Fact-checked by Grok 2 weeks ago

PD-L1

Programmed death-ligand 1 (PD-L1), also known as CD274 or B7-H1, is a type I transmembrane protein encoded by the CD274 gene located on chromosome 9p24.1 in humans. As an immune checkpoint ligand, PD-L1 binds to the programmed cell death protein 1 (PD-1) receptor on activated T cells, delivering inhibitory signals that suppress T cell proliferation, cytokine production, and cytotoxic activity, thereby promoting immune tolerance and preventing excessive immune responses. This interaction is essential for maintaining immune homeostasis in normal tissues, where PD-L1 is expressed at low levels in organs such as the placenta, heart, lungs, and skeletal muscle to regulate self-tolerance and limit autoimmunity. In the context of , PD-L1 plays a critical role in modulating adaptive immune responses, particularly by inducing T cell exhaustion and anergy in chronic infections or inflammatory conditions. Its expression can be upregulated on antigen-presenting cells, endothelial cells, and other non-immune cells in response to inflammatory cytokines like interferon-gamma (IFN-γ), which activates signaling pathways such as JAK-STAT to enhance PD-L1 transcription. Beyond its canonical role, PD-L1 exhibits diverse functions, including tumor-intrinsic signaling that promotes cancer cell survival and proliferation through pathways like and RAS-MAPK, independent of its immune inhibitory effects. In cancer, PD-L1 overexpression is a hallmark of immune evasion, frequently observed in solid tumors such as non-small cell lung cancer (NSCLC), , , and hematological malignancies like (AML). Tumor cells and stromal cells in the upregulate PD-L1 to interact with PD-1 on , leading to suppressed antitumor immunity and poorer clinical outcomes, including and therapy resistance. This aberrant expression is driven by oncogenic signaling (e.g., mutations), epigenetic modifications, and microenvironmental factors, correlating with advanced disease stages in various cancers. Therapeutically, PD-L1 has become a pivotal target in , with blocking antibodies such as , , and approved for treating multiple malignancies by restoring T cell function and enhancing immune-mediated tumor clearance. PD-L1 expression levels serve as a key for predicting response to these inhibitors, though challenges like acquired resistance and heterogeneous expression necessitate combination strategies with , radiotherapy, or other targeted agents. Ongoing research explores soluble and exosomal forms of PD-L1, as well as small-molecule inhibitors, to broaden therapeutic applications beyond into autoimmune and infectious diseases.

Discovery and History

Initial Identification

PD-L1, also known as B7-H1 or CD274, was first identified in 1999 as a novel member of the B7 family of costimulatory molecules. Researchers led by Lieping Chen cloned the human B7-H1 gene through database searches for B7 homologs and subsequent isolation of full-length cDNA, revealing a type I transmembrane glycoprotein with structural similarity to B7-1 and B7-2. expression studies demonstrated that B7-H1 is constitutively present on antigen-presenting cells, including dendritic cells and monocytes, as well as in non-lymphoid tissues such as the heart and lung, suggesting a role in immune regulation. In 2000, Gordon J. Freeman and colleagues independently characterized B7-H1 as programmed death-ligand 1 (PD-L1), the primary ligand for the immunoinhibitory receptor PD-1, by screening (EST) databases and cloning full-length cDNA from placenta and murine activated T cell libraries. They confirmed its identity with B7-H1 through , showing 70% identity between and murine forms, and used to demonstrate specific binding of PD-1-Ig fusion proteins to PD-L1-transfected cells, with no interaction from CD28-Ig, CTLA-4-Ig, or ICOS-Ig. Expression analysis via further revealed PD-L1 on IFN-γ-stimulated monocytes and activated dendritic cells from both species, highlighting its presence on key antigen-presenting cells. Early functional studies established PD-L1's inhibitory role in T-cell responses. Freeman et al. employed co-stimulation assays where immobilized PD-L1-Ig fusion proteins dose-dependently suppressed T-cell receptor-mediated of purified human + and + T cells, reducing by up to 69% and IFN-γ secretion by 80% when co-ligated with anti-CD3 antibodies. Similar assays with murine splenocytes confirmed inhibition of IL-2 production and , underscoring PD-L1's capacity to negatively regulate through PD-1 engagement. These findings shifted the initial view of B7-H1 from a potential costimulator—based on prior assays showing IL-10 induction—to a key molecule.

Key Research Milestones

In the early 2000s, the nomenclature for PD-L1 evolved significantly following its initial identification. Originally cloned and named B7 homolog 1 (B7-H1) in 1999 due to its structural similarity to B7 family members, it was redesignated as programmed death-ligand 1 (PD-L1) after the 2000 discovery of its interaction with PD-1, with the official gene symbol CD274 assigned by the . A pivotal advancement came in 2002 when et al. demonstrated that PD-L1 binding to PD-1 on T cells directly inhibits T-cell , , and production, establishing the pathway's role in immune and laying the foundation for its study in pathological contexts. Throughout the , research illuminated PD-L1's central role in tumor immune evasion, with key studies showing its overexpression on cancer cells suppresses antitumor T-cell responses and correlates with poor prognosis across multiple malignancies. For instance, a 2012 review in Reviews Cancer synthesized evidence from tumor analyses indicating that PD-L1 expression often predicts adverse outcomes by enabling immune escape, influencing subsequent prognostic biomarker development. The pathway's therapeutic potential gained global recognition in 2018 when the Nobel Prize in Physiology or Medicine was awarded to Tasuku Honjo and James P. Allison for their discoveries of negative immune regulation mechanisms, including the PD-1/PD-L1 axis, which revolutionized cancer immunotherapy by inspiring checkpoint inhibitors. From 2023 to 2025, clinical translation accelerated with several FDA approvals enhancing PD-L1-targeted approaches in non-small cell lung cancer (NSCLC). In 2023, the agency approved expanded use of PD-L1 immunohistochemistry (IHC) companion diagnostics, such as the VENTANA PD-L1 (SP263) Assay, to guide cemiplimab therapy selection based on tumor PD-L1 expression levels. Subsequent approvals included subcutaneous formulations like atezolizumab (Tecentriq Hybreza) in September 2024 for advanced NSCLC across prior indications. By 2025, further approvals encompassed pembrolizumab subcutaneous injection (Keytruda Qlex) in September 2025 for NSCLC and other solid tumors.

Molecular Structure and Expression

Gene Organization

The PD-L1 gene, officially known as CD274, is located on the short arm of human chromosome 9 at position 9p24.1, spanning approximately 20 kb from nucleotide 5,450,542 to 5,470,554 (GRCh38.p14 assembly). This locus is notable for its frequent amplification in various cancers, contributing to immune evasion mechanisms. The gene consists of seven exons that encode the primary CD274 transcript (NM_014143.3), which produces a 3.6 kb mRNA translating to a 290-amino-acid protein. Exon 1 contains the 5' untranslated region (UTR), exon 2 includes the start codon and signal peptide, exons 3 and 4 encode the extracellular immunoglobulin-like domains, exon 5 the transmembrane domain, exon 6 part of the cytoplasmic tail, and exon 7 the remainder of the cytoplasmic domain plus the 3' UTR. Alternative splicing generates multiple isoforms, including a shorter variant lacking exon 2 (PD-L1Δex2), which results in a secreted form lacking the signal peptide and potentially altering immune regulatory functions. At least four transcript variants have been identified, with three being protein-coding. The promoter region of CD274 features a CpG island and includes two putative interferon-stimulated response elements (ISREs), which facilitate responsiveness to signaling, alongside binding sites. These elements contribute to the gene's inducibility under inflammatory conditions. Basal expression of PD-L1 is low in resting T cells and B cells but higher in professional antigen-presenting cells such as macrophages and dendritic cells, reflecting its role in . In non-immune s, notable expression occurs in the , particularly on trophoblastic cells, where it supports maternal-fetal . Overall tissue profiling shows highest levels in the (RPKM ~4.5) and appendix (RPKM ~6.7), with broad but variable distribution across hematopoietic and epithelial s.

Protein Structure and Domains

PD-L1, also known as CD274 or B7-H1, is a type I transmembrane glycoprotein encoded by the CD274 gene, with the precursor protein consisting of 290 amino acids and an unglycosylated molecular weight of approximately 33 kDa. The mature protein, following cleavage of the N-terminal signal peptide (residues 1-18), spans residues 19-290 and comprises three main structural domains: an extracellular domain, a transmembrane helix, and a short cytoplasmic tail. The extracellular domain (residues 19-238) is responsible for ligand interactions and features two immunoglobulin-like subdomains: an N-terminal IgV-like domain (residues 19-127) critical for binding to PD-1 and a C-terminal IgC-like or C2-type domain (residues 133-225) that acts as a structural spacer. The transmembrane domain forms a hydrophobic helix (residues 239-259) that anchors PD-L1 in the cell membrane, while the cytoplasmic tail (residues 260-290) is a short, 31-amino-acid sequence lacking canonical immunoreceptor tyrosine-based inhibitory (ITIM) or activation (ITAM) motifs, limiting its direct involvement in intracellular signaling. PD-L1 undergoes extensive post-translational modifications, particularly N-linked at four conserved sites in the extracellular domain: residues 35, 192, 200, and 219. These events add significant mass (increasing the apparent molecular weight to ~45-55 kDa on ) and play key roles in , stability, trafficking to the cell surface, and to proteasomal . For instance, at N192, N200, and N219 predominantly features complex glycans, including poly-LacNAc structures, which enhance PD-L1's immunosuppressive function by promoting its membrane localization and dimerization. Disruption of these sites, such as through , leads to reduced surface expression and impaired stability, underscoring their functional importance. Crystal structures of the PD-L1 extracellular domain, determined in the , have provided atomic-level insights into its architecture and potential for homodimerization. Early structures, such as the 2010 dimeric PD-L1 (PDB: 3FN3), revealed a symmetric homodimer mediated by hydrophobic interactions between the IgV-like domains, suggesting that dimerization may regulate accessibility or stability in the absence of PD-1. Subsequent high-resolution complexes, including those with small-molecule inhibitors (e.g., PDB: 5N2F, 2017; PDB: 5O4Y, 2017), confirmed the dimer's prevalence and showed how binding or influences the conformational dynamics at the dimer , potentially modulating PD-1 engagement. These structural data highlight PD-L1's adaptability, with the IgV-like domain's front β-sheet serving as the primary PD-1 while the dimer exposes complementary surfaces.

Ligand-Receptor Interactions

Binding to PD-1

PD-L1 primarily binds to its receptor PD-1 through the extracellular immunoglobulin variable-like (IgV) domains of both proteins, forming a stable complex that inhibits T cell activation. The binding interface is dominated by hydrophobic interactions between the C′CFG β-strands, creating a flat, extensive contact surface with a buried area of approximately 1,970 Ų. Key residues contributing to this hydrophobic core include Ile54, Tyr56, Met115, Ala121, and Tyr123 on PD-L1, which engage Val64, Ile126, Leu128, Ala132, and Ile134 on PD-1; for instance, Tyr123 of PD-L1 forms an alkyl-π interaction with Ile134 of PD-1. Polar contacts, such as hydrogen bonds between Tyr68 of PD-1 and Asp122 of PD-L1, and a between Glu136 of PD-1 and Arg113 of PD-L1, further stabilize the interface. The structural basis of this interaction was elucidated by of the human PD-1/PD-L1 complex at 2.45 , revealing a roughly orthogonal (90-degree) at the interface that mimics the pairing of variable domains in antibody-antigen recognition. The complex adopts a 1:1 in solution and in the , contrasting with a 2:1 (PD-1:PD-L1) arrangement observed in some murine structures. This geometry supports both cis engagement (PD-1 and PD-L1 on the same cell surface) and trans engagement (between adjacent cells), enabling context-dependent immune modulation. The binding affinity is moderate, with a dissociation constant (Kd) of approximately 600 nM, as determined by measurements. In physiological settings, surface-expressed PD-L1 on antigen-presenting cells (APCs) and tumor cells engages PD-1 on activated T cells, particularly in , to dampen T cell responses and maintain ; cis interactions on APCs can sequester PD-L1, limiting its availability for trans inhibition.

Binding to Other Partners

PD-L1 interacts with (also known as B7-1), a costimulatory molecule expressed on antigen-presenting cells and activated T cells, with a moderate binding affinity of approximately 1.4 μM as determined by analysis. This interaction competes with the binding of to and CTLA-4, thereby modulating T cell activation and contributing to immune regulation. The PD-L1–CD80 binding was first demonstrated in functional assays showing inhibition of T cell responses, with initial identification through co-immunoprecipitation and binding studies in murine systems in 2007, followed by confirmation in human cells. Soluble forms of PD-L1, produced via alternative mRNA splicing or ectodomain shedding, retain the capacity to bind PD-1 with affinities comparable to membrane-bound PD-L1 (Kd in the nanomolar to low micromolar range). These circulating soluble PD-L1 molecules can engage PD-1 on distant T cells, promoting systemic by dampening immune responses beyond local tumor microenvironments. Elevated levels of soluble PD-L1 have been detected in from cancer patients, correlating with disease progression and reduced efficacy of PD-1 blockade therapies. The on PD-L1 for binding overlaps significantly with the PD-1 interaction site but includes distinct residues that allow for selective competition, as revealed by crystallographic structures and mutational analyses. For instance, key residues in the IgV-like domain of PD-L1, such as those in the loop, contribute to both interactions but confer differential affinities, enabling to sterically hinder PD-1 engagement in configurations on the same cell surface. This structural distinction underscores the role of PD-L1 in fine-tuning costimulatory and inhibitory signals within immune synapses.

Signaling Pathways

Effects on T-Cell Signaling

Upon engagement with PD-L1, PD-1 on T cells recruits the protein phosphatases SHP-1 and SHP-2 to its cytoplasmic immunoreceptor -based inhibitory motif (ITIM) and immunoreceptor -based switch motif (ITSM). This recruitment occurs following phosphorylation of the ITIM ( 223) and ITSM ( 248) residues by Src family kinases during T-cell receptor (TCR) activation. The recruited SHP-1 and SHP-2 then dephosphorylate key proximal signaling components of the TCR complex, including the CD3ζ chain and the kinase ZAP-70, thereby dampening early TCR . Downstream of this proximal inhibition, PD-1 signaling selectively suppresses the PI3K-Akt and Ras-MAPK pathways in activated T cells. The blockade of PI3K-Akt reduces activation of transcription factors such as and NFAT, leading to diminished production of interleukin-2 (IL-2) and impaired T-cell proliferation. Similarly, inhibition of the Ras-MAPK pathway limits extracellular signal-regulated kinase (ERK) phosphorylation, further contributing to arrest by upregulating cyclin-dependent kinase inhibitors like p15 and p27 while suppressing Cdc25A expression. These effects collectively attenuate effector functions, with PD-L1 ligation resulting in up to a 70% reduction in interferon-γ (IFN-γ) secretion by PD-1-expressing T cells. In chronic stimulation environments, such as tumors, sustained PD-1/PD-L1 interactions promote T-cell exhaustion by inducing expression of additional inhibitory receptors, including TIM-3 and LAG-3. This exhaustion is characterized by progressive loss of proliferative capacity and , reinforcing immune suppression through compounded co-inhibitory signaling.

Effects on Antigen-Presenting Cells

PD-1 is expressed on the surface of antigen-presenting cells (APCs), including macrophages and dendritic cells, enabling autocrine and when engaged by PD-L1 expressed on the same or adjacent cells. This interaction activates inhibitory signaling cascades within APCs, promoting the of the immunosuppressive IL-10 while suppressing the expression of pro-inflammatory such as TNF-α and IL-6. Such modulation shifts APCs toward a tolerogenic , limiting excessive and supporting immune . PD-L1 expression on APCs, through interactions with PD-1 on responding T cells, further enhances the differentiation of regulatory T cells (Tregs) by activating pathways like signaling, which upregulates tolerogenic factors in APCs. This APC-mediated mechanism fosters an environment conducive to Treg expansion and suppressive function, thereby amplifying peripheral . Engagement of PD-L1 on dendritic cells triggers intracellular reverse signaling that induces metabolic reprogramming, promoting a semi-mature, tolerogenic state in dendritic cells, reducing their capacity to prime pro-inflammatory responses.

Regulation of Expression

Transcriptional Regulation

The transcriptional regulation of PD-L1 (encoded by the CD274 gene) is primarily driven by inducible pathways responsive to inflammatory and environmental cues, enabling rapid adaptation in immune and tumor contexts. The interferon-γ (IFN-γ)/JAK-STAT1 pathway serves as a central mechanism for PD-L1 upregulation. IFN-γ binding to its receptor activates Janus kinase 1 (JAK1) and JAK2, leading to phosphorylation and nuclear translocation of signal transducer and activator of transcription 1 (STAT1), which forms a complex with STAT2 and interferon regulatory factor 9 (IRF9) to bind interferon-stimulated response elements (ISREs) in the CD274 promoter. This results in rapid transcriptional activation, with PD-L1 mRNA levels increasing significantly—often by 10- to 100-fold—within hours of IFN-γ exposure in various cell types, including melanoma and ovarian cancer cells. Other cytokines contribute to PD-L1 transcription through distinct transcription factors. Tumor necrosis factor-α (TNF-α), often secreted by tumor-associated macrophages, activates the pathway by promoting -mediated and nuclear entry of the p65 subunit, which directly binds the CD274 promoter to enhance expression. This mechanism links chronic inflammation to PD-L1 induction in cancers such as and ovarian tumors. Similarly, in tumor cells harboring mutations or amplifications—common in non-small cell —EGFR signaling activates via the interleukin-6 (IL-6)/JAK pathway, where phosphorylated STAT3 binds the CD274 promoter to drive PD-L1 transcription, promoting immune evasion. In hypoxic tumor microenvironments, hypoxia-inducible factor-1α (HIF-1α) plays a key role by stabilizing under low oxygen conditions and binding hypoxia response elements (HREs) in the CD274 proximal promoter, thereby upregulating PD-L1 transcription on myeloid-derived suppressor cells and tumor cells to suppress T-cell activation. This pathway is particularly relevant in solid tumors where hypoxia fosters immune tolerance. Interferon regulatory factor 1 (IRF1), a pivotal transcription factor downstream of IFN-γ signaling, further amplifies PD-L1 expression by directly binding ISREs (also termed IRF response elements) in the promoter, with studies in hepatocellular carcinoma cells confirming dose-dependent activation and functional IRE sites essential for this regulation. IRF1's activator role contrasts with IRF2's competitive repression, fine-tuning PD-L1 levels in response to interferons.

Post-Transcriptional and Epigenetic Regulation

Post-transcriptional regulation of PD-L1 primarily occurs through microRNAs (miRNAs) that bind to its mRNA, modulating stability and . miR-513 directly targets the 3' (3'UTR) of PD-L1 mRNA, suppressing its in IFN-γ-stimulated cells such as biliary epithelial cells and cholangiocytes. Similarly, miR-570 binds the PD-L1 3'UTR to inhibit expression, contributing to reduced PD-L1 levels in tumor cells. These miRNAs establish a feedback loop to fine-tune PD-L1 in immune responses. The miR-200 family also downregulates PD-L1 by targeting its 3'UTR, particularly in cancers where miR-200 expression is diminished, leading to elevated PD-L1 and enhanced immune evasion. In non-small cell lung cancer, miR-34a reduces PD-L1 expression by direct 3'UTR binding, linking p53-mediated tumor suppression to control; 2015 studies confirmed this axis in patient samples and models, showing miR-34a overexpression decreases PD-L1 protein and mRNA levels. Epigenetic modifications provide long-term control of PD-L1 expression. at CpG islands and shores in the PD-L1 promoter inversely correlates with its expression, silencing the gene in some tumors while hypomethylation promotes upregulation in others like colorectal and . acetylation, particularly at H3K27 on the promoter, enhances PD-L1 transcription; HDAC inhibitors increase this acetylation, boosting expression in and other malignancies by dissociating repressive complexes like DNMT3A-HDAC1. Pan-HDAC inhibitors further promote acetylation at the PD-L1 locus, leading to sustained gene activation. At the protein level, and ubiquitination regulate PD-L1 stability and half-life. N-linked on PD-L1 extracellular domains prevents GSK3β phosphorylation, thereby inhibiting recruitment of the E3 ubiquitin ligase β-TrCP and subsequent proteasomal degradation. β-TrCP-mediated ubiquitination targets unglycosylated PD-L1 for lysosomal and proteasomal breakdown, reducing surface expression in tumor cells. This interplay ensures dynamic control of PD-L1 availability during immune interactions.

Physiological Roles

Role in Immune Tolerance

PD-L1 plays a crucial role in maintaining by being expressed on various non-lymphoid tissues, where it inhibits autoreactive T cells through interaction with PD-1. In tissues such as and cardiac myocytes, constitutive PD-L1 expression limits the activation and infiltration of self-reactive T cells, thereby protecting these organs from autoimmune attack under steady-state conditions. This localized inhibitory signaling helps preserve self-tolerance by dampening effector T cell responses without broadly suppressing immunity. In mechanisms, PD-L1 expression on antigen-presenting cells and endothelial cells in lymph nodes and mucosal surfaces further restricts excessive T cell activation. By engaging PD-1 on effector T cells, PD-L1 attenuates their proliferation and production, preventing overzealous responses to self-antigens and promoting function in these sites. This contributes to the overall balance that avoids while allowing appropriate immune surveillance. A prominent example of PD-L1's tolerogenic function occurs during , where it is upregulated on cells at the feto-maternal . PD-L1 on syncytiotrophoblasts and extravillous s interacts with PD-1 on maternal regulatory T cells, inducing their expansion and suppressive activity to foster fetal-maternal and prevent rejection of the semi-allogeneic . This pathway ensures immune throughout . Knockout studies in PD-L1-deficient mice reveal its essential role in , as these animals display enhanced T cell responses, increased production, and heightened susceptibility to induced autoimmune conditions, indicative of mild dysregulated immunity akin to early lupus-like features. Such phenotypes underscore PD-L1's protective function against spontaneous autoimmunity in peripheral tissues.

Role in Pathogen Defense

PD-L1 plays a crucial role in defense by modulating immune responses during active infections, ensuring effective control of while mitigating the risk of excessive and . Upregulated on various types, including antigen-presenting cells and infected cells, PD-L1 interacts with PD-1 on T cells to dampen their , , and production, thereby terminating prolonged responses in chronic settings and preventing . This regulatory function is particularly evident in viral infections, where PD-L1 helps balance antiviral immunity against potential harm. In viral infections such as lymphocytic choriomeningitis virus (LCMV) and human immunodeficiency virus (HIV), PD-L1 expression is induced on infected cells and immune cells to limit chronic T-cell responses. During the late phase of acute LCMV infection, PD-L1 is strongly upregulated, inhibiting terminal differentiation and effector functions of CD8+ T cells to avert immunopathology from overzealous responses. In chronic LCMV models, sustained PD-L1 signaling contributes to T-cell exhaustion, allowing viral persistence but protecting against lethal inflammation; experimental blockade of PD-1/PD-L1 enhances viral clearance yet increases tissue damage in PD-1-deficient mice, underscoring its protective role against immunopathology. Similarly, in HIV infection, PD-L1 on virus-infected cells and antigen-presenting cells promotes CD8+ T-cell exhaustion, correlating with viral latency and reduced immune-mediated damage, though this also hinders effective clearance. For hepatitis C virus (HCV), PD-L1 regulation is essential for resolving acute infections by transiently limiting T-cell hyperactivity to prevent liver damage, but in chronic cases, persistent upregulation drives exhaustion and viral persistence. In bacterial infections, PD-L1 on macrophages modulates Th1 responses to curb excessive that could lead to sepsis-like conditions. For instance, during infection, PD-L1 expression on macrophages and dendritic cells supports initial Th1 priming for bacterial clearance but also dampens overactivation to avoid ; blockade studies reveal reduced T-cell expansion and impaired protective immunity, highlighting its balanced role in defense.

Clinical Significance

Applications in Cancer

PD-L1 is frequently upregulated on tumor cells in response to interferon-gamma (IFN-γ) secreted by activated T cells within the , enabling cancer cells to evade immune detection by inhibiting T-cell activity. This adaptive expression mechanism contributes to immune escape and is associated with poor clinical outcomes, as high PD-L1 levels correlate with reduced survival in a substantial proportion of solid tumors, where expression is observed in 30-50% of cases depending on the cancer type and detection threshold. In diagnostics, PD-L1 serves as a key predictive biomarker for inhibitor therapy, with (IHC) assays quantifying expression to guide patient selection. Common scoring systems include the tumor proportion score (), which measures PD-L1 positivity on tumor cells (e.g., ≥1% or ≥50% for non-small cell [NSCLC]), and the combined positive score (), which incorporates immune cells (e.g., ≥1 for certain indications like head and neck ). These thresholds are used to identify responders in NSCLC and , where PD-L1-positive patients show higher response rates to anti-PD-L1 therapies; recent 2024 guidelines emphasize standardized PD-L1 testing to refine selection, particularly in heterogeneous tumors. Therapeutically, PD-L1 blockade with monoclonal antibodies has transformed cancer treatment, notably atezolizumab (approved by the FDA in 2016 for advanced NSCLC based on the OAK trial) and durvalumab (approved for NSCLC and urothelial carcinoma). These agents disrupt PD-L1/PD-1 interactions, reinvigorating T-cell responses; when combined with chemotherapy, they achieve objective response rates of 20-40% in frontline settings for NSCLC and small cell lung cancer, outperforming chemotherapy alone in progression-free and overall survival. As of 2025, phase III trials for bispecific PD-L1/VEGF antibodies like HB0025 and IMM2510 are advancing, showing promising efficacy in refractory solid tumors such as endometrial cancer and NSCLC. Additionally, subcutaneous formulations of PD-1 inhibitors, such as pembrolizumab, were approved by the FDA in September 2025 for multiple indications, enhancing administration convenience. Despite these advances, resistance to PD-L1 inhibitors remains a challenge, often driven by loss of class I (MHC-I) expression on tumor cells, which impairs to T cells, or mutations in JAK1, which abrogate IFN-γ signaling and downstream PD-L1 induction. To address , ongoing 2025 clinical trials are evaluating bispecific antibodies that simultaneously target PD-L1 and other pathways, such as VEGF (e.g., HB0025 and IMM2510) or OX40, showing promising early in refractory solid tumors like and NSCLC.

Applications in Autoimmunity and Infection

In autoimmune diseases, enhancing PD-L1 signaling through agonists or mimetics has emerged as a therapeutic strategy to restore by suppressing autoreactive T cells. Recombinant PD-L1 Fc-fusion proteins, which mimic natural ligand-receptor interactions, have shown promise in preclinical models of ; for instance, administration of PD-L1 Ig fusion protein reversed in a model by inhibiting autoreactive + T cells and promoting activity. Similarly, in experimental cerebral , PD-L1 fusion protein treatment reduced T cell-mediated and improved survival by dampening excessive inflammation. Clinical translation includes PD-1 agonists, which activate the PD-1/PD-L1 pathway, such as peresolimab in ; in a phase 2 trial, peresolimab treatment led to an ACR20 response in 67.5% of patients versus 29.4% on , indicating substantial reduction in disease activity and flares. As of 2025, emerging PD-1 agonists like rosnilimab are in phase 2b trials for . Other PD-1 agonists like BMS-986019 and AMP-224 are under investigation in phase II trials for autoimmune conditions, including , with preliminary data suggesting modulation of pathogenic T cell subsets. In models, PD-L1 overexpression has demonstrated protective effects on pancreatic s by inhibiting autoreactive T cell infiltration and destruction. Genetic engineering of hematopoietic stem/progenitor cells to overexpress PD-L1 reversed new-onset diabetes in non-obese diabetic mice by restoring tolerance and suppressing insulitis, highlighting potential for approaches. Pharmacological restoration of PD-L1 similarly halted loss and maintained euglycemia in preclinical studies, underscoring its role in preventing autoimmune progression. For systemic lupus erythematosus, PD-1 agonists are under investigation in early-phase trials, targeting hyperactive T follicular helper cells to reduce production and disease flares. In infectious diseases, PD-L1 modulation strategies differ by context, with blockade often used to reinvigorate exhausted T cells in chronic infections, while enhancement via agonists may temper acute hyperinflammation. For chronic viral infections such as (HBV), PD-1/PD-L1 blockade reverses T cell exhaustion, restoring antiviral effector functions and reducing viral loads in preclinical and early clinical studies; in HBV-infected patients, anti-PD-L1 therapy enhanced intrahepatic HBV-specific T cell responses and promoted HBeAg . This approach has informed trials combining PD-1 inhibitors with antivirals to improve outcomes in chronic HBV. In , where early storms drive hyperinflammation followed by late , PD-L1 agonists could theoretically curb excessive immune activation, though most evidence supports timed blockade to counteract exhaustion. Preclinical models indicate that PD-L1 upregulation during promotes tolerance to limit tissue damage, but therapeutic agonists remain exploratory; conversely, PD-L1 blockade in late-stage models improved survival by reversing dysfunction and , emphasizing the need for stage-specific interventions. In bacterial infection models like , PD-L1 knockout enhanced T cell-mediated clearance and survival by boosting innate and adaptive responses, but this came at the cost of increased risk due to unchecked effector activity, mirroring PD-1 pathway deficiencies. These findings highlight PD-L1's dual role in balancing pathogen defense against self-tolerance, informing targeted therapies.

References

  1. [1]
    PD-L1 importance in malignancies comprehensive insights into the ...
    Apr 3, 2025 · Programmed cell death ligand-1 (PD-L1), also called B7-H1 or CD274, functions as an immune checkpoint ligand, naturally present in tissues ...
  2. [2]
    PD-L1 is a diverse molecule regulating both tumor-intrinsic signaling ...
    PD-L1 serves as an 'immune checkpoint' and it binds to receptor programmed death-1 (PD-1) to regulate immune responses among antigen presenting cells and T- ...Pd-L1 Is A Diverse Molecule... · Pd-L1 Expression In Cell... · Role Of Autophagy<|control11|><|separator|>
  3. [3]
    B7-H1, a third member of the B7 family, co-stimulates T ... - PubMed
    Ligation of B7-H1 co-stimulated T-cell responses to polyclonal stimuli and allogeneic antigens, and preferentially stimulated the production of interleukin-10.
  4. [4]
    Engagement of the PD-1 immunoinhibitory receptor by a novel B7 ...
    Engagement of PD-1 by PD-L1 leads to the inhibition of T cell receptor-mediated lymphocyte proliferation and cytokine secretion.Missing: 1999 discovery H1
  5. [5]
    Engagement of the Pd-1 Immunoinhibitory Receptor by a Novel B7 ...
    Human PD-L1 with 290 residues is identical to B7-H1, which was reported to have T cell stimulation activities 16. Murine PD-L1 cDNA encodes a polypeptide ...
  6. [6]
    PD-1, PD-L1 (B7-H1) and Tumor-Site Immune Modulation Therapy
    Jan 25, 2017 · In 1999, Chen cloned the first B7 homolog, the human B7-H1 gene, and identified its inhibitory activity on T cells by inducing IL-10 [47]. ...
  7. [7]
    PD-L1 (B7-H1) and PD-1 Pathway Blockade for Cancer Therapy
    PD-1 has been subsequently identified as a counter-receptor for B7-H1 (18), and B7-H1 is therefore also known as PD-L1 to emphasize this receptor–ligand ...Missing: history nomenclature
  8. [8]
    The blockade of immune checkpoints in cancer immunotherapy
    Mar 22, 2012 · Since that report, analyses of various tumours have suggested that the PDL1 status can either correlate with poor prognosis, better prognosis ...
  9. [9]
    The 2018 Nobel Prize in Physiology or Medicine - Press release
    Oct 1, 2018 · In 1992, a few years before Allison's discovery, Tasuku Honjo discovered PD-1, another protein expressed on the surface of T-cells.
  10. [10]
    Oncology (Cancer)/Hematologic Malignancies Approval Notifications
    On September 19, 2025, the Food and Drug Administration approved pembrolizumab and berahyaluronidase alfa-pmph (Keytruda Qlex, Merck) for subcutaneous injection ...FDA approves darolutamide... · FDA grants accelerated... · FDA approves belzutifan
  11. [11]
    [PDF] 2024 New Drug Therapy Approvals Annual Report - FDA
    Tecentriq Hybreza is the first anti-PD-L1 antibody administered subcutaneously. • Voquezna (vonoprazan) tablets for relief of heartburn associated with non ...
  12. [12]
    2024 FDA approvals - Nature
    Jan 2, 2025 · Checkpoint Therapeutics' PDL1-targeted cosibelimab (Unloxcyt) was granted the eleventh approval for a PD1 or PDL1-blocking antibody.
  13. [13]
    29126 - Gene ResultCD274 CD274 molecule [ (human)] - NCBI
    Aug 23, 2025 · This gene encodes an immune inhibitory receptor ligand that is expressed by hematopoietic and non-hematopoietic cells, such as T cells and B cells and various ...Missing: nomenclature | Show results with:nomenclature
  14. [14]
    Gene code CD274/PD-L1: from molecular basis toward cancer ...
    Dec 17, 2018 · PD-L1 is encoded by the CD274 gene (HGNC accession number: 17635; Ensembl Gene accession: ENSG00000120217), which is located in chromosome 9p24.Missing: ISRE | Show results with:ISRE
  15. [15]
  16. [16]
    Immunomodulatory Gene-Splicing Dysregulation in Tumorigenesis
    Aug 10, 2023 · PD-L1/CD274 gene consists of seven exons ... An illustration of PD-L1 splice variants generated by alternative splicing and their isoforms.
  17. [17]
    Regulation of PD-L1 Expression by NF-κB in Cancer - Frontiers
    PD-L1 promoter contains two putative IFN-stimulated response elements (ISRE) along with NF-κB binding sites. Once activated, p65 forms the canonical p50/p65 ...
  18. [18]
    PD-L1-deficient mice show that PD-L1 on T cells, antigen ... - PNAS
    Because wild-type resting B, T, macrophages, and DCs express low levels of PD-L1, activated cells were evaluated for PD-L1 expression. Activated wild type ...
  19. [19]
    Tissue expression of CD274 - Summary - The Human Protein Atlas
    Summary of CD274 (B7-H, B7-H1, B7H1, PD-L1, PDCD1LG1, PDL1) expression in human tissue. High expression in subsets of immune cells and trophoblastic cells.<|control11|><|separator|>
  20. [20]
    PD-L1 Expression in Human Placentas and Gestational ... - NIH
    This study describes a novel mechanism for trophoblastic cells to create a tolerogenic feto-maternal interface by upregulating PD-L1 in syncytiotrophoblast and ...Missing: macrophages | Show results with:macrophages
  21. [21]
    Gene code CD274/PD-L1: from molecular basis toward cancer ... - NIH
    Dec 17, 2018 · Here we summarize the current knowledge of PD-L1 gene modifications at genetic and epigenetic levels in different tumors.Cd274/pd-L1 Gene Structure · Cd274/pd-L1 Promoter Region · Mirna Targeting Pd-L1
  22. [22]
  23. [23]
    Regulation of PD-L1 Trafficking from Synthesis to Degradation
    PD-L1: Cellular Distribution. PD-L1 is a single-pass transmembrane protein with a 31 amino acid C-terminal cytoplasmic domain and a 220 amino acid ectodomain ...Pd-L1 Trafficking · Pd-L1 Degradation · Pd-L1: Cellular Distribution
  24. [24]
    Secreted PD-L1 variants mediate resistance to PD-L1 blockade ...
    Mar 14, 2019 · PD-L1 has been reported to harbor four N-glycosylation sites in the extracellular region: N35, N192, N200, and N219. Three of these, N192, N200 ...Missing: mature | Show results with:mature
  25. [25]
    Resveratrol targets PD-L1 glycosylation and dimerization to ...
    Resveratrol increases the electrophoretic mobility of PD-L1 protein. PD-L1 is a type I transmembrane glycoprotein with an apparent molecular weight of ~45 kDa.Missing: length | Show results with:length
  26. [26]
    PD-L1 glycosylation and its impact on binding to clinical antibodies
    We demonstrate that PD-L1 on the surface of breast cancer cells carries mostly complex glycans with high proportion of polyLacNAc structures at the N219 sequon.Missing: amino | Show results with:amino
  27. [27]
    Glycosylation and stabilization of programmed death ligand-1 ...
    Aug 30, 2016 · Here we show that the immunosuppression activity of PD-L1 is stringently modulated by ubiquitination and N-glycosylation.Missing: sources | Show results with:sources
  28. [28]
    A dimeric structure of PD-L1: functional units or evolutionary relics?
    PD-L1 is a member of the B7 protein family, most of whose members so far were identified as dimers in a solution and crystalline state.Missing: PDB | Show results with:PDB
  29. [29]
    Structural Biology of the Immune Checkpoint Receptor PD-1 and Its ...
    Aug 1, 2017 · PD-1 has two naturally occurring ligands: PD-L1 (B7-H1, CD274) and PD-L2 (B7-DC, CD273). Both were identified over a decade ago, and their role ...
  30. [30]
  31. [31]
    Revisiting the PD-1 pathway | Science Advances
    (A) According to the canonical PD-1/PD-L1 interaction, PD-L1 expressed on APCs or tumor cells interacts with PD-1 expressed on T cells in trans to attenuate ...
  32. [32]
    PD-1/PD-L1 binding studies using microscale thermophoresis - Nature
    Dec 15, 2017 · We describe a protocol using MST to determine the binding affinity of the PD-1/PD-L1 couple, which is involved in tumour escape processes.Missing: Kd | Show results with:Kd
  33. [33]
    Soluble PD-1 and PD-L1: predictive and prognostic significance in ...
    This systematic review discusses the production of sPD-1/PD-L1 and evaluates the physiological and pathological significance of their levels in human blood.
  34. [34]
    Soluble PD-L1 with PD-1-binding capacity exists in the plasma of ...
    Soluble PD-L1 with PD-1-binding capacity (bsPD-L1) was detected in 29 of 75 plasma samples from non-small cell lung cancer patients using a new ELISA system.Missing: Kd | Show results with:Kd
  35. [35]
    Blockade of trans PD-L1 interaction with CD80 augments antitumor ...
    Apr 10, 2023 · The blockade of PD-L1/CD80 interactions in mice with both cis and trans interactions or with only trans interactions augments antitumor immunity.Sign Up For Pnas Alerts · Results · Discussion
  36. [36]
    Strength of PD-1 signaling differentially affects T-cell effector functions
    T cells expressing high levels of PD-1 were largely nonfunctional when stimulated by aAPCs expressing PD-L1, suggesting that PD-1 expression sufficiently ...Strength Of Pd-1 Signaling... · Results · T-Cell Proliferation Is...<|control11|><|separator|>
  37. [37]
    Dual blockade of IL-10 and PD-1 leads to control of SIV viral ...
    Sep 12, 2024 · We hypothesized that IL-10 and PD-1 blockade would lead to control of viral rebound following analytical treatment interruption (ATI).
  38. [38]
    Programmed Cell Death Ligand 1 (PD-L1) Signaling Regulates ...
    Treatment of mouse and human macrophages with PD-L1 antibodies increased spontaneous macrophage proliferation, survival, and activation.
  39. [39]
    The Role Of PD-1/PD-L1 Axis In Treg Development And Function - NIH
    Oct 15, 2019 · The latest studies have suggested that Tregs might be involved in the treatment of PD-1/PD-L1 blockade and PD-1/PD-L1 axis could influence Treg differentiation ...
  40. [40]
  41. [41]
    Energy status dictates PD-L1 protein abundance and anti-tumor ...
    Jun 3, 2021 · These results suggest that glucose starvation or AMPK activation promotes PD-L1 degradation through S283 phosphorylation, which in turn ...Missing: engagement glycolysis
  42. [42]
    AMPK activation induces RALDH+ tolerogenic dendritic cells by ...
    Aug 8, 2024 · In the present study, we find that drug-induced AMPK activation in human moDCs is sufficient to promote their tolerogenicity, as evidenced by an ...Introduction · Results · Discussion · Materials and methods
  43. [43]
  44. [44]
    Interferon Receptor Signaling Pathways Regulating PD-L1 ... - NIH
    PD-L1 and PD-L2 are ligands for the PD-1 immune inhibiting checkpoint that can be induced in tumors by interferon exposure, leading to immune evasion.
  45. [45]
    Regulatory mechanisms of PD-L1 expression in cancer cells - PMC
    IFN-γ induces expression of PD-L1 through the JAK/STAT1/IRF1 pathway. IL-27 induces PD-L1 expression via the JAK/STAT3 pathway. b Constitutive expression ...
  46. [46]
  47. [47]
    Regulation of PD-L1 Expression by NF-κB in Cancer - PMC
    Nov 25, 2020 · The present review discusses how NF-κB, a master transcription factor of inflammation and immunity, is emerging as a key positive regulator of PD-L1 expression ...
  48. [48]
  49. [49]
    The EGFR pathway is involved in the regulation of PD-L1 ...
    Jul 26, 2016 · Our research indicates that EGFR is involved in the regulation of PD-L1 expression and cell proliferation via the IL-6/JAK/STAT3 signaling pathway in NSCLC.
  50. [50]
    Genetic, transcriptional and post-translational regulation of ... - Nature
    May 16, 2018 · At the transcriptional level, a number of transcriptional factors seem to regulate PD-L1 expression including HIF-1, STAT3, NF-κΒ, and AP-1.
  51. [51]
    PD-L1 is a novel direct target of HIF-1α, and its blockade under ... - NIH
    In the present study, we showed that hypoxia via hypoxia-inducible factor-1α (HIF-1α) selectively up-regulated PD-L1 on MDSCs, but not other B7 family members, ...
  52. [52]
  53. [53]
    Intrinsic and extrinsic control of expression of the immunoregulatory ...
    miR-513 directly regulated PD-L1 mRNA and protein expression by targeting the PD-L1 3-untranslated region in human biliary epithelial cells stimulated by IFN-γ ...Review · Introduction · Immune Regulation By...Missing: original paper<|control11|><|separator|>
  54. [54]
    Regulation of PD-L1: Emerging Routes for Targeting Tumor Immune ...
    May 21, 2018 · Recent research has shed new light into the mechanisms of PD-L1 regulation at genetic, epigenetic, transcriptional, translational, and posttranslational levels.
  55. [55]
    PDL1 Regulation by p53 via miR-34 - PMC - PubMed Central - NIH
    Nov 17, 2015 · PDL1 and miR-34a expression were analyzed in samples from patients with NSCLC and mutated p53 vs wild-type p53 tumors from The Cancer Genome ...
  56. [56]
    Examination of the Functional Relationship between PD-L1 DNA ...
    The methylation status of CpG sites in the PD-L1 CpG island shores is inversely correlated with PD-L1 expression in acute myeloid leukemia and colorectal, ...
  57. [57]
    Dissociation of the DNMT3A-HDAC1 Repressor Complex Induces ...
    Nov 13, 2019 · Taken together, we suggest that mutant DNMT3A induces higher H3K27 acetylation along with PD-L1 expression due to a looser complex between HDAC1 ...
  58. [58]
    HDAC3 Inhibition Upregulates PD-L1 Expression in B-Cell ...
    May 1, 2019 · Our aforementioned results showed that pan-HDAC inhibitors promoted PD-L1 transcription by increasing histone acetylation at the PD-L1 promoter ...
  59. [59]
    PD-L1 degradation pathway and immunotherapy for cancer - NIH
    Nov 6, 2020 · PD-L1 undergoes ubiquitination and degradation by E3 ubiquitin ligases, including STUB1, Cullin3SPOP, and β-TrCP, which is abolished by CMTM4/6, ...
  60. [60]
    Tissue expression of PD-L1 mediates peripheral T cell tolerance
    Apr 10, 2006 · Our data indicate that PD-L1 plays a critical role in tissue tolerance by protecting islets from self-reactive T cells. To confirm the role that ...
  61. [61]
    PD-1 and Its Ligands in Tolerance and Immunity - PMC
    PD-1 and its ligands, PD-L1 and PD-L2, deliver inhibitory signals that regulate the balance between T cell activation, tolerance, and immunopathology.
  62. [62]
    The PD-1 Pathway in Tolerance and Autoimmunity - PMC
    IL-10 also induces PD-L1 on monocytes. Interferons, IL-4, and granulocyte ... Further studies have revealed three other cytokines, IL-10, TGF-β and IL ...Missing: autocrine | Show results with:autocrine
  63. [63]
    The role of the PD‐1 pathway in autoimmunity and peripheral ...
    Jan 28, 2011 · This paper examines the role of PD-1 in limiting autoreactivity and establishing self-tolerance and discusses the hypothesis that PD-1 ligand (PD-L) expressionIntroduction · PD-1 in peripheral tolerance... · Imaging dynamic T cell motility...
  64. [64]
    The importance of the PD-1/PD-L1 pathway at the maternal-fetal ...
    Feb 19, 2019 · Our goal with this study was to investigate the contribution of PD-1/PD-L1 immune-checkpoint pathway to maternal immunotolerance mechanisms.
  65. [65]
    Human inherited PD-L1 deficiency is clinically and immunologically ...
    Apr 18, 2024 · Thus, contrary to the initial study of PD-L1-deficient mice reporting increases in the production of IFN-γ and severe autoimmunity (Latchman ...
  66. [66]
    The PD-1/PD-L1 Axis and Virus Infections: A Delicate Balance - PMC
    Thus, in the late phase of acute viral infection, PD-L1 is strongly upregulated thereby downregulating terminal differentiation of CD8+ T cells and preventing ...
  67. [67]
    PD-1 Signaling Promotes Control of Chronic Viral Infection by ...
    Nov 26, 2019 · Here, we show that PD-1-mediated restriction of immune responses is essential for durable control of chronic LCMV infection in mice.Results · Pd-1 Signaling Protects Host... · Star Methods
  68. [68]
    Reinvigorating exhausted HIV-specific T cells via PD-1–PD-1 ligand ...
    Sep 25, 2006 · Recent work has shown that PD-1 is highly expressed on exhausted T cells during chronic lymphocytic choriomeningitis virus (LCMV) infection in mice.
  69. [69]
    A potential new pathway for PD-L1 costimulation of the CD8-T cell ...
    Feb 11, 2013 · We analyzed the role of PD-L1 in CD8-T cell responses to infection with Listeria monocytogenes (LM) or vesicular stomatitis virus (VSV). PD ...Missing: dampens Th1
  70. [70]
    PDL-1 blockade impedes T cell expansion and protective immunity ...
    These results indicate that for immunity to intracellular bacterial infection, PDL-1 plays an important stimulatory role for priming and expansion of ...Missing: sepsis | Show results with:sepsis
  71. [71]
    Mechanisms controlling PD-L1 expression in cancer - PMC - NIH
    Immune checkpoint programmed death-ligand 1 (PD-L1) plays a critical role in facilitating tumor immune evasion. Cha et al. discuss the mechanisms regulating PD- ...Missing: immunology | Show results with:immunology
  72. [72]
    PD-L1 expression in human cancers and its association with clinical ...
    PD-L1 is an immunoinhibitory molecule that suppresses the activation of T cells, leading to the progression of tumors. Overexpression of PD-L1 in cancers ...Missing: immunology | Show results with:immunology
  73. [73]
    PD-L1 testing by immunohistochemistry in immuno-oncology - PMC
    Taube et al. [41] demonstrated that PD-L1 expression on TC was a predictive biomarker for anti-PD-1 drug Nivolumab in 41 patients with advanced solid tumors, ...
  74. [74]
    Programmed Death Ligand 1 (PD-L1) as a Predictive Biomarker for ...
    The antibody-specific scoring used to assess the prevalence of PD-L1 positivity is the tumor proportion score (TPS) evaluated as percentage of PD-L1-positive ...
  75. [75]
    Predictive Value of Combined Positive Score and Tumor Proportion ...
    TPS and CPS Cutoff Thresholds​​ In clinical practice, the TPS cutoff values of 1% and 50% are used for PD-L1 scoring in NSCLC and the determination of treatment ...Missing: melanoma selection
  76. [76]
    FDA Approval Summary: Atezolizumab for the Treatment of Patients ...
    Based on the preliminary clinical evidence showing a confirmed response rate of 50% (95% CI: 29.3%, 70.7%) in 20 patients with high PD‐L1 expression as ...
  77. [77]
    FDA Approval Summary: Atezolizumab and Durvalumab in ...
    Although the median DoR was 5.1 months in both groups, the proportion of patients estimated as having continued response at 12 months after the onset of ...
  78. [78]
    Outcomes of Combined Atezolizumab Plus Chemotherapy in Non ...
    Jan 3, 2025 · In the 141 patients treated with atezolizumab plus chemotherapy, the response rate (CR + PR) was 56.0% (CR, n=3; PR, n=76). Forty-four patients ...
  79. [79]
    Mechanisms of MHC-I Downregulation and Role in Immunotherapy ...
    Downregulation of MHC-I has been described as a mechanism of intrinsic and acquired resistance to immunotherapy in patients with cancer.
  80. [80]
    Primary resistance to PD-1 blockade mediated by JAK1/2 mutations
    We propose that JAK1/2 loss-of-function mutations are a genetic mechanism of lack of reactive PD-L1 expression and response to interferon gamma, leading to ...
  81. [81]
    A phase II efficacy and safety study of HB0025 (a PD-L1/VEGF ...
    May 28, 2025 · A multicentre, randomized, double-blind, controlled phase III trial will commence in 2025. Clinical trial information: NCT06758557. This is an ...
  82. [82]
    PD-L1 reverses depigmentation in Pmel-1 vitiligo mice by ... - Nature
    Jan 25, 2018 · Interestingly, the PD-L1 fusion protein is effective in inhibiting T cell activity in skin autoimmunity disease. Thus, we have a strong ...Introduction · Pd-L1 Fusion Protein... · Treg Assay In Vitro
  83. [83]
    PDL1 Fusion Protein Protects Against Experimental Cerebral ...
    Jan 13, 2019 · Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity(1999) ...
  84. [84]
    A Phase 2 Trial of Peresolimab for Adults with Rheumatoid Arthritis
    May 17, 2023 · Peresolimab showed efficacy in a phase 2a trial in patients with rheumatoid arthritis. These results provide evidence that stimulation of the PD-1 receptor has ...
  85. [85]
    PD-1 signaling uncovers a pathogenic subset of T cells in ...
    Other PD-1 agonists in clinical trials for autoimmune diseases include BMS-986019, BGB-A317, and TPI-201. The results of these clinical trials are still ...
  86. [86]
    PD-L1 genetic overexpression or pharmacological restoration in ...
    Genetically engineered or pharmacologically modulated HSPCs overexpressing PD-L1 inhibited the autoimmune response in vitro, reverted diabetes in newly ...
  87. [87]
    PDL1 restoration reverses T1DM | Nature Reviews Endocrinology
    Dec 1, 2017 · Genetic overexpression or pharmacological restoration of PDL1 is a viable immunotherapy strategy to reverse T1DM.
  88. [88]
    The effect of combining PD-1 agonist and low-dose Interleukin-2 on ...
    So far, the PD-1 agonists developed for autoimmune diseases are in Phase II and III clinical trials ( Table 1 ). These drugs can stimulate the physiological ...
  89. [89]
    Is PD-1 blockade a potential therapy for HBV? - PMC - NIH
    HBeAg seroconversion is associated with a more effective PD-L1 blockade during chronic hepatitis B infection.
  90. [90]
    Antiviral Intrahepatic T-Cell Responses Can Be Restored by ...
    We studied T-cell exhaustion and the effects of PD-1/PD-L1 blockade on intrahepatic and circulating HBV-specific T cells in patients with chronic hepatitis B.
  91. [91]
    Role of regulation of PD-1 and PD-L1 expression in sepsis - Frontiers
    Mar 8, 2023 · PD-1 and PD-L1 have critical roles in sepsis, indicating that their regulation may be a potential therapeutic target for sepsis.
  92. [92]
    PD-1 on dendritic cells impedes innate immunity against bacterial ...
    Virtually no DKO mice could survive more than 15 days after LM infection (Figure 1C). In contrast, 50% PD-1 KO/B6 mice achieved long-term survival (Figure 1A).
  93. [93]
    Blockade of endogenous B7-H1 suppresses antibacterial protection ...
    Expression of B7-H1 and programmed death (PD)-1 in response to Listeria monocytogenes infection. ... Survival was monitored for 14 days after infection for four ...