Fact-checked by Grok 2 weeks ago

Tumor necrosis factor

Tumor necrosis factor (TNF), also known as TNF-α, is a pleiotropic primarily secreted by activated macrophages, T cells, and other immune cells in response to or , serving as a key mediator of and immune regulation. Originally discovered in 1975 by Lloyd J. Old and colleagues at Memorial Sloan-Kettering Cancer Center as a soluble factor from macrophages capable of inducing hemorrhagic necrosis and regression of certain tumors in mice, TNF was initially named for this antitumor activity but later recognized for its broader roles in host defense and pathology. Structurally, TNF exists as a homotrimer in its active form, with each featuring a compact β-sandwich fold stabilized by bonds, and it is initially synthesized as a 26 kDa transmembrane precursor that is proteolytically cleaved to yield the soluble 17 kDa form. In physiological contexts, TNF contributes to essential processes such as acute inflammatory responses, fever induction, of infected or damaged cells, and orchestration of adaptive immunity, thereby aiding in clearance and . However, dysregulated TNF production underlies numerous pathological conditions, including autoimmune diseases like and , chronic infections, and even cancer progression, where it can paradoxically promote tumor growth and through sustained . TNF exerts its diverse effects by binding to two receptors—TNFR1, ubiquitously expressed and involved in pro-inflammatory and pro-apoptotic signaling, and TNFR2, primarily on immune cells and linked to cell survival and proliferation—activating pathways such as , MAPK, and cascades. Due to its central role in , TNF has become a prime therapeutic target, with biologic inhibitors like monoclonal antibodies (e.g., ) revolutionizing treatment for TNF-driven diseases since the 1990s.

History and Discovery

Early Observations

In 1893, surgeon William B. Coley conducted pioneering experiments by injecting live streptococci from into cancer patients with inoperable sarcomas, observing spontaneous tumor regression in some cases accompanied by hemorrhagic necrosis. These initial successes prompted Coley to refine the approach using heat-killed mixtures of and filtrates, which he termed "Coley toxins," to induce similar anti-tumor effects without the risks of live infection. Over the following decades, Coley treated more than 1,000 patients, documenting tumor shrinkage and occasional cures, particularly in sarcomas, though the mechanism remained unclear and attributed to immune stimulation. Nearly a century later, in 1975, Elizabeth A. Carswell and colleagues at identified a soluble factor responsible for endotoxin-induced tumor necrosis in mice. In their experiments, mice primed with Bacillus Calmette-Guérin (BCG) and then challenged with bacterial (LPS, a component of endotoxin) developed containing a heat-stable, non-dialyzable protein (molecular weight 30,000–50,000) that caused hemorrhagic necrosis in transplanted tumors when injected into naive animals. This factor, later named tumor necrosis factor (TNF), was absent in unprimed mice or those treated with LPS alone, highlighting the role of prior immune activation. Early observations revealed TNF's dual nature: potent anti-tumor activity alongside systemic toxicity. The factor selectively induced necrosis in certain tumors, such as the LPS-sensitive Meth A , while sparing normal tissues, but it also triggered , manifested as profound and wasting in treated animals. Key experiments demonstrated that LPS-stimulated macrophages from BCG-primed mice were the primary source of TNF production, releasing the factor into circulation within hours of endotoxin exposure, which then mediated against Meth A cells both in vitro and . These findings established TNF as a macrophage-derived mediator linking bacterial to tumor regression, echoing Coley's earlier empirical observations.

Purification and Characterization

In the mid-1980s, independent efforts by research groups led to the purification of tumor necrosis factor (TNF) to homogeneity, marking a key milestone in its molecular identification. The group at Memorial Sloan-Kettering Cancer Center, led by Lloyd J. Old, purified TNF from the culture supernatant of the human B-lymphoblastoid LuK II cell line using a sequential chromatographic approach that included Blue Sepharose , DEAE-Sephacel ion-exchange , G-100 gel filtration, and phenyl-Sepharose hydrophobic interaction . Concurrently, the team, including Bharat B. Aggarwal and Peter W. Gray, isolated TNF from serum-free supernatants of phorbol ester-stimulated HL-60 promyelocytic cells (a model for macrophage-derived TNF) through controlled-pore glass adsorption, DEAE-cellulose ion-exchange , Mono Q fast-protein liquid , and reverse-phase . These purifications, building on partial sequencing for cDNA reported in late , confirmed TNF as a novel 17 kDa polypeptide. Early characterization revealed TNF's biochemical properties through sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and amino-terminal sequencing. Under reducing conditions, purified TNF migrated as a 17 kDa band, while non-reducing SDS-PAGE showed a predominant 46 kDa species, indicating a homotrimeric structure composed of three identical subunits essential for biological activity. Amino-terminal analysis of the HL-60-derived TNF demonstrated no glycosylation, as the observed molecular weight matched the predicted sequence from cDNA without post-translational modifications, though minor heterogeneity was noted in LuK II-derived preparations possibly due to incomplete purification or cell-specific variants. Cytotoxicity was assessed using the standard L929 mouse fibrosarcoma cell assay, where TNF induced cell death in a dose-dependent manner, with actinomycin D enhancing sensitivity to mimic tumor conditions. The specific activity of purified TNF reached approximately 5 × 10^7 per mg protein for the HL-60 form and up to 1.5 × 10^8 per mg for the LuK II form, establishing its potency as a agent far exceeding crude preparations. These values were determined by against L929 cells, defining one as the amount causing 50% cell killing under standardized conditions. Initial studies also distinguished TNF-α, primarily produced by macrophages and HL-60-like cells, from TNF-β (lymphotoxin), derived from activated lymphocytes such as in RPMI 1788 B-cell lines; while both exhibited similar trimeric structures and L929 , TNF-β showed distinct sequences and slightly higher molecular weight (~25 kDa monomer) upon purification and . This separation highlighted TNF-α's role in acute and , contrasting with TNF-β's lymphocyte-specific expression.

Nomenclature

The term "tumor necrosis factor" (TNF) originated from experiments conducted in 1975, where a serum factor induced by bacterial endotoxin in Bacillus Calmette-Guérin-primed mice was found to cause hemorrhagic necrosis in transplantable tumors, distinct from previously identified agents like interferon. This factor was initially characterized for its cytotoxic effects on tumor cells in vitro, particularly against L-929 mouse fibrosarcoma cells, leading to its naming based on the observed in vivo tumor regression. In the mid-1980s, following the and sequencing of distinct cytotoxic factors, the was refined to distinguish TNF-α, the predominant form derived from macrophages and monocytes in response to stimuli like , from TNF-β, also known as lymphotoxin-α, which is primarily produced by activated T cells. This differentiation arose from observations that the two proteins shared structural (approximately 30-50% identity) and bound the same receptors but originated from different cellular sources. Concurrently, TNF-α was identified as identical to cachectin, a macrophage-derived protein implicated in and metabolic disturbances during chronic infections, resolving earlier uncertainties about their relationship. The broader TNF superfamily nomenclature emerged in the late 1980s and early as additional homologous ligands and receptors were discovered, with formal standardization occurring through international efforts in the late to assign systematic names reflecting genetic and functional relationships. This classification includes TNF receptors such as TNFR1 (also designated CD120a or p55) and TNFR2 (CD120b or p75), which were cloned and characterized in and mediate signaling through distinct pathways. Currently, TNF is classified within the TNF ligand superfamily (TNFSF), comprising 19 members that form homotrimers or heterotrimers to engage 29 receptors in the (TNFRSF), coordinating diverse immune and inflammatory responses.

Evolutionary Aspects

Origins in Prokaryotes

While true tumor necrosis factor (TNF) proteins are absent in prokaryotes, bacterial genomes encode proteins with structural motifs resembling the TNF homology domain, suggesting ancient evolutionary precursors to the TNF superfamily. For instance, the opportunistic pathogen produces BC2L-C, a soluble featuring an N-terminal domain that adopts a trimeric β-jellyroll fold strikingly similar to that of eukaryotic TNF family members, enabling carbohydrate binding and potentially contributing to host-pathogen interactions. This structural highlights how prokaryotic proteins may have laid groundwork for the trimeric architecture central to TNF signaling in eukaryotes. In prokaryotes like , TNF-like functions manifest through stress response and autolysis mechanisms that parallel TNF's role in . Proteins involved in these processes share sequence similarities with the death domain of TNF receptors, facilitating self-regulated under environmental stress, which aids bacterial and survival. A notable functional exists with bacterial holins, phage-encoded proteins that membrane permeabilization and cell in a timed manner, akin to how TNF induces via receptor oligomerization and activation in metazoans. Similarly, toxin-antitoxin systems such as MazEF in mediate in response to stressors like nutrient deprivation, where the alarmone ppGpp acts as a "death " comparable to TNF, promoting community-level in bacterial biofilms. Genomic and bioinformatics analyses from the reveal that precursors to the TNF superfamily predate metazoan , indicating an in early eukaryotic lineages rather than prokaryotes proper. These studies underscore conserved motifs, such as death domain-like structures (e.g., TRADD-N domains), present in diverse and linked to immunity and regulation, bridging prokaryotic signaling to the more complex TNF pathways observed in vertebrates.

Conservation in Vertebrates

Tumor necrosis factor (TNF) exhibits significant sequence conservation across species, reflecting its fundamental role in immune responses. In mammals, TNF-α shares approximately 80% sequence identity with orthologs in such as mice, while identities drop to around 35-45% in birds like chickens and further to 30-40% in teleost fish such as . Despite these variations, critical functional motifs remain highly preserved, including the trimerization interface essential for TNF oligomerization and specific receptor-binding residues that mediate interactions with TNFR1 and TNFR2. These conserved elements ensure structural integrity and signaling efficacy across distant vertebrate lineages. Functional conservation is evident in the ability of TNF to induce , a process mechanistically similar in non-mammalian vertebrates and mammals. In models, TNF signaling activates a caspase-8-dependent apoptotic pathway involving , mirroring the extrinsic apoptosis cascade observed in mammalian cells and highlighting evolutionary preservation of this cytotoxic function over more than 450 million years. The expansion of the TNF superfamily in jawed vertebrates (gnathostomes) arose from whole-genome duplication events approximately 500 million years ago, which facilitated the divergence of TNF-α and TNF-β (also known as lymphotoxin-α). This duplication contributed to the specialization of TNF ligands in adaptive immunity, with TNF-α focusing on pro-inflammatory and apoptotic roles, while TNF-β supports lymphoid . Comparative genomic analyses have identified preserved TRAF-binding domains in TNF receptor signaling pathways from amphibians to humans, underscoring the conservation of downstream adaptor interactions that link TNF activation to and JNK pathways critical for immune regulation. These domains, including the TRAF-C region, maintain structural and functional integrity across classes, as revealed by phylogenetic reconstructions of TRAF family genes.

Molecular Biology

Gene Organization and Location

The human TNF gene, encoding tumor necrosis factor alpha (TNF-α), is located on the short arm of at cytogenetic band 6p21.3, within the class III region of the (MHC). This positioning places it amid a cluster of immune-related genes, approximately 250 kb telomeric to the class II region and 850 kb centromeric to the class I region. The gene itself spans approximately 2.8 kb of genomic DNA and is organized into four s interrupted by three introns, where 1 encodes the 5' untranslated region and the majority of the , 2 encodes the remainder of the , 3 encodes the N-terminal portion of the mature protein, and 4 encodes the C-terminal portion of the mature protein and the 3' untranslated region. The TNF gene forms part of a compact gene cluster in the MHC class III region, closely flanked by the (LTA, also known as TNF-β) gene upstream and the lymphotoxin beta (LTB) gene downstream, with the entire LTA-TNF-LTB locus spanning about 10 kb. This cluster is embedded within a broader ~220-280 kb segment of the MHC that includes adjacent HLA-linked genes such as HLA-DRA (centromeric) and HLA-B (telomeric), facilitating coordinated regulation of inflammatory responses. No functional of TNF are present in the , though genomes contain processed pseudogene copies that may influence evolutionary divergence in expression patterns. The proximal promoter of the human TNF gene, located upstream of exon 1, features a TATA box at approximately -30 bp relative to the transcription start site and multiple NF-κB binding sites (including κB1, κB2, κ3, and others) between -50 and -650 bp, which are critical for inducible transcription in immune cells. A notable genetic variant in this promoter is the single nucleotide polymorphism (SNP) rs1800629 (-308 G/A), where the A allele is associated with enhanced transcriptional activity and higher TNF-α protein expression levels compared to the G allele, influencing susceptibility to autoimmune and infectious diseases. This polymorphism occurs within a functional NF-κB-like site, underscoring its role in modulating basal and stimulated gene output.

Transcription and Regulation

The transcription of the TNF gene is primarily regulated at the level of initiation through its core promoter region, spanning approximately -1250 to +80 base pairs relative to the transcription start site. This region contains multiple binding sites for key transcription factors, including SP-1 sites that facilitate basal transcription, AP-1 sites that respond to stress signals, and sites that drive inducible expression. Activation of these elements occurs rapidly in response to proinflammatory stimuli such as (LPS) from bacterial sources or interleukin-1 (IL-1), which trigger translocation to the nucleus and subsequent binding to the promoter, resulting in enhanced recruitment and TNF mRNA synthesis. Inducible regulation of TNF transcription involves both positive and negative mechanisms to fine-tune inflammatory responses. Positive regulation is mediated by (MAPK) and c-Jun N-terminal kinase (JNK) pathways, which phosphorylate and activate AP-1 family members (e.g., c-Jun and c-Fos), enabling their binding to the promoter and synergistic interaction with to amplify transcription. In contrast, negative regulation is exerted by , which bind to the (GR) and interact with glucocorticoid response elements (GREs) in the promoter or indirectly suppress and AP-1 activity through protein-protein interactions, thereby repressing TNF expression to resolve . Tissue-specific control of TNF transcription is achieved through distal enhancers, particularly in immune cells like macrophages, where the family transcription factor PU.1 binds to conserved enhancer regions upstream of the promoter. PU.1 recruitment facilitates and cooperative binding with , enabling high-level, cell-type-restricted TNF expression in response to microbial signals. This mechanism ensures that TNF production is prioritized in professional antigen-presenting cells during innate immune activation. To model these dynamics, a basic differential equation describes the rate of TNF mRNA accumulation as dependent on NF-κB activity: \frac{d[\text{TNF mRNA}]}{dt} = k_{\text{trans}} \cdot [\text{NF-κB}] - \delta \cdot [\text{TNF mRNA}] Here, k_{\text{trans}} represents the transcription rate constant modulated by promoter occupancy, and \delta is the mRNA degradation rate, highlighting how transient NF-κB nuclear levels dictate the temporal profile of TNF expression.

Post-Transcriptional Control

Post-transcriptional regulation of tumor necrosis factor (TNF) expression ensures tight control over its levels, preventing excessive while allowing rapid responses to stimuli. This occurs primarily through mechanisms affecting mRNA and efficiency, with the 3' untranslated region (3' UTR) playing a central role in decay pathways. The TNF mRNA contains multiple AU-rich elements () in its 3' UTR, which serve as binding sites for destabilizing factors that promote rapid degradation. The tristetraprolin (TTP) specifically recognizes these , recruiting deadenylation enzymes and the exosome complex to shorten the poly(A) tail and trigger 5'-3' exonucleolytic decay, resulting in a short mRNA of approximately 30 minutes in lipopolysaccharide-stimulated macrophages. This instability limits TNF production to brief bursts following transcriptional activation. In contrast, during inflammatory conditions, the HuR binds to the same , competing with TTP to stabilize the mRNA and extend its , thereby sustaining TNF expression; studies have shown HuR translocation to the upon stimulation, enhancing binding and protection from decay. Additionally, alternative sites in the TNF 3' UTR can generate isoforms with varying lengths, influencing the inclusion of and miRNA binding sites to modulate , with HuR promoting usage of proximal sites that shorten the UTR and potentially enhance persistence in inflammatory contexts. MicroRNAs (miRNAs) further fine-tune TNF post-transcriptionally by targeting its mRNA in immune cells. In macrophages, miR-125b directly binds the TNF 3' UTR, suppressing translation and promoting mRNA degradation to dampen pro-inflammatory responses during bacterial infections. Similarly, miR-146a, upregulated in response to signaling, inhibits TNF expression by targeting upstream regulators like IRAK1 and TRAF6, indirectly reducing TNF mRNA levels and providing negative feedback in activated macrophages.

Protein Structure and Variants

Structure of TNF-α and TNF-β

Tumor necrosis factor alpha (TNF-α) is initially synthesized as a 233-amino acid type II transmembrane precursor protein with a molecular weight of approximately 26 kDa. Proteolytic cleavage by a zinc metalloprotease, such as ADAM17, releases the mature soluble form, which consists of 157 amino acids and has a monomer molecular weight of about 17 kDa. The mature TNF-α monomer features a compact jelly-roll β-sandwich fold, characterized by two antiparallel β-sheets composed of eight β-strands, and contains two cysteine residues (at positions 69 and 101) forming an intramolecular disulfide bond. The homotrimer is stabilized primarily by hydrophobic interactions at the monomer interfaces, with a total molecular weight of roughly 52 kDa and exhibiting threefold rotational symmetry in a bell-shaped quaternary structure. The crystal structure of the human TNF-α homotrimer, determined at 2.6 Å resolution, reveals a central pore along the symmetry axis and reveals how the trimer's surface facilitates biological activity. TNF-α undergoes limited post-translational modifications in its mature form; it possesses a potential N-linked glycosylation site at asparagine residue 7 (Asn83 in the precursor), though glycosylation is not always observed and may vary by cellular context. Tumor necrosis factor beta (TNF-β), also known as lymphotoxin alpha (LT-α), is produced as a 205-amino acid precursor, with the mature secreted form comprising 171 amino acids after removal of a 34-residue signal peptide. Similar to TNF-α, the LT-α monomer adopts a jelly-roll β-sandwich topology with two β-sheets formed by β-strands, lacking disulfide bonds and cysteine residues in the mature form. LT-α primarily exists as a soluble homotrimer, analogous to TNF-α, with a structure determined by X-ray crystallography at 1.9 Å resolution for residues 24–171 of the mature protein, highlighting its bell-shaped trimeric assembly with threefold symmetry. Unlike TNF-α, LT-α does not form a transmembrane precursor but associates with lymphotoxin beta (LT-β) to create a membrane-bound heterotrimer (LT-α1β2), where one LT-α subunit pairs with two LT-β subunits anchored via LT-β's transmembrane domain; this heterotrimer is expressed on the surface of activated lymphocytes and exhibits distinct functional properties. The LT-α homotrimer shares approximately 30% sequence identity with TNF-α, contributing to their structural homology within the TNF superfamily.

Receptor Binding Domains

The receptor binding domains of tumor necrosis factor alpha (TNF-α) are located within the TNF homology domain of its trimeric structure, enabling high-affinity interactions with TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). These domains facilitate the assembly of signaling complexes by contacting the extracellular cysteine-rich domains (CRDs) of the receptors. The TNF-α trimer adopts a bell-shaped fold with three identical subunits, each contributing residues to the binding interfaces, though receptor engagement often results in asymmetric complexes due to steric and affinity differences. Binding to TNFR1 primarily occurs through residues in the CD loop (connecting strands C and D) and DE loop (connecting strands D and E) of the , which interact with CRD2 and CRD3 of the receptor. Key residues such as Arg32 in the CD loop and Tyr59 in the DE loop form critical hydrogen bonds and hydrophobic contacts with the receptor, stabilizing the trimer-receptor interface. These interactions are essential for the initial recognition and clustering of TNFR1 molecules on the cell surface. In contrast, TNF-α exhibits specificity for TNFR2 through a broader that incorporates the AA' (connecting the N-terminal extension to strand A) alongside elements of the CD and DE loops, allowing for more extensive contacts across CRD1-3 of the receptor. This contributes to , with a (Kd) of approximately 20–100 for the TNF-α-TNFR1 complex compared to about 100–400 for TNF-α-TNFR2; TNFR1 thus has higher for soluble TNF-α. Cryo-EM structures of the TNF-α trimer bound to TNFR1 and TNFR2 reveal asymmetric arrangements, where receptors bind at an angle to the trimer axis, promoting oligomerization without full symmetry. Mutations within these domains significantly alter receptor selectivity and function. For instance, the R32W substitution in the CD loop disrupts interactions with TNFR2 while preserving TNFR1 binding, resulting in variants that selectively activate TNFR1-mediated pathways and abolish TNFR2 engagement, as demonstrated in binding assays showing no detectable affinity for TNFR2. Such mutations also impact reverse signaling, where transmembrane TNF-α acts as a for adjacent cells, highlighting the domains' role in bidirectional communication.

Signaling Mechanisms

TNFR1 Pathway

Upon binding of tumor necrosis factor (TNF) to TNFR1, the receptor trimerizes, enabling the recruitment of the adaptor protein tumor necrosis factor receptor type 1-associated death domain (TRADD) through interactions between their respective death domains (DD). TRADD subsequently serves as a scaffold to recruit receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and TNF receptor-associated factor 2 (TRAF2), forming the core TNFR1 signaling complex at the plasma membrane. This assembly is crucial for initiating downstream signaling events. The TNFR1 pathway bifurcates into pro-survival/inflammatory and branches. In the pro-survival arm, TRAF2 recruits the (IKK) complex, leading to activation, which promotes transcription of anti-apoptotic and inflammatory genes. Conversely, the death pathways involve activation, resulting in , or, under conditions where are inhibited, phosphorylation of RIPK3 by , followed by mixed lineage kinase domain-like protein (MLKL) activation to execute necroptosis. These outcomes depend on post-translational modifications and complex stability. Key signaling complexes distinguish these fates: Complex I, comprising TNFR1, TRADD, , TRAF2, cellular inhibitors of apoptosis proteins (cIAP1/2), and the linear ubiquitin chain assembly complex (LUBAC), assembles at the membrane to drive pro-inflammatory signaling. Upon disassembly, dissociates to form Complex II, which includes TRADD, , and for , or , RIPK3, and MLKL for necroptosis. LUBAC contributes to Complex I stability by mediating linear ation of at residues such as K377, which anchors signaling adaptors and prevents death complex formation. Regulation of pathway branching is tightly controlled, particularly by cIAP1/2, which mediate K63-linked ubiquitination of to recruit NEMO and activate , while silencing signaling.

TNFR2 Pathway

The (TNFR2), also known as TNFRSF1B, is characterized by the absence of a (DD) in its cytoplasmic tail, distinguishing it from TNFR1 and precluding direct initiation of or necroptosis pathways. Instead, TNFR2 possesses four cysteine-rich in its extracellular region for ligand binding and a TRAF-binding in the intracellular domain that facilitates direct recruitment of TNF receptor-associated factors (TRAFs), primarily TRAF2 and TRAF5, upon trimerization with TNF-α. This recruitment initiates non-canonical signaling cascades, including activation of the canonical pathway through IKK complex phosphorylation and the alternative pathway via stabilization, as well as MAPK pathways such as JNK and p38 that promote gene transcription for survival and proliferation. TNFR2 signaling prominently activates the PI3K/Akt pathway, which is more robust than in TNFR1 and drives anti-apoptotic effects by phosphorylating Bad and FoxO transcription factors, thereby enhancing cell survival and inhibiting caspase activation. In T cells, particularly regulatory T cells (Tregs), TNFR2 engagement promotes expansion and suppressive function through PKCθ-mediated activation of and MAPK, fostering and without inducing . Unlike TNFR1, which can share TRAF2 recruitment for overlapping signals, TNFR2's pathways emphasize regenerative outcomes, such as tissue repair and responses. TNFR2 exhibits tissue-specific expression, notably on endothelial cells, where it drives by upregulating VEGF and adhesion molecules via , supporting vascular remodeling without apoptotic contributions. This receptor does not induce , focusing instead on pro-survival and proliferative signals that contrast with TNFR1's pro-apoptotic potential. Regulation of TNFR2 involves rapid ligand-induced and lysosomal , limiting sustained signaling, and it preferentially binds membrane-bound TNF over soluble forms due to higher for the transmembrane ligand, enhancing localized cell-cell interactions.

Reverse and Transmembrane Signaling

Transmembrane TNF-α (tmTNF-α), a 26 kDa , is primarily expressed on the surface of activated immune cells including monocytes, macrophages, and T lymphocytes, where it serves dual roles as both a for forward signaling and a receptor for reverse signaling. Reverse signaling is initiated when tmTNF-α is engaged or crosslinked by TNF receptors (TNFRs) on neighboring s or by therapeutic anti-TNF antibodies, transmitting signals from the extracellular environment into the of the tmTNF-α-expressing . This bidirectional communication distinguishes tmTNF-α from its soluble counterpart and allows for fine-tuned regulation of immune responses in cell-cell interactions. The intracellular domain of tmTNF-α lacks canonical death domains but contains motifs that facilitate recruitment of adaptor proteins, leading to activation of key signaling cascades in immune cells. In monocytes and macrophages, reverse signaling prominently activates (PKC) isoforms and the (MAPK) family, including ERK and p38 pathways, which promote production and endotoxin tolerance. For example, crosslinking of tmTNF-α induces MAPK/ERK signaling, resulting in lipopolysaccharide (LPS) resistance by downregulating proinflammatory responses. In T cells, reverse signaling enhances activation by co-stimulating proliferation and secretion, such as interferon-γ, thereby amplifying adaptive immunity. Regulation of reverse signaling is tightly controlled by proteolytic shedding of tmTNF-α, primarily mediated by the metalloprotease ADAM17 (also known as TACE), which cleaves the extracellular stalk region to release soluble TNF-α and diminish surface tmTNF-α levels. This cleavage event modulates the intensity and duration of reverse signals, preventing excessive activation in inflammatory contexts; inhibition of ADAM17 enhances tmTNF-α-mediated reverse signaling and associated effects in macrophages. The 26 kDa tmTNF-α form on not only delivers forward signals to induce in adjacent target cells via TNFR1 engagement but also, through reverse signaling, supports monocyte survival and polarization toward an phenotype. A notable application of these mechanisms is observed in models of (GvHD), where reverse signaling through tmTNF-α on donor T cells promotes their survival and persistence, contributing to disease . Experimental blockade of tmTNF-α reverse signaling in murine GvHD models reduces T cell expansion and ameliorates symptoms, highlighting its pro-survival role independent of forward signaling pathways.

Physiological Roles

Immune System Modulation

Tumor necrosis factor (TNF) plays a pivotal role in innate immunity by activating macrophages, which are key effectors in the early response to pathogens. Upon binding to TNF receptors on macrophages, TNF-α induces the production of pro-inflammatory cytokines such as interleukin-1 (IL-1) and IL-6, enhances phagocytic activity, and promotes the expression of nitric oxide synthase, thereby amplifying antimicrobial defenses. This activation is crucial for coordinating the initial inflammatory cascade, as evidenced by studies showing that TNF-deficient macrophages exhibit reduced responsiveness to bacterial stimuli. Additionally, TNF facilitates neutrophil recruitment to infection sites by upregulating intercellular adhesion molecule-1 (ICAM-1) on endothelial cells and neutrophils, enabling firm adhesion and transmigration through the vascular wall. In adaptive immunity, TNF contributes to T-cell costimulation, enhancing activation and proliferation of CD4+ T cells during . Through TNFR2 signaling, TNF provides a secondary signal that sustains T-cell responses and promotes effector functions, particularly in Th1 differentiation. For B cells, TNF supports to IgA, an isotype essential for mucosal immunity. This occurs through TNF/iNOS-producing dendritic cells that induce and IL-6, promoting class-switch recombination involving activation-induced cytidine deaminase (). Furthermore, TNF synergizes with IL-1 and IL-6 to drive the acute phase response, stimulating hepatocytes to produce acute phase proteins like and , which bolster systemic defenses against infection. TNF maintains immune homeostasis, as demonstrated in TNF knockout mice, which display impaired granuloma formation and increased susceptibility to mycobacterial infections due to disorganized macrophage aggregation and reduced containment of pathogens. Within cytokine networks, TNF amplifies Th1 responses by enhancing interferon-γ production and skewing T-cell toward pro-inflammatory profiles, while the balance between TNFR1 and TNFR2 signaling fine-tunes these effects to prevent excessive .

Inflammation and Apoptosis

Tumor necrosis factor (TNF), primarily TNF-α, acts as a potent pro-inflammatory by stimulating the expression of adhesion molecules and on endothelial and immune cells, thereby promoting leukocyte recruitment to sites of . Specifically, TNF-α upregulates vascular cell adhesion molecule-1 () on endothelial cells, facilitating the adhesion and transmigration of monocytes and lymphocytes. Similarly, TNF-α induces the production of such as CXCL8 (also known as interleukin-8), which attracts neutrophils to amplify the inflammatory response. In addition to local effects, TNF-α contributes to by inducing fever through the synthesis of (PGE2) in the , where it acts on thermoregulatory neurons to elevate body temperature. TNF-α also triggers apoptosis, a form of , through both extrinsic and intrinsic pathways, enabling the elimination of infected or damaged cells. In the extrinsic pathway, TNF-α binds to TNF receptor 1 (TNFR1), leading to the recruitment of adaptor proteins such as TRADD and to form the death-inducing signaling complex (), which activates and initiates cascade amplification. The intrinsic pathway is engaged when cleaved Bid (tBid) from the extrinsic signal translocates to mitochondria, promoting Bax oligomerization and insertion into the outer mitochondrial membrane, resulting in release and activation of the . The balance between pro-survival and pro-death signals determines whether TNF-α promotes cell survival or , with significant crosstalk between and JNK pathways playing a central role. Activation of by TNF-α induces anti-apoptotic genes that suppress sustained JNK activation, thereby inhibiting and favoring survival; conversely, unchecked JNK signaling enhances mitochondrial dysfunction and death receptor-mediated . This crosstalk is modulated by (ROS), which amplify JNK activity when is inhibited. In physiological contexts, TNF-α-mediated and are beneficial for clearance, as they enhance immune infiltration and eliminate infected s to limit microbial spread. However, dysregulated TNF-α signaling can drive detrimental chronic , contributing to tissue damage in autoimmune and persistent inflammatory conditions through sustained production and unresolved apoptotic events.

Central Nervous System Functions

Tumor necrosis factor (TNF), particularly TNF-α, exerts neuroprotective effects in the (CNS) at physiological or low concentrations, primarily through TNFR2 signaling. Low-dose TNF-α enhances (LTP) in the , a key process for and memory formation, by modulating excitatory synaptic transmission. This enhancement is evident in hippocampal slices where TNF-α potentiates LTP following hypoxic stress, promoting neuronal survival and recovery. Astrocyte-derived TNF-α contributes to these effects by upregulating such as glial-derived neurotrophic factor (GDNF) and (BDNF), which support neuronal viability and synaptic function. Astroglial TNFR2 signaling is essential for astrocyte-neuron communication, regulating hippocampal in a sex-dependent manner and providing against . In , TNF-α drives activation, which can exacerbate CNS pathology in models of autoimmune disorders. In experimental autoimmune encephalomyelitis (EAE), a model for , TNF-α signaling via TNFR2 in modulates immune responses, with microglial TNFR2 promoting anti-inflammatory phenotypes that limit disease progression, while macrophagic TNFR2 has opposing pro-inflammatory effects. Sustained TNF-α production by CNS-infiltrating macrophages increases blood-brain barrier () permeability by disrupting tight junctions, facilitating immune cell infiltration and amplifying neuroinflammatory cascades. Activated release TNF-α, further promoting BBB breakdown and contributing to synaptic dysfunction in inflammatory conditions. During CNS development, TNF-α supports myelination and neuronal organization through TNFR2-mediated pathways. TNF-α promotes the proliferation and maturation of progenitors, essential for sheath formation and axonal insulation. It also influences neuronal and spatial organization in neurogenic zones, such as the , where TNF deficiency leads to accelerated maturation and altered positioning of neurons. TNF mice exhibit disrupted neurogenic zone architecture, with reduced expression of WNT signaling proteins critical for neuronal and . Regarding sensitivity, TNF and TNFR1 null mice display to and stimuli, indicating that basal TNF-α signaling normally suppresses nociceptive responses in the CNS. Conversely, TNFR1 or TNFR2 reduces and following , highlighting TNF's dual role in modulating pathways. Recent post-2020 research links persistent TNF-α elevation to neurofog in , stemming from prolonged storms after infection. Elevated TNF-α contributes to chronic , impairing cognitive function through sustained microglial activation and disruption. Studies show that profiles in patients feature persistent TNF-α alongside IL-6 and IL-1β, correlating with brain fog symptoms via ongoing immune dysregulation in the CNS. This persistence of TNF-α-driven post-infection underscores its role in long-term neurological sequelae.

Clinical Implications

Autoimmune and Inflammatory Diseases

Tumor necrosis factor (TNF) plays a central pathogenic role in several autoimmune and inflammatory diseases by promoting chronic inflammation and tissue damage through its interactions with immune cells and resident tissues. In (RA), TNF is markedly elevated in and tissue, where it drives the production of matrix metalloproteinases (MMPs) by fibroblast-like synoviocytes, leading to degradation and destruction. Studies have shown that synovial levels of TNF correlate positively with disease activity, as measured by the Disease Activity Score 28 (DAS28), indicating its utility as a biomarker for RA severity. In (IBD), particularly , TNF is overexpressed in the intestinal mucosa, where it disrupts epithelial barrier integrity by inducing dysfunction and increasing permeability, thereby exacerbating inflammation and ulceration. Elevated mucosal TNF levels contribute to the recruitment of inflammatory cells and perpetuation of tissue damage. Clinical data indicate that approximately 58% of patients with achieve a primary response to anti-TNF therapies, highlighting TNF's key role in disease , though response rates vary based on baseline production. Anti-TNF treatment has been shown to restore intestinal epithelial barrier function in responsive patients. TNF also contributes to the inflammatory cascade in , where it is produced by and immune cells, amplifying a proinflammatory loop with interleukin-17 (IL-17). Keratinocyte-derived TNF synergizes with IL-17A to upregulate psoriasis-associated genes and cytokines, promoting epidermal hyperproliferation and plaque formation. This axis sustains chronic skin inflammation, with TNF inhibitors effectively targeting the pathway. Recent reviews confirm that TNF biosimilars demonstrate comparable efficacy and safety to originators in psoriasis management, with sustained clinical responses observed in post-2023 studies. Genetic variations further modulate TNF's involvement in these diseases. The -308 G/A polymorphism in the TNF promoter region (rs1800629) is associated with increased TNF production and heightened risk for autoimmune conditions, including , IBD, and , as the A allele enhances transcriptional activity. Additionally, tumor necrosis factor receptor-associated periodic (TRAPS), an autoinflammatory , arises from dominant mutations in TNFRSF1A (encoding TNFR1), leading to dysregulated TNF signaling, recurrent fevers, and independent of binding. These genetic factors underscore TNF pathway dysregulation as a common thread in immune-mediated diseases.

Oncology and Tumor Biology

Tumor necrosis factor (TNF) exhibits a paradoxical role in , initially identified for its capacity to induce tumor but later recognized for promoting tumor progression under certain conditions. Discovered in 1975 for its ability to cause hemorrhagic in Meth A sarcomas in mice, TNF was named for this anti-tumor activity, which involves direct and indirect immune-mediated effects. In tumor , TNF's anti-tumor effects primarily stem from vascular damage, where it selectively disrupts tumor endothelium by upregulating adhesion molecules like and , leading to hemorrhagic and ischemia in sensitive tumors such as sarcomas. Additionally, TNF can trigger in tumor cells via TNFR1 signaling, particularly in early-stage or TNF-sensitive malignancies, by activating cascades through death-inducing signaling complex formation. Clinical exploration of TNF's anti-tumor potential began in the with recombinant human TNF-α (rhTNF-α) administered systemically in phase I and II trials for advanced solid tumors, including sarcomas. These trials demonstrated partial responses in some patients, with objective regressions observed in , , and soft tissue sarcomas, but overall efficacy was limited by severe dose-dependent toxicities such as fever, , and , restricting the maximum tolerated dose to levels insufficient for broad anti-tumor activity. Isolated limb perfusion techniques with rhTNF-α combined with showed higher response rates (up to 70% in sarcomas) by concentrating the agent locally, minimizing systemic exposure, though this approach remains niche due to procedural risks. Conversely, in established tumors, TNF often exerts pro-tumor effects by sustaining chronic that fosters a permissive microenvironment. TNF produced by tumor-associated macrophages (TAMs) drives through induction of (VEGF) expression in endothelial and tumor cells, enhancing vascularization and nutrient supply to support tumor growth. TAM-derived TNF also facilitates by promoting remodeling via matrix metalloproteinases and epithelial-mesenchymal transition in cancer cells, as observed in breast and colorectal cancers. This shift underscores TNF's context-dependent biology, where low-level chronic exposure overrides cytotoxic effects. The dual nature of TNF signaling via its receptors TNFR1 and TNFR2 further explains this paradox. In early-stage cancers, TNFR1 predominates in mediating pro-apoptotic signals through recruitment of and , potentially limiting tumor initiation in TNF-sensitive cells. However, in advanced tumors, TNFR1 often activates pro-survival pathways like to confer resistance to , while TNFR2 exclusively promotes cell survival, proliferation, and migration in tumor cells and immunosuppressive regulatory T cells, exacerbating progression and immune evasion. This receptor imbalance contributes to TNF's net pro-tumorigenic role in late-stage disease. Recent investigations (2024–2025) highlight TNF's integration into strategies for , where its blockade enhances PD-1 inhibitor efficacy. Preclinical models show that TNF inhibition reduces resistance to anti-PD-1 therapy by alleviating immunosuppressive signatures in the , improving T-cell infiltration and response rates. Ongoing clinical trials, such as NCT05867004, are evaluating TNF blockers in with PD-1 inhibitors for advanced , aiming to overcome resistance while mitigating immune-related adverse events like . These approaches build on TNF's historical anti-tumor rationale but leverage its blockade to potentiate modern checkpoint inhibition.

Infectious Diseases and Sepsis

Tumor necrosis factor (TNF) exhibits dual roles in infectious diseases, providing essential protection against certain bacterial pathogens while contributing to pathology in severe systemic infections like . In , TNF is critical for the formation and maintenance of , which encapsulate and prevent bacterial dissemination. Studies in TNF gene-targeted mice demonstrate structural deficiencies in granuloma organization, resulting in increased susceptibility to aerosolized M. tuberculosis infection and higher mortality rates. Similarly, TNF signaling via its receptor TNFR1 is vital for host defense against ; mice deficient in TNFR1 are highly susceptible to this intracellular bacterium, rapidly succumbing to infection due to impaired activation and bacterial clearance. In sepsis and endotoxemia, however, dysregulated TNF production drives harmful . (LPS) from binds (TLR4) on immune cells, initiating the LPS-TLR4 axis that triggers massive TNF release, leading to characterized by , , and high mortality. Experimental models show that excessive TNF in endotoxemia directly mediates vascular leakage and myocardial depression, underscoring its pathological role in acute inflammatory crises. Recent reviews highlight TNF's divergent effects in , where neutralization can mitigate in some contexts but exacerbate infection control in others. TNF also plays a key part in viral infections, particularly through storms that amplify lung injury. In infections, TNF-α exacerbates by promoting excessive and immune cell recruitment, contributing to severe respiratory distress despite its antiviral effects. During the (2020-2025), elevated TNF-α levels in plasma were strongly associated with (ARDS) severity and mortality; meta-analyses confirmed that higher TNF-α concentrations independently predicted poor outcomes in hospitalized patients. In , persistent TNF-driven persists for up to two years post-infection, correlating with ongoing systemic symptoms and immune dysregulation in affected individuals.

Metabolic and Liver Diseases

Tumor necrosis factor (TNF), particularly TNF-α, plays a significant role in the development of , a hallmark of metabolic disorders such as and . In obese individuals, TNF-α expression is elevated in , contributing to systemic insulin resistance by promoting the serine of insulin receptor substrate-1 (IRS-1). This phosphorylation inhibits IRS-1's ability to mediate insulin signaling, thereby impairing and metabolic . Studies in murine models have demonstrated that neutralizing TNF-α in obese animals restores insulin sensitivity, highlighting its causal involvement. In nonalcoholic fatty liver disease (NAFLD), now reclassified as metabolic dysfunction-associated steatotic liver disease (MASLD), TNF-α exacerbates hepatic steatosis and progression to nonalcoholic steatohepatitis (NASH). Hepatocytes exposed to TNF-α exhibit increased lipid accumulation through upregulation of lipogenic pathways and downregulation of fatty acid oxidation, driven by activation of nuclear factor kappa B (NF-κB) signaling. Circulating TNF-α levels are elevated in NAFLD patients, correlating with disease severity and insulin resistance. Recent 2023 studies in diet-induced mouse models of non-obese MASLD have identified TNF-α as a primary trigger of hepatic inflammation, linking it to early steatohepatitis development independent of overt obesity. TNF-α also contributes to liver fibrosis in metabolic contexts by activating hepatic stellate cells (HSCs) primarily through TNFR1 signaling. Upon binding TNFR1, TNF-α induces HSC proliferation, survival, and extracellular matrix production, including collagen and matrix metalloproteinase-9 (MMP-9), which perpetuate fibrotic remodeling. In animal models of chronic liver injury, such as bile duct ligation, TNFR1 knockout mice exhibit significantly reduced HSC activation and liver fibrosis compared to wild-type controls, with decreased expression of profibrotic genes like pro-collagen-α1(I) and TIMP-1. These findings underscore TNF-α's pro-fibrogenic role in advancing MASLD to cirrhosis.

Neurological and Muscular Disorders

Tumor necrosis factor alpha (TNF-α) plays a significant role in neurodegeneration, particularly in (AD), where it is produced by activated surrounding amyloid-beta (Aβ) plaques. Microglial cells, upon recognizing Aβ aggregates, release TNF-α, which exacerbates and contributes to synaptic dysfunction and neuronal loss. Elevated levels of TNF-α have been consistently observed in the (CSF) and brain tissue of AD patients, correlating with disease severity and cognitive decline. This pro-inflammatory promotes the compaction of plaques while impairing clearance mechanisms, shifting microglia from a protective to a detrimental state over time. In major depressive disorder (MDD), TNF-α contributes to pathophysiology by activating the hypothalamic-pituitary-adrenal (HPA) axis, leading to dysregulated cortisol responses and sustained inflammation. Elevated peripheral and central TNF-α levels are associated with MDD symptoms, potentially worsening treatment resistance through interference with neurotransmitter signaling and neuroplasticity. Clinical trials exploring anti-TNF therapies, such as etanercept, have shown preliminary antidepressant effects in treatment-resistant cases, suggesting that blocking TNF-α can normalize HPA axis activity and alleviate depressive symptoms. These findings highlight TNF-α's role in the inflammatory subtype of depression, where cytokine inhibition may offer adjunctive therapeutic benefits. TNF-α is a key mediator of muscle wasting in cancer , driving through activation of the ubiquitin-proteasome pathway in . In this condition, tumor-derived signals induce TNF-α expression, which upregulates E3 ubiquitin ligases like MuRF1 and MAFbx, leading to rapid breakdown of myofibrillar proteins and loss of muscle mass. This catabolic process contributes to , , and reduced survival in cancer patients, with TNF-α inhibition partially reversing in preclinical models. Paradoxically, exercise-induced TNF-α in can exert protective effects by promoting adaptive and rather than degradation. Moderate triggers localized TNF-α release from myocytes, which supports muscle repair and without activating the full ubiquitin-proteasome cascade seen in . This controlled elevation helps mitigate and enhances resilience to subsequent stressors, underscoring TNF-α's context-dependent role in muscular . Emerging research from 2024-2025 implicates TNF-α in (ALS), where it contributes to loss via microglial and astrocytic in the . In ALS models, TNF-α hinders neuroprotective pathways, such as those involving fibroblast growth factor 4, accelerating dysfunction and neuronal degeneration. Additionally, recent studies link elevated TNF-α in salivary and lingual tissues to taste alterations () in inflammatory disorders, including those associated with cancer and neurodegeneration, through disruption of integrity and signaling. These findings suggest TNF-α as a potential therapeutic target for mitigating sensory and motor deficits in these conditions.

Other Clinical Associations

Tumor necrosis factor receptor-associated periodic syndrome (TRAPS) is a rare autosomal dominant autoinflammatory disorder caused by mutations in the TNFRSF1A gene, which encodes the 55-kDa (TNFR1). These mutations lead to misfolding of the TNFR1 protein, resulting in its retention in the and activation of the unfolded protein response, which triggers recurrent episodes of characterized by high-grade fever, , , and , typically lasting 1–3 weeks and occurring every 4–6 weeks. Unlike (FMF), which is driven by gene mutations, TRAPS represents an MEFV-independent form of autoinflammation, highlighting the distinct role of TNFR1 signaling dysregulation in its pathogenesis. In the context of physical activity, tumor necrosis factor (TNF) exhibits dynamic regulation that influences physiological adaptation. Acute bouts of moderate-to-intense exercise transiently elevate circulating TNF levels, promoting responses and facilitating muscle repair and metabolic adaptations through enhanced immune cell recruitment and crosstalk. This short-term rise supports and prevents excessive post-exercise. In contrast, chronic in athletes is associated with persistently lower baseline TNF concentrations, which correlates with reduced and a protective effect against chronic inflammatory conditions. Tumor necrosis factor also modulates taste perception, particularly during inflammatory states, by altering the function and structure of lingual epithelia housing . Elevated TNF levels, as seen in conditions like or acute , contribute to reduced taste bud abundance and impaired renewal of taste cells, leading to diminished sensitivity to flavors—perceived through detection of like glutamate. This effect is evident in patients with acute inflammatory diseases, who report lesser perception of umami in foods such as roasted meats, potentially exacerbating by altering appetite and food preferences. TNF expression within themselves further influences sensory signaling, though its primary impact on umami appears indirect via broader epithelial disruption. Emerging research implicates TNF in cardiovascular and skeletal pathologies beyond traditional inflammatory contexts. In , TNF drives adverse myocardial remodeling by promoting cardiomyocyte , , and extracellular matrix degradation, thereby exacerbating ventricular dilation and dysfunction. Similarly, TNF accelerates loss through direct activation of osteoclasts, enhancing their and resorptive activity independent of receptor activator of nuclear factor kappa-B ligand () in some cases, which contributes to and inflammatory bone erosion. These associations underscore TNF's broader role in tissue remodeling across multiple systems.

Therapeutic Targeting

TNF Inhibitors and Blockers

TNF inhibitors, also known as anti-TNF agents, are biologic drugs designed to neutralize the pro-inflammatory tumor necrosis factor (TNF), thereby mitigating excessive immune responses in various autoimmune and inflammatory conditions. These agents primarily soluble TNF and, to varying degrees, membrane-bound TNF, preventing their binding to TNF receptors (TNFR1 and TNFR2) on surfaces. By blocking TNF signaling, they reduce , joint damage, and tissue destruction, with clinical applications including (RA), , and . Monoclonal antibodies represent a major class of TNF inhibitors. , a chimeric composed of constant regions and murine variable regions, binds with high affinity to both soluble and transmembrane forms of TNF-α, neutralizing its biologic activity and inducing (ADCC) against TNF-expressing cells. , a fully , similarly binds to soluble and membrane-bound TNF-α, inhibiting receptor activation and promoting regulatory T-cell expansion through interactions with transmembrane TNF on monocytes. Both agents are administered via intravenous () or subcutaneous () routes and demonstrate comparable binding avidities to TNF. In contrast, functions as a soluble receptor decoy. This dimeric consists of the extracellular ligand-binding portion of the human TNFR2 fused to the region of human IgG1, which competitively binds soluble TNF-α (and to a lesser extent lymphotoxin-α) with high , preventing its interaction with cell-surface receptors. Unlike the monoclonal antibodies, has reduced binding to transmembrane TNF, limiting certain effector functions such as ADCC or induction in TNF-expressing cells. It is administered subcutaneously and has a shorter compared to the antibodies. The U.S. (FDA) approved the first TNF inhibitors in 1998: in August for moderate-to-severe in combination with methotrexate, followed by etanercept in November for the same indication. received FDA approval in December 2002 for . These approvals marked a pivotal advancement in therapy, with subsequent expansions to other indications like and . In clinical trials, TNF inhibitors achieve American College of Rheumatology 20% improvement (ACR20) response rates of approximately 60-70% in patients after 6-12 months, significantly outperforming and reducing radiographic progression of joint damage. Despite their efficacy, TNF inhibitors carry risks, including reactivation of (TB) due to TNF's role in maintenance and immune containment of mycobacteria. The incidence of TB is elevated 2- to 4-fold with these agents, particularly with monoclonal antibodies like and compared to , prompting FDA-mandated screening for latent TB prior to initiation. Other adverse effects include serious infections, , and demyelinating disorders, necessitating vigilant monitoring. Biosimilars of have expanded access and affordability since 2016. The first infliximab biosimilar, -dyyb (Inflectra), was FDA-approved in April 2016, followed by (Erelzi) and adalimumab biosimilars like -abbm (Cyttezo) in 2016 and 2017, respectively. By 2025, over 20 biosimilars are approved globally across regions like the , , and , with uptake driven by patent expirations and cost reductions of 20-50% compared to originators, improving treatment accessibility in low- and middle-income countries despite regulatory and reimbursement challenges.

TNF-Inducing Agents

TNF-inducing agents represent a class of therapeutics designed to upregulate tumor necrosis factor (TNF) production or delivery primarily for anticancer applications, leveraging TNF's cytotoxic effects on tumor cells while aiming to minimize systemic exposure. Historically, one of the earliest examples is OK-432, a lyophilized preparation derived from Streptococcus pyogenes, which was developed in the 1970s and approved in Japan in 1975 as an immunomodulatory anticancer agent. OK-432 stimulates the release of TNF from immune cells such as macrophages and monocytes, leading to tumor lysis through direct cytotoxicity and enhanced immune activation. In early clinical studies, intratumoral administration of OK-432 at doses of 100 KE to patients with advanced gastric cancer induced detectable TNF activity in sera, as measured by L929 fibroblast cytotoxicity assays, with partial neutralization by anti-TNF monoclonal antibodies confirming the cytokine's involvement. Similarly, intracavitary doses of 10 KE in patients with malignant effusions resulted in TNF elevation in pleural or peritoneal fluids, correlating with reduced tumor burden and improved local control. These findings established OK-432's role in promoting TNF-mediated tumor necrosis, particularly in gastric and head-and-neck cancers, where preoperative intratumoral injections improved 5-year survival rates in randomized trials. In modern , Toll-like receptor (TLR) have emerged as key TNF inducers by activating innate immune pathways in and dendritic cells, thereby boosting local TNF production to enhance antitumor immunity. , a synthetic TLR7 approved by the FDA in 1997 for external , with later approvals in 2004 for and superficial , promotes TNF-α secretion from tumor-associated , which reprograms the immunosuppressive and induces autophagic in cancer cells. This effect is particularly pronounced in skin cancers like , where topical application increases TNF levels, leading to tumor regression via both direct and recruitment of adaptive immune responses. When combined with , such as or , TLR7 like amplify TNF-mediated and reduce tumor recurrence; preclinical models show synergistic efficacy in and cancers, with elevated TNF contributing to enhanced polarization toward an M1 antitumor phenotype. Clinical trials in the 2010s and 2020s have validated this approach, demonstrating improved response rates in solid tumors when TLR are integrated with standard regimens, though optimal dosing remains under investigation to balance efficacy and inflammation. Gene therapy approaches, particularly adenoviral vectors delivering , offer a targeted method for locoregional TNF upregulation in cancer, bypassing the toxicity of systemic recombinant TNF administration. (AdGV.EGR..11D), a replication-deficient adenovirus encoding -α under an Egr-1 promoter responsive to , was evaluated in phase I trials for recurrent head-and-neck cancers. Intratumoral injections combined with chemoradiotherapy resulted in dose-dependent , with biopsies confirming hemorrhagic and viable tumor reduction in 83% of patients, alongside a median survival of 9.6 months. These trials, conducted in the early , highlighted the strategy's ability to achieve high local TNF concentrations—up to 100-fold greater than systemic levels—without significant or organ toxicity, as the vector's localized expression limited spillover. Similar adenoviral constructs have been tested in locoregional settings for sarcomas and melanomas, showing induction via endothelial damage and immune infiltration while maintaining tolerability. Despite these advances, TNF-inducing agents carry risks, primarily dose-limiting (CRS), characterized by fever, hypotension, and multi-organ inflammation due to excessive TNF and secondary cytokines like IL-6. In early OK-432 studies, high doses occasionally triggered transient CRS, necessitating careful monitoring, while adenoviral TNF trials reported grade 3-4 flu-like symptoms in up to 20% of patients at escalating doses. In the , research has shifted toward targeted inducers, such as nanoparticle-encapsulated TLR agonists or tumor-specific promoters in viral vectors, to confine TNF elevation to the tumor site and mitigate CRS incidence below 10% in preclinical models. These innovations aim to harness TNF's antitumor potential—such as vascular disruption and —while addressing safety concerns through precision delivery.

Future Directions in Therapy

Emerging research in TNF-targeted therapies emphasizes selective modulation to minimize adverse effects associated with broad TNF inhibition, such as . TNFR1-specific monoclonal antibodies (mAbs) are under to block the pro-inflammatory signaling of TNFR1 while preserving the protective, functions of TNFR2. These agents show promise in preclinical models of , where TNFR1 drives neuronal damage and microglial activation without the broad immune suppression seen in non-selective TNF blockers. For instance, selective TNFR1 inhibition has demonstrated reduced disease severity in experimental autoimmune encephalomyelitis (EAE), a model for , by attenuating inflammation in the . Nanodelivery systems represent another innovative approach to harness TNF's cytotoxic potential in while mitigating systemic toxicity. TNF-loaded gold nanoparticles conjugated with targeting peptides, such as CALNN, enable site-specific delivery to tumor cells, enhancing in cancer lines like AMJ13 while reducing off-target effects on healthy tissues. These nanoparticles exploit the in tumors, allowing localized TNF release that amplifies antitumor activity without widespread . Preclinical studies indicate that such formulations inhibit tumor more effectively than free TNF, paving the way for safer cancer immunotherapies. Gene editing technologies, particularly CRISPR-Cas9, offer precise strategies for downregulating TNF expression in inflammatory conditions like (IBD). Preclinical studies using CRISPR-Cas9 to target TNF-related pathways in IBD models show potential to reduce pro-inflammatory responses and promote mucosal healing. These approaches target TNF-producing cells directly, potentially providing long-term remission without repeated dosing. Ongoing investigations explore CRISPR interference to modulate TNF receptor pathways, showing efficacy in autoimmune models by selectively inhibiting inflammatory cascades. Personalized medicine in TNF therapy is advancing through biomarker-driven strategies and modeling to predict treatment responses and optimize dosing. Circulating TNF levels and genetic variants in TNFRSF1A serve as biomarkers to identify responders to TNF inhibitors, enabling tailored initiation of therapy in conditions like . algorithms, integrating multi-omics data and clinical parameters, have achieved high accuracy in forecasting anti-TNF efficacy, with models predicting individual responses based on biomarkers from clinical trials. For dosing, pharmacogenomic tools generate individualized plans to maintain therapeutic trough levels, reducing variability and improving outcomes in personalized regimens.