infected individuals boosts polyfunctional Th1/Th17 responses, enhancing protection against through sustained IFN-γ and IL-2 production. Post-2015 studies on adjuvanted BCG formulations, such as those incorporating IL-12 or type I interferons, have demonstrated amplified Th1 polarization, leading to reduced bacterial burdens in preclinical models. Dysregulation of type 1 immunity, particularly excessive Th1 responses, contributes to autoimmune conditions like , where unchecked IFN-γ production drives β-cell destruction in the . In non-obese diabetic () models, of TGF-β signaling in T cells results in hyperactive Th1 differentiation and rapid onset due to dysregulated IFN-γ and impaired homeostasis. Human studies corroborate this link, showing elevated Th1-associated chemokines like IP-10 in patients, correlating with disease progression and .

Cell-Mediated Hypersensitivity

Cell-mediated , also known as , represents a delayed mediated by sensitized T lymphocytes rather than antibodies, distinguishing it from the antibody-dependent types I-III hypersensitivities. This reaction typically manifests 24 to 72 hours after antigen exposure, as it requires T cell activation, , and recruitment of inflammatory cells, leading to tissue damage through release and direct . Unlike immediate reactions driven by IgE or immune complexes, type IV responses involve + helper T cells and + cytotoxic T cells that recognize presented by MHC molecules on antigen-presenting cells. Type IV hypersensitivity is further subdivided into subtypes IVa through IVd based on the predominant T cell subsets and cytokines involved. Subtype IVa is characterized by Th1 cells producing interferon-gamma (IFN-γ), interleukin-1 (IL-1), and IL-2, which activate monocytes and macrophages, as seen in classic delayed-type hypersensitivity (DTH) reactions. Subtype IVb involves Th2 cells secreting IL-4, IL-5, and IL-13, promoting recruitment and activation with IgE production. Subtype IVc features CD8+ T cells exerting direct via perforin and granzymes, while subtype IVd is driven by Th17 cells releasing IL-17 and , inducing infiltration. These subtypes highlight the diverse profiles that underpin the inflammatory pathology in cell-mediated responses. Key mechanisms of include antigen-specific T cell sensitization followed by effector phase inflammation. In , such as that induced by in , hapten antigens penetrate the skin, conjugate to self-proteins, and are presented to T cells, primarily + effectors that mediate epidermal damage through . The skin test exemplifies a controlled DTH response, where of purified protein derivative from elicits induration due to IFN-γ-producing Th1 cells recruiting macrophages. These processes underscore the role of memory T cells in amplifying reactions upon re-exposure. Clinical examples of type IV hypersensitivity include (GVHD), where donor T cells in transplanted tissue recognize host antigens as foreign, leading to multi-organ inflammation via + and + mediated cytotoxicity. In celiac disease, peptides presented by or DQ8 molecules activate gluten-specific + T cells, triggering intestinal epithelial damage through IFN-γ and IL-21 secretion. Such responses illustrate how dysregulated cell-mediated immunity can target self or environmental antigens, resulting in chronic pathology. Therapeutic modulation of type IV hypersensitivity often targets T cell activation pathways. Calcineurin inhibitors like cyclosporine bind to cyclophilin, inhibiting the calcineurin-NFAT pathway and thereby suppressing IL-2 production and T cell proliferation, which is effective in managing severe contact dermatitis and preventing GVHD.

Physiological Roles and Dysregulation

Protection Against Intracellular Pathogens

Cell-mediated immunity (CMI) plays a pivotal role in defending against intracellular pathogens, including viruses, bacteria, and parasites that evade humoral responses by residing within host cells. CD4+ and CD8+ T cells orchestrate this defense by recognizing infected cells via antigen presentation on MHC molecules, leading to targeted elimination and activation of other immune effectors. This process is crucial for containing infections that could otherwise disseminate systemically, preventing severe disease outcomes. In viral infections, CD8+ T cells are primary effectors, identifying and eliminating virus-infected cells through mechanisms such as perforin-mediated lysis and cytokine secretion. For instance, during (HBV) infection, HBV-specific CD8+ T cells can clear the virus from hepatocytes without necessarily killing the host cell, demonstrating non-cytolytic control via interferon-gamma (IFN-γ) production. A notable example is seen in HIV elite controllers, a rare subset of individuals who maintain undetectable viral loads without antiretroviral therapy, primarily due to robust, polyfunctional CD8+ T cell responses that suppress viral replication in infected CD4+ T cells. These responses target conserved epitopes, sustaining long-term control through both cytolytic and non-cytolytic effects. For intracellular bacteria like (Mtb), CMI facilitates formation, structured aggregates of immune cells that isolate and limit bacterial growth in the lungs. CD4+ T cells, particularly Th1 subsets, produce IFN-γ to activate macrophages within granulomas, enhancing their bactericidal activity and preventing dissemination, as evidenced by spatial transcriptomic analyses of human TB lesions showing coordinated T cell-macrophage interactions. Similarly, in infection, + T cells employ to lyse infected host cells, with perforin-deficient models exhibiting impaired bacterial clearance and increased mortality, underscoring the granule pathway's necessity. Parasitic infections, such as those caused by , are controlled through IFN-γ-driven activation of macrophages and other innate cells, which deploy like Irgb6 to disrupt the parasitophorous vacuole and eliminate intracellular tachyzoites. Genome-wide screens in IFN-γ-primed macrophages have identified over 350 T. gondii genes influencing parasite fitness, confirming the parasite's reliance on evading this CMI mechanism for survival. Live-attenuated vaccines, such as the , harness CMI by mimicking natural infection to generate long-lived memory T cells; the Edmonston strain induces durable CD4+ and CD8+ T cell responses against measles virus epitopes, contributing to lifelong protection and preventing complications like . Recent studies from the 2020s highlight CMI's involvement in persistence, where dysregulated T cell responses correlate with prolonged symptoms. In post-acute sequelae of (PASC), individuals exhibit altered T cell subset distributions, including exhausted + T cells and persistent , potentially driven by residual viral antigens that hinder effective clearance. Longitudinal analyses show that while spike-specific + T cells may wane, cytotoxic + T cells persist in some cases, yet their dysfunction contributes to symptom chronicity, as seen in cohorts followed up to three years post-infection. These findings emphasize CMI's dual role in resolution and potential during viral persistence.

Involvement in Autoimmunity and Cancer

Cell-mediated immunity (CMI) plays a paradoxical role in autoimmunity, where failure of self-tolerance mechanisms allows autoreactive T cells to drive chronic inflammation and tissue damage. In rheumatoid arthritis (RA), Th17 cells, a subset of CD4+ T helper cells, promote joint inflammation by secreting pro-inflammatory cytokines such as IL-17 and IL-22, exacerbating synovial hyperplasia and cartilage destruction.30146-8) Similarly, in multiple sclerosis (MS), CD8+ T cells infiltrate the central nervous system (CNS), recognizing myelin antigens and contributing to demyelination through cytotoxic mechanisms and cytokine release, with clonal expansions of these cells observed in MS lesions. This breakdown in tolerance is often linked to genetic factors, including strong associations with human leukocyte antigen (HLA) alleles; for instance, HLA-DRB104:01 increases RA risk by enhancing presentation of arthritogenic peptides to autoreactive T cells, while HLA-DRB115:01 is a major susceptibility factor for MS by influencing CD8+ T cell responses in the CNS. In cancer, CMI contributes to immune surveillance by enabling T cells to detect and eliminate transformed cells expressing neoantigens. Tumor-infiltrating lymphocytes (TILs), primarily CD8+ cytotoxic T cells, accumulate within tumors and exert anti-tumor effects by recognizing tumor-specific antigens and inducing apoptosis in malignant cells, with higher TIL density correlating to improved prognosis in cancers such as melanoma and breast cancer. However, tumors evade this surveillance through upregulation of immune checkpoints, notably the PD-1/PD-L1 axis, where PD-L1 expression on tumor cells binds PD-1 on activated T cells, inhibiting their cytotoxic function and promoting T cell exhaustion to facilitate immune escape. Therapeutic strategies harnessing CMI have transformed treatment landscapes for both conditions. In autoimmunity, targeting dysregulated CMI pathways, such as Th17 inhibition with IL-17 blockers, has shown efficacy in RA, while in cancer, immune checkpoint inhibitors like ipilimumab, a CTLA-4 monoclonal antibody approved by the FDA in 2011 for metastatic melanoma, enhance T cell activation to boost anti-tumor responses. Post-2020 advancements include bispecific T cell engagers (BiTEs), engineered antibodies that simultaneously bind tumor antigens and CD3 on T cells to redirect cytotoxic activity, with agents like blinatumomab (approved for hematologic malignancies) and emerging solid tumor candidates demonstrating durable responses by overcoming checkpoint-mediated evasion. Regulatory T (Treg) cells, characterized by Foxp3 expression, maintain a delicate balance in CMI by suppressing autoreactive T cells to prevent , yet their accumulation in the inhibits anti-tumor TILs through mechanisms like IL-10 and TGF-β secretion, thereby promoting that favors tumor progression. This duality underscores the therapeutic challenge of selectively modulating Treg function to enhance anti-cancer immunity without triggering autoimmune flares.

References

  1. [1]
    Physiology, Immune Response - StatPearls - NCBI Bookshelf - NIH
    The immune response is the body's ability to stay safe by protecting against harmful agents. The response involves lines of defense against most microbes.
  2. [2]
    T Cell-Mediated Immunity - Immunobiology - NCBI Bookshelf - NIH
    Cell-mediated immune responses involve the destruction of infected cells by cytotoxic T cells, or the destruction of intracellular pathogens by macrophages ( ...
  3. [3]
    Cell-Mediated Immunity and Vaccines - PMC - NIH
    Cell-mediated immunity (CMI) is one of the important effector arms during immunological responses to infection and vaccine development.
  4. [4]
    An introduction to immunology and immunopathology - PMC - NIH
    Sep 12, 2018 · Cell-mediated immunity is directed primarily at microbes that survive in phagocytes as well as those that infect non-phagocytic cells.
  5. [5]
    Evolution of adaptive immunity in vertebrates - PMC - PubMed Central
    The adaptive immune system in all of the jawed vertebrates is featured by antigen-specific cellular and humoral immunity, which together provide durable ...
  6. [6]
    The Humoral Immune Response - Immunobiology - NCBI Bookshelf
    The humoral immune response is mediated by antibody molecules that are secreted by plasma cells. Antigen that binds to the B-cell antigen receptor signals B ...
  7. [7]
    Antibody-Mediated Immunity against Intracellular Pathogens
    The view that antibody-mediated immunity protects against extracellular pathogens and cell-mediated immunity protects against intracellular pathogens has been ...
  8. [8]
    Humoral and Cell-Mediated Immunity to the Intracellular Pathogen ...
    Cell-mediated immunity has long been believed to be critical for protection and the importance of humoral immunity is also now recognized. Furthermore, synergy ...
  9. [9]
    Koch's Discovery of the Tubercle Bacillus - CDC
    In 1890, he announced the discovery of tuberculin, a substance derived from tubercle bacilli, which he thought was capable of arresting bacterial development ...
  10. [10]
    The history of tuberculosis: from the first historical records to ... - NIH
    The famous scientist Robert Koch was able to isolate the tubercle bacillus and presented this extraordinary result to the society of Physiology in Berlin on 24 ...
  11. [11]
    Delayed type hypersensitivity: Current theories with a historic ...
    It was not until 1942 that Landsteiner and Chase demonstrated that the DTH reaction could be transferred by a "cell only" fraction.[3] The basis for the ...
  12. [12]
    Peter Medawar – Nobel Lecture - NobelPrize.org
    The term first came to be used in the context of tissue transplantation immunity, i.e. of the form of immunity that usually prohibits the grafting of ...
  13. [13]
    Delayed type hypersensitivity: current theories with an historic ...
    Delayed type hypersensitivity ... Landsteiner and Chase proved that the reaction was mediated by the cellular and not the humoral arm of the immune system.Missing: transfer 1942
  14. [14]
    A human T cell-specific cDNA clone encodes a protein ... - Nature
    Mar 8, 1984 · We have cloned and sequenced a human mRNA specific for mammalian T-lymphoid cells. The message was found to be expressed in human and murine T lymphoblasts.
  15. [15]
    Human T cell development, localization, and function throughout life
    Feb 20, 2019 · T lymphocytes originate from bone marrow progenitors that migrate to the thymus for maturation, selection, and subsequent export to the ...
  16. [16]
    T-Cell Lineage Determination - PMC - NIH
    T cells originate from hematopoietic stem cells (HSCs) in the bone marrow but complete their development in the thymus. HSCs give rise to a variety of ...
  17. [17]
    Different Subsets of T Cells, Memory, Effector Functions, and CAR-T ...
    Mar 15, 2016 · This review is focused on different subsets of T cells: CD4 and CD8, memory and effector functions, and their role in CAR-T therapy.2. Cd4 Cell Subsets · 4. Cd8 Cell Subsets And Cell... · Figure 6<|control11|><|separator|>
  18. [18]
    Histology, T-Cell Lymphocyte - StatPearls - NCBI Bookshelf
    CD4+ T cells recognize MHC 2 bound antigens, while CD8+ T cells recognize MHC 1 bound antigens. Both CD4+ T cells and CD8+ T cells have the TCR (and the co- ...
  19. [19]
    CD4 and CD8 molecules can physically associate with the same T ...
    CD4 and CD8 function by binding to class II and class I MHC molecules on the antigen-presenting cell (APC), thereby increasing the adhesion between the T cell ...<|control11|><|separator|>
  20. [20]
    Qualitative differences between naïve and memory T cells - PMC
    In general, these studies have shown that the lifespan of naïve CD4+ and CD8+ T cells is shortened in the absence of MHC class II or class I, respectively.
  21. [21]
    Naive and memory T cells show distinct pathways of lymphocyte ...
    In this report, we have addressed two questions concerning immunological memory: the way in which naive and memory T cells recirculate through the body, ...
  22. [22]
    CD4+ T Lymphocytes count in sickle cell anaemia patients attending ...
    Thymus-derived (T) lymphocytes play an important role in cellular immunity. In the blood, T lymphocytes constitute 60-70% of peripheral lymphocytes and are ...Missing: comprise | Show results with:comprise
  23. [23]
    Natural Killer Cells and Cytotoxic T Cells - PubMed Central - NIH
    They constitute about 5–15% of all lymphocytes circulating in the blood, which makes them the third largest lymphocyte population, after T and B cells. Humans ...3. Cytotoxic Cd8 T Cells Or... · 3.1. Important Cd8 T-Cell... · 5. How Nk Cells And...
  24. [24]
    Functions of natural killer cells | Nature Immunology
    Apr 18, 2008 · Natural killer (NK) cells are effector lymphocytes of the innate immune system that control several types of tumors and microbial infections.
  25. [25]
    Natural killer cells in antitumour adoptive cell immunotherapy - Nature
    Jul 25, 2022 · Natural killer (NK) cells comprise a unique population of innate lymphoid cells endowed with intrinsic abilities to identify and eliminate virally infected ...
  26. [26]
    Comprehensive snapshots of natural killer cells functions, signaling ...
    Nov 8, 2024 · Natural killer (NK) cells were first described as killer lymphocytes that induce rapid leukemia cell death without requiring soluble factors and ...
  27. [27]
    Natural killer cells in antiviral immunity | Nature Reviews Immunology
    Jun 11, 2021 · Natural killer (NK) cells play an important role in innate immune responses to viral infections. Here, we review recent insights into the role of NK cells in ...
  28. [28]
    γδ T cells: origin and fate, subsets, diseases and immunotherapy
    Nov 22, 2023 · They can rapidly respond to various pathogens through their innate-like receptors, allowing for early immune defense even in the absence of ...
  29. [29]
    Macrophages in immunoregulation and therapeutics - Nature
    May 22, 2023 · Macrophages are crucial in innate immunity by regulating several homeostatic and evolutionary host defense immune responses.
  30. [30]
    Mucosal dendritic cells shape mucosal immunity - Nature
    Mar 14, 2014 · Dendritic cells (DCs) are key modulators that shape the immune system. In mucosal tissues, DCs act as surveillance systems to sense infection.
  31. [31]
  32. [32]
  33. [33]
  34. [34]
    T-cell activation by dendritic cells in the lymph node - Nature
    Interactions between T cells and dendritic cells (DCs) in the lymph nodes are crucial for initiating cell-mediated adaptive immune responses.
  35. [35]
    Harnessing IL-2 for immunotherapy against cancer and chronic ...
    Sep 2, 2024 · IL-2 is a pivotal cytokine the plays a role in the clonal expansion of activated T cells and enhances the cytotoxic activity of CD8 + T cells ...
  36. [36]
    Similarities and differences in CD4+ and CD8+ effector and memory ...
    Sep 1, 2003 · This review compares and contrasts how naive CD4 + and CD8 + T cells make the transition to effector and/or memory cells and discusses the implications of ...
  37. [37]
    CD8+ T Cells and NK Cells: Parallel and Complementary Soldiers of ...
    CD8+ T cells and NK cells have different mechanisms of target recognition and signaling cascades to achieve very similar goals: to kill infected and transformed ...
  38. [38]
    Blockade of the granzyme B/perforin pathway through ... - PNAS
    Target-cell killing by CTLs involves the induction of apoptosis by two major mechanisms: through death receptors and the perforin/granzyme B (GrB) pathway.
  39. [39]
    Cytotoxic CD8 + T cells in cancer and cancer immunotherapy - Nature
    Sep 15, 2020 · Cytotoxic T cells expressing cell-surface CD8 are the most powerful effectors in the anticancer immune response and form the backbone of current successful ...
  40. [40]
    NK-mediated antibody-dependent cell-mediated cytotoxicity in solid ...
    Our review will focus on NK cells, mAbs, possible mechanisms to enhance ADCC and preclinical and clinical data focusing on ADCC in vivo.
  41. [41]
    Article Cytotoxic Cell Granule-Mediated Apoptosis: Perforin Delivers ...
    The mechanism underlying perforin (PFN)-dependent delivery of apoptotic granzymes during cytotoxic cell granule-mediated death remains speculative.Missing: paper | Show results with:paper
  42. [42]
    The many roles of FAS receptor signaling in the immune system - PMC
    The role of FASL-FAS induced apoptosis in normal as well as pathological killing of target cells. Both classical CD8+ cytotoxic T cells as well as a portion of ...Missing: paper | Show results with:paper
  43. [43]
    Cytotoxic T lymphocytes kill multiple targets simultaneously ... - PNAS
    Jul 18, 2006 · Cytotoxic T lymphocytes kill multiple targets simultaneously via ... The combination of “serial” and multiple killing can amplify cytotoxic ...
  44. [44]
    Cytotoxic T lymphocytes kill multiple targets simultaneously via ...
    Jul 18, 2006 · Cytotoxic T lymphocytes kill multiple targets simultaneously via spatiotemporal uncoupling of lytic and stimulatory synapses ... multiple killing ...Missing: serial paper
  45. [45]
    Th1 cytokines, true functional signatures for protective immunity ...
    Nov 20, 2017 · Among these subsets, IFN-γ-producing Th1 cells are accepted as the major population that mediates protective immunity against TB. Indeed, mice ...
  46. [46]
    Harnessing innate immune pathways for therapeutic advancement ...
    Mar 25, 2024 · The innate immune pathway is receiving increasing attention in cancer therapy. This pathway is ubiquitous across various cell types, ...
  47. [47]
    Cytokines in clinical cancer immunotherapy | British Journal of Cancer
    Nov 9, 2018 · Interleukin-2. IL-2 is viewed as a key cytokine in promoting the expansion of natural killer (NK) cells and T lymphocytes. Thus, it is widely ...
  48. [48]
    T cells in health and disease | Signal Transduction and Targeted ...
    Jun 19, 2023 · T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus.
  49. [49]
    Humoral Innate Immunity and Acute-Phase Proteins
    Feb 1, 2023 · We review key aspects of the production, structure, and function of selected acute-phase proteins, which continue to represent pillar diagnostic tools.
  50. [50]
    Cytokines in Febrile Diseases - Mary Ann Liebert, Inc.
    This article reviews the pathophysiological characteristics of fever and the cytokines related to fever (IL-2, 4, 6, 8, 10, IFN, TNF, etc.).
  51. [51]
    Immune mechanisms of granuloma formation in sarcoidosis ... - JCI
    Jan 2, 2024 · In this Review, we perform a mechanistic interrogation of the immune activities that contribute to granuloma formation in sarcoidosis and compare these ...
  52. [52]
    Deep insight into cytokine storm: from pathogenesis to treatment
    Apr 16, 2025 · Additionally, TNF and IFN-γ are two important pro-inflammatory cytokines that can activate kinases of the JAK family, particularly JAK1. This ...
  53. [53]
    C-terminal truncation of IFN-γ inhibits proinflammatory macrophage ...
    Jun 20, 2018 · Proinflammatory macrophages are classically activated by interferon-γ (IFN-γ) secreted from type 1 T helper (Th1) cells and natural killer (NK) ...
  54. [54]
    Classical versus alternative macrophage activation - Nature
    Jul 30, 2014 · IFN-γ production by Th1-type T cells and NK cells can stimulate M1 macrophage activation in a signal transducer and activator of transcription ( ...
  55. [55]
    A Novel Transcription Factor, T-bet, Directs Th1 Lineage Commitment
    Here, we report the isolation of T-bet, a Th1-specific T box transcription factor that controls the expression of the hallmark Th1 cytokine, IFNγ.
  56. [56]
    T-bet and GATA3 orchestrate Th1 and Th2 differentiation ... - Nature
    Dec 11, 2012 · Differentiation of CD4+ T cells into Th1 and Th2 effector cells is controlled by the transcription factors T-bet and GATA3, respectively. T-bet ...
  57. [57]
    The TH1 cell lineage-determining transcription factor T-bet ... - NIH
    Apr 19, 2022 · In CD4+ T cells, T-bet dictates differentiation of the TH1 lineage, whereas GATA3 drives differentiation of the alternative TH2 ...
  58. [58]
    IFN-γ-independent immune markers of Mycobacterium tuberculosis ...
    May 20, 2019 · We show that 'resisters' possess IgM, class-switched IgG antibody responses and non-IFN-γ T cell responses to the Mtb-specific proteins ESAT6 and CFP10.
  59. [59]
    Different antibody isotypes against tuberculosis: what we know and ...
    The five human antibody isotypes (IgG, IgM, IgA, IgE, and IgD) mediate an array of functional activities against bacterial infections, including neutralization ...
  60. [60]
    The transcription factors T-bet and GATA-3 control alternative ...
    T-bet is known to directly activate expression of IFN-γ to promote Th1 differentiation (4, 17–23). To determine whether the human T-bet gene targets were ...
  61. [61]
    BCG revaccination boosts adaptive polyfunctional Th1/Th17 and ...
    The strong evidence we provide that BCG revaccination can boost polyfunctional Th1/Th17 and innate effectors in subjects living with latent TB in the absence ...
  62. [62]
    Promising Cytokine Adjuvants for Enhancing Tuberculosis Vaccine ...
    IFN-α enhances BCG's immune response by promoting Th1-type cytokines and reducing Th2-type cytokines [28]. In another study, consecutive boosts of IFN-α in BCG- ...1. Introduction · Table 1 · 2. Type I Ifns
  63. [63]
    Excessive Th1 responses due to the absence of TGF-β signaling ...
    Excessive Th1 responses due to the absence of TGF-β signaling cause autoimmune diabetes and dysregulated Treg cell homeostasis.
  64. [64]
    Excessive Th1 Responses Due to the Absence of TGF-β Signaling ...
    Here we showed that abrogation of TGF-β signaling in thymic T cells led to rapid type 1 diabetes (T1D) development in NOD mice transgenic for the BDC2.5 T-cell ...
  65. [65]
    Elevated Serum IP-10 Levels Observed in Type 1 Diabetes
    Mar 1, 2001 · Here, we focused on interferon-inducible protein-10 (IP-10), a chemokine that promotes the migration of activated T-helper 1 (Th1) cells and ...
  66. [66]
    Type IV Hypersensitivity Reaction - StatPearls - NCBI Bookshelf - NIH
    May 4, 2025 · Type IV hypersensitivity reactions are T-cell–mediated immune responses that typically develop 48 to 72 hours or longer after exposure to an antigen.
  67. [67]
    Mechanisms of Hypersensitivity - PMC - PubMed Central - NIH
    Most hypersensitivities to drugs manifest as type I or type IV reactions. Type II and type III drug hypersensitive reactions are far less often seen and are ...
  68. [68]
    [Type IV of hypersensitivity and its subtypes] - PubMed
    IVa with Th1 and monocyte directed and cytokines: IFNgamma, IL-1, IL-2, 2. IVb with Th2 and eosinophils directed and cytokines: L-5, IL-4, IL-13, 3. IVc ...
  69. [69]
    CD8+ T cells are the effectors of the contact dermatitis induced by ...
    CD8+ T cells are the effectors of the contact dermatitis induced by urushiol in mice and are regulated by CD4+ T cells.
  70. [70]
    Osmosis: Graft-versus-host disease (GvHD) Flashcards | Quizlet
    -is a type IV hypersensitivity reaction -is a transplant rejection disorder that involves grafted immunocompetent T cells that proliferate in the ...
  71. [71]
    T cells in celiac disease - PMC - PubMed Central - NIH
    It was determined that gluten specific T cells are present in biopsies of all untreated CD patients at a frequency of 0.1–1.2% of CD4 T cells, and, the results ...Missing: hypersensitivity | Show results with:hypersensitivity
  72. [72]
    Therapeutics in Allergic Contact Dermatitis, when Avoidance Fails
    Sep 17, 2014 · Cyclosporine is a systemic calcineurin inhibitor that is often effective at inducing a rapid resolution of the signs and symptoms of ACD.
  73. [73]
    Th17 cell pathogenicity in autoimmune disease - Nature
    Sep 1, 2025 · T helper 17 (Th17) cells have been implicated in numerous inflammatory autoimmune diseases. Clinical benefits from targeting Th17 ...
  74. [74]
    Pathogenic CD8 T Cells in Multiple Sclerosis and Its Experimental ...
    Lymphocytes in MS plaques are biased toward the CD8 lineage, and MS patients harbor CD8 T cells specific for multiple central nervous system (CNS) antigens.
  75. [75]
    Myelin-specific CD8 + T cells exacerbate brain inflammation in ... - JCI
    CD8+ T cells are the predominant lymphocyte seen in CNS lesions of MS patients (13, 14). Clonal expansion is more commonly observed among CD8+ compared with CD4 ...
  76. [76]
    HLA Association with Autoimmune Diseases - NCBI - NIH
    The HLA-DRB1*15:01 and HLA-DQB1*06:02 alleles, which are in LD, are the main alleles associated with risk for MS in Caucasians and Latin Americans (18).
  77. [77]
    Associations of human leukocyte antigens with autoimmune diseases
    Aug 20, 2015 · (a) HLA associations with autoimmune diseases may be explained by the selective presentation of disease-relevant self-peptides by the disease ...
  78. [78]
    Tumor-infiltrating lymphocytes in cancer immunotherapy
    Tumor-infiltrating lymphocytes (TILs) are a diverse population of immune cells that play a central role in tumor immunity and have emerged as critical mediators ...
  79. [79]
    Regulatory mechanisms of PD-1/PD-L1 in cancers - Molecular Cancer
    May 18, 2024 · PD-1, a checkpoint receptor expressed on T cells, causes T cell exhaustion and prevents excessive immune stimulation when it receives signals ...
  80. [80]
    Immune Checkpoint Inhibitors - NCI - National Cancer Institute
    Apr 7, 2022 · The binding of PD-L1 to PD-1 keeps T cells from killing tumor cells in the body (left panel). Blocking the binding of PD-L1 to PD-1 with an ...
  81. [81]
    FDA Approves YERVOY™ (ipilimumab) for the Treatment of Patients ...
    Mar 25, 2011 · YERVOY is the first and only therapy to demonstrate a statistically significant overall survival benefit in patients with unresectable or ...Missing: checkpoint inhibitor
  82. [82]
    Current landscape of T-cell engagers in early-phase clinical ... - NIH
    Oct 6, 2025 · These molecules bridge T lymphocytes with tumor cells, generating an immunologic synapse that leads to potent immune-mediated tumor destruction.
  83. [83]
    Bispecific T cell engagers: an emerging therapy for management of ...
    May 3, 2021 · Recent studies have focused on improving the efficacy of BiTEs by optimizing treatment regimens and refining the molecular structures of BiTEs.
  84. [84]
    Regulatory T Cells in Autoimmunity and Cancer - PubMed
    Sep 3, 2021 · Regulatory T (Treg) cells, possess a strategic role in the maintenance of immune homeostasis, and their function has been closely linked to ...
  85. [85]
    Regulatory T Cells in Autoimmunity and Cancer - Frontiers
    We review the literature on the Treg cell plasticity with lessons learned in autoimmunity and cancer and discuss challenges and open questions.