Fact-checked by Grok 2 weeks ago

CD80

CD80, also known as B7-1 or BB1, is a transmembrane glycoprotein encoded by the CD80 gene located on the long arm of human chromosome 3 at position 3q13.3. It belongs to the B7 family of immune co-stimulatory molecules and is primarily expressed on the surface of antigen-presenting cells, including dendritic cells, activated B lymphocytes, and macrophages. CD80 plays a pivotal role in adaptive immunity by delivering a co-stimulatory signal essential for T-cell activation; upon binding to the CD28 receptor on naive T cells, it augments the primary antigen-specific signal from the T-cell receptor (TCR)-major histocompatibility complex (MHC) interaction, thereby promoting T-cell proliferation, differentiation, cytokine production, and survival. Additionally, CD80 can bind to the inhibitory receptor CTLA-4 (CD152) on activated T cells, which competes with CD28 for binding and delivers a negative signal to prevent excessive immune responses and induce peripheral tolerance. Beyond its canonical role in T-cell modulation, CD80 contributes to innate immune responses by activating signaling in macrophages and can indirectly enhance anti-tumor immunity through interactions with . In pathological contexts, aberrant CD80 expression has been implicated in autoimmune diseases, , and cancer, where it serves as a target for immunotherapies such as CD80-Fc fusion proteins designed to boost anti-tumor T-cell responses. As of 2025, CD80-based therapies, including fusion proteins like GI-102 (CD80-IL2v3), are in phase 1/2 clinical trials for advanced solid tumors. The protein's structure features an extracellular domain with two immunoglobulin-like domains (IgV and IgC), a transmembrane region, and a short cytoplasmic tail lacking intrinsic signaling motifs, relying instead on ligand-induced clustering for function.

Molecular Characteristics

Gene

The CD80 gene, with the official symbol CD80 and full name CD80 molecule, is located on the long arm of human at the cytogenetic band 3q13.33, with genomic coordinates NC_000003.12 (119524293..119559614, complement). This gene spans approximately 35 kb and comprises 7 exons, encoding a primary transcript that produces a protein of 288 . of the CD80 pre-mRNA generates multiple isoforms, including membrane-bound forms and soluble variants that lack the , resulting in differences in localization and potential function. The primary protein product of the CD80 gene is a type I transmembrane . The CD80 gene exhibits evolutionary conservation across mammalian species, reflecting its fundamental role in immune regulation, and shares approximately 25% amino acid sequence identity with the closely related gene. Orthologs are present in various mammals, such as mice (Cd80 on ) and rats, with high sequence similarity in key functional domains. Regulation of CD80 gene expression occurs primarily through its promoter region, which is responsive to inducible signals via transcription factors including and PU.1, allowing rapid upregulation in antigen-presenting cells during immune activation. This transcriptional control mechanism ensures that CD80 expression is tightly linked to inflammatory and stimulatory cues, such as exposure.

Protein Structure

CD80 is a 33 kDa glycoprotein consisting of 288 amino acids. The protein features an extracellular domain composed of two immunoglobulin-like subdomains: an N-terminal V-set (IgV-like) domain that facilitates ligand binding and a membrane-proximal C2-set (IgC-like) domain that provides , followed by a 21-amino-acid transmembrane and a 25-amino-acid cytoplasmic tail. The protein predominantly adopts a dimeric through non-covalent hydrophobic interactions involving residues in the β-strands of the IgV domain, although monomeric forms are also observed and display lower ligand-binding . CD80 contains multiple N-linked sites at (Asn) residues, such as Asn-19, Asn-55, and Asn-64 in the extracellular domains, which increase its apparent molecular weight beyond the predicted 33 kDa and enhance conformational stability.90335-2/fulltext) Insights into the three-dimensional structure of CD80 derive from crystallographic studies, including the soluble extracellular domain (PDB ID: 1DR9), which depicts a rigid, elongated homodimer, and complexes with ligands such as CTLA-4 (PDB ID: 1I8L), highlighting the binding interface on the AGFCC′C″ face of the IgV domain for interactions with and CTLA-4.

Expression Patterns

CD80 exhibits constitutive low-level expression on professional antigen-presenting cells (APCs), including dendritic cells, macrophages, and activated B cells, but is absent from resting T cells. This expression pattern extends to inducible upregulation on non-professional APCs, such as endothelial cells and fibroblasts, in response to inflammatory signals. For instance, cytokines like interferon-gamma (IFN-γ) and (TNF-α) induce de novo CD80 surface expression on microvessel endothelial cells. Similarly, the combination of IFN-γ and TNF-α triggers regulated CD80 expression on murine fibroblasts, while ligands promote its upregulation on these cell types during immune activation. In terms of tissue distribution, CD80 is predominantly found in lymphoid organs such as the and lymph nodes, where it localizes to APCs, and at inflamed tissues; under homeostatic conditions, its presence is minimal in non-immune tissues. The molecule's expression is tightly regulated at the transcriptional level by proinflammatory cytokines, enabling rapid surface upregulation within hours following cellular activation to support timely immune responses.

Physiological Functions

Costimulatory Role

CD80, expressed primarily on antigen-presenting cells (APCs) such as dendritic cells, macrophages, and activated B cells, plays a pivotal role in T cell by binding to on naive T cells. This interaction delivers the essential second signal that synergizes with the (TCR) signal 1 triggered by MHC-peptide complexes, preventing T cell anergy and promoting full activation. The CD80- binding, facilitated by the IgV-like domain of CD80, activates downstream signaling pathways including PI3K and , which are critical for T cell survival and effector functions. The costimulatory signal from CD80-CD28 induces robust IL-2 , T cell , and enhanced cell , enabling clonal expansion of antigen-specific T cells. This pathway is indispensable for preventing in activated T cells and ensuring sustained immune responses. Furthermore, CD80 promotes Th1 by favoring the of pro-inflammatory such as IFN-γ and TNF-α, while also driving cytotoxic T lymphocyte (CTL) activation and function through amplified secretion and signals. In , CD80 is essential for formation and B cell help, as CD28 on T follicular helper (Tfh) cells receives costimulatory signals from CD80 on APCs, supporting class switching and affinity maturation. Studies in CD80/CD86 double knockout mice demonstrate that the absence of these ligands leads to profound defects in T cell activation, resulting in anergy, reduced , and of responding T cells, underscoring their non-redundant yet cooperative roles in adaptive immunity.

Regulatory Interactions

CD80 plays a critical role in immune regulation by binding with high affinity to cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), an inhibitory receptor expressed on activated regulatory T cells (Tregs) and effector T cells, with a (Kd) of approximately 0.2 μM. This interaction allows CTLA-4 to outcompete the costimulatory receptor for CD80 binding, thereby dampening T cell activation and preventing excessive immune responses that could lead to . The higher of CTLA-4 for CD80 compared to ensures that CTLA-4 preferentially engages CD80 on antigen-presenting cells (APCs), limiting the duration of costimulatory signals. In the , CD80 forms clusters with and CTLA-4, where CTLA-4 modulates signal duration through transendocytosis, a process in which CTLA-4 captures and internalizes CD80 from the surface into the T cell, leading to lysosomal degradation of the . This cell-extrinsic mechanism depletes CD80 on APCs, reducing the availability of costimulatory ligands for other T cells and thereby attenuating overall immune activation. Additionally, CTLA-4 engagement with CD80 triggers intracellular signaling that promotes of key (TCR) pathway components, such as CD3 and , inhibiting downstream activation of pathways like GRB2-RAS and suppressing T cell proliferation and cytokine production. CD80 also participates in indirect crosstalk with programmed death-ligand 1 (PD-L1) through cis-heterodimerization on the APC surface, which selectively weakens CD80-CTLA-4 interactions while preserving CD80-CD28 binding, thereby fine-tuning the balance between inhibition and activation in the immune checkpoint network. Primarily, however, CTLA-4-mediated effects dominate regulatory outcomes. Through these interactions, CD80 contributes to peripheral tolerance by supporting Treg expansion and the production of immunosuppressive cytokines like IL-10, which further promotes an anti-inflammatory environment and prevents autoreactive T cell responses. This regulatory function of CD80 contrasts with its role in T cell priming via CD28, highlighting its dual capacity to balance immunity and tolerance.

Pathophysiological Roles

In Autoimmune Diseases

Dysregulated CD80 expression on antigen-presenting cells (APCs), including B cells and dendritic cells, contributes to the breakdown of in autoimmune diseases by enhancing excessive T cell costimulation. In (MS), increased CD80 expression on peripheral blood B cells correlates with active disease phases and higher disability scores, promoting proinflammatory T cell responses that exacerbate inflammation. Similarly, in systemic lupus erythematosus (SLE), upregulated CD80 on B cells and monocytes drives hyperactivation of autoreactive T cells, leading to enhanced production and through polyclonal B cell responses. This overexpression disrupts the normal balance of costimulatory signaling required for T cell tolerance, favoring pathogenic effector responses over regulatory mechanisms. Genetic variations in the CD80 gene further heighten susceptibility to autoimmune conditions by altering costimulatory thresholds. Specific polymorphisms in CD80, such as those in the promoter region, have been linked to increased risk and earlier disease onset, likely through enhanced APC-T cell interactions that amplify autoreactive responses. In (RA), CD80 variants contribute to joint inflammation by promoting sustained T cell activation in synovial tissues, while similar polymorphisms associate with progression via intensified beta-cell-targeted . These genetic factors underscore CD80's role in genetically predisposed loss of self-tolerance across diverse autoimmune etiologies. Elevated levels of soluble CD80 (sCD80) in the of SLE patients reflect ongoing immune dysregulation and B cell hyperactivity. sCD80 concentrations are significantly higher in SLE compared to healthy controls, indicating shedding from activated APCs that perpetuates and correlates with markers of B cell activation, such as hypergammaglobulinemia. This soluble form may act as a decoy , yet in excess, it sustains aberrant , linking directly to heightened disease activity and autoantibody-driven pathology in SLE. Preclinical evidence from animal models highlights CD80's pathogenic contribution and potential for . In experimental autoimmune encephalomyelitis (EAE), a model of , blockade of CD80-CD28 interactions reduces disease severity by shifting cytokine profiles toward anti-inflammatory Th2 responses and limiting encephalitogenic T cell infiltration into the CNS. These findings demonstrate that inhibiting CD80 signaling can restore immune balance and attenuate autoimmune-mediated demyelination without broadly suppressing adaptive immunity.

In Cancer

CD80 exhibits paradoxical roles in tumor immunity, where its expression on tumor cells or tumor-associated antigen-presenting cells (APCs) can promote by preferentially engaging CTLA-4 on T cells, leading to T cell exhaustion and reduced antitumor responses. This CTLA-4 dominance occurs because CTLA-4 binds CD80 with higher affinity than the costimulatory receptor , thereby dampening T cell activation and proliferation in the (TME). In cancers such as and , this mechanism contributes to immune evasion, as tumor-expressed CD80 shifts the balance toward inhibitory signaling, enhancing Treg-mediated suppression and limiting cytotoxic T lymphocyte (CTL) infiltration. Low levels of CD80 expression on tumor cells have been associated with poorer prognosis in various malignancies, including , where reduced CD80 correlates with decreased immune activation and higher tumor aggressiveness. In , CD80 expression is notably lower in certain subtypes and is suppressed on tumor-associated macrophages (TAMs) by , contributing to an immunosuppressive TME and worse clinical outcomes. Upregulation of CD80, particularly on tumor-associated APCs, can reverse this by enhancing costimulatory signals; for instance, oncolytic viruses induce CD80 expression on dendritic cells and other APCs, thereby boosting natural killer () cell recruitment and CTL-mediated tumor killing through improved and inflammatory cytokine release. Within the , CD80-positive exhibit dual functionality, promoting both and antitumor inflammation depending on their state. CD80+ M1-like drive proinflammatory responses that can enhance CTL and NK cell activity against tumors, yet in the hypoxic TME, they may also secrete factors like , supporting that aids tumor progression. This ambivalence underscores CD80's context-dependent role in modulating behavior. Recent studies from 2022 onward have linked variations in CD80 expression profiles—analyzed through sequencing—to progression, including altered immune gene networks and increased risk. For example, elevated CD80 in correlates with enriched immune activation pathways but also heightened metastatic potential via interactions with inflammatory and leukocyte migration genes, highlighting its role in shaping tumor heterogeneity.

In Infectious Diseases

CD80, a key costimulatory expressed on antigen-presenting (APCs), plays a critical role in enhancing adaptive immune responses by providing the second signal necessary for T activation via interaction with on T . During infection, CD80 expression is upregulated on certain APC subsets, such as plasmacytoid dendritic , in response to viral TLR ligands, which facilitates + T activation and thereby promotes viral spread by increasing susceptible target . However, this upregulation also supports cytotoxic T lymphocyte (CTL) responses by aiding + T priming against infected . Additionally, elevated levels of soluble CD80 in have been associated with immune dysregulation and correlate with disease progression in patients, potentially reflecting ongoing T exhaustion and viral persistence. In sepsis, excessive CD80 expression on circulating monocytes contributes to hyperinflammation by amplifying proinflammatory cytokine production, such as IL-6 and IL-1β, through NF-κB signaling pathways, leading to exacerbated organ dysfunction. This heightened CD80 also promotes T cell dysfunction by altering costimulatory signals, resulting in impaired adaptive immunity and increased mortality in both human patients and mouse models of polymicrobial sepsis. Studies in CD80-deficient mice demonstrate improved survival and reduced inflammatory responses following cecal ligation and puncture, underscoring CD80's detrimental role in septic hyperinflammation. CD80 exhibits a protective function in bacterial infections, such as those caused by , by enhancing Th1 responses through the production of IFN-γ and IL-2 from CD4+ T cells. In mouse models, blockade of CD80 impairs Listeria-specific secretion and indicates a role for CD80-mediated in Th1 polarization, though it does not affect clearance. Recent research highlights CD80's involvement in , where upregulated CD80 on inflammatory drives excessive T cell activation and contributes to storms via enhanced costimulatory signaling. In severe cases, these CD80-high , often exhibiting a hyperinflammatory , correlate with poor outcomes, and studies suggest that PI3K pathways amplify this CD80-dependent T cell overstimulation, leading to immune exhaustion. Targeting the CD80 axis has been proposed as a strategy to mitigate hyperinflammation in , based on observations of altered expression patterns in infected patients.

Therapeutic Targeting

Immunotherapy Strategies

CD80, a key costimulatory , contributes to cancer immune evasion when its expression is downregulated on tumor cells or antigen-presenting cells (APCs), limiting effective T cell activation. strategies targeting CD80 modulation aim to restore or enhance this costimulatory signal to potentiate antitumor immunity, particularly in solid tumors like and . One established approach involves combining CD80 modulation with , an anti-CTLA-4 that blocks the inhibitory CTLA-4-CD80 interaction, thereby promoting T cell priming and effector function. This relieves CD80-mediated suppression by CTLA-4 on regulatory T cells and exhausted effector T cells, enhancing overall immune activation. In advanced , the combination of ipilimumab with nivolumab demonstrated superior efficacy in the phase 3 CheckMate-067 trial, with a 10-year overall survival rate of 52% compared to 46% for nivolumab monotherapy and 23% for ipilimumab alone, highlighting improved response rates through synergistic checkpoint blockade involving CD80 pathways. CD80-transfected tumor vaccines represent a promising preclinical strategy to augment APC function and T cell priming in solid tumor models. By genetically engineering tumor cells to express CD80, these vaccines mimic professional APCs, delivering both tumor antigens and costimulatory signals to naïve T cells. In mouse models of , transfection of syngeneic tumor cells with CD80 alongside genes efficiently activated antigen-specific + T cells, leading to robust antitumor immunity and tumor regression without systemic toxicity. Similar preclinical studies in models using CD80-overexpressing tumor variants have shown enhanced cytotoxic T lymphocyte responses and improved survival, underscoring CD80's role in priming durable adaptive immunity. Emerging strategies include engineering oncolytic viruses to express or upregulate for localized immune activation within the , particularly in immunologically "cold" tumors like . Oncolytic viruses selectively replicate in tumor cells, lysing them and releasing antigens while modulating immunity; incorporating CD80 expression amplifies this by providing direct costimulation to infiltrating T cells. In , oncolytic treatment has been shown to promote intratumoral T cell infiltration and improve therapeutic outcomes in murine models. CD80 also serves as an adjunct in chimeric antigen receptor (CAR) T cell therapy, where its costimulatory signaling via CD28 enhances CAR-T persistence and antitumor efficacy in solid tumors. CAR-T cells engineered with CD28 domains respond to endogenous or supplemented CD80 ligands, sustaining proliferation and cytokine production in immunosuppressive environments. Preclinical studies indicate that CD80-mediated costimulation integrates with 4-1BB signals to improve CAR-T persistence, leading to better tumor rejection kinetics and long-term memory formation in vivo. This adjunctive role is particularly valuable for solid tumors, where CAR-T exhaustion limits durability, and CD80 provision via vaccine or viral co-delivery could optimize therapeutic persistence.

Inhibitor and Fusion Protein Developments

Recent advancements in molecular engineering have focused on CD80-Fc fusion proteins to enhance antitumor immunity by preferentially activating CD28 costimulation while minimizing CTLA-4-mediated inhibition. Davoceticept (ALPN-202), a tri-specific fusion protein comprising an engineered CD80 variant fused to an IgG Fc domain, binds CD28 with high affinity (KD = 9.9 nM) to promote T cell activation and also antagonizes PD-1/PD-L1 and CTLA-4/CD80-CD86 pathways through interactions with PD-L1 (KD = 505 nM) and CTLA-4 (KD = 1.87 nM). In preclinical models, including immune checkpoint inhibitor (ICI)-resistant B16-F10/hPD-L1 tumors, davoceticept induced significant tumor regression when combined with anti-PD-1 therapy, outperforming monotherapies by enhancing CD8+ T cell infiltration, cytokine production (e.g., IFNγ and IL-2), and natural killer (NK) cell activity. Clinical development of davoceticept was terminated in October 2022 following two patient deaths related to cardiogenic shock in early-phase trials, despite promising preclinical data. Similarly, a recombinant CD80-Fc protein combined with discoidin domain receptor 1 inhibition showed robust T cell activation and tumor growth inhibition in ICI-poor responder models as of 2025. Small-molecule inhibitors targeting the IgV domain of CD80 represent an emerging strategy to dampen excessive in autoimmune conditions like (RA) and (MS). These compounds, identified through and structural optimization, disrupt CD80-CD28 binding with high selectivity, aiming to reduce T cell hyperactivation without broad . Heterocyclic small molecules and engineered monobodies (e.g., CFN13-Fc variants) have demonstrated potent inhibition of CD80 interactions in preclinical assays, offering potential advantages in oral and tissue penetration over biologics like . As of 2025, these inhibitors remain in early per patent analyses, with no phase I trials reported for RA or MS applications, though ongoing optimizations focus on and specificity to advance toward clinical testing. Soluble CD80 decoys have been investigated as a means to sequester CTLA-4 and counteract in chronic infections, leveraging CD80's bidirectional interactions with and CTLA-4. These recombinant soluble forms bind CTLA-4 on exhausted T cells, potentially freeing CD80 ligands for engagement and restoring antiviral responses. In models of chronic viral persistence, soluble CD80 delayed disease progression by modulating T cell function without direct suppression via CTLA-4, suggesting a mechanism that limits inhibitory signaling. Although primarily studied as biomarkers of , therapeutic soluble CD80 constructs show promise in preclinical settings for infections like , where CTLA-4 upregulation contributes to T cell exhaustion.