Fact-checked by Grok 2 weeks ago

Cytotoxic T cell

Cytotoxic T cells, also known as or cytotoxic T lymphocytes (CTLs), are a specialized of T lymphocytes that function as key effectors in the , primarily responsible for recognizing and eliminating virus-infected cells, intracellular pathogens, tumor cells, and other abnormal cells through targeted induction of . These cells originate from hematopoietic stem cells in the and mature in the , where they undergo positive and negative selection to ensure they can bind to ( molecules while avoiding self-reactivity. Upon maturation, naïve cytotoxic T cells circulate in the and lymphoid tissues until activation, after which they differentiate into effector cells capable of rapid cytotoxicity. Activation of cytotoxic T cells requires two signals: the first involves the (TCR) complex, including CD3 and , recognizing antigenic peptides presented by molecules on antigen-presenting cells or infected target cells, and the second is a costimulatory signal via binding to B7 molecules (/) on the presenting cell. This process forms an at the contact site, enabling polarized release of cytotoxic granules and preventing damage to bystander cells. Once activated, these cells proliferate and migrate to sites of infection or tumors, where they exert their effector functions. The primary mechanisms of cytotoxicity employed by these cells include the release of perforin and granzymes from lytic granules, as well as the expression of (FasL). Perforin polymerizes to form pores in the target , allowing granzymes to enter and activate , which dismantle the cell through , a process that can occur within minutes of contact. Alternatively, FasL on the cytotoxic T cell binds to receptors on the target, triggering an extrinsic pathway via activation. In addition to direct killing, activated cytotoxic T cells secrete cytokines such as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), which enhance antiviral states in neighboring cells and activate other immune components like macrophages. Cytotoxic T cells are pivotal in immune surveillance against intracellular threats and malignancies, contributing to long-term immunity through the formation of + T cells that provide faster responses upon re-exposure to antigens. However, in chronic infections or tumors, they can become exhausted, marked by upregulated inhibitory receptors like PD-1 and CTLA-4, reducing their efficacy—a phenomenon targeted by inhibitors in , which have shown durable responses in over 50% of patients with metastatic . Dysregulation of these cells also underlies certain autoimmune diseases, such as , where they erroneously attack self-tissues.

Basic Biology

Definition and Characteristics

Cytotoxic T cells, also known as CD8+ T lymphocytes, are a subset of T cells in the specialized for recognizing and eliminating virus-infected cells, tumor cells, and other damaged or aberrant cells through the detection of antigens presented on ( molecules. These cells express the co-receptor, which binds to the invariant region of , enhancing the affinity of the (TCR) for the peptide-MHC complex and facilitating upon antigen encounter. The TCR on cytotoxic T cells is typically composed of α and β chains, forming a heterodimer that specifically recognizes the antigenic in the context of self-. Additionally, these cells express adhesion molecules such as leukocyte function-associated antigen 1 (LFA-1), an that promotes stable interactions with target cells and antigen-presenting cells during immune surveillance. The primary functions of cytotoxic T cells include inducing in target cells to prevent the spread of intracellular pathogens or , as well as secreting pro-inflammatory cytokines to amplify immune responses. Key cytokines produced include interferon-gamma (IFN-γ), which activates macrophages and enhances expression on nearby cells, and tumor necrosis factor-alpha (TNF-α), which contributes to and direct cytotoxic effects. These effector roles position cytotoxic T cells as central mediators of , bridging innate and adaptive responses. The discovery of cytotoxic T cells emerged in the through cytotoxicity assays demonstrating lymphocyte-mediated killing of target cells, initially observed in allograft rejection models. Pivotal advancements in the 1970s came from studies by Rolf Zinkernagel and Peter Doherty, who elucidated the of cytotoxic T cell responses using virus-infected mouse models, revealing that T cells recognize antigens only in the context of self-MHC molecules—a finding that earned them the 1996 in Physiology or Medicine.

Distinction from Other Immune Cells

Cytotoxic T cells, also known as CD8+ T cells, are distinguished from CD4+ helper T cells primarily by their antigen recognition and functional roles in the immune response. While cytotoxic T cells recognize antigens presented by major histocompatibility complex class I (MHC I) molecules on nearly all nucleated cells, enabling them to target intracellular pathogens like viruses or abnormal cells such as tumors, helper T cells interact with MHC class II molecules expressed mainly on professional antigen-presenting cells to orchestrate broader immune responses through cytokine secretion and activation of other immune effectors. This MHC restriction reflects their complementary functions: cytotoxic T cells focus on direct cell killing via perforin and granzymes, whereas helper T cells primarily coordinate humoral and cellular immunity without direct cytotoxicity as their primary mechanism. In contrast to natural killer (NK) cells, which belong to the and exhibit constitutive without prior sensitization, cytotoxic T cells are part of the adaptive and require antigen-specific priming for . cells recognize targets through germline-encoded receptors that detect stress ligands or antibody-coated cells via mechanisms like , lacking the (TCR) specificity for peptide-MHC complexes that defines cytotoxic T cell responses. Furthermore, while cells patrol tissues continuously and respond rapidly to infections or malignancies, cytotoxic T cells undergo clonal expansion upon encountering specific antigens, generating long-lived populations for enhanced secondary responses. Cytotoxic T cells differ from γδ T cells in their TCR composition and paradigms, with the former predominantly expressing αβ TCRs restricted by classical MHC I molecules for precise detection. In comparison, γδ T cells utilize γδ TCRs that often operate independently of classical , recognizing non- such as phosphoantigens or stress-induced molecules on infected or transformed cells, which positions them as a bridge between innate and adaptive immunity. This MHC-independent allows γδ T cells broader, less specific reactivity, whereas cytotoxic T cells' αβ TCR-MHC I interaction ensures high specificity but dependence on and presentation. A unique feature of cytotoxic T cells among lymphocytes is their into distinct subsets based on homing receptor expression, such as central (T_CM) cells marked by CCR7+ CD45RA- , which recirculate through lymphoid organs for rapid upon re-encountering , and effector (T_EM) cells characterized by CCR7- CD45RA- expression, enabling direct migration to inflamed peripheral tissues for immediate effector functions. These subsets, identified through surface marker analysis, underscore cytotoxic T cells' capacity for long-term surveillance and tissue-specific responses, differing from the more uniform effector profiles in or γδ T cells.

Development and Maturation

Thymic Development

Cytotoxic T cells, also known as CD8+ T cells, originate from hematopoietic stem cells in the that differentiate into early T cell progenitors and migrate to the for further development. These progenitors enter the at the double-negative (DN) stage, characterized by the absence of and coreceptors, and constitute a small fraction of thymocytes that undergo and commitment to the under the influence of signaling and cytokines such as IL-7. During thymic development, DN thymocytes progress through substages (DN1 to DN4) where V(D)J recombination occurs to generate a diverse T cell receptor (TCR) repertoire. TCR β-chain rearrangement happens primarily at the DN3 stage, forming a pre-TCR complex with pre-Tα and CD3 that drives survival and proliferation to the DN4 and subsequently double-positive (DP) stage, where CD4 and CD8 are co-expressed. At the DP stage, TCR α-chain rearrangement completes the αβ TCR assembly, resulting in a stochastic generation of approximately 10^15 possible TCR specificities capable of recognizing diverse peptide-MHC complexes. Positive selection occurs in the thymic cortex, where DP thymocytes interact with low-affinity self-peptides presented by molecules on cortical thymic epithelial cells (cTECs) via their TCR and coreceptor. Thymocytes receiving appropriate survival signals upregulate to evade and differentiate toward the + lineage, ensuring the selection of T cells capable of recognizing self-. The expression of tissue-specific self-antigens in the , facilitated by the AIRE in medullary thymic epithelial cells (mTECs), contributes to the overall context of selection for self-tolerance, though positive selection primarily relies on cTEC-presented peptides. Negative selection follows in the thymic medulla, eliminating and early single-positive thymocytes with high-affinity recognition of self-antigens presented by on mTECs, dendritic cells, or other antigen-presenting cells to prevent . This process induces through activation of the pro-apoptotic protein Bim, a BH3-only member of the , which is upregulated by strong TCR signaling and commits self-reactive cells to . Surviving CD8+ single-positive (SP) thymocytes mature into naive T cells, downregulating while maintaining CD8 and TCR expression, and are exported from the to the periphery as recent thymic emigrants to join the circulating T cell pool. This export marks the completion of intrathymic development, yielding a self-tolerant repertoire of cytotoxic T cells ready for encounter.

Peripheral Maturation and Homeostasis

Naive cytotoxic T cells, also known as CD8+ T cells, circulate continuously through the blood and secondary lymphoid organs, such as lymph nodes, to surveil for foreign antigens presented by major histocompatibility complex class I molecules. This recirculation is facilitated by the expression of L-selectin (CD62L) on their surface, which mediates tethering and rolling on high endothelial venules for entry into lymph nodes, in conjunction with the chemokine receptor CCR7. Following thymic export, these naive cells maintain a quiescent state while undergoing slow, tonic signaling through their T cell receptors interacting with self-peptide/MHC complexes to support survival. Homeostasis of the naive CD8+ T cell pool is preserved through cytokine-driven self-renewal, particularly in lymphopenic environments where cell numbers are low, prompting compensatory to restore . Interleukin-7 (IL-7) is essential for this , promoting survival and basal by upregulating anti-apoptotic proteins like , while interleukin-15 (IL-15) provides additional support, especially for enhancing division rates in depleted settings. In healthy adults, these mechanisms sustain a diverse peripheral pool of naive CD8+ T cells, preventing exhaustion and ensuring repertoire diversity. Following antigen-driven activation (see Activation Process section), some naive + T cells differentiate into effector cells and, upon resolution, into long-lived memory subsets such as central memory (T_CM) and effector memory (T_EM) cells, distinguished by their migratory patterns and functional properties. T_CM cells, characterized by expression of CCR7 and CD62L, home to lymphoid tissues for rapid secondary responses, while T_EM cells downregulate these receptors and patrol peripheral tissues. Senescence in CD8+ T cells is marked by progressive telomere shortening due to repeated divisions and diminished telomerase activity, ultimately restricting replicative potential and inducing cell cycle arrest via inhibitors like p16 and p21. Co-expression of killer cell lectin-like receptor G1 (KLRG1) further signals this senescent state, particularly in terminally differentiated effector subsets, by inhibiting proliferation through tyrosine-based motifs that dampen signaling pathways such as AKT. With advancing age, progressively diminishes naive + T cell output, shifting reliance to peripheral homeostatic mechanisms that favor memory cell accumulation. This leads to oligoclonal expansions of specific + clones, often driven by chronic viral antigens like , resulting in reduced repertoire diversity and impaired responses to new pathogens in the elderly.

Activation Process

Antigen Recognition

Cytotoxic T cells recognize antigens through their (TCR), which binds to peptide-major complex class I (pMHC I) complexes displayed on the surface of target cells. The peptides presented by molecules are typically derived from intracellular proteins degraded by the and are usually 8-10 in length, allowing them to fit snugly into the MHC binding groove. This interaction enables cytotoxic T cells to detect infected or abnormal cells expressing foreign or altered peptides. MHC class I molecules consist of a polymorphic heavy chain with three extracellular domains (α1, α2, and α3) non-covalently associated with the invariant β2-microglobulin (β2m) light chain. The α1 and α2 domains form a closed -binding groove that accommodates the antigenic , while β2m stabilizes the overall of the complex. In humans, alleles such as , , and exhibit polymorphism primarily in the α1 and α2 domains, influencing specificity and binding affinity. The of revealed this , establishing the foundation for understanding pMHC I . Although typically presents endogenous peptides from intracellular sources, certain antigen-presenting cells like dendritic cells can cross-present exogenous antigens on via specialized pathways, such as vacuolar or cytosolic routes that redirect internalized proteins for proteasomal processing. This process, first demonstrated in the context of minor histocompatibility antigen responses, allows cytotoxic T cells to initiate immunity against extracellular pathogens or tumor antigens not produced within the presenting cell itself. The of TCR for pMHC I is generally low, with dissociation constants (K_D) ranging from approximately 1 to 100 μM, which contributes to the specificity and of . To amplify signaling despite this low , the serial engagement model posits that a single pMHC I complex can sequentially bind and trigger multiple TCRs on the T cell surface, facilitating rapid signal propagation. This mechanism, observed through measurements of TCR internalization and calcium flux, reconciles the paradox of weak individual interactions driving robust T cell activation. The coreceptor plays a crucial role in enhancing recognition by binding to the invariant α3 of , thereby stabilizing the TCR-pMHC I interaction and increasing overall . Additionally, recruits the to the , initiating downstream events essential for T cell activation. This dual function of ensures efficient detection of low-abundance .

Co-stimulation and Signaling

Upon engagement of the (TCR) with peptide-MHC class I complexes, a second signal is required for full activation of naive cytotoxic T cells, primarily provided by co-stimulatory molecules. The key co-stimulatory receptor on T cells binds to (B7-1) or (B7-2) ligands expressed on antigen-presenting cells (APCs), delivering a signal that enhances T cell survival, proliferation, and cytokine production, particularly interleukin-2 (IL-2). This interaction is essential for inducing IL-2 gene expression and secretion, which acts in an autocrine manner to sustain T cell responses; without co-stimulation, TCR signaling alone leads to anergy or . Counterbalancing this activation, CTLA-4 serves as an inhibitory co-receptor that competes with for binding to and , but with higher affinity, thereby dampening T cell responses to prevent excessive activation. CTLA-4 engagement recruits phosphatases such as PP2A, which dephosphorylate key signaling molecules, inhibiting downstream pathways and limiting IL-2 production and . This inhibitory role is critical for maintaining immune and is upregulated following initial T cell activation. TCR and CD28 co-stimulation initiate intricate intracellular signaling cascades that integrate these signals for T cell commitment. TCR engagement phosphorylates CD3 ITAMs, recruiting and activating the kinase Zap-70, which in turn phosphorylates the adaptor protein LAT and SLP-76, forming a signalosome that activates phospholipase Cγ1 (PLCγ1). PLCγ1 hydrolyzes PIP2 to generate IP3 and DAG; IP3 induces calcium flux from intracellular stores, activating to dephosphorylate NFAT, which translocates to the to drive transcription of activation genes like IL-2. Concurrently, DAG activates PKCθ, leading to activation, while the Ras-MAPK/ERK pathway promotes AP-1 (c-Fos/c-Jun) formation, collectively enabling for and . The IL-2 produced binds to its high-affinity heterotrimeric receptor (IL-2Rαβγ) on activated T cells, triggering via JAK1/JAK3 kinases that phosphorylate STAT5. STAT5 dimers translocate to the nucleus, promoting expression of genes like for survival and cyclins for progression, driving clonal expansion. In response to , naive + T cells can undergo up to a 10,000-fold increase in numbers through this IL-2-dependent proliferation, peaking around 7-10 days post-activation before contraction. These signals also direct differentiation of activated + T cells into cytotoxic effectors, mediated by transcription factors such as Runx3, which enforces the cytotoxic lineage by repressing helper T cell genes (e.g., ) and promoting expression of effectors like perforin and granzymes. Runx3 cooperates with T-box factors like T-bet to establish accessibility at cytotoxic loci, ensuring commitment to the effector program. In chronic stimulation settings, such as persistent or tumors, inhibitory checkpoints like PD-1 emerge to modulate these pathways and prevent overactivation. PD-1 on T cells binds (or PD-L2) on target cells or APCs, recruiting SHP-1/2 phosphatases that dephosphorylate CD3ζ and inhibit PI3K/Akt and /ERK signaling, thereby dampening calcium flux, NFAT activation, and proliferation while promoting exhaustion-like states. This interaction fine-tunes responses in prolonged exposure, balancing efficacy and tolerance.

Effector Functions

Cytotoxic Mechanisms

Activated cytotoxic T cells, also known as cytotoxic T lymphocytes (CTLs), primarily induce target through two major pathways: granule and death receptor signaling. These mechanisms are initiated upon recognition of antigenic peptides presented by class I molecules on the target cell surface, following CTL . The processes occur at the of the , a specialized structure that ensures directed delivery of cytotoxic effectors. The perforin-granzyme pathway represents the dominant mechanism of rapid CTL-mediated . Upon synaptic contact, CTLs release preformed lytic granules containing perforin and granzymes from their cytotoxic granules. Perforin, a pore-forming protein, polymerizes in the target to create transmembrane pores approximately 10-20 nm in diameter, facilitating the entry of serine proteases such as into the . then cleaves and activates effector (e.g., caspase-3 and -7) and Bid, triggering both caspase-dependent and mitochondrial outer membrane permeabilization, leading to rapid target within minutes. This pathway is perforin-dependent and accounts for the majority of CTL killing , as demonstrated in perforin-deficient models where is severely impaired.80014-2) In parallel or alternatively, CTLs employ the Fas-Fas ligand (FasL) interaction for target cell , particularly when granule is limited. FasL, expressed on the CTL surface or released via extracellular vesicles, binds to (CD95) receptors on the target cell, recruiting the adaptor protein and activating through the death-inducing signaling complex (). This extrinsic pathway initiates caspase cascades independent of perforin and granzymes, though it is slower (typically 1-2 hours) and can synergize with the granzyme pathway for enhanced killing. The Fas-FasL mechanism is crucial for eliminating activated immune cells and certain virally infected targets resistant to granule-mediated death.80014-2) Effective requires stable cell-cell contact mediated by the . This structure forms through adhesion molecules such as LFA-1 (on CTL) binding (on target), stabilizing the interface and polarizing the CTL's microtubule-organizing center toward the synapse for directed granule release. The synapse segregates signaling (central supramolecular activation cluster) from secretory domains (peripheral), ensuring precise delivery of cytotoxic molecules while minimizing bystander damage.81716-6) To prevent self-inflicted damage during effector release, CTLs express protective mechanisms including inhibitors. Cytoplasmic PI-9 (serpin B9) specifically inhibits , blocking its autocatalytic activity and activation within the CTL itself. Additionally, low pH in endolysosomal compartments and binding to serglycin inactivate perforin, confining its activity to the target membrane. These safeguards enable sustained CTL function without . Quantitatively, a single activated CTL can sequentially eliminate up to 10-20 target cells over several hours, replenishing lytic granules between killings via cycles. This serial killing capacity amplifies immune responses, allowing efficient clearance of infected or malignant cells .

Cytokine Production and Modulation

Cytotoxic T cells, particularly those of the Tc1 subset, primarily secrete interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) as key that coordinate broader immune responses. IFN-γ activates macrophages to enhance and activity while upregulating major histocompatibility complex class I (MHC I) expression on target cells, thereby amplifying and immune surveillance. TNF-α contributes to by recruiting additional immune cells and directly induces in infected or malignant cells through receptor-mediated signaling. In contrast to Th2-associated cytokines, Tc1 cells produce only minimal amounts of interleukin-4 (IL-4) and IL-10, reinforcing their pro-inflammatory, type 1 immune bias that prioritizes clearance over suppression. The production of these cytokines is driven by transcriptional polarization following T cell activation, with the T-box transcription factor T-bet playing a central role in committing CD8+ T cells to the Tc1 phenotype. T-bet expression is induced by signals such as IL-12 and promotes the necessary for IFN-γ and TNF-α gene transcription, ensuring robust effector responses. This mechanism allows cytotoxic T cells to rapidly amplify their secretory output upon encounter, integrating direct with soluble factors that shape the immune microenvironment. Through secretion, cytotoxic T cells modulate the activity of other immune components, including enhancing natural killer () cell via IFN-γ signaling that boosts NK and target cell killing. Indirectly, IFN-γ from these T cells promotes class switching toward IgG2a and IgG3 isotypes by inducing T-bet expression in B cells, thereby supporting against intracellular pathogens. Additionally, cytotoxic T cells secrete chemokines such as CXCL9 and , which bind on effector T cells and NK cells to recruit further reinforcements to infection or tumor sites. Cytokine profiles exhibit variability across cytotoxic T cell subsets, with short-lived effector cells generally producing higher levels of IFN-γ compared to long-lived cells, reflecting their role in acute, high-intensity responses. cells, while capable of rapid IFN-γ recall upon re-exposure, maintain lower baseline production to support persistence and self-renewal. This subset-specific modulation ensures balanced immunity, preventing excessive while preserving effector potential.

Roles in Immunity

Antiviral and Antibacterial Defense

Cytotoxic T cells, also known as + T cells, play a pivotal role in antiviral defense by recognizing and eliminating virus-infected cells through antigen-specific mechanisms. In acute infections such as , , and (CMV), these cells rapidly expand and target viral antigens presented on class I (MHC-I) molecules, leading to the of infected host cells and thereby limiting viral replication. For instance, during acute infection, HLA-A2-restricted cytotoxic T lymphocytes (CTLs) recognize epitopes derived from the gag protein, such as the SLYNTVATL , enabling precise targeting of infected cells and contributing to the initial decline in . Similarly, in infection, + T cells directed against conserved epitopes facilitate clearance by inducing in infected respiratory epithelial cells. In CMV infections, HCMV-specific + CTLs are essential for controlling viral replication and preventing disseminated disease, particularly in immunocompromised individuals, through sustained surveillance and effector responses. Overall, the activation of these epitope-specific + T cells during acute viral infections can reduce viral loads by 2-3 logs, marking a critical phase of immune control before potential transition to chronicity or resolution. Beyond viruses, cytotoxic T cells contribute to defense against intracellular bacterial pathogens, such as and , by deploying granzyme-mediated killing to disrupt bacterial survival within host cells. In infections, + T cells infiltrate infected tissues and release granzymes that degrade bacterial factors secreted into the , thereby attenuating pathogen spread and promoting bacterial clearance from macrophages and hepatocytes. For , human + CTLs express granulysin and , which target intracellular mycobacteria by perforating phagosomal membranes and inducing infected , thus restricting bacterial persistence in granulomas. These mechanisms highlight the versatility of cytotoxic T cells in combating bacteria that evade by residing within host cells. A key aspect of cytotoxic T cell-mediated protection is the formation of long-lived memory CD8+ T cells, which provide rapid recall responses upon re-exposure to the same . These memory cells persist for decades after primary or , maintaining a poised state that allows for 10-100 times faster proliferation compared to naive T cells, enabling swift differentiation into effectors that control reinfection. This memory function is exemplified in resolved viral infections like , where memory CD8+ T cells confer heterosubtypic immunity by rapidly expanding to limit viral titers upon challenge with variant strains. The implications of cytotoxic T cell responses extend to vaccine design, particularly live-attenuated vaccines that mimic natural infection to induce robust T cell immunity. The 17D vaccine (YF-17D), one of the most efficacious vaccines available with over 99% rates, elicits a broad and polyfunctional memory T cell response targeting multiple viral epitopes, which correlates with long-term protection against severe disease. This vaccine-induced T cell compartment persists for years, providing a model for developing similar responses against other intracellular pathogens.

Antitumor Surveillance

Cytotoxic T cells play a pivotal role in antitumor surveillance by recognizing and eliminating malignant cells that arise from transformed host tissues. These cells, primarily + T lymphocytes, patrol peripheral tissues and detect aberrant proteins expressed by tumors through class I (MHC I) molecules on the surface of cancer cells. This process is essential for preventing the outgrowth of nascent tumors and maintaining immune control over established malignancies. Tumor antigens recognized by cytotoxic T cells include neoantigens, which are novel peptides generated from somatic mutations in cancer cells and presented via MHC I to activate antigen-specific T cell responses. For instance, in cancers driven by oncogenic viruses, such as human papillomavirus (HPV)-associated , viral oncoproteins like and E7 are processed and displayed on MHC I, enabling cytotoxic T cells to target infected and transformed cells. This recognition triggers T cell activation and subsequent cytotoxic responses, highlighting the specificity of antitumor immunity. Once activated, cytotoxic T cells infiltrate tumors as tumor-infiltrating lymphocytes (TILs), guided by chemokine receptors such as , which binds to ligands like CXCL9 and produced in the to facilitate homing and accumulation at the site of . Upon infiltration, TILs exert perforin-dependent , releasing perforin and granzymes to induce in target cancer cells, thereby promoting tumor regression. This mechanism is critical for the direct elimination of antigen-expressing tumor cells within solid tumors.30190-6) In the process, cytotoxic T cells contribute to an phase where chronic immune recognition keeps tumors in a dormant state, suppressing their progression through ongoing surveillance and selective pressure on antigen-expressing cells. This phase precedes potential tumor escape, where less immunogenic variants may emerge. Evidence from mouse models supports this role; for example, MHC I-deficient tumors exhibit accelerated growth and increased incidence in immunocompetent hosts, underscoring the necessity of MHC I presentation for T cell-mediated control.01199-0)00230-9) Human studies further validate these findings, as adoptive transfer of TILs in patients has led to objective tumor regressions, with durable responses correlating to the persistence of neoantigen-specific cytotoxic T cells. However, challenges persist due to tumor heterogeneity, which can generate loss variants that evade T cell recognition, allowing immune escape and tumor progression despite initial surveillance.00413-8)00225-8)

Involvement in Pathology

Autoimmune and Inflammatory Diseases

Dysregulated cytotoxic T cells, or + T cells, contribute to autoimmune and inflammatory diseases by escaping central tolerance mechanisms, such as negative selection in the , allowing autoreactive clones to persist and infiltrate target tissues. These autoreactive + T cells recognize self-antigens presented by molecules, leading to direct against healthy cells through pathways like perforin/granzyme release or Fas-Fas (FasL) interactions. In , for instance, + T cells specific for β-islet cell antigens escape negative selection and mediate β-cell destruction via FasL-induced , as demonstrated in mouse models and human studies. In rheumatoid arthritis, cytotoxic CD8+ T cells infiltrate the synovium, where they target citrullinated antigens in an HLA class I-dependent manner, promoting joint inflammation and tissue damage through granzyme B and granulysin expression. Similarly, in multiple sclerosis, CD8+ T cells predominate in central nervous system lesions, often outnumbering CD4+ T cells with ratios up to approximately 6-fold in some progressive cases, and drive demyelination by recognizing myelin-derived peptides and inducing axonal transection via MHC class I-restricted cytotoxicity. In celiac disease, gliadin-specific CD8+ T cells, restricted by HLA class I alleles like HLA-A2, activate in the intestinal mucosa, releasing granzyme B and IFN-γ to induce enterocyte apoptosis and exacerbate villous atrophy. Cytotoxic T cells amplify in diseases like through IFN-γ production, which disrupts epithelial barriers, promotes , and enhances permeability, often in synergy with + T cells. Genetic factors, including HLA associations, further predispose to + T cell-mediated pathology; for example, HLA-B27-restricted autoreactive + T cells recognizing self-epitopes correlate with , contributing to spinal . Overall, + T cells play a substantial role in organ-specific , often collaborating with + T cells to drive disease progression in conditions affecting 5-10% of the population worldwide.

Transplant Rejection and Immunodeficiencies

Cytotoxic T cells, also known as + T cells, play a central role in acute through direct allorecognition, where they recognize intact foreign class I (MHC I) molecules on donor graft cells, triggering rapid cytotoxic responses that damage the allograft. This process is mediated by the binding to allogeneic MHC I-peptide complexes on graft endothelial and parenchymal cells, leading to perforin and granzyme release that induces in target cells. In parallel, indirect allorecognition occurs when host antigen-presenting cells (APCs) process and present donor-derived peptides on self-MHC I molecules to + T cells, amplifying the and contributing to graft infiltration and dysfunction during the early post-transplant period. These mechanisms dominate cellular rejection episodes, often occurring within the first year after transplantation. In , cytotoxic T cells contribute to progressive vascular damage, particularly in heart and allografts, through sustained perforin-mediated endothelial , which promotes (TVD) characterized by intimal thickening and luminal narrowing. Perforin release from CD8+ T cells and natural killer cells targets vascular , leading to and that impair long-term graft function, with TVD being a primary cause of late allograft failure in solid organ transplants. This perforin-dependent pathway underscores the persistent alloimmune activity of cytotoxic T cells in maintaining low-grade rejection over years. In immunodeficiencies, cytotoxic T cell function and numbers are severely compromised; for instance, in , gp120-mediated mechanisms induce in activated + T cells via interactions with receptors like on macrophages, resulting in progressive reduction of naive + T cell counts during the asymptomatic phase. In (SCID), thymic developmental defects, such as those in genes regulating T cell maturation, lead to profound reductions in circulating + T cells due to impaired positive selection and differentiation in the . Advanced progression to AIDS further exacerbates this through functional exhaustion of remaining + T cells, marked by impaired production, reduced proliferative capacity, and upregulation of inhibitory receptors like PD-1, diminishing their antiviral and cytotoxic potential despite initial expansions. Diagnostic evaluation of cytotoxic T cell involvement in these contexts includes assays, which quantify alloreactive + T cell frequencies by measuring interferon-gamma secretion in response to donor antigens, serving as a predictive tool for acute rejection risk in transplant candidates. In primary immunodeficiencies, + lymphopenia is a key marker reflecting underlying thymic or developmental impairments that can be confirmed via . Cytotoxic T cells dominate cellular rejection mechanisms, with CD8+ responses driving the majority of T cell-mediated allograft damage, contributing to acute rejection rates of 10-20% within the first post-transplant year that, if untreated, progress to chronic failure. This CD8+ dominance highlights their critical pathological role, as elevated alloreactive frequencies correlate with poorer long-term outcomes in kidney and heart transplants.

Clinical and Therapeutic Applications

T Cell Exhaustion and Regulation

T cell exhaustion represents a progressive loss of effector functions in (+ T cells) during chronic infections or cancer, characterized by diminished production, reduced proliferation, and impaired in response to persistent stimulation. This state is marked by the upregulation of inhibitory receptors such as PD-1, TIM-3, and LAG-3 on the T cell surface, which transmit signals that dampen T cell activation and effector responses. At the molecular level, exhaustion is driven by epigenetic and transcriptional reprogramming, with the TOX playing a central role in establishing and maintaining this dysfunctional state. TOX induces accessibility changes that promote the expression of exhaustion-associated genes, including those encoding inhibitory receptors, while simultaneously reducing responsiveness to cytokines and growth factors like IL-2. This leads to a self-reinforcing loop where exhausted T cells exhibit altered metabolic profiles, favoring survival over robust effector activity. Regulation of cytotoxic T cell activity to prevent overactivation involves multiple mechanisms, including suppression by regulatory T cells (Tregs), which express CTLA-4 to compete with effector T cells for binding to and on antigen-presenting cells, thereby depriving effectors of costimulatory signals. Tregs also secrete immunosuppressive such as IL-10 and TGF-β, which further inhibit effector T cell proliferation and secretion through feedback loops involving and SMAD signaling pathways. Strategies to reverse T cell exhaustion have focused on epigenetic modifiers, with recent highlighting the role of TET2 in influencing exhaustion propensity by altering patterns that affect T cell differentiation trajectories. In preclinical models, TET2 deficiency has been shown to delay exhaustion onset and enhance antitumor efficacy in engineered T cells, suggesting potential therapeutic targeting to restore function in chronic settings. Clinically, T cell exhaustion is commonly observed in chronic hepatitis B virus (HBV) and (HCV) infections, where it contributes to viral persistence and is associated with treatment failure in the majority of cases by limiting effective immune clearance.

Immunotherapies and CAR-T Cells

Immunotherapies leveraging cytotoxic T cells have revolutionized by enhancing their antitumor activity. Checkpoint inhibitors, such as anti-PD-1 antibodies, block inhibitory signals that lead to T cell exhaustion, thereby reinvigorating cytotoxic T cells against tumors. , approved by the FDA in 2014 for advanced , demonstrated objective response rates of 21-34% in patients with refractory to targeted therapies. In non-small cell with PD-L1 TPS ≥1%, achieved an objective response rate of 27% as first-line therapy (KEYNOTE-042), with 45% in PD-L1 TPS ≥50% patients (KEYNOTE-024), and durable responses observed in PD-L1-positive patients. These therapies have shown response rates ranging from 20-40% across and indications, highlighting their role in prolonging by unleashing endogenous cytotoxic T cell responses. Chimeric antigen receptor (CAR) T cell therapy represents a of adoptive , where patient-derived T cells are genetically engineered to express CARs that redirect cytotoxic T cells to tumor s. The CAR structure typically includes a (scFv) for recognition, such as CD19 in B-cell malignancies, linked to intracellular signaling domains like CD3ζ for activation and co-stimulatory domains such as 4-1BB for enhanced persistence. , the first CAR-T therapy approved by the FDA in 2017 for relapsed or refractory B-cell (B-ALL) in patients up to 25 years old, targets and has induced remissions in over 80% of eligible pediatric and young adult patients, with complete remissions in 60% (ELIANA trial). This approach has transformed outcomes in hematologic cancers by enabling precise, potent cytotoxic killing of -expressing cells. Adoptive cell transfer using (TILs) involves extracting, expanding, and reinfusing tumor-specific cytotoxic T cells from patient tumors, often following lymphodepleting to enhance engraftment. Clinical trials post-2010 have reported objective response rates of up to 50% in patients with metastatic treated with TIL , with some achieving durable complete remissions. In February 2024, the FDA approved lifileucel (LN-144), the first TIL for solid tumors, for adult patients with unresectable or metastatic previously treated with a PD-1 and, if applicable, BRAF V600-targeted , based on a 31.4% ORR from the phase 2 C-144-01 trial. This method harnesses naturally occurring cytotoxic T cells reactive to tumor neoantigens, offering a personalized strategy particularly effective in immunogenic tumors like . Recent advances extend cytotoxic T cell therapies to broader applications, including T cell receptor (TCR)-engineered T cells and bispecific antibodies. TCR-engineered T cells targeting cancer-testis antigens like NY-ESO-1 have shown promise in phase I/II trials for solid tumors, such as and , with objective responses observed in over 50% of NY-ESO-1-positive patients across studies involving more than 100 participants. Bispecific T cell engagers, exemplified by , a CD19/CD3 bispecific approved for B-ALL, redirect cytotoxic T cells to lyse CD19-positive tumor cells by simultaneously binding tumor antigens and CD3 on T cells, achieving complete remission rates of 40-45% in relapsed patients. These innovations address limitations in solid tumors by improving T cell trafficking and activation. Despite their efficacy, cytotoxic T cell therapies face challenges including (CRS), a potentially life-threatening inflammatory response managed primarily with , an IL-6 that rapidly resolves severe CRS without impairing T cell function. In hematologic malignancies, CAR-T therapies have led to 5-year overall improvements of up to 50% in select B-ALL and cohorts, underscoring their transformative impact while necessitating vigilant toxicity monitoring.

References

  1. [1]
    Histology, Cytotoxic T Cells - StatPearls - NCBI Bookshelf - NIH
    Sep 20, 2023 · Cytotoxic cells are involved in directly killing intracellular and extracellular pathogens and eliminating mutated and cancerous cells.
  2. [2]
    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 ...
  3. [3]
    T cell-mediated cytotoxicity - Immunobiology - NCBI Bookshelf - NIH
    Cytotoxic T cells kill target cells bearing specific antigen while sparing neighboring uninfected cells. All the cells in a tissue are susceptible to lysis by ...8-21. Cytotoxic T cells can... · 8-22. Cytotoxic effector...
  4. [4]
    Structure, function, and immunomodulation of the CD8 co-receptor
    CD8+ T cells detect pathogens, cancer, and autoimmunity towards eliminating diseased cells through T cell antigen receptor (TCR) recognition of antigens ...<|separator|>
  5. [5]
    T cell receptor (TCR) signaling in health and disease - Nature
    Dec 13, 2021 · This review provides a comprehensive snapshot of the various molecules involved in regulating T cell receptor signaling, covering both enzymes and adaptors.
  6. [6]
    A novel intracellular pool of LFA-1 is critical for asymmetric CD8+ T ...
    LFA-1 mediates T cell migration and their stable interaction with antigen-presenting cells, but how intracellular LFA-1 dynamically and spatially regulates ...
  7. [7]
    Cytotoxic T Cell Overview | Thermo Fisher Scientific - US
    Cytotoxic T lymphocytes (CTLs) often called CD8 + T cells, are a critical component of the adaptive immune system and play an important role in immune defense.Cd8 T Cell--Mediated... · Isolation Of Cd8 T Cells · Table 4. Cellular Markers...Missing: definition | Show results with:definition
  8. [8]
  9. [9]
    An early history of T cell-mediated cytotoxicity - PubMed
    After 60 years of intense fundamental research into T cell-mediated cytotoxicity, we have gained a detailed knowledge of the cells involved.
  10. [10]
    The Nobel Prize in Physiology or Medicine 1996 - Press release
    4. Zinkernagel RM, Doherty PC. MHC restricted cytotoxic T cells: Studies on the biological role of polymorphic major transplantation antigens determining T cell ...
  11. [11]
    Rules of engagement: the discovery of MHC restriction - PMC
    ... cytotoxicity assays—they demonstrated that the T cells recovered from the cerebrospinal fluid of infected mice were antigen-specific cytotoxic T cells (CTLs) ...
  12. [12]
    Helper and Cytotoxic T Cells - British Society for Immunology
    There are two types of MHC: MHC class I and MHC class II. MHC class I presents to cytotoxic T cells; MHC class II presents to helper T cells.
  13. [13]
    CD4 vs CD8 T Cells - Danaher Life Sciences
    CD4 T cells recognize antigens presented on MHC class II molecules by antigen-presenting cells, whereas CD8 T cells detect antigens on MHC class I molecules ...Activation Of Cd4 And Cd8 T... · Role Of Cd4 In Immunology · Cd8 T Cell Differentiation...Missing: distinctions | Show results with:distinctions
  14. [14]
    A guide to cancer immunotherapy: from T cell basic science ... - Nature
    May 20, 2020 · In this Review, we emphasize the role of T cells in modern cancer immunotherapies and discuss three different categories of immunotherapeutic ...
  15. [15]
    Natural Killer Cells and Cytotoxic T Cells - PubMed Central - NIH
    While the NK cell is part of the innate immune system, the CD8+ cell is one of the major components of adaptive immunity. Still, these two very different types ...
  16. [16]
    NK cells at the interface between innate and adaptive immunity
    Jun 1, 2007 · In this review, we will highlight recent studies on the mechanisms by which NK cells are recruited to inflamed tissues and respond to pathogens.Nk Cell Migration Into... · Nk Cells And Dcs Meet In... · Nk Cell-Mediated...
  17. [17]
    Hidden talents of natural killers: NK cells in innate and adaptive ...
    Sep 4, 2009 · Herein, we discuss recent studies that indicate that NK cells span the conventional boundaries between innate and adaptive immunity.<|separator|>
  18. [18]
    Function of γδ T cells in tumor immunology and their application to ...
    Mar 12, 2021 · T cells of the γδ lineage are unconventional T cells with functions not restricted to MHC-mediated antigen presentation.
  19. [19]
    αβ and γδ T cell receptors: Similar but different - Morath - 2020
    Jan 29, 2020 · Review compares the αβ and γδ T cell Ag receptor (TCR) complexes, with a particular focus on their molecular signaling mechanisms.
  20. [20]
    The Role of Human γδ T Cells in Anti-Tumor Immunity and Their ...
    γδ T cells have no MHC restriction on tumor antigen recognition and have strong cytotoxic effects on tumor cells. Importantly, they can be easily expanded in ...
  21. [21]
    Two subsets of memory T lymphocytes with distinct homing ... - Nature
    Oct 14, 1999 · The CCR7+ and CCR7- T cells, which we have named central memory (TCM) and effector memory (TEM), differentiate in a step-wise fashion from naive ...
  22. [22]
    Two subsets of memory T lymphocytes with distinct homing ...
    The CCR7+ and CCR7- T cells, which we have named central memory (TCM) and effector memory (TEM), differentiate in a step-wise fashion from naive T cells, ...
  23. [23]
    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.
  24. [24]
    Positive and negative selection of the T cell repertoire
    Indeed, thymic B cells have repeatedly been found to be capable of inducing negative selection. Most convincingly, myelin oligodendrocyte glycoprotein (MOG)- ...
  25. [25]
    AIRE in the Thymus and Beyond - PMC - PubMed Central
    AIRE is an Autoimmune Regulator that promotes self-tolerance by inducing tissue-specific antigens, especially in the thymus, and drives ectopic expression of ...
  26. [26]
    T cell-specific inhibition of multiple apoptotic pathways blocks ...
    However, evidence supporting an essential role for negative selection is limited. Loss of Bim, a Bcl-2 BH3-only protein essential for thymocyte apoptosis, ...
  27. [27]
  28. [28]
    Naive, effector and memory CD8 T-cell trafficking - PubMed Central
    L-selectin (CD62L) is expressed by all naive CD8 T cells and certain subsets ... Acquisition of selectin binding and peripheral homing properties by CD4(+) and ...
  29. [29]
    Normal T cell homeostasis: the conversion of naïve cells into ...
    This article reviews recent work on the factors controlling the survival of naïve T cells and how some of these cells are induced to switch to MP cells.
  30. [30]
    Homeostasis of Naive and Memory T Lymphocytes - PMC
    In this review, we discuss the generation and homeostasis of naive T cells and these two types of memory T cells, focusing on their relative interaction with ...
  31. [31]
    Understanding subset diversity in T cell memory - PMC
    Even the circulating effector memory pool can be further subdivided: like Tcm, many cells in the TEM pool express CXCR3 and Eomes, but some express CX3CR1 (and ...Localization And Trafficking · Resident Memory T Cells... · Epigenetic Control Of T Cell...
  32. [32]
    Markers of T Cell Senescence in Humans - PMC - PubMed Central
    Aug 10, 2017 · In this review, we will focus on T cells and discuss the surface and molecular markers that are associated with T cell senescence.
  33. [33]
    Contributions of Age-Related Thymic Involution to ... - PubMed Central
    Jan 20, 2020 · We discuss the research progress uncovering how age-related thymic involution contributes to immunosenescence and inflammaging, as well as their intersection.
  34. [34]
    Rethinking peptide supply to MHC class I molecules - Nature
    CD8+ T cells prevent the spread of intracellular pathogens through the recognition of pathogen-derived peptides (epitopes), generally 8–10 amino acids in length ...
  35. [35]
    Structural Comparison Between MHC Classes I and II - Frontiers
    Jun 13, 2021 · MHC-I molecules, within the groove—formed by their membrane-distal domains—which is closed at both ends, typically bind peptides of 8-11 amino ...
  36. [36]
    Structure of the human class I histocompatibility antigen, HLA-A2
    Oct 8, 1987 · The class I histocompatibility antigen from human cell membranes has two structural motifs: the membrane-proximal end of the glycoprotein contains two domains ...
  37. [37]
    Current Concepts of Antigen Cross-Presentation - Frontiers
    Although many cells are able to present extracellular antigens on MHC I, DCs are considered to be the most prominent and most relevant cross-presenting cells.
  38. [38]
    Crystal structures of high affinity human T-cell receptors bound to ...
    Rather, T-cells are subject to positive and negative selection such that only those expressing TCRs with low affinities (Kd = 1–100 µM) for pMHC survive (van ...Results · Biacore Binding Analysis · X-Ray Structural Analysis
  39. [39]
    CD4 and CD8 binding to MHC molecules primarily acts to enhance ...
    Main Function of Both CD4 and CD8 Is to Enhance Lck Recruitment, Not Stabilize TCR–pMHC Interactions. Although CD4 does not stabilize TCR–pMHC interactions ...
  40. [40]
    CD28 co-stimulation in T-cell homeostasis: a recent perspective - PMC
    Here, we discuss recent insights into the molecular events underlying CD28-mediated co-stimulation, its impact on gene regulation, and the differential role of ...Missing: paper | Show results with:paper
  41. [41]
    The role of CTLA-4 in the regulation of T cell immune responses
    In the present review, evidence for the proposed inhibitory role of CTLA-4 is examined and a model suggesting a role for CTLA-4 in both early and late stages ...
  42. [42]
    Recent insights of T cell receptor-mediated signaling pathways for T ...
    May 21, 2020 · In this review, we summarize current knowledge on the pathways that govern how the TCR complex transmits signals into cells and the roles of effector molecules ...
  43. [43]
    Biology of IL‐2 and its therapeutic modulation: Mechanisms and ...
    Mar 9, 2018 · We review the underlying basic science related to IL-2 and its biologic actions, discuss the current state of IL-2-based immunotherapy, and focus on emerging ...4 Regulation Of Il-2 And... · 5 High-Dose Il-2 Therapy In... · 7 Il-2--Anti-Il-2 Antibody...<|separator|>
  44. [44]
    Interleukin-2 shapes the cytotoxic T cell proteome and immune ... - NIH
    A quintessential biological function of IL-2 is to control the clonal expansion of antigen receptor-activated T cells. The commitment of T cells to DNA ...
  45. [45]
    Runx3 and T-box proteins cooperate to establish the transcriptional ...
    Runx3 initially induces and then cooperates with T-box transcription factors to regulate gene transcription in differentiating CTLs.Missing: Ascl1 | Show results with:Ascl1
  46. [46]
    The transcription factor Runx3 guards cytotoxic CD8+ effector T cells ...
    Runx3-deficient CD8 + T EFF cells aberrantly upregulated genes characteristic of follicular helper T (T FH ) cell lineage, including Bcl6, Tcf7 and Cxcr5.Missing: Ascl1 | Show results with:Ascl1
  47. [47]
    Reinvigorating Exhausted T Cells by Blockade of the PD-1 Pathway
    Programmed cell death-1 (PD-1) is a major regulator of T-cell exhaustion, and blocking the PD-1 pathway restores T-cell function and improves pathogen control ...
  48. [48]
    The Expanding Arsenal of Cytotoxic T Cells - PMC - NIH
    Apr 20, 2022 · Cytotoxic T cells (CTLs) are the main cellular mediators of the adaptive immune defenses against intracellular pathogens and malignant cells.Missing: definition | Show results with:definition
  49. [49]
    Real-time visualization of perforin nanopore assembly - PubMed
    Perforin can self-assemble into oligomeric pores of 10-20 nm inner diameter in the membranes of virus-infected and cancerous cells.
  50. [50]
    Epigenetic Control of Interferon-Gamma Expression in CD8 T Cells
    This review will focus on the chromatin status of Ifng promoter in CD8 T cells and possible influences of epigenetic modifications in Ifng gene and conserved ...
  51. [51]
    CD8 T-cell subsets: heterogeneity, functions, and therapeutic potential
    Nov 1, 2023 · Thymic mature CD8 T cells, known as naïve CD8 T cells, are poised to be activated after encountering specific antigens. Recent studies have ...
  52. [52]
    T cell–derived tumor necrosis factor induces cytotoxicity ... - JCI Insight
    Nov 9, 2021 · We show that TNF released by T cells is necessary and sufficient to activate RIPK1-dependent cell death in target cells and thereby mediate target cell ...
  53. [53]
    Human Tc1 and Tc2/Tc0 CD8 T-cell clones display distinct cell ...
    We investigated the effects of IL-4 and IL-12 on the proliferation and cytokine production of these subsets. In addition, the 3 subsets were compared for the ...
  54. [54]
    Antigen-driven effector CD8 T cell function regulated by T-bet | PNAS
    Here, we report that the transcription factor T-bet, known to regulate Th cell differentiation, also controls the generation of the CD8 + cytotoxic effector ...
  55. [55]
    Early T-bet promotes LFA1 upregulation required for CD8 + effector ...
    Nov 29, 2022 · The T-box transcription factor T-bet is regarded as a “master regulator” of CD4+ Th1 differentiation and IFN-γ production.
  56. [56]
    Interferon-γ derived from cytotoxic lymphocytes directly enhances ...
    Jun 1, 2017 · Pre-incubation of CD8 T cells with IFNγ directly enhanced CD8 T-cell cytolysis independent of its effects on target cells. CD8 T cells not ...
  57. [57]
    CD8+ T Cells and NK Cells: Parallel and Complementary Soldiers of ...
    CD8 + T cells and NK cells are both cytotoxic effector cells of the immune system, but the recognition, specificity, sensitivity, and memory mechanisms are ...
  58. [58]
    IFN-γ produced by CD8 T cells induces T-bet–dependent and - PNAS
    Sep 20, 2010 · We now report that CD8 T cells also respond to alum proteins, proliferating extensively and producing IFN-γ, a key Th1 cytokine.
  59. [59]
    CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation - NIH
    The CXCL9, -10, -11/CXCR3 axis regulates immune cell migration, differentiation, and activation, leading to tumor suppression (paracrine axis).
  60. [60]
    Inflammation Directs Memory Precursor and Short-Lived Effector ...
    Aug 24, 2007 · Conceptually, effector CD8+ T cells can be divided into at least two subsets, memory precursor effector cells (MPECs) that can become long-lived ...
  61. [61]
    Differential interferon‐γ production by naive and memory‐like CD8 T ...
    May 18, 2020 · IFN-γ production by CD8 + T cells depends on NFAT1 transcription factor and regulates Th differentiation. J Immunol. 2005; 175: 5931-5939 ...
  62. [62]
    Memory CD8 T cells are vulnerable to chronic IFN-γ signals but not ...
    May 28, 2024 · We propose that chronic IFN-γ signals deplete memory CD8 T cells by compromising their long-term survival and by diverting self-renewing CD127 high cells ...
  63. [63]
    Neoantigens: promising targets for cancer therapy - Nature
    Jan 6, 2023 · Even in some tumors with low TMB and limited immune infiltration, neoantigens generated by gene fusion are still able to activate cytotoxic T ...
  64. [64]
    Infiltrating T-cell markers in cervical carcinogenesis - Nature
    Dec 1, 2020 · ... E6 and E7, both of which are markers of transforming HPV infection. ... cytotoxic and regulatory T lymphocytes in cervical cancer tissue.
  65. [65]
    T-cell responses to human papillomavirus type 16 among women ...
    Jun 28, 2005 · We have investigated T-cell responses to five HPV16 proteins (E6, E7, E4, L1 and L2) in women with CIN or cervical carcinoma directly ex vivo.Results · T-Cell Reactivity Was... · Cd8 T-Cell Responses To...
  66. [66]
    Neoantigen-specific CD8 T cells with high structural avidity ... - Nature
    Jun 6, 2023 · Identification of neoantigens relies on in silico prediction of antigen binding avidity to MHC ... Cytotoxic T cells · Immunological surveillance ...
  67. [67]
    Radiotherapy remodels the tumor microenvironment for enhancing ...
    Oct 13, 2023 · The predominant role of cytotoxic or effector T cells is to secrete perforins and granzymes that kill infected and mutated cells [17]. They are ...
  68. [68]
    Mutational and putative neoantigen load predict clinical benefit of ...
    Nov 23, 2017 · Cellular immunotherapy, namely adoptive T-cell therapy (ACT) using tumor infiltrating lymphocytes (TILs), has demonstrated very high objective ...
  69. [69]
    Cytotoxic T lymphocytes and autoimmunity - PubMed
    The purpose of this review is to discuss recent studies that shed light on the implication of cytotoxic T lymphocytes in several autoimmune disorders.
  70. [70]
  71. [71]
    The role of Fas-FasL in CD8+ T-cell-mediated insulin ... - PubMed
    The role of Fas-FasL in CD8+ T-cell-mediated insulin-dependent diabetes mellitus (IDDM) ... J Clin Immunol. 2001 Jan;21(1):15-8. doi: 10.1023/a:1006780629564.
  72. [72]
    Cytotoxic CD8+ T cells target citrullinated antigens in rheumatoid ...
    Jan 19, 2023 · Characterization of effector memory CD8+ T cells in the synovial fluid of rheumatoid arthritis. J. Clin. Immunol. 32, 709–720 (2012) ...
  73. [73]
    Involvement of CD8+ T Cells in Multiple Sclerosis - Frontiers
    The injection of in vitro activated CD8+ T cells bearing a TCR specific for HA led to CNS inflammation and demyelination. Of note, the activated CD8+ T cells ...
  74. [74]
  75. [75]
    New Insights on CD8+ T Cells in Inflammatory Bowel Disease and ...
    Oct 11, 2021 · Recent data suggest that cytotoxic CD8+ T cells (Tc1) and interleukin (IL) 17-producing CD8+ (Tc17) cells contribute to the pathogenesis of IBD.
  76. [76]
    CD8 + T-cell autoreactivity to an HLA-B27–restricted self-epitope ...
    HLA-B27 is highly associated with ankylosing spondylitis (AS), but the mechanism is unknown. Among the HLA-B27 alleles, B*2709, which differs by one amino ...
  77. [77]
    CD8 T-cell recognition of acquired alloantigen promotes ... - PNAS
    Cytotoxic CD8 T-cell responses against mismatched MHC class I alloantigen are the principal arm of the cellular response against a transplanted organ.
  78. [78]
    T cell Allorecognition Pathways in Solid Organ Transplantation - PMC
    T cells can recognize alloantigen by at least two distinct pathways: as intact MHC alloantigen on the surface of donor cells via the direct pathway; and as ...
  79. [79]
    Perforin Mediates Endothelial Cell Death and Resultant Transplant ...
    T cell-induced endothelial injury is an important event in the development of transplant vascular disease (TVD), the leading expression of chronic rejection ...
  80. [80]
    Granzymes and perforin in solid organ transplant rejection - Nature
    Oct 30, 2009 · In this review, I discuss clinical data implicating granzymes and perforin in acute and chronic solid organ transplant rejection.
  81. [81]
    Fas ligand-mediated depletion of CD4 and CD8 lymphocytes by ...
    We found that gp 120 can deplete both CD4 and CD8 T cells when they have recently been activated and are exposed to IL-2-deficient conditions. Bioassay of the ...
  82. [82]
    Inborn errors of immunity associated with defects of thymic ...
    Severe Combined Immune Deficiency (SCID) includes a variety of disorders in which T-cells are absent or present in very low numbers. Unless immune ...
  83. [83]
    Functional Restoration of Exhausted CD8 T Cells in Chronic HIV-1 ...
    CD8 T cell exhaustion is a hallmark of HIV-1 infection, characterized by phenotypic and functional CD8 T cell abnormalities that persist despite years of ...Abstract · Introduction · Results · Discussion
  84. [84]
    Alloreactive T cells to Assess Acute Rejection Risk in Kidney ... - NIH
    Apr 20, 2023 · Pretransplant donor-specific interferon-gamma ELISPOT assay predicts acute rejection episodes in renal transplant recipients. Transplant Proc.
  85. [85]
    Evaluation of Primary Immunodeficiency Disease in Children - AAFP
    Jun 1, 2013 · The overall incidence in the United States is 10.3 per 100,000 person-years; the median interval from onset of symptoms to diagnosis is 2.7 ...
  86. [86]
    Circulating cell-free nucleosomes as biomarker for kidney transplant ...
    Feb 11, 2021 · Background. Approximately 10–20% of kidney transplant recipients experiences acute rejection (AR) within the first year after transplantation [1] ...
  87. [87]
    The current state and future of T-cell exhaustion research - PMC
    Introduction. The exhaustion of cytotoxic T-cells was originally described in the context of chronic lymphocytic choriomeningitis virus (LCMV) infections of ...
  88. [88]
    T Cell Exhaustion - Annual Reviews
    Jun 28, 2024 · Here we review the process of T cell exhaustion, detailing the key characteristics and drivers as well as highlighting our current understanding ...
  89. [89]
    Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion ...
    Sep 6, 2010 · Exhausted T cells fail to proliferate and exert effector functions such as cytotoxicity and cytokine secretion in response to antigen ...Results · Tim-3 And Pd-1 Co Expression... · Discussion
  90. [90]
    TOX for tired T cells | Nature Reviews Immunology
    Jun 26, 2019 · Six studies describe a role for TOX in promoting an 'exhausted' CD8+ T cell phenotype in settings of chronic antigenic stimulation, ...Missing: basis | Show results with:basis
  91. [91]
    Treg-expressed CTLA-4 depletes CD80/CD86 by trogocytosis ... - NIH
    Jul 23, 2021 · Thus, Tregs are able to inhibit the T cell stimulatory activity of APCs by reducing their CD80/CD86 expression via CTLA-4–dependent trogocytosis ...
  92. [92]
    TGF-β Regulation of T Cells - Annual Reviews
    Apr 26, 2023 · Integrin αvβ8-mediated TGF-β activation by effector regulatory T cells is essential for suppression of T-cell-mediated inflammation.
  93. [93]
    Regulatory T cells in homeostasis and disease - Nature
    Oct 14, 2025 · Key molecules such as IL-10, TGF-β, and CTLA-4 mediate Treg-mediated suppression of pro-inflammatory cells, oxidative stress, and plaque ...
  94. [94]
  95. [95]
    TET2 regulates early and late transitions in exhausted CD8+ T cell ...
    Nov 13, 2024 · TET2 regulates early and late transitions in exhausted CD8 + T cell differentiation and limits CAR T cell function.Missing: 2020s | Show results with:2020s
  96. [96]
    T-cell exhaustion in chronic hepatitis B infection - NIH
    In humans, chronic HBV infection often shows weak or absent virus-specific T-cell reactivity, which is described as the 'exhaustion' state characterized by poor ...Missing: prevalence | Show results with:prevalence
  97. [97]
    T cell responses during HBV and HCV infections - PubMed Central
    Here, we review recent advances defining similarities and differences in T cell evasion by HBV and HCV, and the potential for reversal following antiviral ...
  98. [98]
    Functional Exhaustion of HBV-Specific CD8 T Cells Impedes PD-L1 ...
    Higher levels of functionally exhausted HBV-specific CD8 T cells are associated with a lack of response that cannot be restored by blocking the PD-1:PD-L1 axis.