Fact-checked by Grok 2 weeks ago

Natural killer cell

Natural killer (NK) cells are large granular lymphocytes that form a critical component of the , specializing in the rapid recognition and destruction of virus-infected cells, tumor cells, and other abnormal targets without requiring prior antigen-specific sensitization or (MHC) restriction. Originating from hematopoietic stem cells in the , NK cells undergo multilineage through a series of developmental stages, including common lymphoid progenitors and NK cell precursors, maturing primarily in the and secondary lymphoid tissues before entering circulation. Their effector functions are governed by a dynamic balance of activating and inhibitory receptors on the cell surface, such as natural cytotoxicity receptors (e.g., NKp46, NKp30) for and killer cell immunoglobulin-like receptors (KIRs) for inhibition, allowing NK cells to distinguish healthy "self" cells from stressed or altered targets via the "missing-" . In humans, NK cells are phenotypically divided into two main subsets: the cytokine-producing CD56bright population, which predominates in lymph nodes and supports adaptive immunity through interferon-gamma (IFN-γ) secretion, and the highly cytotoxic CD56dimCD16+ subset, which circulates in blood and mediates (ADCC). Beyond direct killing via perforin and granzymes, NK cells bridge innate and adaptive responses by interacting with dendritic cells, T cells, and macrophages, while dysregulation of NK cell activity is implicated in viral infections, autoimmune disorders, and cancer progression.

Discovery and history

Early observations

In the , pioneering experiments in transplantation using irradiated mice established the feasibility of rescuing lethally irradiated animals through bone marrow infusion, laying the groundwork for understanding hematopoietic reconstitution. However, subsequent studies revealed complexities, including unexpected resistance to allograft engraftment in certain settings, highlighting an innate mechanism of rejection independent of adaptive immunity. These findings distinguished the effectors from macrophages, which primarily phagocytose rather than lyse target cells, and from T cells, which require prior . The phenomenon of hybrid resistance was first systematically documented in the early 1960s by Georges Cudkowicz, who reported that semi-allogeneic mice rejected parental grafts despite lacking classical (MHC) barriers that typically permit graft acceptance. Later collaborations with Milton Bennett further explored this. In these studies, irradiated s resisted transplantation of as few as 10^4 parental marrow cells, a resistance that developed rapidly without prior immunization and was radioresistant, further differentiating it from T cell-mediated immunity. This H-2-linked but non-classical rejection mechanism provided early evidence of specialized innate effectors capable of surveilling hematopoietic cells. Concurrent 1960s experiments in mice uncovered spontaneous, non-antibody-dependent against tumor cells, particularly lymphomas, in unprimed and peripheral blood populations. Eva Klein and colleagues observed that normal mouse lysed certain syngeneic and allogeneic tumor targets without complement or , a activity resistant to treatments that depleted T cells or macrophages, such as anti-theta or silica particles. These observations underscored an innate population distinct from adaptive effectors, active against transformed cells lacking normal regulatory signals. In humans, similar non-antibody-dependent killing was noted in the late through assays showing peripheral blood lymphocytes spontaneously lysing cultured tumor cell lines, such as those derived from Burkitt's lymphoma, without prior exposure or humoral factors. These human studies paralleled murine findings and suggested a conserved innate cytotoxic pathway, later recognized as precursors to formal NK cell identification.

Identification and nomenclature

Natural killer (NK) cells were first identified in 1975 through independent studies in mice and s, marking a pivotal moment in understanding innate immune cytotoxicity. In mice, Rolf Kiessling, Eva Klein, and Hans Wigzell at the described a population of bone marrow-derived lymphoid cells capable of spontaneously lysing Moloney virus-induced tumor cells without prior immunization or antigen-specific priming. Concurrently, Ronald B. Herberman and colleagues at the reported similar spontaneous cytotoxic activity in human peripheral blood lymphocytes against a human cell line, distinguishing these effectors from conventional T and B lymphocytes. These early discoveries characterized the cells as "null lymphocytes" or "null cells," a term reflecting their lack of established surface markers for T cells (such as theta antigen in mice or E-rosette formation in humans) or B cells (such as surface immunoglobulin). The null cell designation arose from functional assays showing enrichment of in lymphocyte fractions depleted of T and B cells via rosetting techniques or antibody panning, confirming their distinct identity within the lymphoid lineage. The nomenclature "natural killer" was coined in these 1975 studies to emphasize the cells' innate, non-adaptive killing mechanism, which occurred spontaneously against tumor targets without the need for or deliberate . This term gained widespread acceptance by the late , as evidenced in key publications reviewing the phenomenon, and it highlighted the cells' role in immediate host defense. During the 1980s, phenotypic identification advanced with the recognition of specific surface markers: (FcγRIII, the low-affinity IgG ) was established as a defining feature of NK cells in 1983, enabling flow cytometric isolation of cytotoxic effectors. Subsequently, CD56 () was identified in 1986 as another key marker, allowing delineation of NK cell subsets such as CD16+ CD56dim and CD56bright populations based on expression levels. These markers, validated through studies, solidified the formal identification of NK cells distinct from other lymphocytes.

Origin and development

Hematopoietic lineage

Natural killer (NK) cells originate from hematopoietic stem cells (HSCs) within the bone marrow, where they derive specifically from common lymphoid progenitors (CLPs). These CLPs represent a committed stage in lymphoid differentiation, giving rise to all lymphoid lineages, including NK cells, but NK cell precursors diverge early by committing to the NK lineage without the need for antigen receptor gene rearrangements characteristic of T and B cell development. This distinction ensures that NK cells remain part of the innate immune system, bypassing the adaptive processes of V(D)J recombination required for T cell receptors and B cell immunoglobulins. Commitment to the NK cell lineage is tightly regulated by key transcription factors, including E4BP4 (also known as NFIL3), which is essential for initiating NK progenitor specification from CLPs by promoting the expression of downstream genes critical for development. TOX, a member of the high-mobility group box family, further supports this commitment by enabling the survival and differentiation of early NK progenitors, independent of pathways leading to T or B cells. Additionally, Id2, a helix-loop-helix of basic helix-loop-helix transcription factors, is indispensable for maintaining NK cell fate by suppressing alternative differentiation programs, such as those toward B cells.00533-6) During embryonic development, NK cell origins differ from those in adults, with the fetal liver serving as the primary site of hematopoiesis and NK progenitor emergence as early as gestational week 9 in humans. Fetal liver-derived progenitors contribute significantly to the initial pool of circulating NK cells, transitioning to dominance postnatally as the primary hematopoietic niche for sustained NK cell production in adults. This shift reflects the sequential colonization of hematopoietic sites during , ensuring robust innate immunity from fetal stages onward. NK cells develop through thymus-independent pathways, primarily within the microenvironment.

Maturation and education

Natural killer (NK) cells arise from common lymphoid progenitors derived from hematopoietic cells and undergo maturation primarily within specialized niches to acquire functional competence. In humans, NK cell maturation follows a sequential progression from an immature stage characterized by high expression of CD56 (CD56bright NK cells) to a mature stage with low CD56 expression (CD56dim NK cells). CD56bright NK cells represent an early developmental phase, featuring high levels of CD94/NKG2 receptors and potent production capacity but limited cytotoxic potential. As maturation advances, CD56dim NK cells emerge, marked by acquisition of (FcγRIII) for enhanced and increased expression of perforin and granzymes for direct killing. This transition also involves downregulation of CD117 (c-Kit) and upregulation of maturation markers like CD57. In mice, NK cell maturation parallels the human process but is delineated by CD27 and CD11b expression levels, with immature stages identified as CD27high CD11blow and mature stages as CD27low CD11bhigh, the latter expressing the NK1.1 marker in relevant strains. Immature murine NK cells exhibit interferon-γ production similar to CD56bright cells, while mature ones gain robust akin to CD56dim counterparts. Maturation occurs predominantly in the bone marrow, where NK cell precursors interact with stromal cells and cytokines like IL-15 to support differentiation, though secondary lymphoid tissues such as lymph nodes, , and tonsils also serve as critical niches for further development and terminal maturation in both humans and mice. These peripheral sites provide additional microenvironmental cues, including IL-15 trans-presentation, to refine NK cell subsets. A pivotal aspect of NK cell maturation is the education or licensing , which ensures self-tolerance by calibrating responsiveness through interactions between germline-encoded inhibitory receptors and self-major histocompatibility complex (MHC) class I molecules. In humans, killer-cell immunoglobulin-like receptors (KIRs) and CD94/NKG2A recognize specific HLA class I alleles, while in mice, Ly49 receptors interact with H-2 MHC class I; this engagement during development "licenses" NK cells to respond effectively to cells lacking self-MHC (missing-self ). Unlicensed NK cells, which fail to engage self-MHC due to mismatched inhibitory receptors, remain hyporesponsive or anergic, thereby preventing but limiting their effector functions. This rheostat-like calibration tunes NK cell potency proportionally to the strength of self-MHC interactions.

Characteristics and subtypes

Morphological features

Natural killer (NK) cells are a subset of large granular lymphocytes (LGLs), distinguished by their medium-to-large size and prominent cytoplasmic inclusions. In humans and mice, these cells typically range from 12 to 15 μm in diameter, larger than typical small lymphocytes (7–10 μm), with a round to reniform nucleus occupying about half the cell volume and eccentric placement. The abundant pale blue , visible under light microscopy, contains 5–20 azurophilic granules that stain positively with Wright-Giemsa, reflecting their lysosomal origin. These granules primarily store perforin, which forms pores in target cell membranes, and granzymes, serine proteases that induce . NK cells are phenotypically defined by the absence of T cell receptor (TCR) and B cell receptor (BCR) complexes, lacking CD3 expression that is characteristic of T lymphocytes. In humans, mature NK cells express CD56 (neural cell adhesion molecule) as a pan-marker, with subsets further delineated by CD16 (FcγRIII) expression: CD56bright CD16 cells predominate in lymphoid tissues and emphasize cytokine production, while CD56dim CD16+ cells are more abundant in blood and exhibit enhanced cytotoxicity. In mice, NK cells are identified by CD3 expression combined with NK1.1 (in certain strains like C57BL/6) or DX5 (CD49b, an integrin α2 subunit), markers that highlight their innate lymphoid identity without antigen-specific receptors.01326-4/fulltext) Ultrastructural analysis via electron microscopy reveals distinctive cytoplasmic features that underpin NK cell function. The is rich in free ribosomes and polyribosomes, facilitating rapid protein synthesis, alongside a well-developed Golgi apparatus and rough involved in the and trafficking of cytokines and lytic granule components. Mitochondria are sparse but functional, supporting energy demands during activation. These organelles collectively enable NK cells to form and release cytotoxic granules efficiently, contributing to their role in innate immune surveillance.

Tissue-resident populations

Tissue-resident natural killer (NK) cells represent specialized subsets that persist in non-lymphoid organs, distinct from circulating NK cells, and exhibit adaptations to local microenvironments for sustained immune surveillance. These populations maintain tissue-specific phenotypes and functions, often self-renewing locally without reliance on continuous input. In the liver, tissue-resident NK cells (LrNK) are notable for their expression of tumor necrosis factor-related apoptosis-inducing ligand (), which enables them to target and eliminate activated hepatic stellate cells, thereby regulating liver progression. This TRAIL-mediated is NKG2D-dependent and plays a protective role against fibrotic diseases. LrNK cells constitute a significant proportion of hepatic NK cells, displaying enhanced IFN-γ production and longevity compared to circulating counterparts. Salivary gland-resident NK cells form a unique subset characterized by hyporesponsiveness to viral infections, with distinct surface markers like low and high expression, allowing them to prioritize tolerance in this mucosal site while retaining cytotoxic potential. In the gut, intraepithelial NK cell variants reside within the epithelial layer, exhibiting heightened cytolytic activity in early life and contributing to barrier immunity; these cells often overlap with ILC1-like profiles but maintain classical NK features such as perforin and granzyme expression.02231-6/fulltext) Transcriptionally, tissue-resident NK cells differ from circulating NK cells, often showing lower Eomesodermin (Eomes) expression, which correlates with immature yet tissue-adapted states, particularly in murine models where liver trNK cells exhibit Eomes^low T-bet^high profiles. This transcriptional divergence supports their residency and specialized effector functions, such as localized over systemic responses. Epigenetically, these cells display distinct patterns and accessibility at loci regulating tissue-specific genes, including those for molecules like CD49a, ensuring stable residency and functional plasticity in diverse organs. Developmentally, many tissue-resident NK populations originate during fetal stages from progenitors in the or fetal liver, seeding organs early in . For instance, liver-resident NK cells emerge from fetal liver hematopoietic waves independent of adult contributions, enabling lifelong self-maintenance through local proliferation. While conventional NK cells mature primarily in the , tissue-resident subsets like those in the liver and gut can develop via hematopoietic stem cell-independent pathways in the , highlighting their ontogenic divergence.00388-5)

Receptors and recognition

Activating receptors

Natural killer (NK) cells express a variety of activating receptors that recognize stress-induced or altered self-ligands on target cells, such as virally infected or transformed cells, thereby triggering cytotoxic responses and production. These receptors initiate signaling cascades that promote NK cell activation, including and target cell lysis. Key families include the natural cytotoxicity receptors (NCRs) and other germline-encoded receptors like , DNAM-1, and CD16. The NCRs, comprising NKp30 (NCR3), NKp44 (NCR2), and NKp46 (NCR1), are immunoglobulin-like transmembrane glycoproteins primarily expressed on NK cells and are critical for natural cytotoxicity against tumor and virus-infected cells. NKp46, the first identified NCR, binds to ligands such as viral hemagglutinins on influenza-infected cells and proteoglycans on tumor cells, facilitating direct recognition and lysis. NKp30 engages tumor-associated ligands like B7-H6, a member of the B7 family expressed on various malignancies, and certain viral proteins, contributing to antitumor and antiviral responses. NKp44, predominantly found on activated or decidual NK cells, interacts with (PCNA) on tumor cells, which inhibits NK cell-mediated killing, and hemagglutinins from , which enhances NK cell-mediated killing. Additional activating receptors include , a C-type lectin-like receptor that pairs with the DAP10 in humans, binding to class I-related molecules such as , MICB, and the UL16-binding proteins (ULBPs), which are upregulated on stressed or malignant cells. DNAM-1 (CD226), an member, recognizes nectin-like molecules including CD112 (nectin-2) and ( receptor), which are overexpressed on tumor cells, thereby promoting NK cell adhesion and cytotoxicity. (), a low-affinity , mediates (ADCC) by binding the Fc portion of IgG antibodies coating target cells, enabling NK cells to lyse opsonized pathogens or tumors. Upon ligand engagement, these receptors transduce signals through immunoreceptor tyrosine-based activation motifs (ITAMs) or other motifs that recruit proteins. For instance, NCRs and certain others with DAP12, which contains ITAMs that, upon , recruit Syk family kinases, leading to activation of Cγ and downstream pathways. NKG2D and DNAM-1 primarily signal via DAP10, which lacks ITAMs but engages (PI3K), resulting in AKT activation and cytoskeletal reorganization for . Collectively, these pathways culminate in calcium mobilization, granule , and perforin/granzyme release, enabling target cell elimination.

Inhibitory receptors

Inhibitory receptors on natural killer (NK) cells deliver negative signals upon engagement with self-major histocompatibility complex ( molecules, thereby suppressing NK cell activation to preserve immune and self-tolerance. These receptors dominate NK cell regulation in both humans and mice, with their ligation overriding activating signals to prevent . In humans, killer-cell immunoglobulin-like receptors (KIRs) constitute the primary family of MHC-specific inhibitory receptors expressed on NK cells. Inhibitory KIRs such as KIR2DL1, KIR2DL2, and KIR2DL3 possess three immunoglobulin-like domains and bind to distinct epitopes on allotypes: KIR2DL1 recognizes HLA-C alleles with a at position 80 (C2 group), while KIR2DL2 and KIR2DL3 bind HLA-C alleles with at position 80 (C1 group). Additionally, leukocyte immunoglobulin-like receptor subfamily B member 1 (LILRB1), also known as CD85j or ILT-2, functions as an inhibitory receptor on a subset of NK cells by binding , a nonclassical HLA class I molecule expressed at immune-privileged sites like the . In mice, the orthologous inhibitory receptors include members of the Ly49 family, which are C-type lectin-like proteins encoded in the natural killer complex on chromosome 6. Inhibitory Ly49 receptors, such as Ly49A, Ly49C/I, and Ly49G2, specifically recognize alleles of classical molecules (H-2D, H-2K, or H-2L), with each subtype exhibiting distinct specificities that calibrate responses during . The NKG2A/CD94 heterodimer, a conserved inhibitory receptor across , binds to the nonclassical MHC molecule in humans (Qa-1^b in mice), which displays peptides derived from the leader sequences of other HLA class I proteins. The cytoplasmic tails of these inhibitory receptors contain one or more immunoreceptor tyrosine-based inhibitory motifs (ITIMs). Ligand binding induces ITIM tyrosine phosphorylation by Src family kinases, enabling recruitment of Src homology 2 (SH2) domain-containing protein tyrosine phosphatases SHP-1 (PTPN6) and SHP-2 (PTPN11). SHP-1 predominantly dephosphorylates key activatory signaling intermediates, such as Vav1 and phospholipase Cγ, while SHP-2 modulates ERK and PI3K pathways; together, they counteract proximal signals from activating receptors, inhibiting NK cell cytotoxicity and cytokine release. This inhibitory signaling underpins missing-self recognition, whereby NK cells are licensed to target MHC-deficient cells while sparing healthy self-expressing ones.

Mechanisms of action

Direct cytotoxicity

Natural killer (NK) cells mediate direct cytotoxicity against virus-infected and transformed cells primarily through two antibody-independent pathways: the release of cytotoxic granules containing perforin and granzymes, and the expression of death receptor ligands such as Fas ligand (FasL) and TNF-related apoptosis-inducing ligand (TRAIL). These mechanisms enable rapid elimination of aberrant cells without prior sensitization, distinguishing NK cells from adaptive cytotoxic lymphocytes. The granule-mediated pathway predominates in mature NK cells, accounting for the majority of lysis in many experimental models, while death ligand engagement provides an alternative or complementary route, particularly against certain resistant targets. In the perforin/granzyme pathway, upon recognition of a target, NK cells polarize their lytic granules toward the immunological synapse and release their contents via exocytosis. Perforin, a pore-forming protein discovered in the early 1980s, oligomerizes in the target cell membrane to create transmembrane pores approximately 10-20 nm in diameter, allowing entry of serine proteases like granzyme B. Once inside, granzyme B cleaves and activates Bid, leading to mitochondrial outer membrane permeabilization, and directly processes caspases such as caspase-3 and -7 to induce apoptosis. This process is calcium-dependent and highly efficient, with perforin-deficient NK cells exhibiting severely impaired cytotoxicity in vivo. Studies in perforin-knockout mice have demonstrated that this pathway is essential for NK cell control of certain viral infections and tumors, highlighting its non-redundant role in innate immunity. The death ligand pathway involves surface expression or soluble release of FasL and TRAIL by activated NK cells, which engage corresponding receptors on target cells to trigger extrinsic apoptosis. FasL binds Fas (CD95) to recruit the death-inducing signaling complex (DISC), activating caspase-8 and downstream effector caspases, while TRAIL interacts with death receptors DR4 and DR5 to similarly initiate caspase cascades. Seminal work identified functional FasL expression on freshly isolated human NK cells, enabling lysis of Fas-sensitive targets independently of granules. Likewise, NK cells were established as major producers of TRAIL, with its expression upregulated upon activation and contributing to cytotoxicity against TRAIL-sensitive cells like certain tumor lines. This pathway is particularly relevant for targets lacking susceptibility to granzymes, such as some immature dendritic cells or virally infected cells expressing death receptors. Target selection for direct cytotoxicity relies on the integration of activating and inhibitory signals, where stressed cells upregulate ligands for NK activating receptors while downregulating to evade inhibition. For instance, , a key activating receptor, binds stress-inducible ligands such as and MICB, which are expressed on infected or transformed cells due to DNA damage or oncogenic stress. This recognition, first demonstrated in 1999, triggers NK cell activation and or death ligand upregulation, ensuring selective killing of unhealthy cells. Cytokines like IL-15 can briefly prime resting NK cells, enhancing receptor expression and granule content for more potent responses.

Antibody-dependent cytotoxicity

Antibody-dependent cytotoxicity (ADCC) is a key mechanism by which natural killer (NK) cells eliminate antibody-opsonized target cells, such as virus-infected or tumor cells coated with (IgG). This process is primarily mediated by the low-affinity Fcγ receptor IIIa (FcγRIIIa, also known as ), which is expressed on the surface of most human NK cells and binds to the Fc domain of IgG antibodies bound to target cells. Upon ligation, CD16 clusters and initiates signaling cascades that activate NK cell . The signaling pathway downstream of CD16 involves immunoreceptor tyrosine-based activation motifs (ITAMs) present in associated adaptor proteins, specifically the CD3ζ chain and the FcεRIγ chain. These adaptors form homo- or heterodimers with , and of their ITAMs by Src family kinases recruits and activates spleen tyrosine kinase (Syk) and ζ-chain-associated protein kinase 70 (ZAP-70), leading to downstream events such as calcium mobilization, cytoskeletal reorganization, and . This targeted activation bridges adaptive and innate immunity, enhancing the efficiency of antibody-mediated responses against pathogens and malignancies. A functional polymorphism in the FCGR3A encoding influences ADCC potency; the V158 variant exhibits higher affinity for the IgG Fc domain compared to the F158 variant, resulting in more robust cell activation and greater therapeutic efficacy in antibody-based treatments like rituximab for . Individuals homozygous for V158 (V/V) show enhanced ADCC responses, which has implications for personalized strategies. Engineered cell lines expressing the high-affinity V158 , such as haNK cells, demonstrate superior cytotoxicity against antibody-coated targets in preclinical models. In addition to standalone ADCC, CD16 engagement synergizes with NK cell direct cytotoxicity mechanisms on antibody-opsonized targets, amplifying overall killing efficiency against virus-infected or tumor cells through enhanced granule exocytosis containing perforin and granzymes. This cooperative action underscores ADCC's role in bolstering antitumor and antiviral defenses.

Cytokine secretion

Natural killer (NK) cells play a key role in innate immunity through the secretion of immunomodulatory cytokines, which help shape the early immune response. The primary cytokine produced by NK cells is interferon-gamma (IFN-γ), which enhances macrophage activation by upregulating their antimicrobial activity and promotes Th1-biased responses by favoring the differentiation of T helper 1 cells. This IFN-γ production is potently induced by synergistic stimulation with interleukin-12 (IL-12) and interleukin-18 (IL-18), cytokines often released by antigen-presenting cells during infection. In addition to IFN-γ, activated NK cells secrete tumor necrosis factor-alpha (TNF-α) and (GM-CSF), both of which contribute to the initiation and amplification of early by recruiting and activating other immune cells. TNF-α promotes pro-inflammatory signaling, while GM-CSF supports the and of myeloid cells involved in the inflammatory milieu. Cytokine secretion is particularly pronounced in the CD56bright subset of human NK cells, which prioritizes immunomodulatory functions over direct , leading to higher output of , , and GM-CSF compared to the CD56dim subset. This subset bias enables CD56bright NK cells to rapidly modulate the immune environment upon activation.

Physiological roles

Antiviral defense

Natural killer (NK) cells serve as a critical component of the innate immune system's early response to viral infections, rapidly eliminating infected cells and limiting viral dissemination through direct cytotoxicity and cytokine production. Many viruses, such as herpesviruses, downregulate class I (MHC-I) molecules on infected cells to evade recognition by cytotoxic T cells; however, this "missing-self" phenotype renders the cells susceptible to NK cell-mediated killing via activating receptors like and DNAM-1, which detect stress-induced ligands upregulated on infected surfaces. Additionally, NK cells produce interferon-gamma (IFN-γ) and other cytokines that induce an antiviral state in neighboring cells, inhibit , and promote the and of adaptive immune components. This dual mechanism enables NK cells to bridge innate and adaptive immunity during the initial 24–48 hours post-infection, before antigen-specific responses mature. A prominent example of NK cell specificity in antiviral defense is observed in mice infected with murine cytomegalovirus (MCMV), where genetic resistance in strains like is conferred by the activating receptor Ly49H on NK cells, which binds the viral MHC-I homolog m157 expressed on infected s. This recognition triggers selective proliferation and IFN-γ production by Ly49H-positive NK cells, leading to efficient control of viral replication in the and liver during acute infection; mice lacking Ly49H exhibit markedly increased viral titers and mortality. In humans, analogous interactions occur with human (HCMV), where killer immunoglobulin-like receptors (KIRs) on NK cells interact with HLA class I alleles to enhance antiviral activity; for instance, activating KIR2DS2 in combination with HLA-C1 ligands is associated with better control of HCMV replication in transplant recipients, reducing through enhanced NK cell and secretion. NK cell licensing, or education, further refines their antiviral efficacy by calibrating responsiveness based on self-MHC-I recognition during . Licensed NK cells, which express inhibitory receptors (e.g., Ly49 or KIR) that bind self-MHC-I, exhibit heightened functionality against MHC-I-low targets like virus-infected cells, displaying superior and IFN-γ production compared to unlicensed counterparts.00289-4) In the context of infections, this education ensures that licensed NK subsets preferentially target infected cells with downregulated MHC-I while sparing healthy tissues; studies in MCMV-infected mice demonstrate that unlicensed NK cells drive more effective viral clearance than licensed Ly49+ ones, particularly when MHC-I modulation is prominent. This process underscores the adaptive-like of NK cells for precise antiviral responses.

Tumor surveillance

Natural killer (NK) cells play a critical role in tumor surveillance by recognizing and eliminating malignant cells through a balance of activating and inhibitory signals. This process allows NK cells to detect early signs of cellular , such as oncogenic , before tumors establish themselves. Unlike adaptive immune cells, NK cells can act rapidly without prior , providing an innate barrier against cancer and progression. A key mechanism in NK-mediated tumor surveillance involves the upregulation of stress-induced ligands on transformed cells, which engage activating receptors on NK cells. Specifically, major histocompatibility complex class I-related chain A (MICA) and unique long 16-binding proteins (ULBPs) are frequently overexpressed on the surface of tumor cells due to cellular stress from DNA damage or oncogenic signaling. These ligands bind to the NKG2D receptor on NK cells, triggering degranulation and cytokine release to induce target cell apoptosis. This NKG2D-MICA/ULBP axis is particularly effective against a broad range of solid and hematological malignancies, where ligand expression correlates with enhanced NK cytotoxicity. Another pivotal aspect of NK tumor surveillance is the "missing-self" recognition, where NK cells target cells that have downregulated class I (MHC-I) molecules. Many tumors reduce MHC-I expression to evade cytotoxic T lymphocytes, but this loss removes inhibitory signals from NK cell receptors like KIRs and NKG2A, thereby licensing NK activation and attack. This mechanism ensures that MHC-I-deficient variants, common in cancers such as and colorectal , are selectively eliminated by NK cells, preventing immune escape. Seminal studies in MHC-deficient mouse models confirmed that NK cells reject such tumor cells , underscoring the hypothesis's relevance to oncogenesis. In controlling , liver-resident NK cells are instrumental in intercepting circulating tumor cells (CTCs) that disseminate from primary tumors. These NK cells patrol the hepatic sinusoids and rapidly lyse CTCs expressing stress ligands or lacking MHC-I, thereby reducing the seeding of distant . Experimental models demonstrate that depletion of liver NK cells significantly increases and liver in mice challenged with or colon cells, highlighting their frontline role in limiting systemic tumor spread. This surveillance is enhanced by the unique microenvironment of the liver, where NK cells constitute up to 15% of lymphocytes and exhibit heightened cytotoxic potential against blood-borne malignancies.

Reproductive immunology

Uterine natural killer (uNK) cells represent a specialized subset of cells that predominate in the during early , comprising 50-90% of decidual lymphocytes. These cells are primarily of the CD56bright , which distinguishes them from peripheral blood CD56dim NK cells by their high expression of CD56 and low cytotoxic potential, enabling supportive roles in reproductive processes. Abundant in the decidua basalis, uNK cells accumulate during the implantation window and peak in number during the first , facilitating key events in placental formation. uNK cells contribute to embryo implantation and placental development by promoting and vascular remodeling at the maternal-fetal interface. They secrete angiogenic factors such as (VEGF) and (PlGF), which stimulate endothelial and spiral artery modification to ensure adequate nutrient and oxygen supply to the developing . These cytokines enhance decidual vascularization, supporting trophoblast invasion and preventing shallow . To maintain maternal-fetal tolerance, cells interact with non-classical HLA class I molecules, particularly expressed on extravillous s, through inhibitory receptors like LILRB1. This engagement suppresses uNK , preventing attack on fetal cells while allowing controlled migration. The LILRB1-HLA-G axis promotes an immunosuppressive environment, balancing immune surveillance with protection of the semi-allogeneic . Dysfunctions in cells are associated with reproductive disorders, including and . In , reduced numbers of decidual NK cells correlate with impaired spiral artery remodeling and placental ischemia. Similarly, alterations in uNK populations, such as decreased levels of specific subsets like uNK1, have been observed in , contributing to implantation failure and loss.

Pathological involvement

Immune deficiencies and disorders

Natural killer (NK) cell deficiencies represent a group of rare primary immunodeficiencies characterized by impaired NK cell numbers, maturation, or function, leading to increased susceptibility to viral infections and malignancies. Classical NK cell deficiency, also known as NK cell deficiency type 1, is defined by the selective absence or severe reduction of circulating NK cells, often resulting in recurrent or severe herpesviral infections such as or , as well as higher rates of papillomavirus-associated and certain cancers like . A key genetic cause is heterozygous germline mutations in the GATA2 gene, which encodes a essential for hematopoietic development; these mutations lead to a profound loss of the CD56bright NK cell subset while sparing the CD56dim subset to varying degrees, disrupting NK cell maturation and homeostasis. Patients with GATA2 mutations often present with a broader MonoMAC syndrome, including monocytopenia, B-cell deficiency, and myelodysplasia, but the NK cell defect contributes specifically to the infectious vulnerability. Classical NK cell deficiency type 2 (CNKD2) is caused by biallelic mutations in the MCM4 gene, which encodes a DNA replication component; this leads to partial depletion particularly of the CD56dim NK cell subset, severe functional impairment, and associated features such as growth retardation and , predisposing to life-threatening viral infections like disseminated varicella or severe EBV disease. Beyond numerical deficiencies, functional NK cell defects occur in conditions like type 1 (XLP1), a severe triggered primarily by Epstein-Barr virus infection. XLP1 is caused by mutations in the SH2D1A gene, which encodes the adaptor protein (signaling lymphocytic activation molecule-associated protein), essential for signaling through SLAM family receptors on NK cells. In affected individuals, NK cells exhibit impaired against EBV-infected B cells due to defective 2B4 receptor signaling, as SAP fails to associate properly with this activating receptor, leading to , , or dysgammaglobulinemia upon viral challenge. NK cell numbers may be normal, but their functional impairment underscores the role of SAP in NK-mediated immune surveillance. NK cell dysfunction also manifests in autoimmune disorders such as systemic lupus erythematosus (SLE), where reduced NK cell numbers and activity contribute to immune dysregulation. In SLE patients, peripheral blood NK cell counts are significantly decreased compared to healthy controls, accompanied by diminished cytotoxic function and impaired responsiveness, which may exacerbate production and tissue damage. This low NK cell activity correlates with disease activity, potentially due to increased or sequestration in inflamed tissues, thereby hindering the clearance of apoptotic cells and apoptotic debris that drive . In contrast to hyperactivation states, these deficiencies highlight NK cells' regulatory role in preventing excessive immune responses.

Hyperactivation and autoimmunity

Hyperactivation of natural killer (NK) cells can contribute to severe pathological conditions, including life-threatening hyperinflammatory syndromes and disorders, where uncontrolled and production lead to tissue damage and immune dysregulation. In (HLH), a rare but fatal syndrome, NK cells exhibit excessive activation alongside T cells and macrophages, driving a characterized by elevated levels of interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6). This hypercytokinemia is often fueled by dysregulated signaling through IL-2 and IL-15, that potently stimulate NK cell , survival, and effector functions, resulting in uncontrolled immune responses that overwhelm regulatory mechanisms and cause multiorgan failure. In adult-onset HLH, NK cells display an activated phenotype with preserved cytotoxic capacity, contrasting with the NK deficiencies typical in familial forms, and contribute to the syndrome's hallmark hemophagocytosis and hyperinflammation. In autoimmune diseases such as (RA), NK cell overactivation can promote pathology by targeting self-tissues, particularly in the synovium where inflammatory subsets accumulate. Activated NK cells in RA joints produce pro-inflammatory mediators like (GM-CSF), (M-CSF), and (RANKL), which exacerbate synovial inflammation, activation, and joint destruction. This overactivation arises from an imbalance in NK cell receptor signaling, where diminished inhibitory receptor function (e.g., via killer-cell immunoglobulin-like receptors) fails to restrain responses to self-antigens, leading to aberrant against autologous cells and amplification of adaptive autoimmune responses through IFN-γ secretion. Although peripheral NK cell numbers may be reduced in RA, the tissue-resident activated NK populations drive chronic , highlighting their dual role in both protection and pathogenesis. Rare clonal expansions of NK cells manifest as NK-type large granular lymphocyte (LGL) leukemia, a chronic lymphoproliferative disorder characterized by persistent of mature NK cells, often exceeding 2 × 10^9/L in peripheral blood. This clonal overgrowth leads to hyperactivation-like symptoms, including cytopenias, recurrent infections, and associated (e.g., or pure red cell aplasia), driven by somatic mutations in genes like that enhance NK survival and responsiveness. In NK-LGL leukemia, the expanded cells exhibit a mature immunophenotype (CD3^− ^+ CD56^+) with constitutive activation markers, contributing to immune dysregulation through excessive IFN-γ production and impaired immune surveillance. This condition underscores how dysregulated NK can mimic hyperactivation, bridging lymphoproliferation and autoimmune features.

Evasion strategies by targets

Pathogens and tumor cells employ various mechanisms to evade detection and elimination by natural killer (NK) cells, which rely on the balance between activating and inhibitory signals for target . One prominent strategy used by viruses involves mimicry, where human (HCMV) encodes the glycoprotein UL18, a structural homolog of molecules that binds to inhibitory receptors such as leukocyte immunoglobulin-like receptor 1 (LILRB1) on cells. This interaction delivers inhibitory signals that suppress cell activation and cytotoxicity against HCMV-infected cells, thereby promoting viral persistence. UL18's high-affinity binding to LILRB1 mimics the engagement of self-, exploiting the cell's "missing-self" paradigm to avoid . Tumor cells counteract NK-mediated surveillance through the proteolytic shedding of activating ligands, particularly releasing soluble forms of chain-related protein A (s) from the cell surface. This soluble binds to the receptor on NK cells, leading to its internalization and downregulation, which impairs NK cell recognition and cytotoxic responses against the tumor. Elevated levels of sMICA in the of cancer patients correlate with reduced expression and diminished antitumor immunity, facilitating tumor escape. In the tumor microenvironment, immunosuppressive factors like transforming growth factor-β (TGF-β) further hinder NK cell function by inhibiting their maturation and effector capabilities. TGF-β signaling suppresses the development of mature NK cells, maintaining them in an immature state with reduced and production, a process exacerbated by high TGF-β levels in solid tumors. This also directly impairs perforin and granzyme expression in mature NK cells, contributing to an overall immunosuppressive milieu that shields tumors from NK attack.

Therapeutic applications

Adoptive NK cell therapies

Adoptive natural killer () cell therapies involve the expansion and infusion of NK cells to harness their innate cytotoxic potential against malignancies, particularly in hematologic cancers like (AML). These therapies typically utilize unmodified NK cells derived from the patient (autologous) or donors (allogeneic), with allogeneic approaches preferred due to enhanced alloreactivity against tumors lacking inhibitory ligands. In autologous infusions, NK cells are isolated from the patient's peripheral , activated and expanded using cytokines such as interleukin-2 (IL-2), and reinfused following lymphodepleting to reduce endogenous immune suppression and improve engraftment. However, autologous NK therapies have shown limited antitumor efficacy in clinical settings, primarily due to the patient's immunosuppressive , with phase I trials in solid tumors reporting safety but modest response rates below 20%. Allogeneic adoptive NK cell therapy, particularly from haploidentical donors, has emerged as a more promising strategy for AML, leveraging mismatched killer immunoglobulin-like receptor (KIR) interactions to promote NK cell licensing and tumor targeting. Seminal work demonstrated that IL-2-activated haploidentical NK cells, infused after lymphodepleting regimens like and , could expand for up to 12 days and mediate complete remissions in 5 of 19 poor-prognosis AML patients (26%), with no observed. Subsequent trials refined this approach by incorporating recombinant human IL-15 (rhIL-15) to support NK cell survival without the toxicity of high-dose IL-2, achieving remissions in 35% of AML patients. Haploidentical donors are selected for KIR incompatibility to maximize alloreactive potential, and cells are typically expanded overnight or for short periods to preserve functionality. Clinical trials have highlighted the utility of adoptive NK cell therapy in the post-hematopoietic transplant (HSCT) setting for relapse prevention or . In a phase II trial, haploidentical NK cells infused after haploidentical HSCT in pediatric AML patients resulted in event-free survival improvements, with response rates around 50% in intermediate-risk cases. Similarly, donor-derived NK cells administered post-HSCT in AML achieved complete remission in approximately 57% of patients at one month, underscoring their role in bridging to long-term remission. Overall, across multiple trials in AML, response rates range from 20% to 50%, with higher efficacy in settings, though durable responses often require subsequent allogeneic HSCT. These outcomes establish adoptive NK as a safe bridge , with infusion-related toxicities minimal and primarily limited to transient release. Despite these advances, challenges persist in adoptive NK cell therapies, notably the short in vivo persistence of infused cells, typically lasting only 1-3 weeks without sustained support, which limits long-term tumor control and contributes to rates exceeding 50% in cases. Lymphodepleting is essential to enhance NK cell homing to and lymphoid tissues by depleting regulatory T cells and competing lymphocytes, thereby creating an immunological niche; without it, engraftment is negligible. Heterogeneity in donor NK cell quality and patient responses further complicates outcomes, prompting ongoing efforts to optimize protocols and briefly explore engineered enhancements for improved . Future refinements aim to address these hurdles through better supportive care to extend NK cell functionality.

Engineered NK cells

Engineered natural killer () cells involve genetic modifications to enhance their specificity, persistence, and antitumor activity, primarily through the introduction of chimeric antigen receptors (), use of immortalized cell lines, or bispecific antibody constructs that redirect NK cell killing. These approaches aim to overcome limitations of primary NK cells, such as short lifespan and variable activation, while maintaining their inherent safety profile, including reduced risk of compared to T cell therapies. Early clinical data indicate that engineered NK cells can achieve complete remissions in cancers without severe adverse events. CAR-NK cells express synthetic receptors that recognize tumor-associated antigens, enabling targeted cytotoxicity independent of restriction. A prominent example targets , a marker on B-cell malignancies like and , where CAR-NK therapy has shown objective response rates of up to 73% in phase I/II trials involving 11 patients with relapsed or refractory disease, with no instances of GVHD or observed. This GVHD resistance stems from the alloreactive nature of NK cells and lack of T cell contamination in NK preparations, contrasting with CAR-T cells that often require lymphodepleting . Preclinical studies further demonstrate that CD19-CAR-NK cells, often derived from or induced pluripotent stem cells, exhibit potent of CD19-positive tumor cells while sparing healthy tissues. The NK-92 cell line, derived from a with , serves as a renewable source for engineered NK therapies due to its perpetual proliferation and high baseline . To mitigate risks of uncontrolled growth, NK-92 cells are irradiated before , rendering them replication-incompetent while preserving effector functions for off-the-shelf without patient-specific . In clinical settings, irradiated NK-92 cells expressing against targets like HER2 or have been tested in solid tumors, including and , demonstrating safety with no significant side effects and partial responses in some patients across multiple phase I trials. For instance, NK-92-CD33-CAR therapy in achieved stable disease in 6 out of 9 (67%) treated individuals. Bispecific engagers enhance NK cell targeting by simultaneously binding activating receptors on NK cells and tumor antigens, forming an that triggers and release. Bispecific killer engagers (BiKEs), such as those linking (the FcγRIII receptor) to tumor markers like or HER2, have shown preclinical efficacy in redirecting NK cells against and cells, with enhanced compared to monoclonal antibodies alone. -Fc fusion proteins, which dimerize NKG2D ligands overexpressed on stressed tumor cells to activate NKG2D receptors, promote NK-mediated tumor clearance and depletion of immunosuppressive cells like myeloid-derived suppressor cells in the . These constructs, often Fc-optimized for prolonged half-life, have demonstrated antitumor activity in models without off-target effects on healthy cells. Ongoing trials explore BiKEs and TriKEs (trispecific variants incorporating IL-15 for NK expansion) in hematologic and solid malignancies, highlighting their potential for rapid deployment. These engineered strategies are increasingly combined with checkpoint inhibitors or cytokines to further boost efficacy, though standalone applications remain the focus of current advancements. Recent 2025 data from early-phase trials, such as the off-the-shelf CAR-NK SENTI-202, have shown complete remissions in relapsed or cancers, including AML, in a subset of patients (e.g., 4 out of 7), underscoring ongoing progress as of April 2025.

Combination with immunomodulators

Natural killer (NK) cells can be enhanced through combination therapies involving immunomodulators such as cytokines, checkpoint inhibitors, and Toll-like receptor (TLR) agonists, which amplify their cytotoxic activity and persistence in clinical settings. Cytokine preconditioning with interleukin-15 (IL-15) and interleukin-21 (IL-21) promotes NK cell expansion and activation by stimulating proliferation and enhancing effector functions like cytotoxicity and cytokine production. IL-15 supports NK cell survival and memory-like differentiation, while IL-21 synergistically boosts expansion yields up to eightfold when combined with IL-15, leading to improved antitumor responses. In clinical protocols for neuroblastoma, ex vivo preconditioning of NK cells with IL-15 and IL-21 has been employed to generate highly cytotoxic populations for adoptive transfer, demonstrating safety and preliminary efficacy in relapsed/refractory patients. Checkpoint inhibitors targeting inhibitory receptors on NK cells, such as killer-cell immunoglobulin-like receptors (KIRs), relieve suppression and enhance NK-mediated killing. The anti-KIR monoclonal antibody lirilumab blocks multiple KIRs (e.g., KIR2DL1, KIR2DL2/3, KIR3DL2), promoting NK cell activation against HLA-expressing targets without causing significant toxicity. When combined with PD-1 inhibitors like nivolumab, lirilumab augments NK cell function in solid tumors, such as non-small cell lung cancer, by concurrently disrupting PD-1/PD-L1 and KIR/HLA inhibitory axes, resulting in improved tumor control in preclinical models and early-phase trials. TLR agonists like CpG oligodeoxynucleotides (CpG-ODN) and polyinosinic:polycytidylic acid (poly I:C) indirectly activate NK cells by inducing cytokine storms, including type I s and IL-12, which drive NK maturation and . CpG, a TLR9 agonist, triggers NK cell and IFN-γ production via plasmacytoid dendritic cells, enhancing antitumor immunity in models of infection and cancer. Poly I:C, a TLR3 agonist, similarly promotes NK cell activation through signaling, with combined CpG/poly I:C formulations showing synergistic effects on NK against tumors and virus-infected cells, though careful dosing is required to mitigate excessive release.

Emerging research

Adaptive and memory-like properties

Natural killer (NK) cells exhibit memory-like properties characterized by epigenetic modifications following human cytomegalovirus (HCMV) infection, enabling enhanced functional responses upon re-encounter with the . These changes involve and modifications that silence genes encoding signaling adapters such as FcRγ, SYK, and EAT-2, leading to a distinct adaptive cell subset with altered signaling pathways and improved (ADCC). In HCMV-seropositive individuals, this epigenetic reprogramming results in the selective expansion of NK cells with reduced expression of these adapters, promoting a memory-like state that persists long-term. Memory-like NK cells demonstrate enhanced responses, producing higher levels of interferon-γ (IFN-γ) and other cytokines upon secondary stimulation compared to naive NK cells. This capacity is particularly evident in HCMV-specific contexts, where pre-exposed NK cells exhibit amplified effector functions, contributing to improved control of viral reinfection. Epigenetic inheritance of these modifications ensures clonal expansion and maintenance of the , as shown in mouse models of infection where encounter drives pronounced remodeling. Adaptive NK cells, a specialized subset, are defined by the absence of FcRγ expression and are predominantly found in HCMV-seropositive individuals. These FcRγ-negative NK cells display expanded populations expressing multiple killer-cell immunoglobulin-like receptors (KIRs), particularly those with inhibitory specificities for self-HLA class I alleles, which correlate with heightened ADCC against HCMV-infected targets. The expansion of KIR+ FcRγ-negative NK cells is driven by HCMV infection and results in a reprogrammed signaling axis that favors antibody-mediated responses over natural . NK cell licensing, or education through self-MHC class I interactions, plays a critical role in enabling memory formation, with pre-educated (licensed) cells showing antigen-specific boosting upon exposure. Licensed cells, which have received inhibitory signals via KIR or NKG2A during , exhibit enhanced responsiveness to cytokines like IL-12, IL-15, and IL-18, leading to amplified IFN-γ production in recall scenarios. This pre-education ensures that only functional cells contribute to memory-like responses, linking classical licensing to adaptive features without altering core inhibitory receptor expression.

Tissue-specific functions

Natural killer (NK) cells exhibit specialized functions tailored to the unique immunological demands of different tissues, reflecting adaptations in their activation, production, and interactions with local cell types. In the liver, gut, and , distinct NK cell subsets contribute to , defense, and tissue repair, often through organ-specific mechanisms that modulate and . In the liver, NK cells play a critical anti-fibrotic role by targeting activated hepatic stellate cells (HSCs), the primary producers of during , through direct and secretion of interferon-gamma (IFN-γ). This IFN-γ production inhibits HSC activation and proliferation while promoting their , thereby limiting fibrotic progression in models of chronic liver injury induced by toxins or . Liver-resident NK cells, which constitute a significant proportion of hepatic lymphocytes, further enhance this function by expressing high levels of activating receptors like , enabling rapid responses to fibrogenic stimuli. Additionally, these cells provide immune surveillance against hepatic tumors, such as , by recognizing stress-induced ligands on malignant hepatocytes and eliminating them via perforin- and granzyme-mediated , which helps control early tumor dissemination in the hepatic microenvironment. In the gut, ILC1-like NK cells, characterized by their expression of T-bet and production of IFN-γ, contribute to regulation by influencing epithelial barrier integrity and modulating bacterial translocation. These cells respond to microbial signals at the mucosal interface, promoting the production of and cytokines that maintain symbiotic balance while preventing dysbiosis-associated . At the intestinal mucosa, NK cells exert potent antiviral effects by recognizing infected epithelial cells through natural cytotoxicity receptors and (ADCC), leading to the clearance of pathogens like noroviruses and , which is particularly vital in the where viral entry occurs frequently. In the brain, NK cells interact closely with microglia during neuroinflammation, where they infiltrate the central nervous system (CNS) and modulate microglial activation via IFN-γ secretion, which can amplify pro-inflammatory responses in conditions like multiple sclerosis or infection but also facilitate debris clearance. These interactions occur primarily in the perivascular space and parenchyma, with NK cells upregulating to home to inflamed sites and influencing microglial toward an M1-like state that enhances control. Post-stroke, NK cells contribute to repair processes by promoting and in the peri-infarct region; brain endothelial cells secrete to recruit protective NK subsets that release growth factors and limit excessive infiltration, thereby supporting vascular remodeling and neuronal recovery in ischemic lesions. Liver, gut, and brain NK cells largely originate from precursors that seed tissues during development, with local sustaining resident pools.

Clinical trial advancements

Recent advancements in chimeric antigen receptor (CAR)-NK cell therapies have progressed to phase II trials targeting lymphomas since 2023, demonstrating improved safety profiles and response rates compared to earlier phases. For instance, off-the-shelf CAR-NK therapies incorporating logic-gated mechanisms have elicited complete remissions in patients with relapsed or refractory blood cancers, including lymphomas, as reported at the 2025 AACR meeting. To address antigen escape and relapse, multi-antigen targeting strategies in CAR-NK designs, such as dual CD19/CD20 approaches, have shown enhanced antitumor activity and persistence in preclinical and early clinical models for B-cell lymphomas. Allogeneic NK cell therapies derived from have expanded into trials for tumors, with phase I studies in 2023-2024 reporting favorable safety and preliminary efficacy in advanced pediatric tumors when expanded . These off-the-shelf products avoid HLA matching requirements, enabling broader application, and ongoing trials as of 2024 include combinations with checkpoint inhibitors for and other malignancies. In 2024-2025, the FDA granted fast-track designations to several allogeneic NK combinations, Biomarkers like KIR-HLA mismatch have emerged as predictors of response in NK cell therapies, with mismatched donor-recipient pairs correlating to heightened antitumor activity in recent analyses of adoptive transfer trials. Specifically, inhibitory KIR ligand mismatches have been linked to reduced relapse risk post-therapy in 2024 retrospective studies. Complementing this, AI-driven modeling for patient selection, including machine learning-derived NK cell signatures, has improved prognostic accuracy in trial enrollment for NK therapies from 2023 onward, enabling personalized matching based on features.

References

  1. [1]
    Human natural killer cells - PMC - NIH
    Natural killer (NK) cells were discovered more than 30 years ago. NK cells are large granular lymphocytes that belong to the innate immune system because ...
  2. [2]
    Natural killer cells and cancer - PubMed - NIH
    Natural killer (NK) cells are lymphocytes that were first identified for their ability to kill tumor cells without deliberate immunization or activation.
  3. [3]
    Human natural killer cells: form, function, and development - NIH
    NK cell effector function is dictated by a balance of activating and inhibitory receptors, including the killer immunoglobulin-like receptors (KIRs), the low ...
  4. [4]
    Natural Killer Cells: Development, Maturation, and Clinical Utilization
    A common schema of human natural killer (NK) cell development in the bone marrow and lymph nodes. A total of six distinct developmental stages have been ...
  5. [5]
    Functions of natural killer cells - PubMed
    Natural killer (NK) cells are effector lymphocytes of the innate immune system that control several types of tumors and microbial infections.Missing: definition | Show results with:definition
  6. [6]
    Histology, Natural Killer Cells - StatPearls - NCBI Bookshelf
    Feb 6, 2023 · Morphologically, NK cells are large, granular lymphocytes that are defined ... CD56bright natural killer (NK) cells: an important NK cell subset.
  7. [7]
    Human NK cells: From development to effector functions - PMC - NIH
    Human decidual natural killer cells are a unique NK cell ... Melanoma cells inhibit natural killer cell function by modulating the expression of activating ...
  8. [8]
    Natural killer cells as an initial defense against pathogens - PMC
    Natural killer (NK) cells serve as a crucial first line of defense against tumors and a diverse range of pathogens.
  9. [9]
    Mouse Models of Bone Marrow Transplantation - PMC - NIH
    Bone marrow transplantation (BMT) was conceived in the early 1950s out of rodent studies focusing on the effects of radiation. These now classical ...
  10. [10]
    History of hematopoietic cell transplantation: challenges and progress
    Oct 9, 2020 · Other studies addressed transfusion-induced sensitization to minor histocompatibility antigens, which often resulted in marrow graft rejection ...The 21st Century · Current Trends · Future Directions<|control11|><|separator|>
  11. [11]
    HYBRID RESISTANCE TO PARENTAL MARROW GRAFTS - PubMed
    All the tested H-2 heterozygotes resisted the transplantation of 10(4) parental marrow cells, whereas H-2 homozygotes were nonresistant, in spite of ...Missing: Bennett 1960s NK
  12. [12]
    Hybrid Resistance to Parental Marrow Grafts: Association with the K ...
    All the tested H-2 heterozygotes resisted the transplantation of 106 parental marrow cells, whereas H-2 homozygotes were nonresistant, in spite of ...
  13. [13]
    Eva Klein (1925–2025) | Nature Immunology
    Mar 7, 2025 · In the early 1960s, Eva suggested researching a human tumor. The choice fell on Burkitt's lymphoma, later shown to carry Epstein–Barr virus ...
  14. [14]
    An Historical Overview: The Discovery of How NK Cells Can Kill ...
    Jun 19, 2019 · NK cells were originally defined as effector lymphocytes of innate immunity characterized by the unique ability of killing tumor and virally infected cells.
  15. [15]
    Natural Killer Activity: Early Days, Advances, and Seminal ... - NIH
    This manuscript describes the early history of NK cell discovery, with emphasis on the events in the first decade of NK cell studies, 1972–1982.Missing: 1950s 1960s
  16. [16]
    Five decades of natural killer cell discovery - PMC - NIH
    Jun 6, 2024 · NK cells from wildtype mice kill immune cells from B2m-deficient mice in vitro and reject syngeneic B2m-deficient bone marrow grafts. These ...
  17. [17]
    NK Cell Precursors in Human Bone Marrow in Health and ... - NIH
    Aug 28, 2019 · Natural Killer (NK) cells are innate lymphoid cells (ILC) with potent cytotoxic effector activity, due to their constitutive expression of ...
  18. [18]
    Identification of the earliest natural killer cell–committed progenitor ...
    Nov 17, 2011 · Natural killer (NK) cells develop in the bone marrow and are known to gradually acquire the ability to eliminate infected and malignant ...
  19. [19]
    Natural Killer Cells: Development, Maturation, and Clinical Utilization
    CLPs give rise to Pro-B, Pre-T, innate lymphoid cells (ILCs), lymphoid tissue inducers, and CD122+ Pre-T/early NK cell progenitor (NKP) lineages. Expression of ...
  20. [20]
    The transcription factor E4bp4/Nfil3 controls commitment to the NK ...
    We generated a panel of candidates to be regulated by E4bp4. Eomes, T-bet, Id2, Gata3, Gfi1, Ets1, CEBPγ, IRF2, MEF, and Tox were included because of the ...
  21. [21]
    Transcription factors involved in the regulation of natural killer cell ...
    ... Tox and Id2 is required for the regulation of NK cell development. In addition, Tox has also been shown to regulate NK cell differentiation from ...
  22. [22]
    Human natural killer cells: a unique innate immunoregulatory role ...
    All resting CD56bright NK cells have high expression of CD94/NKG2 C-type lectin receptors.14 A small percentage (less than 10%) expresses killer cell ...
  23. [23]
    Human CD56 dim CD16 dim Cells As an Individualized Natural ...
    Jun 18, 2017 · Human natural killer (NK) cells can be subdivided in several subpopulations on the basis of the relative expression of the adhesion molecule CD56 and the ...
  24. [24]
    Molecular Regulation of NK Cell Maturation - PMC - NIH
    Aug 11, 2020 · In mice, CD27/CD11b divides NK cell maturation into three stages. In human, CD56/CD57 divides NK cell maturation into three stages. NK Cell ...
  25. [25]
    Characterization of the IL-15 niche in primary and secondary ... - PNAS
    Jan 21, 2014 · IL-15 is a cytokine critical for development, maintenance, and response of T cells, natural killer (NK) cells, NK T cells, and dendritic cells.
  26. [26]
    Natural Killer Cell Education and Tolerance - PMC - PubMed Central
    Not all NK cells express inhibitory receptors for self-MHC class I; thus, other tolerance mechanisms are necessary to prevent NK cell-mediated autoimmunity.
  27. [27]
    Human NK Cell Education by Inhibitory Receptors for MHC Class I
    We show here that NK cells lacking inhibitory KIR for self MHC class I molecules are present in human peripheral blood.
  28. [28]
    MHC class I–deficient natural killer cells acquire a licensed ...
    Thus, engagement of self–MHC-specific receptor “licenses” NK cells to be functionally competent to be triggered through their activation receptors. Although ...
  29. [29]
    Adaptations of Natural Killer Cells to Self-MHC Class I - Frontiers
    Licensing enables NK cells to respond to occasional aberrant cells that have lost MHC class I molecules (missing-self recognition). NK cell education is also ...
  30. [30]
    Large Granular Lymphocyte - an overview | ScienceDirect Topics
    Large granular lymphocytes (LGL) refer to medium to large lymphocytes ... μm diameter with prominent azurophilic cytoplasmic granules and round to irregular- ...
  31. [31]
    Clinical Features, Pathogenesis, and Treatment of Large Granular ...
    Large granular lymphocytes (LGLs) are large lymphocytes with azurophilic granules in their cytoplasm ... A subset of natural killer cells in peripheral ...
  32. [32]
    Developmental pathways that generate natural-killer-cell diversity in ...
    Immature NK cells express CD161 and natural-killer group 2, member D (NKG2D) in both mice and humans, as well as receptors required for growth and survival. NK- ...
  33. [33]
    Five decades of natural killer cell discovery
    Jun 6, 2024 · Cudkowicz and Bennet in 1971 reported the rejection of parental bone marrow grafts in F1 recipient mice, a process they referred to as “hybrid ...
  34. [34]
    Liver natural killer cells: subsets and roles in liver immunity - PMC
    Dec 7, 2015 · ... Golgi apparatus, free ribosomes, and few small mitochondria. ... T cell- and B cell-independent adaptive immunity mediated by natural killer cells ...
  35. [35]
    Historical overview on the morphological characterization of large ...
    Historical overview on the morphological characterization of large granular lymphocytes/natural killer cells. Author links open overlay panel. Domenico ...
  36. [36]
    Tissue-Resident NK Cells: Development, Maturation, and Clinical ...
    Here, we summarize the molecular basis, phenotypic signatures, and functions of tissue-resident NK cells and compare them with conventional NK cells.
  37. [37]
    Liver-resident NK cells and their potential functions - PMC - NIH
    Natural killer cells ameliorate liver fibrosis by killing activated stellate cells in NKG2D-dependent and tumor necrosis factor-related apoptosis-inducing ...
  38. [38]
    Salivary Gland NK Cells Are Phenotypically and Functionally Unique
    Here, we identify a distinct salivary gland resident NK cell population, which detects the infection but remains mostly hyporesponsive. Peripheral NK cells ...
  39. [39]
    The functional diversity of tissue‐resident natural killer cells against ...
    Jun 25, 2022 · Consistent with this finding, infant gut-resident NK cells were shown to be more cytolytic than adult gut-resident NK cells [102]. According to ...Liver-Resident Nk Cells · Uterine Nk Cells · Lung Tissue Nk Cells
  40. [40]
    Tissue-resident natural killer (NK) cells are cell lineages distinct from ...
    Apr 8, 2014 · Natural killer (NK) cells belong to the innate immune system; they can control virus infections and developing tumors by cytotoxicity and ...Cd49a Dx5 Trnk Cells In The... · Cd49a + Dx5- Trnk Cells Of... · Article And Author...Missing: salivary | Show results with:salivary<|control11|><|separator|>
  41. [41]
    Unique transcriptional and protein-expression signature in human ...
    Aug 26, 2019 · We identify NK cell subsets with a gene and protein-expression profile consistent with that of tissue-resident lymphocytes, e.g., cell surface ...
  42. [42]
    Localization Matters: Epigenetic Regulation of Natural Killer Cells in ...
    Here, we review how NK cell functionality and plasticity are regulated at the epigenetic level in different tissue microenvironments and within tumor ...
  43. [43]
    Embryonic and neonatal waves generate distinct populations of ...
    Sep 2, 2022 · Analysis of human liver-resident NK cells further reveals a shared signature with murine ILC1s, indicating fetal origin. Our study provides a ...
  44. [44]
    Establishment of tissue-resident immune populations in the fetus
    Yolk sac macrophages, fetal liver, and adult monocytes can colonize an empty niche and develop into functional tissue-resident macrophages. Immunity. 2016 ...
  45. [45]
    Mechanisms of natural killer cell‐mediated cellular cytotoxicity - Prager
    May 20, 2019 · NK cells can mediate their cytotoxic activity via 2 distinct pathways. They can release cytotoxic granules containing perforin and granzymes, or ...Missing: seminal | Show results with:seminal
  46. [46]
    All About (NK Cell-Mediated) Death in Two Acts and an Unexpected ...
    Upon activation, NK cells can kill target cells through two complex mechanisms: the release of cytotoxic granules containing perforin, granzymes and granulysin ...Introduction · First Act: Judges and hangmen... · Second Act: The Execution...
  47. [47]
    Natural Killer (NK) Cell–mediated Cytotoxicity: Differential Use of ...
    Mature natural killer (NK) cells use Ca2+-dependent granule exocytosis and release of cytotoxic proteins, Fas ligand (FasL), and membrane-bound or secreted.Materials And Methods · Results And Discussion · Acknowledgments
  48. [48]
    [PDF] FUNCTIONAL SIGNIFICANCE OF THE PERFORIN/GRANZYME ...
    Granzymes are necessary for triggering apoptosis in target cells,but they depend on being appropriately delivered by perforin.The cytotoxic T lymphocytes (CTLs) ...<|control11|><|separator|>
  49. [49]
    Activation of NK Cells and T Cells by NKG2D, a Receptor for Stress ...
    A receptor for MICA was detected on most γδ T cells, CD8 + αβ T cells, and natural killer (NK) cells and was identified as NKG2D.Missing: paper | Show results with:paper
  50. [50]
    Activation of NK cells and T cells by NKG2D, a receptor for ... - PubMed
    A receptor for MICA was detected on most gammadelta T cells, CD8+ alphabeta T cells, and natural killer (NK) cells and was identified as NKG2D.
  51. [51]
    CD16 is indispensable for antibody-dependent cellular cytotoxicity ...
    Sep 27, 2016 · CD16, the FcγRIIIA, is essential for ADCC by NK cells, and is also expressed by a subset of human blood monocytes. We found that human CD16− ...
  52. [52]
    The CD3ζ adaptor structure determines functional differences ...
    Mar 23, 2022 · In human NK cells, CD16 associates with homo- or heterodimers of FcεR1γ and CD3ζ adaptor molecules which contain immunoreceptor tyrosine-based ...Introduction · Results · Discussion · Materials and methods
  53. [53]
    Specificity and affinity of human Fcγ receptors and their polymorphic ...
    A higher efficiency of IgG1 anti-CD20 therapy was observed in hFcγRIIIA V/V158 patients than in F/F158 patients, and peripheral blood lymphocytes from V/V158 ...<|control11|><|separator|>
  54. [54]
    ADCC employing an NK cell line (haNK) expressing the high affinity ...
    NK-92 cells were genetically modified to produce IL-2 in an autocrine loop, as well as to express a high affinity variant (V158) of the CD16 FcγRIII, and have ...
  55. [55]
    Direct and antibody-dependent cell-mediated cytotoxicity of head ...
    Feb 1, 2019 · High affinity natural killer cells (haNKs) are a cell therapy product capable of mediating both direct and antibody-dependent cell-mediated cytotoxicity (ADCC).
  56. [56]
    Metabolic Regulation of Natural Killer Cell IFN-γ Production - PMC
    Functions of IFN-γ include supporting Th1 differentiation, boosting macrophage ... NK cells respond to activation and controlling IFN-γ translation directly.
  57. [57]
    Murine Macrophages Secrete Interferon γ upon Combined ...
    IL-12 induces NK cells to rapidly secrete IFN-γ, which then acts back to activate macrophages early in an immune response. Furthermore, IL-12 induces IFN-γ ...
  58. [58]
    IL-12 synergizes with IL-18 or IL-1β for IFN-γ production from human ...
    IL-1β is required for IL-12 to induce production of IFN-γ by NK cells. A role for IL-1β in the T cell-independent mechanism of resistance against ...Abstract · Introduction · Methods · Results
  59. [59]
    IκBζ is essential for natural killer cell activation in response to IL-12 ...
    IκBζ was essential for the production of IFN-γ production and cytotoxic activity in NK cells in response to IL-12 and/or IL-18 stimulation.
  60. [60]
    The Molecular Mechanism of Natural Killer Cells Function and Its ...
    Sep 13, 2017 · Effector Immune Response of NK Cells​​ Activated NK cells secrete a wide variety of cytokines such as IFN-γ, TNF-α, GM-CSF, IL-10, IL-5, and IL- ...
  61. [61]
    Comprehensive snapshots of natural killer cells functions, signaling ...
    Nov 8, 2024 · In 1975, the term NK “Natural Killer” was coined (Fig. 1), and the discovery of IL-2 the same year, later revolutionized NK cell studies. In ...
  62. [62]
    NK cell–derived GM-CSF potentiates inflammatory arthritis and is ...
    Feb 25, 2020 · Synovial NK cells promote a neutrophilic inflammatory cell infiltrate, and persistent arthritis, via GM-CSF production, as deletion of NK cells, ...
  63. [63]
    STATs in NK-Cells: The Good, the Bad, and the Ugly - PMC
    CD56bright NK-cells are mainly found in lymph nodes, produce cytokines upon activation, and possess only minor cytotoxic potential. Upon maturation to CD56dim ...
  64. [64]
    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 ...Missing: defense | Show results with:defense
  65. [65]
    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.
  66. [66]
    Specific and nonspecific NK cell activation during virus infection
    Sep 10, 2001 · We show here that NK cell proliferation and production of interferon-γ (IFN-γ) was not dependent on Ly49H expression during early MCMV infection.
  67. [67]
    KIR and HLA interactions are associated with control of primary ...
    A retrospective, single-center cohort study was performed to study the interaction of KIR genotype and primary control of CMV infection after transplantation.
  68. [68]
    Licensing delineates helper and effector NK cell subsets during viral ...
    May 18, 2017 · Those expressing inhibitory receptors capable of binding to self-MHC class I molecules are deemed licensed (16), educated (17), or functional NK ...
  69. [69]
    Natural killer cell licensing during viral infection - PubMed
    Surprisingly, these licensed NK cells fail to control viral infection. During mouse cytomegalovirus (MCMV) infection, SHP-1 signaling downstream of inhibitory ...
  70. [70]
    New aspects of natural-killer-cell surveillance and therapy of cancer
    Nov 1, 2002 · Summary. Natural-killer (NK) cells are important components of the innate immune system. There is evidence of tumour rejection by NK cells.
  71. [71]
    Natural killer group 2D receptor and its ligands in cancer immune ...
    Feb 27, 2019 · In this review, we describe the mechanisms of natural killer group 2D (NKG2D) receptor and NKG2D ligand (NKG2DL) in tumor immune responses.
  72. [72]
    Leveraging NKG2D Ligands in Immuno-Oncology - Frontiers
    MICA/B expressed on the cell surface of tumor cells can be recognized by NK cells through NKG2D and promote a cytotoxic response that leads to tumor cell ...
  73. [73]
    In search of the 'missing self': MHC molecules and NK cell recognition
    According to the 'missing self' hypothesis, one function of NK cells is to recognize and eliminate cells that fail to express self major histocompatibility ...Missing: cancer | Show results with:cancer
  74. [74]
    Inducible down-regulation of MHC class I results in natural killer cell ...
    Dec 17, 2018 · These results show that MHC-I down-regulation can induce either NK cell tolerance or killing in vivo and that inflammation promotes missing-self reactivity.Nk Cell Missing-Self... · Nk Cells Adapt To Global... · Nk Cell Tolerance To...
  75. [75]
    Control of Metastasis by NK Cells - ScienceDirect.com
    Aug 14, 2017 · Malignant cells avoid recognition and elimination by natural killer (NK) cells via multiple mechanisms that operate directly on NK cells, upon ...
  76. [76]
    Human Decidual Natural Killer Cells Are a Unique NK Cell Subset ...
    Natural killer cells constitute 50–90% of lymphocytes in human uterine decidua in early pregnancy. Here, CD56bright uterine decidual NK (dNK) cells were ...
  77. [77]
    Decidual Natural Killer Cells: A Good Nanny at the Maternal-Fetal ...
    May 11, 2021 · Decidual natural killer (dNK) cells are the tissue-resident and major subpopulation of NK cells at the maternal-fetal interface.Abstract · Introduction · The Functional Dialogue... · dNK Cells and Pathological...
  78. [78]
    Cytokine modulation and immunoregulation of uterine NK cells in ...
    The production of growth factors by uNK cells, such as vascular endothelial growth factor (VEGF) and placental growth factor (PlGF), is essential for promoting ...
  79. [79]
    Uterine natural killer cells and angiogenesis in recurrent ...
    Oct 3, 2008 · uNK cells produce angiogenic growth factors and are potential regulators of decidual angiogenesis in early pregnancy. The final common mechanism ...
  80. [80]
    The HLA-G cycle provides for both NK tolerance and immunity at the ...
    HLA-G acts mainly to prevent NK cell cytotoxicity (4–6) and T-cell cytotoxicity (7) and induce tolerance (8, 9). The maternal–fetal interface is a unique ...
  81. [81]
    Maternal natural killer cells at the intersection between reproduction ...
    Apr 26, 2021 · Higher sHLA-G levels might modulate the immune response and prevent fetal loss but can also lead to congenital infection. HLA-G may supress NK ...
  82. [82]
    Role of Decidual Natural Killer Cells in Human Pregnancy and ...
    This review is focused on the role of dNK cells in normal pregnancy and pathological pregnancy, including preeclampsia, recurrent spontaneous abortion, ...<|control11|><|separator|>
  83. [83]
    Uterine NK cells underexpress receptors recognizing HLA-C2 and ...
    Nov 25, 2022 · Taken together, our findings suggest that reproductive failure may be caused by global reduction in expression of uNK receptors important for ...
  84. [84]
    Natural killer cell deficiency - PMC - NIH
    What is also presently unclear in patients with GATA2 mutations is whether or not the NK cell deficiency occurs first, is a hardwired component of the mutation, ...
  85. [85]
    X-linked lymphoproliferative disease due to SAP/SH2D1A deficiency
    X-linked lymphoproliferative disease (XLP1) is a rare immunodeficiency characterized by severe immune dysregulation and caused by mutations in the ...
  86. [86]
    Natural killer cell in systemic lupus erythematosus. Defects in ... - JCI
    Spontaneous cytotoxicity mediated by natural killer (NK) cells is impaired in several human diseases including systemic lupus erythematosus (SLE).
  87. [87]
    Natural Killer Cell Count in Systemic Lupus Erythematosus Patients
    Oct 12, 2023 · In SLE patients, the number of CD56(+) NK cells decreases as compared to healthy controls; this is caused by an imbalance in the creation and ...
  88. [88]
    Hemophagocytic Lymphohistiocytosis - Allen Press
    Aug 4, 2021 · HLH is characterized by uncontrolled activation of cytotoxic T lymphocytes, natural killer cells, and macrophages that can lead to a cytokine ...
  89. [89]
    Hemophagocytic lymphohistiocytosis: current treatment advances ...
    Nov 7, 2024 · T cells, NK cells and macrophages are predominantly responsible for the increased secretion of these inflammatory cytokines. However, despite ...
  90. [90]
    NK cells: energized yet exhausted in adult HLH - ASH Publications
    Jul 30, 2020 · The investigators find that natural killer (NK) cells from adults with HLH exhibit an activated phenotype and normal cytotoxic capacity.
  91. [91]
    Natural killer cells in inflammatory autoimmune diseases - PMC
    Feb 1, 2021 · Current evidence shows that synovial NK cells aggravate arthritis through the production inflammatory mediators such as GM‐CSF, M‐CSF and RANKL, ...Missing: overactivation | Show results with:overactivation
  92. [92]
    The Role of Natural Killer Cells in Autoimmune Diseases - Frontiers
    Natural killer (NK) cells, the large granular lymphocytes differentiated from the common lymphoid progenitors, were discovered in early 1970's.Introductıon · Long-Standıng Members of the... · Natural Killer Maturation...
  93. [93]
    Natural Killer Cells and Their Role in Rheumatoid Arthritis - NIH
    In this review, we focus on current knowledge regarding NK cells and NK cell receptors in human autoimmune diseases such as RA. 2. Natural Killer Cells. Natural ...
  94. [94]
    NK large granular lymphocytic leukemia - Pathology Outlines
    Feb 27, 2025 · NK large granular lymphocytic leukemia (NK LGLL) is characterized by a persistent increase of monoclonal peripheral blood NK cells (usually > 2 x 10 9 /L)
  95. [95]
    Large granular lymphocytic leukemia: a brief review - PMC - NIH
    LGL leukemia is a rare chronic lymphoproliferative disorder of cytotoxic lymphocytes which can be immunophenotypically either T cell or NK cell-derived.
  96. [96]
    Genomic landscape characterization of large granular lymphocyte ...
    Feb 7, 2017 · Large granular lymphocyte (LGL) leukemia is a rare clonal disease characterized by a persistent increase in the number of CD8+ cytotoxic T cells or CD16/56+ ...<|control11|><|separator|>
  97. [97]
    Latest Advances in the Diagnosis and Treatment of Large Granular ...
    May 23, 2018 · LGL leukemia is characterized by a clonal expansion of cytotoxic T or NK lymphocytes. Pathogenesis is still incompletely understood, but ...
  98. [98]
    Function and Structure of Cytomegalovirus MHC-I-Like Molecules in ...
    How the Virus Outsmarts the Host: Function and Structure of Cytomegalovirus MHC-I-Like Molecules in the Evasion of Natural Killer Cell Surveillance · 1.2.3. Nk Receptors In Viral... · 2.4. Nkg2d · 2.5. Ly49 Receptors
  99. [99]
    Structure of UL18, a peptide-binding viral MHC mimic, bound ... - NIH
    One strategy is to down-regulate cell surface expression of host class I MHC molecules, thereby allowing HCMV-infected cells to avoid recognition by virus- ...
  100. [100]
    The Natural Selection of Herpesviruses and Virus-Specific NK Cell ...
    ... NK cell inhibition could represent an immune evasion mechanism of HCMV. ... The HCMV-encoded UL18 glycoprotein has structural homology to MHC class I ...Ly49h And Mcmv · Ly49p And Mcmv · Cd94-Nkg2c And Hcmv
  101. [101]
    Downregulation and/or Release of NKG2D Ligands as Immune ...
    Recently, it has been reported that soluble MICA (sMICA) and MICB are released in sera from patients with different malignancies and downregulate effector cell- ...
  102. [102]
    NKG2D and MICA/B shedding: a 'tag game' between NK cells ... - NIH
    Dec 22, 2020 · MICA/B shedding by cancer cells causes effective escape from NKG2D recognition and allows the development of cancers.
  103. [103]
    TGF-β is responsible for NK cell immaturity during ontogeny and ...
    We demonstrate that NK cells could complete maturation early in life if TGF-βR signaling was blocked, indicating plasticity of NK cell maturation during ...
  104. [104]
    Arrested development: suppression of NK cell function in the tumor ...
    Jan 10, 2021 · TGF‐β impairs NK cell function directly by limiting NK cell antibody‐dependent cellular cytotoxicity (ADCC) and IFN‐γ production through ...
  105. [105]
    Phase I clinical trial of autologous NK cell therapy using novel ...
    In this phase I clinical trial consisting of advanced digestive cancer patients, we explored the safety and feasibility of adoptive transfer of NK cells ...
  106. [106]
    Successful adoptive transfer and in vivo expansion of ... - PubMed
    These findings suggest that haploidentical NK cells can persist and expand in vivo and may have a role in the treatment of selected malignancies.
  107. [107]
    First-in-human trial of rhIL-15 and haploidentical natural killer cell ...
    Jul 2, 2019 · Haploidentical NK-cell infusions given with rhIL-15 achieved remission in 35% of patients with refractory acute myeloid leukemia.
  108. [108]
    A phase II clinical trial of adoptive transfer of haploidentical natural ...
    Mar 20, 2019 · To determine whether haploidentical NK cell therapy prolongs event-free survival in children with intermediate-risk AML, we conducted a phase 2 ...
  109. [109]
    Adoptive NK cell therapy in AML: progress and challenges - PMC
    Jan 17, 2025 · This review provides a comprehensive analysis of clinical outcomes and toxicity profiles provided from clinical trials, clinical studies and ...
  110. [110]
    Prospects and Advances in Adoptive Natural Killer Cell Therapy for ...
    The reported efficacies have so far been moderate and of short duration due to problems with homing, in vivo persistence, lack of expansion and ...
  111. [111]
    Engineered natural killer cells for cancer therapy - Cell Press
    Oct 23, 2025 · Allogeneic natural killer (NK) cell immunotherapy is emerging as a promising and scalable, off-the-shelf platform for treating relapsed and ...
  112. [112]
    Chimeric antigen receptor-engineered NK cells - NIH
    Nov 2, 2022 · Many preclinical studies on CAR-engineered NK (CAR-NK) cells have shown their remarkable efficacy in cancer therapy and their superiority over autologous CAR-T ...Missing: BiKEs | Show results with:BiKEs
  113. [113]
    The clinical landscape of CAR NK cells - PMC - PubMed Central
    Mar 27, 2025 · Here we explore the current clinical landscape of CAR NK cells, and their application in hematologic malignancies and solid cancers, as well as their potential ...Missing: BiKEs | Show results with:BiKEs
  114. [114]
    Gene-edited and CAR-NK cells: Opportunities and challenges ... - NIH
    Engineered NK cell therapy has so far mostly shown results in hematological tumors, with a phase I/II trial of CD19-CAR-NK cells in acute myeloid leukemia ...
  115. [115]
    Chimeric antigen receptor engineered natural killer cells for cancer ...
    Aug 10, 2023 · In this review, we present the favorable profile of NK cells as a potential platform for CAR-based engineering and then summarize the outcomes ...Missing: BiKEs | Show results with:BiKEs
  116. [116]
    The Natural Killer Cell Line NK-92 and Its Genetic Variants - NIH
    Jun 13, 2025 · The unmodified NK-92 cells have completed several clinical trials confirming no significant side effects and clinical responses. Its CAR ...Missing: BiKEs | Show results with:BiKEs
  117. [117]
    Application and prospects of genetic engineering in CAR-NK cell ...
    ... engineered NK-92 cells (20). Additionally, CAR-NK cells utilizing endogenous promoters have demonstrated promising anti-tumor efficacy and safety in ...Missing: BiKEs | Show results with:BiKEs
  118. [118]
    Principles and current clinical landscape of NK cell engaging ...
    Sep 29, 2023 · Some groups have named these bispecific antibodies “bispecific killer cell engagement (BiKEs).” These BiKEs work by bringing NK cells and target ...
  119. [119]
    NKG2D-Fc fusion protein promotes antitumor immunity through the ...
    May 28, 2020 · We explore NKG2D-Fc as a modality to modulate antitumor immunity through the depletion of immunosuppressive MDSCs and Tregs in the TME.
  120. [120]
    Fc-Engineered NKG2D-IgG1 Fusion Proteins Target Leukemia Cells ...
    Nov 18, 2011 · Our results demonstrate that Fc-engineered NKG2D-Fc-ADCC fusion proteins can effectively target NKG2DL-expressing leukemia cells for NK anti-tumor reactivity.
  121. [121]
    Natural Killer Cell Engagers (NKCEs): a new frontier in cancer ... - NIH
    NKCEs, such as BiKE (bispecific killer cell engager) or TriKE (trispecific killer cell engager), provide several advantages over the earlier two strategies.
  122. [122]
    Engineered NK Cells Against Cancer and Their Potential ... - NIH
    Here, we will discuss the potential of NK cells in CAR-based therapies focusing on the applications of CAR-NK cells in cancer therapy and beyond.Missing: BiKEs | Show results with:BiKEs
  123. [123]
    NK cell-based cancer immunotherapy: from basic biology to clinical ...
    Jan 6, 2021 · The natural cytotoxic receptor NKp46 is also often used to identify mouse and human NK cells in combination with the absence of CD3 expression.Nk Cell Biology · Car-Nk Cell As An... · Nk Cells ``priming''...
  124. [124]
    The Synergistic Use of IL-15 and IL-21 for the Generation of NK ...
    Our study revealed that NK cells have a significantly higher cytotoxic potential to combat NB than CIK cell products, especially following the synergistic use ...Missing: preconditioning | Show results with:preconditioning
  125. [125]
    The Synergistic Use of IL-15 and IL-21 for the Generation of NK ...
    Our study revealed that NK cells have a significantly higher cytotoxic potential to combat NB than CIK cell products, especially following the synergistic use ...Missing: preconditioning | Show results with:preconditioning
  126. [126]
    A phase 1 study of lirilumab (antibody against killer immunoglobulin ...
    This phase 1 trial demonstrated the satisfactory safety profile of lirilumab up to doses that enable full and sustained blockade of KIR.
  127. [127]
    The combination of anti-KIR monoclonal antibodies with anti-PD-1 ...
    Apr 24, 2018 · The inhibitory HLA/KIR could combine with the PD-1/PD-L1 signaling pathway negatively regulating NSCLC tumor immunity. Keywords: non-small cell ...
  128. [128]
    Safety of the natural killer (NK) cell-targeted anti-KIR antibody ...
    1086P - Safety of the natural killer (NK) cell-targeted anti-KIR antibody, lirilumab (liri), in combination with nivolumab (nivo) or ipilimumab (ipi) in two ...
  129. [129]
    CpG and double-stranded RNA trigger human NK cells by Toll ... - NIH
    CpG and double-stranded RNA trigger human NK cells by Toll-like receptors: Induction of cytokine release and cytotoxicity against tumors and dendritic cells.Missing: agonists storm
  130. [130]
    TLR‐mediated activation of NK cells and their role in bacterial/viral ...
    Dec 24, 2013 · NK cells possess receptors allowing them to sense and respond to viral and bacterial patterns, including Toll-like receptors (TLRs).
  131. [131]
    Activation of NK cell cytotoxicity by aerosolized CpG-ODN/poly(I:C ...
    In a melanoma lung metastasis model, aerosolized TLR9 and TLR3 agonists have been reported to induce antitumor immunity through NK cells activation. In the ...Missing: cytokine storm
  132. [132]
    Clonal expansion and epigenetic inheritance of long-lasting NK cell ...
    Oct 26, 2022 · Competitive clonal expansion after antigen encounter is accompanied by pronounced epigenetic remodeling, forming a repertoire of memory cells ...
  133. [133]
    Cytokine activation induces human memory-like NK cells | Blood
    Dec 6, 2012 · We show that human NK cells preactivated briefly with cytokine combinations including IL-12, IL-15, and IL-18 followed by a 7- to 21-day rest have enhanced IFN ...
  134. [134]
    AACR 2025: Off-the-shelf CAR natural killer cell therapy with logic ...
    Apr 30, 2025 · AACR 2025: Off-the-shelf CAR natural killer cell therapy with logic gates elicits complete remissions in relapsed or refractory blood cancers.
  135. [135]
    Dual antigen-targeted off-the-shelf NK cells show durable ... - PubMed
    Dec 8, 2022 · iDuo NK cells combined with therapeutic antibodies represent a promising approach to prevent relapse due to antigen loss and tumor heterogeneity in patients ...
  136. [136]
    A phase 1 dose escalation trial of ex-vivo expanded allogeneic cord ...
    May 31, 2023 · Allogeneic ex-vivo expanded NK cells immunotherapy demonstrated favorable safety profile in patients with advanced pediatric solid tumors.
  137. [137]
    Mapping the global clinical landscape of NK cell therapies for solid ...
    Oct 4, 2025 · (C) NK cell therapy trials, by status, as of December 31, 2024. (D) ... Allogeneic NK cells are mainly obtained from umbilical cord blood ...
  138. [138]
    Pipeline: NK Cell Therapy for Autoimmune Disease and Cancer
    AlloNK is an allogeneic, non-genetically modified NK cell therapy in clinical trials for the treatment of autoimmune disease and cancer.Missing: II | Show results with:II
  139. [139]
    Development of NK cell-based cancer immunotherapies through ...
    Mar 5, 2024 · Natural killer (NK) cell-based immunotherapies are attracting increasing interest in the field of cancer treatment.
  140. [140]
    Impact of inhibitory KIR ligand mismatch and other variables ... - NIH
    Dec 16, 2024 · We evaluated the impact of inhibitory killer immunoglobulin-like receptor (KIR)/HLA mismatch, alongside patient, donor, and transplant factors, on clinical ...
  141. [141]
    Machine learning-derived natural killer cell signature predicts ...
    A NK cells-related risk model was established via the machine learning-based integrative procedure, which could provide reliable prognostic information.