Fact-checked by Grok 2 weeks ago

Programmed cell death protein 1

Programmed cell death protein 1 (PD-1), also known as CD279 and encoded by the PDCD1 located on 2q37.3 in humans, is a type I transmembrane belonging to the that serves as a key receptor. Expressed primarily on the surface of activated T cells, B cells, natural killer (NK) cells, and other immune cells, PD-1 negatively regulates immune responses by inhibiting T cell activation, proliferation, secretion, and upon binding, thereby promoting peripheral and preventing . Originally discovered in through studies on apoptotic T cell hybridomas, PD-1 was identified as a novel member of the immunoglobulin superfamily induced during , highlighting its role in regulation. Structurally, PD-1 consists of an extracellular immunoglobulin variable-like (IgV) domain, a transmembrane region, and an intracellular tail containing an immunoreceptor tyrosine-based inhibitory motif (ITIM) and an immunoreceptor tyrosine-based switch motif (ITSM), which recruit phosphatases like SHP-1 and SHP-2 to transduce inhibitory signals. Its primary ligands are programmed cell death ligand 1 (PD-L1, also CD274) and PD-L2 (PDCD1LG2), which are upregulated on antigen-presenting cells, endothelial cells, and various tumor cells in response to inflammatory cytokines such as interferon-gamma. The interaction between PD-1 and its ligands dampens effector T cell functions, maintaining immune homeostasis but also enabling pathogens and tumors to evade immune surveillance by inducing T cell exhaustion in chronic infections and cancer microenvironments. In oncology, the PD-1/PD-L1 axis has emerged as a critical target for immunotherapy, with monoclonal antibodies such as nivolumab and pembrolizumab—FDA-approved since 2014—blocking this pathway to reinvigorate antitumor T cell responses and achieving durable remissions in malignancies including melanoma, non-small cell lung cancer, and Hodgkin lymphoma. Beyond cancer, PD-1 dysregulation contributes to autoimmune diseases like rheumatoid arthritis and systemic lupus erythematosus, where enhancement of its signaling is under investigation for therapeutic modulation. Ongoing research continues to explore PD-1's expression on non-T cells, such as regulatory T cells and myeloid-derived suppressor cells, and its interplay with other checkpoints like CTLA-4 to refine combination therapies.

Discovery and history

Initial identification

The programmed cell death protein 1 (PD-1) was first identified in 1992 by Yoshimasa Ishida, Yutaka Agata, and at through subtractive cDNA hybridization screening. This approach targeted genes upregulated during in two murine cell lines: the antigen-stimulated T-cell hybridoma 2B4.11 and the interleukin-3 (IL-3)-deprived pro-B cell line LyD9, both of which undergo under these conditions. The newly cloned gene was designated PD-1, an abbreviation for " 1," reflecting its strong and transient induction specifically during the early stages of apoptotic in these models, distinguishing it from other apoptosis-related genes. of the PD-1 cDNA revealed it encodes a 288-amino-acid protein predicted to form a type I transmembrane with a molecular weight of approximately 55 kDa upon , featuring an extracellular immunoglobulin variable-like , a transmembrane region, and an intracellular tail with potential signaling motifs. Further insight into PD-1's function emerged in 1999 from experiments with PD-1 mice generated by Tasuku Honjo's group, which spontaneously developed lupus-like autoimmune diseases characterized by , proliferative , and autoantibodies, thereby establishing PD-1's critical role in maintaining and preventing .

Key milestones and recognition

Following the initial identification of PD-1 in 1992 by Tasuku Honjo's group through subtractive hybridization in apoptotic T cells, subsequent research rapidly advanced understanding of its function and interactions. In 2000, Gordon J. Freeman and colleagues identified PD-L1 (also known as B7-H1 or CD274) as the first ligand for PD-1, demonstrating that its engagement inhibits T cell receptor-mediated proliferation and production in T-cell assays. This discovery positioned PD-1 within the CD28/B7 family of immune regulators. In 2001, Yvonne Latchman and colleagues reported PD-L2 (CD273) as a second ligand for PD-1, further confirming its inhibitory role on T cell activation through similar functional assays. These findings from 2000 to 2002 established PD-1's core mechanism as an , suppressing T cell responses to maintain . The groundbreaking contributions to PD-1 and related checkpoint research were recognized with the 2018 in or Medicine, awarded jointly to and for their discoveries of cancer therapy by inhibition of negative immune regulation, including PD-1's role alongside CTLA-4. Therapeutic advancements culminated in the 2023 U.S. Food and Drug Administration (FDA) approval of toripalimab-tpzi (Loqtorzi), a PD-1 inhibitor, in combination with and for first-line treatment of adults with metastatic or recurrent locally advanced , and as monotherapy for previously treated cases. In 2025, a genomic analysis by R. Kondo and colleagues revealed PD-1's evolutionary conservation across jawed vertebrates, including and bony fishes, indicating its ancient origin in adaptive immunity and highlighting conserved structural features with ligands and PD-L2, as well as interacting phosphatases like SHP-2.

Structure and biochemistry

Gene and protein domains

The encodes programmed cell death protein 1 (PD-1), an receptor, and is located on the long arm of human at position 2q37.3. This spans approximately 9 kb and consists of five , with 1 encoding the , 2 the IgV-like domain, 3 the stalk and , 4 the initial cytoplasmic region, and 5 the remainder of the cytoplasmic tail. The homolog, Pdcd1, maps to and shares high sequence similarity, facilitating studies in . The human PD-1 precursor protein comprises 288 , processed into a mature form by cleavage of the N-terminal spanning positions 1-25. The extracellular region features an immunoglobulin variable-like (IgV-like) domain from residues 26-150, which adopts a β-sheet structure typical of the and serves as the primary site for ligand interaction. This is followed by a short transmembrane (residues 171-191) that anchors PD-1 in the membrane and a cytoplasmic tail (residues 192-288) containing key signaling elements, including an immunoreceptor tyrosine-based inhibitory motif (ITIM; sequence VTYAEL at positions approximately 223-228) and an immunoreceptor tyrosine-based switch motif (ITSM; sequence TEYATIV at positions approximately 248-254). These motifs become tyrosine-phosphorylated upon receptor engagement, enabling recruitment of phosphatases like SHP-1 and SHP-2. PD-1 belongs to the CD28/CTLA-4 family of T-cell costimulatory and coinhibitory receptors, sharing structural homology in the extracellular IgV-like domain and overall membrane topology, though PD-1 lacks the covalent dimerization seen in some family members. Post-translational modifications further regulate PD-1 function: N-linked glycosylation occurs at asparagine 49 (Asn49) within the IgV-like domain, contributing to protein stability and ligand binding affinity, while the cytoplasmic tail harbors serine/threonine phosphorylation sites, such as serine 261, that modulate intracellular trafficking and dephosphorylation dynamics. Alternative splicing of PDCD1 produces isoforms, including a soluble form lacking the transmembrane domain and an exon 3-skipped variant that alters inhibitory signaling.

Expression patterns

Programmed cell death protein 1 (PD-1) is primarily expressed on activated immune cells, including CD4⁺ and CD8⁺ T cells, B cells, cells, natural killer T (NKT) cells, activated monocytes, macrophages, and dendritic cells (DCs). Under steady-state conditions, PD-1 expression remains low on naive or resting T cells and B cells, with basal levels detectable on only a small subset of peripheral T cells. However, it is rapidly upregulated following activation, particularly through (TCR) or (BCR) stimulation, often becoming detectable within 24 hours and persisting in response to ongoing antigenic signals. In terms of tissue distribution, PD-1 shows high expression in primary and secondary lymphoid organs, including the , , and lymph nodes, where it is associated with maturing and antigen-experienced lymphocytes in germinal centers and white pulp regions. Expression is inducible in non-lymphoid tissues during , such as at sites of chronic infection (e.g., liver in infection or lungs in ) or within tumor microenvironments, reflecting recruitment of activated PD-1⁺ immune cells to these locales. Developmentally, PD-1 expression is regulated during T-cell maturation in the , where it is largely absent from the earliest progenitor stages but appears at low levels (3–5% of total thymocytes) on double-negative (⁻) subsets during TCRβ rearrangement and the transition to double-positive thymocytes. This thymic expression supports processes like positive selection, but PD-1 levels remain modest overall in the compared to peripheral tissues. In mature peripheral lymphocytes, expression peaks upon encounter and activation, enabling its role in modulating responses in secondary lymphoid organs and inflamed sites. In chronic settings like persistent infections or cancer, sustained high PD-1 expression on these mature cells contributes to T-cell exhaustion.

Ligands and interactions

Primary ligands

The primary ligands of programmed cell death protein 1 (PD-1) are (also known as CD274 or B7-H1) and PD-L2 (also known as PDCD1LG2 or B7-DC), both members of the B7 family of immune-regulatory proteins. is a type I transmembrane consisting of 290 , featuring an extracellular domain with immunoglobulin variable-like (IgV) and constant-like (IgC) regions, a , and a short cytoplasmic tail. It is broadly expressed on antigen-presenting cells such as dendritic cells and monocytes, as well as on tumor cells and endothelial cells, where its upregulation is often induced by inflammatory cytokines like interferon-gamma. binds to PD-1 with a (Kd) of approximately 770 nM, reflecting moderate affinity that contributes to its role in modulating immune responses. In contrast, PD-L2 is a 273-amino acid type I with a similar IgV-IgC extracellular architecture but more restricted expression, primarily on dendritic cells and macrophages, particularly upon activation by stimuli such as interleukin-4 or . Despite its lower baseline expression compared to , PD-L2 exhibits higher binding affinity to PD-1, with a Kd of about 140 nM, enabling more stable interactions under physiological conditions. Both and PD-L2 can exist in soluble forms, generated through alternative mRNA splicing that produces truncated variants lacking the or via proteolytic shedding of the membrane-bound protein by matrix metalloproteinases, allowing these circulating ligands to potentially influence systemic immune regulation. No other major ligands for PD-1 have been identified, though minor interactions between and (B7-1) have been reported, which do not constitute primary binding partnerships.

Binding mechanisms

The binding of programmed cell death protein 1 (PD-1) to its ligands, primarily and PD-L2, occurs through the immunoglobulin -like (IgV) domains of both proteins, forming a 1:1 stoichiometric complex that mimics the fragment (Fv) structure of antibodies. The interface is centered on the front β-sheets and loops of these domains, specifically the CC' and loops of PD-1's IgV domain, which engage the corresponding IgV-like domain of or PD-L2 via hydrophobic and polar contacts. Key residues at this interface include tyrosine 123 (Tyr123) in PD-1's loop, which forms π-π stacking interactions, and 115 (Met115) in , contributing to the hydrophobic core of the binding pocket. Crystallographic studies, such as those deposited in the (PDB) under codes 3BIK (2008, murine PD-1/human PD-L1) and 3BP5 (2008, murine PD-1/PD-L2), reveal that the PD-1/PD-L1 complex adopts a compact, two-domain with extensive buried surface area (~900 Ų), promoting partial dimerization through strand-swapped conformations in the ligands. While the core complex is 1:1, membrane-bound configurations allow for 2:1 stoichiometries, where one ligand molecule can engage two PD-1 molecules in cis (on the same cell) or trans (across cells), enhancing in physiological settings. The binding reflect moderate , with association rates (k_on) on the order of 10^5 M^{-1} s^{-1} and dissociation rates (k_off) around 10^{-1} s^{-1} for human PD-1/PD-L1, yielding equilibrium dissociation constants (K_D) of approximately 0.3–8 μM depending on the experimental system; PD-L2 exhibits 3–4-fold higher affinity due to slower dissociation. Regulation of these interactions is influenced by post-translational modifications and environmental factors. N-linked on , particularly at sites near the (e.g., Asn192, Asn200), sterically modulates accessibility, with deglycosylated forms showing reduced PD-1 efficiency and altered immunosuppressive activity. Additionally, the exhibits pH sensitivity, with enhanced in the acidic (pH ~6.5), where of residues (e.g., His56 in PD-1) stabilizes the , potentially amplifying immune suppression in hypoxic tumors.

Physiological functions

Immune checkpoint role

Programmed cell death protein 1 (PD-1) functions as a key by delivering inhibitory signals to activated T cells, thereby preventing excessive immune responses during antigen-specific interactions. Upon ligation with its ligands, PD-1 attenuates signaling, which inhibits T-cell and reduces the production of pro-inflammatory cytokines such as interferon-gamma (IFN-γ) and interleukin-2 (IL-2). This regulatory mechanism ensures that T-cell activation remains proportional to the antigenic stimulus, limiting potential tissue damage from overzealous immune activity. In peripheral tissues, PD-1 promotes by suppressing the activity of autoreactive T cells that may escape central tolerance mechanisms. By upregulating cell cycle inhibitors like p27 and p15, PD-1 engagement halts the expansion of self-reactive T lymphocytes, thereby maintaining self-tolerance and preventing autoimmune reactions in non-lymphoid organs. This process is particularly evident in scenarios involving exposure, where PD-1 helps balance effector functions with regulatory controls. PD-1 also exerts regulatory effects on B cells, contributing to the suppression of . Expressed on B lymphocytes, PD-1 signaling inhibits B-cell expansion and impairs antibody class switching, particularly in response to T-independent antigens, which limits the generation of long-lived plasma cells and excessive immunoglobulin production. This B-cell intrinsic inhibition helps fine-tune responses to avoid hyperactivation. In mucosal tissues, PD-1 maintains immune by preventing overreactions to commensal . The PD-1 pathway fosters to harmless gut antigens, ensuring that regulatory T cells and effector responses remain balanced to support symbiotic relationships without triggering . Disruption of this checkpoint, such as through PD-1 , can convert mucosal into pathogenic responses against commensals.

Signaling pathways

Upon ligation, PD-1's intracellular tail, featuring an (ITIM) and an (ITSM), undergoes , enabling the recruitment of Src homology 2 (SH2) domain-containing phosphatases SHP-1 and SHP-2. These phosphatases bind primarily to the ITSM ( 248 in humans), with SHP-2 showing stronger affinity, and key proximal (TCR) signaling components, including the zeta chain of the CD3 complex (CD3ζ) and zeta-chain-associated protein kinase (ZAP70). This disrupts the initiation of downstream TCR signals, thereby dampening T-cell activation. The recruited SHP-1 and SHP-2 phosphatases inhibit critical pathways essential for T-cell proliferation and effector functions, particularly the (PI3K)-Akt pathway and the Ras- (MAPK) pathway.30156-X) By dephosphorylating intermediaries, PD-1 signaling reduces Akt activation downstream of PI3K, which is triggered by TCR and , leading to diminished metabolic reprogramming and survival signals in T cells.30156-X) Similarly, inhibition of the -MAPK pathway, likely via SHP-2-mediated suppression of Ras guanine nucleotide exchange, attenuates extracellular signal-regulated kinase (ERK) phosphorylation, further curtailing production and cell growth responses from TCR/ engagement.30156-X) A simplified model of phosphatase recruitment and activity can be represented as: \text{Phospho-PD-1} + \text{SHP-2} \rightarrow \text{Dephospho-PD-1} + \text{p-ZAP70 (dephosphorylated to ZAP70)} This reaction illustrates the transient docking of SHP-2 to phosphorylated PD-1, enabling dephosphorylation of ZAP70 and other substrates, though in practice, SHP-2 acts catalytically on multiple targets. These inhibitory effects culminate in reduced interleukin-2 (IL-2) production by activated T cells, impairing autocrine proliferation signals even in the presence of CD28 costimulation. PD-1 engagement also induces cell cycle arrest at the G0/G1 phase by suppressing cyclin-dependent kinase activity and related progression factors. Additionally, it promotes BIM-mediated apoptosis through upregulation of the pro-apoptotic BH3-only protein BIM, enhancing mitochondrial outer membrane permeabilization and caspase activation in T cells.

Role in diseases

Cancer

In the (TME), PD-1 is upregulated on (TILs), particularly + T cells, leading to their exhaustion and anergy, which impairs effective anti-tumor immunity. This upregulation is driven by chronic antigen exposure and immunosuppressive signals from tumor-repopulating cells, resulting in reduced T-cell proliferation, cytokine production, and . Consequently, PD-1 engagement promotes a state of T-cell dysfunction that allows tumor progression by evading immune surveillance. Cancer cells often overexpress , the primary ligand for PD-1, which is induced by interferon-gamma (IFN-γ) secreted from activated T cells in the TME. This overexpression correlates with poor clinical in various malignancies, including , non-small cell (NSCLC), and (RCC). For instance, elevated PD-L1 levels on tumor cells in NSCLC and RCC are associated with advanced disease stages and reduced overall survival. PD-1 signaling also contributes to the suppressive function of regulatory T cells (Tregs) within the TME, enhancing their ability to inhibit anti-tumor responses from effector T cells. PD-1-expressing Tregs accumulate in tumors and promote an immunosuppressive milieu by limiting CD8+ T-cell activation and infiltration. High PD-1 and PD-L1 expression serves as a prognostic biomarker, predicting response to immunotherapy in approximately 20-40% of patients across PD-1/PD-L1-positive cancers. Tumors with substantial PD-L1 expression on immune or tumor cells are more likely to benefit from checkpoint blockade, highlighting its utility in patient stratification.

Autoimmune disorders

PD-1 plays a critical protective role in maintaining and preventing by inhibiting the activation and proliferation of autoreactive T cells. In genetic models, deficiency of PD-1 leads to spontaneous autoimmune disorders in mice. For instance, PD-1 knockout mice on the background develop a lupus-like syndrome characterized by the production of anti-DNA antibodies, immune complex deposition, and , while those on the background exhibit autoimmune . Additionally, PD-1-deficient mice are prone to resembling , with joint inflammation driven by dysregulated T-cell responses. These phenotypes highlight PD-1's essential function in suppressing self-reactive immune responses. In humans, polymorphisms in the PDCD1 gene, which encodes PD-1, have been associated with increased susceptibility to several autoimmune diseases. Specific variants, such as those in the promoter region or intronic sequences, correlate with higher risk of systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and . For example, the PD-1.3 polymorphism (rs11568821) is linked to SLE in multiple populations, potentially by altering PD-1 expression levels on T cells and reducing inhibitory signaling. Similarly, associations with RA and involve variants that impair PD-1's regulatory capacity, leading to enhanced autoreactivity. The underlying mechanism of PD-1's protective role involves its inhibition of signaling, which prevents the unchecked expansion of autoreactive T cells and subsequent inflammatory cascades. Loss of PD-1 function results in hyperproliferative T cells that produce excessive proinflammatory , such as IFN-γ and IL-17, potentially culminating in cytokine storms that exacerbate tissue damage in autoimmune settings. This dysregulation is evident in PD-1-deficient models where T follicular helper cells and effector T cells fail to be properly restrained, promoting production and . Therapeutically, the recognition of PD-1's anti-autoimmune function has spurred interest in developing PD-1 agonists to restore in autoimmune diseases. Unlike PD-1 inhibitors used in cancer, agonists aim to enhance PD-1 signaling to suppress overactive immune responses. Several candidates, including monoclonal antibodies like peresolimab, have shown promise in preclinical models and early clinical trials for conditions such as and SLE, with phase II and III studies ongoing to evaluate efficacy and safety. However, no PD-1 agonists have received regulatory approval for autoimmune indications as of 2025.

Infectious diseases

In chronic viral infections such as , (HCV), and virus (LCMV), PD-1 is upregulated on virus-specific + T cells, leading to T cell exhaustion characterized by reduced proliferation, production, and , which impairs viral clearance. This exhaustion is driven by sustained PD-1 signaling, resulting in a progressive loss of effector functions and contributing to persistent infection. For instance, in , PD-1 expression correlates with diminished memory + T cell responses in typical progressors, exacerbating viral persistence. In bacterial infections like caused by , PD-1 engagement inhibits antigen-specific + T cell proliferation and interferon-γ production, promoting immune evasion and disease progression. PD-1 blockade in experimental models restores T cell effector functions and enhances macrophage , reducing bacterial replication and improving host control of . In parasitic infections such as caused by species, elevated PD-1 expression on parasite-specific + T cells induces exhaustion, marked by decreased interferon-γ secretion and expression, which limits protective immunity and sustains chronic parasitemia. This PD-1-mediated dysfunction exacerbates blood-stage infection severity, as evidenced by faster parasite clearance and sterile immunity in PD-1-deficient mice compared to wild-type controls. However, PD-1 also restrains excessive T cell activation, thereby mitigating during acute phases. PD-1 exhibits a dual role across infection types: in acute s, it tempers T cell expansion and to limit and support formation, whereas in chronic settings, it enforces exhaustion that hinders pathogen resolution. For example, transient PD-1 signaling during acute LCMV optimizes CD8+ T cell recall responses without inducing dysfunction.

Neurodegenerative conditions

Programmed cell death protein 1 (PD-1) has emerged as a key regulator in neuroinflammatory processes underlying (AD), primarily through its expression on and infiltrating T cells within the . In AD models, PD-1 signaling on , stimulated by astrocytic , enhances the uptake and clearance of amyloid-β (Aβ) peptides, thereby mitigating plaque accumulation and associated . Similarly, PD-1 on T cells promotes interferon-γ (IFN-γ) production, which recruits peripheral monocytes to cross the blood-brain barrier and facilitate Aβ , demonstrating a protective role in early disease stages. These mechanisms highlight PD-1's contribution to immune-mediated debris removal in the brain parenchyma. However, the dual nature of PD-1 signaling becomes apparent in chronic contexts, where prolonged activation shifts toward detrimental effects. While initial PD-1 engagement supports Aβ clearance and limits excessive , sustained signaling fosters a pro-inflammatory microenvironment that exacerbates synaptic loss and neuronal dysfunction. Deletion of microglial PD-1 in models compromises Aβ uptake while triggering heightened inflammatory responses, underscoring the balance required for . In human , elevated PD-1 expression on peripheral T cells and increased levels in correlate with disease severity, including tau hyperphosphorylation and cognitive decline, as observed in postmortem analyses and cohort studies. Research from the has further elucidated PD-1's therapeutic potential through blockade strategies in preclinical models. Administration of anti-PD-1 antibodies in amyloidogenic mice reduces Aβ plaque burden and by enhancing microglial activation and recruitment, leading to improved performance. In models, PD-1 inhibition decreases glycogen synthase kinase-3β activity and hyperphosphorylation, attenuating cognitive impairments. Nonetheless, such interventions carry risks of , including potential from unchecked immune activation, necessitating careful modulation to avoid exacerbating .

Therapeutic targeting

Inhibitory antibodies

Inhibitory antibodies targeting programmed cell death protein 1 (PD-1) represent a cornerstone of , functioning as monoclonal antibodies that disrupt the PD-1 signaling pathway to reinvigorate antitumor immune responses. These agents, primarily human or humanized immunoglobulin G4 (IgG4) subtypes, bind specifically to the extracellular domain of PD-1 on T cells, thereby preventing its interaction with ligands such as and PD-L2. Pembrolizumab (Keytruda) is a humanized IgG4 approved by the U.S. (FDA) in September 2014 for the treatment of unresectable or metastatic in patients with disease progression following and, if BRAF V600 mutation-positive, a BRAF inhibitor. It exerts its therapeutic effect by binding to the extracellular domain of PD-1, thereby blocking the interaction with its ligands and restoring T-cell-mediated against tumor cells. This approval marked pembrolizumab as the first PD-1 inhibitor to receive FDA authorization, based on phase I/II trial data demonstrating objective response rates of approximately 26% in ipilimumab-refractory patients. Nivolumab (Opdivo), a fully IgG4 , received initial FDA approval in December 2014 for unresectable or metastatic and was subsequently expanded in March 2015 to include advanced non-small cell (NSCLC) after platinum-based . Its pharmacokinetic profile includes a of approximately 25 days, enabling dosing regimens of 240 mg every 2 weeks or 480 mg every 4 weeks via intravenous infusion. Like other PD-1 inhibitors, nivolumab prevents ligand binding to PD-1, thereby enhancing T-cell activation and proliferation in the . Cemiplimab (Libtayo), another human IgG4 , was approved by the FDA in September 2018 for metastatic or locally advanced (CSCC) in patients ineligible for curative or . On October 8, 2025, the FDA approved for the treatment of adults with high-risk CSCC at risk of recurrence after and , based on phase 3 C-POST data showing a 68% reduction in the risk of recurrence or death compared to . It shares a similar mechanism with and nivolumab, binding to PD-1 to inhibit engagement and promote antitumor T-cell responses. Clinical trials supporting its initial approval reported objective response rates ranging from 41% to 47% in advanced CSCC, with durable responses observed in a majority of responders; broader data across solid tumors indicate response rates of 15-50% depending on tumor type and expression. The core mechanism of these inhibitory antibodies involves steric blockade of the PD-1/PD-L1 interaction, which normally suppresses signaling and production, leading to T-cell exhaustion in the tumor setting. By alleviating this inhibition, the antibodies restore T-cell and proliferation, enabling effective immune attack on cancer cells. However, this enhanced immune activation can result in immune-related adverse events (irAEs), such as , , , and endocrinopathies, occurring in 10-20% of patients as severe (grade 3-4) events, necessitating vigilant monitoring and management.

Emerging modulators and combinations

Beyond traditional monoclonal antibodies, small-molecule inhibitors targeting the PD-1/PD-L1 pathway offer potential advantages in and tissue penetration. Other candidates, such as INCB086550, an oral PD-L1 small-molecule inhibitor, are in ongoing phase 1/2 trials evaluating safety and in patients with advanced cancers, highlighting improved over biologics. These agents aim to disrupt PD-1 signaling without the infusion requirements of antibodies, though challenges in selectivity and duration of inhibition persist. Bispecific antibodies that simultaneously target PD-1 pathway components and other checkpoints are advancing as multifunctional modulators. KN046, a bispecific antibody against and CTLA-4, has shown favorable efficacy and manageable toxicity in phase 2 trials for metastatic non-small cell (NSCLC), including combinations with that improved . Multiple clinical trials of KN046 are ongoing in NSCLC, small cell (SCLC), and other solid tumors in , the U.S., and as of 2025. This approach enhances immune activation by blocking multiple inhibitory signals, potentially broadening response rates in checkpoint-resistant tumors. Combinatorial strategies integrating PD-1 inhibitors with other therapies have significantly improved outcomes in . The combination of nivolumab (anti-PD-1) and (anti-CTLA-4) in advanced yields a 5-year overall survival rate of 52%, substantially higher than monotherapy options. Similarly, in NSCLC, (anti-PD-1) plus platinum-based increases objective response rates to approximately 48% and extends compared to chemotherapy alone, establishing it as a standard first-line regimen. These synergies leverage complementary immune mechanisms, though increased immune-related adverse events necessitate careful patient selection. Applications of PD-1 modulation extend beyond cancer to autoimmune and infectious diseases. PD-1 agonists, such as S-4321, a bifunctional agonizing PD-1 and FcγRIIb, are in phase 1 trials for autoimmune conditions like systemic lupus erythematosus (SLE), aiming to restore without broad . In infectious diseases, peptide-based PD-1 immunomodulators enhance efficacy by countering PD-1-mediated T-cell exhaustion, demonstrating improved adaptive immunity in preclinical models of viral infections. These developments underscore PD-1's versatile role in immune regulation across disease contexts.

Preclinical research

Animal models of infection

Animal models have been instrumental in elucidating the role of PD-1 in T-cell exhaustion during chronic s, particularly through blockade or knockout strategies that enhance antiviral responses while revealing potential risks of . In models of () , which closely mimic pathogenesis in humans, PD-1 blockade during chronic reduces plasma by approximately 1-2 logs in treated animals and restores proliferative capacity and production in exhausted CD8+ T cells. This intervention expands SIV-specific CD8+ T cells in blood and lymphoid tissues, leading to improved viral control and prolonged survival compared to untreated controls. However, the effects on are often transient without therapies, highlighting PD-1's central role in maintaining exhaustion during lentiviral persistence. The virus (LCMV) model in mice provides a foundational system for studying PD-1-mediated exhaustion in chronic viral infections, where persistent LCMV Clone 13 induces high PD-1 expression on + T cells, impairing their effector functions and allowing viral persistence. Anti-PD-1 or anti-PD-L1 therapy in this model reinvigorates exhausted + T cells, enhancing , secretion (e.g., IFN-γ, TNF-α), and production, which collectively reduce viral titers in tissues like the and liver. These findings demonstrate PD-1's dominance in exhaustion but also the potential for functional restoration, though full clearance often requires combination approaches. In (TB) models, PD-1 knockout (KO) mice infected with exhibit increased bacterial burden in the , with higher colony-forming units compared to wild-type mice, due to impaired CD4+ and CD8+ T-cell responses and reduced IFN-γ production. However, this leads to exacerbated lung pathology, including severe , infiltration, and early mortality, underscoring PD-1's protective role in limiting during bacterial persistence. PD-1 blockade in wild-type mice similarly exacerbates disease in models, increasing susceptibility and risking disseminated . Translational limitations arise from species-specific differences in PD-1/PD-L1 interactions; human PD-1 binds its ligands with higher affinity and inhibitory potency than murine PD-1, potentially leading to overestimation of blockade efficacy in mouse models and challenges in primate translation. These variations affect T-cell suppression thresholds and must be considered when extrapolating preclinical findings to human infectious diseases. Recent 2025 studies confirm human PD-1's stronger inhibitory signaling compared to rodent orthologs.

Models of autoimmunity and transplantation

Studies in PD-1 knockout (KO) mice have been instrumental in demonstrating the protein's role in maintaining and preventing . These mice spontaneously develop a range of autoimmune disorders, including lupus-like , destructive , and , with symptoms typically emerging by 6 months of age and progressing to multi-organ infiltration involving the heart, lungs, liver, and other tissues by 6-12 months, often leading to fatal outcomes. The autoimmune in PD-1 KO mice arises from dysregulated T and proliferation, highlighting PD-1's suppressive function on autoreactive lymphocytes. Notably, low-dose IL-2 therapy has been shown to ameliorate aspects of this by compensating for metabolic shifts in T cells and enhancing function, thereby rescuing the mice from severe disease manifestations. In transplantation models, PD-1 signaling promotes allograft tolerance and survival, while its blockade exacerbates rejection. In murine heart transplant models with fully MHC-mismatched allografts, administration of anti-PD-1 antibodies or PD-1/PD-L1 pathway blockade accelerates acute rejection by enhancing alloimmune responses, particularly in costimulation-deficient settings where CD28 or B7 pathways are blocked. Similarly, in liver transplantation models, PD-1 blockade or deficiency in donor tissue leads to rapid acute rejection, underscoring the pathway's role in mitigating T cell-mediated damage to the graft. Conversely, strategies to enhance PD-1 signaling, such as T cell-specific overexpression of PD-1, prolong allograft survival in cardiac transplant models by fostering tolerance through ICOS-dependent mechanisms and reducing effector T cell responses. PD-1 agonists or mimetics have also been explored to extend survival in various allograft models, supporting their potential to induce long-term tolerance without broad immunosuppression. The collagen-induced arthritis (CIA) model in mice further illustrates PD-1's protective role against autoimmune . PD-1 KO mice exhibit exacerbated severity in CIA, characterized by increased swelling, higher arthritis scores, and enhanced Th17 , confirming PD-1's suppression of pathogenic T responses. with anti-PD-1 antibodies in wild-type CIA mice worsens , promotes proinflammatory production (e.g., IFN-γ and IL-17), and accelerates pathology, reinforcing that PD-1 blockade unleashes autoreactive + and + T cells. These findings support PD-1's function in limiting osteoclastogenesis and erosion in rheumatoid arthritis-like conditions. Insights from these preclinical models have directly informed the development of PD-1 agonists for therapeutic applications in and transplantation. In (RA), phase II clinical trials of the PD-1 agonist peresolimab have demonstrated disease improvement in patients with active RA, with reductions in disease activity scores and no major safety concerns. For transplantation, these guide efforts toward PD-1-enhancing agents to achieve without increasing infection risk, with ongoing preclinical work exploring agonist antibodies and fusion proteins to promote graft tolerance in clinical settings.

References

  1. [1]
    Gene ResultPDCD1 programmed cell death 1 [ (human)] - NCBI
    Aug 19, 2025 · Programmed cell death protein 1 (PDCD1) is an immune-inhibitory receptor expressed in activated T cells; it is involved in the regulation of T-cell functions.
  2. [2]
    Programmed Cell Death Protein 1 (PD-1) and ... - PubMed Central
    Sep 2, 2023 · This article endeavors to present a comprehensive review of the PD-1/PD-L1 immune checkpoint, elucidating its underlying mechanisms and ...
  3. [3]
    Induced expression of PD‐1, a novel member of the immunoglobulin ...
    These results suggest that activation of the PD‐1 gene may be involved in the classical type of programmed cell death.
  4. [4]
  5. [5]
    Programmed Cell Death 1 - an overview | ScienceDirect Topics
    Programmed death-1 (PD-1) is an inhibitory co-receptor expressed on innate and adaptive immune cells that is critical for the regulation of inflammation and ...
  6. [6]
    PD-1: Its Discovery, Involvement in Cancer Immunotherapy ... - NIH
    Jun 1, 2020 · PD-1, induced to be expressed on the surface of activated T cells, forces T cells to ignore cancer cells as one of the authentic 'self' components.
  7. [7]
    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.
  8. [8]
    Engagement of the Pd-1 Immunoinhibitory Receptor by a Novel B7 ...
    We report here that the ligand of PD-1 (PD-L1) is a member of the B7 gene family. Engagement of PD-1 by PD-L1 leads to the inhibition of T cell receptor– ...
  9. [9]
    PD-L2 is a second ligand for PD-1 and inhibits T cell activation
    We identify here PD-1 ligand 2 (PD-L2) as a second ligand for PD-1 and compare the function and expression of PD-L1 and PD-L2. Engagement of PD-1 by PD-L2 ...Missing: 2001-2002 original paper
  10. [10]
    [PDF] LOQTORZI (toripalimab-tpzi) - accessdata.fda.gov
    Initial U.S. Approval: 2023 ... LOQTORZI is a prescription medicine used to treat adults with a kind of cancer called nasopharyngeal carcinoma. (NPC).
  11. [11]
    PD-1 is conserved from sharks to humans: new insights ... - Frontiers
    May 27, 2025 · Programmed cell death protein 1 (PD-1) is an immune checkpoint molecule until recently believed to exist only in tetrapod species.
  12. [12]
    Gene: Pdcd1 (ENSMUSG00000026285) - Summary - Mus_musculus
    Chromosome 1: 93,966,027-93,980,278 reverse strand. ... This gene has 1 transcript (splice variant), 105 orthologues and is associated with 45 phenotypes.Missing: homolog | Show results with:homolog
  13. [13]
    PDCD1 - Programmed cell death protein 1 - Homo sapiens (Human)
    Inhibitory receptor on antigen activated T-cells that plays a critical role in induction and maintenance of immune tolerance to self.
  14. [14]
    The human PD-1 gene: complete cDNA, genomic ... - PubMed - NIH
    Isolation of the genomic locus of the hPD-1 gene reveals that the gene is composed of five exons located on human chromosome 2 at band q37. The 5' flanking ...
  15. [15]
    Interaction of SHP-2 SH2 domains with PD-1 ITSM induces PD ... - NIH
    Mar 17, 2020 · PD-1 has one immunoreceptor tyrosine-based inhibitory motif (ITIM) at Y223 and one immunoreceptor tyrosine-based switch motif (ITSM) at Y248.
  16. [16]
    CTLA-4 and PD-1 Pathways - PubMed Central - NIH
    PD-1 is a member of the B7/CD28 family of costimulatory receptors. · Both CTLA-4 and PD-1 binding have similar negative effects on T-cell activity; however, the ...Ctla-4 Pathway · Pd-1 Pathway · Implications Of Ctla-4 And...
  17. [17]
    Crystal structure of the complex between programmed death-1 (PD ...
    Jul 29, 2008 · Another important feature of PD-1 is its oligomeric state. Both CTLA-4 and CD28 exist as disulfide-linked homodimers. In addition, B7–1 ...
  18. [18]
    Small Molecule Agents Targeting PD-1 Checkpoint Pathway for ...
    May 1, 2022 · PD-1 is heavily glycosylated at Asn49, Asn 58, Asn 74, and Asn 116 in T cells, and Asn 58 is highly critical for PD-1/PD-L1 interaction (89) ...
  19. [19]
    Post-translational Modification of PD-1: Potential Targets for Cancer ...
    Mar 15, 2024 · An additional phosphorylation site at serine 261 (S261) on PD-1 has been identified as an important regulator of PD-1 translocation into the ...Missing: Ser/ | Show results with:Ser/
  20. [20]
    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.Missing: original | Show results with:original
  21. [21]
    Revisiting the PD-1 pathway | Science Advances
    Programmed death–1 (PD-1) was discovered in 1992 by T. Honjo and colleagues in Kyoto University as an apoptosis-associated gene. However, PD-1 overexpression ...
  22. [22]
  23. [23]
    Developmentally regulated expression of the PD-1 protein ... - PubMed
    We examined expression of the PD-1 protein during differentiation of thymocytes in normal adult, fetal and RAG-2(-/-) mice with or without anti-CD3 mAb ...Missing: mature | Show results with:mature
  24. [24]
  25. [25]
    The B7 family of immune-regulatory ligands - Genome Biology
    May 31, 2005 · The B7 family consists of structurally related, cell-surface protein ligands, which bind to receptors on lymphocytes that regulate immune responses.
  26. [26]
    Gene code CD274/PD-L1: from molecular basis toward cancer ... - NIH
    Dec 17, 2018 · CD274/PD-L1 gene structure ... The PD-L1 gene comprises seven exons and encodes a putative type I transmembrane protein of 290 amino acids.
  27. [27]
    Expression and Regulation of the PD-L1 Immunoinhibitory Molecule ...
    Endothelial cells also have the capacity to serve as antigen-presenting cells because they can express both MHC class I and II molecules (23,27,28). It is ...
  28. [28]
    Dissociation constants (K D ) of human PD-1, PD-L1, PD-L2 and...
    Magneto-nanosensor platform for probing low-affinity protein–protein interactions and identification of a low-affinity PD-L1/PD-L2 interaction.<|separator|>
  29. [29]
  30. [30]
    Evolving landscape of PD-L2: bring new light to checkpoint ... - NIH
    Dec 15, 2022 · Programmed death ligand-2 (PD-L2), another ligand of PD-1, was initially discovered to be expressed in macrophages and dendritic cells (DCs).<|separator|>
  31. [31]
    PD-L1 and PD-L2 differ in their molecular mechanisms of interaction ...
    PD-L1 and PD-L2 bound PD-1 with comparable affinities, but striking differences were observed at the level of the association and dissociation characteristics.Figure 1. Pd-L1 And Pd-L2 Do... · Figure 3. Pd-L1 And Pd-L2... · Pd-1.3 Mab Prevents The...
  32. [32]
    Soluble Programmed Death Ligand-1 (sPD-L1): A Pool of ... - MDPI
    The shedding of PD-L1 was not only observed in breast cancer cells (MDA-MB-231, MCF10A), but also with other cell lines derived from prostate (DU-145) or lung ( ...1. The Pd-1/pd-L1 Checkpoint · 1.3. Soluble Pd-L1 · 4. Soluble Pd-L1 Beyond...
  33. [33]
    A snapshot of the PD-1/PD-L1 pathway - Journal of Cancer
    PD-L1 and PD-L2 are ligands of PD-1, and the PD-L1/PD-L2 binding with PD-1 leads to suppression of T-cell activation. PD-L1 also interacts with B7-1 (CD80) on ...Pd-1/pd-L1-Related Cell... · Anti-Pd-1/pd-L1 Therapies In... · Combination Therapy For...
  34. [34]
    3BIK: Crystal Structure of the PD-1/PD-L1 Complex - RCSB PDB
    Feb 26, 2008 · Here we present the crystal structure of murine PD-1 in complex with human PD-L1. PD-1 and PD-L1 interact through the conserved front and side of their Ig ...
  35. [35]
    PD-1-mediated inhibition of T cell activation - PubMed Central - NIH
    Feb 23, 2024 · The PD-1 axis negatively regulates T cell activation and controls the magnitude and duration of T cell responses to limit tissue damage and ...
  36. [36]
    Role of the PD-1 Pathway in the Immune Response - PMC
    PD-1 and its ligands, PD-L1 and PD-L2, have emerged as critical inhibitory signaling pathways that regulate T cell response and maintain peripheral tolerance.
  37. [37]
    PD-1 inhibits T cell proliferation by upregulating p27 and p15 and ...
    The programmed cell death-1 (PD)-1 receptor (CD279) is a potent T cell inhibitor with a critical role in peripheral tolerance, but it can also compromise anti- ...The Pd-1/pd-L Pathway In The... · Effects Of Pd-1 On The... · Pd-1 Promotes...
  38. [38]
    The PD-1 Pathway in Tolerance and Autoimmunity - PMC
    The PD-1:PD-L pathway controls peripheral T-cell tolerance in several ways. This pathway limits the initial phase of activation and expansion of self-reactive T ...
  39. [39]
    PD-1 Suppresses B-1b Cell Expansion and Long-lived IgG ...
    The results of this study demonstrate a key role for the PD-1:PDL regulatory axis in controlling B-1b cell responses and IgG production to TI-2 Ags.
  40. [40]
    PD-1 regulates germinal center B cell survival and the formation and ...
    Thus, PD-1 regulates selection and survival in the GC, impacting the quantity and quality of long-lived PCs. Introduction. Adaptive immune responses generate ...Missing: class | Show results with:class
  41. [41]
    Role of PD-L1 in Gut Mucosa Tolerance and Chronic Inflammation
    Dec 1, 2020 · In this review, we summarize the current knowledge supporting the critical role of PD-L1 in gastrointestinal mucosal tolerance.
  42. [42]
    Intestinal Tolerance Is Converted to Autoimmune Enteritis upon PD ...
    Our results provide evidence that the PD-1:PD-L1 inhibitory pathway plays a critical role in maintaining mucosal tolerance by preventing naive, gut-specific CD ...
  43. [43]
    Co-inhibitory receptor programmed cell death protein 1 targets ... - NIH
    Aug 5, 2017 · Early studies with human T cells showed that phosphatases—SHP-1 and SHP-2, but not SHIP—are recruited by these motifs and that ITSM was crucial ...
  44. [44]
    Biochemical Signaling of PD-1 on T Cells and Its Functional ...
    PD-1 consists of a single N-terminal IgV-like domain, an approximately 20 amino acid stalk separating the IgV domain from the plasma membrane, a transmembrane ...
  45. [45]
    PD1 signal transduction pathways in T cells - PMC - NIH
    Here we review the current knowledge on PD1-dependent intracellular signaling pathways, and the consequences of disrupting PD1 signal transduction.
  46. [46]
    PD‐1:PD‐L inhibitory pathway affects both CD4+ and CD8+ T cells ...
    Feb 12, 2002 · We show that PD-1 engagement by PD-L1 or PD-L2 inhibits proliferation as a result of compromised IL-2 production. Initially, CD28 costimulation ...
  47. [47]
    Selective Effects of PD-1 on Akt and Ras Pathways Regulate ...
    As determined with ethynyl deoxyuridine (EdU) staining, we found that ligation of PD-1 resulted in the cell cycle arrest of CD4+ T cells at the G0-G1 phase (Fig ...
  48. [48]
    The Programmed Death-1 Immune Suppressive Pathway
    This effect occurs because PD-1 ligation decreases T cells survival by upregulating the pro-apoptotic factor Bim (36). PD-1 ligation also facilitates down- ...
  49. [49]
    Development of PD-1/PD-L1 Pathway in Tumor Immune ... - Nature
    Aug 17, 2015 · ... PD-L1 can decrease the apoptosis of T cells and facilitate antitumor immunity. ② Facilitate T cell anergy and exhaustion: A research ...
  50. [50]
    Human breast tumor-infiltrating CD8+ T cells retain polyfunctionality ...
    Oct 16, 2018 · PD-1 expression in T cells involved in chronic infections and tumors such as melanoma often correlates with a state of T-cell exhaustion. Here ...
  51. [51]
    Genetic, transcriptional and post-translational regulation of ... - Nature
    May 16, 2018 · In melanoma cells, NF-κB mediated PD-L1 overexpression induced by IFN-γ. PD-L1 upregulation by NF-κΒ was independent of STAT3 and c-Jun ...
  52. [52]
    The intracellular signalosome of PD-L1 in cancer cells - Nature
    Sep 28, 2018 · Therefore, PD-L1 overexpression in tumors was generally found to be an indicator of progression and poor prognosis in cancer. Most studies have ...
  53. [53]
    PD-L1 Expression is Associated With Poor Prognosis in Renal Cell ...
    PD-L1 Expression is Associated With Poor Prognosis in Renal Cell Carcinoma. Appl Immunohistochem Mol Morphol. 2020 Mar;28(3):213-220. doi: 10.1097/PAI ...
  54. [54]
    PD-L1 signaling selectively regulates T cell lymphatic ... - Nature
    Apr 21, 2022 · Likewise, activated Tregs in tumor-infiltrating lymphocytes (TILs) express higher levels of PD-1 than circulating Tregs. The ratio of Tregs to ...
  55. [55]
    Therapeutic targets and biomarkers of tumor immunotherapy - Nature
    Sep 19, 2022 · The clinical efficacy of PD-1/L1 blocking antibodies was first observed against tumors with high PD-L1 expression, including melanoma, non-small ...<|control11|><|separator|>
  56. [56]
    Decoding the signs of response to cancer immunotherapy - Nature
    Only 20-40% of patients respond to immunotherapy1 and, because these drugs can activate a broad range of immune cells, they can sometimes trigger severe auto- ...
  57. [57]
    Genes, Tolerance and Systemic Autoimmunity - PMC
    PD-1-deficient mice develop spontaneous autoimmune diseases including SLE (nephritis and autoantibodies in C57BL/6 mice and cardiomyopathy in Balb/c mice) ...
  58. [58]
    Targeting the programmed cell death-1 pathway in rheumatoid arthritis
    May 29, 2017 · ... PD-1 knockout mice suggesting a more robust autoimmune response. ... PD-1 knock out animals develop inflammatory arthritis. Inhibitory ...
  59. [59]
    Are programmed cell death 1 gene polymorphisms correlated ... - NIH
    Sep 1, 2017 · PDCD1 gene polymorphisms were found to be correlated with the risk of several autoimmune diseases, such as SLE, RA, diabetes mellitus, and ...
  60. [60]
    PD-1, gender, and autoimmunity - PMC - NIH
    Immunized PD-1 knockout mice had a delayed onset of disease but not complete protection from EAE following administration of E2 [87]. Prior to treatment, PD-1 ...
  61. [61]
    The effect of combining PD-1 agonist and low-dose Interleukin-2 on ...
    This article reviewed the relationship between PD-1 and SLE, and proposed to use PD-1 as a predictive biomarker for SLE disease activity.5. Pd-1 Expression In Sle... · Figure 3 · 7. Pd-1 Agonist MonotherapyMissing: glomerulonephritis | Show results with:glomerulonephritis
  62. [62]
    PD-1/PD-L pathway inhibits M.tb-specific CD4+ T-cell functions and ...
    Dec 7, 2016 · Blockade of the PD-1/PD-L pathway can partly or completely restore effector T-cells. However, the role of the PD-1/PD-L pathway in murine TB ...Missing: improves | Show results with:improves
  63. [63]
  64. [64]
  65. [65]
    A change of PD-1/PD-L1 expression on peripheral T cell subsets ...
    Sep 30, 2022 · Moreover, these findings indicate that PD-1/PD-L1 blockade treatment could be a novel choice to slow AD disease deterioration. Keywords: ...
  66. [66]
    The Synergistic Potential of Combining PD-1/PD-L1 Immune ... - MDPI
    Jun 30, 2023 · Microglial PD-1 stimulation by astrocytic PD-L1 suppresses neuroinflammation and Alzheimer's disease pathology. EMBO J. 2021, 40, e108662 ...<|control11|><|separator|>
  67. [67]
    Programmed Cell Death Protein 1 Blockade Reduces Glycogen ...
    Dec 16, 2021 · The application of PD1-blocking antibody reduced tau hyperphosphorylation and GSK3β activity and prevented memory impairments.
  68. [68]
    Limitations and potential strategies of immune checkpoint blockade ...
    Sep 23, 2024 · In this review we will provide possible evidence behind the use of immunotherapy with MAbs in AD and PD and highlight the recent clinical development landscape.
  69. [69]
    [PDF] KEYTRUDA® (pembrolizumab) for injection - accessdata.fda.gov
    KEYTRUDA® (pembrolizumab) is indicated for the treatment of patients with unresectable or metastatic melanoma and disease progression following ipilimumab and, ...
  70. [70]
    Fundamental Mechanisms of Immune Checkpoint Blockade Therapy
    Here, we review how the negative costimulatory molecules CTLA4 and PD-1 attenuate T-cell activation. We also discuss current dogma and recent conceptual ...Introduction · Mechanisms Of Pd-1--Mediated... · Therapeutic CombinationsMissing: incidence | Show results with:incidence<|control11|><|separator|>
  71. [71]
    Merck Receives Accelerated Approval of KEYTRUDA ...
    Sep 4, 2014 · KEYTRUDA is a humanized monoclonal antibody that works by increasing the ability of the body's immune system to fight advanced melanoma.
  72. [72]
    Pembrolizumab for the treatment of advanced melanoma - PMC - NIH
    Pembrolizumab, a humanized monoclonal antibody directed against programed death receptor 1 has shown significant activity in advanced melanoma resulting in its ...
  73. [73]
    [PDF] OPDIVO (nivolumab) injection Label - accessdata.fda.gov
    See full prescribing information for OPDIVO. OPDIVO (nivolumab) injection, for intravenous use. Initial U.S. Approval: 2014. --------------------------RECENT ...
  74. [74]
    Opdivo (Nivolumab): Second PD-1 Inhibitor Receives FDA Approval ...
    On March 4, 2015, the FDA approved a new indication for nivolumab for the treatment of patients with metastatic squamous non–small-cell lung cancer (NSCLC) that ...Missing: half- | Show results with:half-<|separator|>
  75. [75]
    Nivolumab - StatPearls - NCBI Bookshelf
    Nivolumab received FDA approval in 2014. Depending on the indication, it can ... The drug has a half-life of 25 days. Go to: Administration. Available ...
  76. [76]
    [PDF] OPDIVO® (nivolumab) injection, for intravenous use
    Initial U.S. Approval: 2014. ----------------------------RECENT MAJOR ... The geometric mean elimination half-life (t1/2) is 25 days (77.5%). Specific ...
  77. [77]
    Nivolumab for Treatment of Unresectable or Metastatic Melanoma ...
    Abstract. On December 22, 2014, the FDA granted accelerated approval to nivolumab (OPDIVO ... half-life (t1/2) was 26.7 days. Steady-state concentrations of ...
  78. [78]
    [PDF] Page 1 of 34 - accessdata.fda.gov
    LIBTAYO is indicated for the treatment of patients with metastatic cutaneous squamous cell carcinoma (mCSCC) or locally advanced CSCC (laCSCC) who are not ...
  79. [79]
    Cemiplimab and Cutaneous Squamous Cell Carcinoma - NIH
    The objective response rate was 41.1% illustrating substantial antitumour activity while the duration of response at 8 months was 95% among responding patients.
  80. [80]
    Cemiplimab Becomes First Treatment Approved for Advanced ...
    Sep 29, 2018 · Among 108 total patients, 47% responded to the immunotherapy treatment and 4% experienced complete responses characterized by total tumor ...
  81. [81]
    Management of Immune-Related Adverse Events in Patients ...
    In general, patients receiving anti–PD-1 or PD-L1 antibodies have a lower incidence of any-grade irAEs than those treated with anti–CTLA-4 agents, with ...
  82. [82]
    Adverse effects of immune-checkpoint inhibitors - Nature
    May 15, 2019 · Patients receiving anti-PD-1 or anti-PD-L1 antibodies have a lower incidence of any-grade irAEs than those receiving anti-cytotoxic T lymphocyte ...
  83. [83]
    NCT02812875 | A Study of CA-170 (Oral PD-L1, PD-L2 and VISTA ...
    This is a multi-center, open-label, Phase 1 trial of orally administered CA-170 in adult patients with advanced solid tumors or lymphomas.Missing: 2025 | Show results with:2025
  84. [84]
    Article KN046, a bispecific antibody against PD-L1 and CTLA-4, plus ...
    Mar 19, 2024 · KN046, a bispecific antibody targeting PD-L1 and CTLA-4, presents a promising therapeutic option for metastatic non-small cell lung cancer (NSCLC).
  85. [85]
    KN046-Alphamab Oncology
    KN046 has been granted Orphan Drug Designation by the U.S. Food and Drug Administration (FDA) for the treatment of thymic epithelial tumors. Clinical Research ...
  86. [86]
    Five-Year Survival with Combined Nivolumab and Ipilimumab in ...
    Sep 27, 2019 · Overall survival at 5 years was 52% in the nivolumab-plus-ipilimumab group and 44% in the nivolumab group, as compared with 26% in the ipilimumab group.
  87. [87]
    Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell ...
    Apr 16, 2018 · The addition of pembrolizumab to chemotherapy resulted in significantly higher rates of response and longer progression-free survival than chemotherapy alone ...
  88. [88]
    Novel Peptide-Based PD1 Immunomodulators Demonstrate Efficacy ...
    Aug 6, 2025 · Here, we report the discovery and characterization of a peptide-based class of PD1 checkpoint inhibitors, which have a potent adaptive immunity ...<|control11|><|separator|>
  89. [89]
    Enhancing SIV-specific immunity in vivo by PD-1 blockade | Nature
    Dec 10, 2008 · PD-1 blockade during chronic SIV infection resulted in a rapid expansion of SIV-specific CD8 T cells in the blood of all macaques (Fig. 1a, b).
  90. [90]
    PD-1 blockade during chronic SIV infection reduces ... - PubMed
    Apr 23, 2012 · PD-1 blockade during chronic SIV infection reduces hyperimmune activation and microbial translocation in rhesus macaques · Authors · Affiliation.Missing: load | Show results with:load
  91. [91]
    PD-1 blockade during chronic SIV infection reduces ... - JCI
    Apr 23, 2012 · PD-1 blockade during chronic SIV infection reduces hyperimmune activation and microbial translocation in rhesus macaques. Ravi Dyavar Shetty ...
  92. [92]
    Interplay between regulatory T cells and PD-1 in modulating T cell ...
    Aug 11, 2014 · Our results so far revealed that T reg cell ablation leads to a striking rescue of virus-specific CD8 T cells during chronic LCMV infection.
  93. [93]
    PD-1 blockade exacerbates Mycobacterium tuberculosis infection in ...
    Thus, PD-1 blockade increased bacterial loads in pulmonary granulomas but did not result in disseminated infection. Frequency of Mtb-specific T cells in ...
  94. [94]
    Functional differences between rodent and human PD-1 linked to ...
    Jan 3, 2025 · We found that human PD-1 is more inhibitory than mouse PD-1, owing to stronger interactions with the ligands PD-L1 and PD-L2 and more efficient recruitment of ...<|control11|><|separator|>
  95. [95]
    Development of Lupus-like Autoimmune Diseases by Disruption of ...
    In the present study, we report that PD-1-deficient B6 mice spontaneously developed typical lupus-like glomerulonephritis and destructive arthritis as they age, ...
  96. [96]
    Programmed Cell Death-1 Pathway Deficiency Enhances ...
    Studies have demonstrated that PD-1 gene–knockout (Pdcd1−/−) mice develop lupus-like arthritis, glomerulonephritis, and autoimmune dilated cardiomyopathy. In ...
  97. [97]
    Low-dose IL-2 therapy compensates for metabolic shifts and ... - NIH
    Oct 16, 2020 · Low-dose IL-2 therapy compensates for metabolic shifts and reverses anxiety-like behavior in PD-1 deficiency-induced autoimmunity.Missing: fatal | Show results with:fatal
  98. [98]
    PDL1 Is Required for Peripheral Transplantation Tolerance and ...
    The PD-1 pathway has been shown to play an important role in regulating the alloimmune response in experimental models of skin and heart transplantation (25, 26) ...
  99. [99]
    PD1/B7-H1 Interaction Contribute to the Spontaneous Acceptance of ...
    Blockade of the PD-1/B7-H1 signal by administration of anti-B7-H1mAb or liver donor deficient in B7H1 resulted in the acute rejection of the allograft, ...
  100. [100]
    Overexpression of PD-1 on T cells promotes tolerance in cardiac ...
    Using PD-L1–KO donor allografts, we demonstrated that allograft PD-L1 expression was required for this survival benefit. These results suggest that the ...
  101. [101]
    Role of the PD-1 Pathway in the Immune Response
    Preliminary work in mice from our group suggests that enhanced signaling of PD-1 is able to protect the allograft against chronic rejection and induce long-term ...
  102. [102]
    Program Death-1 Suppresses Autoimmune Arthritis by Inhibiting ...
    To study the role of PD-1 in the pathogenesis of rheumatoid arthritis ... Suppression of immune induction of collagen-induced arthritis in IL-17-deficient mice.Missing: knockout | Show results with:knockout
  103. [103]
    PD-1 inhibitor unleashes pathogenic CD8+ T cells and induce ...
    The severity of arthritis at euthanasia was comparable between mice with PD-1i arthritis and conventional collagen-induced arthritis (CIA) mice. Histological ...
  104. [104]
    opportunities for immune-mediated kidney diseases | Nephrology ...
    Oct 10, 2023 · In addition to CTL4A (discussed above), a phase II study of peresolimab, a PD-1 agonist, led to improvement, without safety signals in ...Adaptive Immunity: Targeting... · Adaptive Immunity: B-Cell... · Innate Immunity: Targeting...