PD-1 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

PD-1: Structure, Function, and Role in Immunity

PD-1 (CD279) is a cell-surface receptor expressed on T cells, B cells, and macrophages. Its primary role is to suppress excessive immune responses, preventing autoimmunity through two mechanisms:

  • Induction of T-cell apoptosis in lymph nodes to eliminate self-reactive T cells.

  • Enhanced survival of regulatory T cells (Tregs) to maintain immune tolerance .

Structural Features (Source: ):

ComponentDescription
ExtracellularImmunoglobulin variable (IgV) domain binding PD-L1/L2.
TransmembraneSingle-pass membrane protein.
IntracellularContains immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and a switch motif (ITSM) that recruit phosphatases (e.g., SHP-1/2) .

PD-1 binds its ligands, PD-L1 (ubiquitously expressed on tumor cells) and PD-L2 (restricted to antigen-presenting cells), to inhibit T-cell activation, cytokine production, and cytotoxicity .

Discovery and Development of PD-1 Inhibitors

PD-1 was discovered in 1992 during a screen for apoptosis-related genes . Early studies in mice revealed its role in preventing autoimmune diseases like lupus and cardiomyopathy . The therapeutic potential of PD-1 blockade was validated in preclinical models, leading to clinical trials in the 2000s.

Key Milestones (Sources: ):

  • Nivolumab (Opdivo) and pembrolizumab (Keytruda) became the first FDA-approved PD-1 inhibitors in 2014–2015.

  • MW11-h317, a novel PD-1 antibody with a distinct glycosylation-dependent epitope (Asn58), demonstrated superior binding affinity in preclinical models .

Mechanism of Action

PD-1 monoclonal antibodies block the PD-1/PD-L1 interaction, reversing immune suppression in the tumor microenvironment. This restores T-cell functions:

  • Proliferation: Increased T-cell expansion and effector differentiation.

  • Cytotoxicity: Enhanced killing of tumor cells via perforin/granzyme secretion .

  • Microenvironment Modulation: Reduced myeloid-derived suppressor cells (MDSCs) and enhanced T-cell infiltration .

Synergistic Effects (Source: ):

Therapy CombinationOutcome
PD-1 + CAR T-cellsEnhanced tumor eradication and reduced MDSCs in preclinical models.
PD-1 + ChemotherapyImproved survival in non-small cell lung cancer (NSCLC) vs. chemo alone .

Approved PD-1 Monoclonal Antibodies

PD-1 inhibitors are classified into two categories: PD-1 antibodies and PD-L1 antibodies. Below are key drugs and their applications:

DrugTargetCancer IndicationsKey Trials
NivolumabPD-1Melanoma, NSCLC, RCC, Hodgkin lymphomaCheckMate 037, 066, 067
PembrolizumabPD-1Melanoma, NSCLC, head/neck cancer, MSI-high tumorsKEYNOTE-001, -024, -040
CemiplimabPD-1Cutaneous squamous cell carcinomaEMPOWER-CSCC 1
AtezolizumabPD-L1NSCLC, bladder cancer, triple-negative breast cancerIMpower150, 130, 010
AvelumabPD-L1Merkel cell carcinoma, urothelial carcinomaJAVELIN Merkel 200

Clinical Efficacy Across Cancer Types

PD-1/PD-L1 inhibitors show variable efficacy depending on tumor type and PD-L1 expression:

Melanoma

  • Pembrolizumab and nivolumab achieve objective response rates (ORRs) of 28–40%, with durable responses exceeding 2+ years .

Renal Cell Carcinoma (RCC)

  • Nivolumab + ipilimumab (CheckMate 214) yields superior ORRs (42%) vs. sunitinib (27%) .

Meta-Analysis Findings (Source: ):

EndpointPD-1/PD-L1 Monotherapy vs. Standard Care
ORR20.21% vs. 10.6% (OR = 1.98)
OS (HR)0.75 (95% CI: 0.67–0.83)

Biomarker Optimization

  • PD-L1 expression correlates with response but is not definitive (e.g., PD-L1-negative tumors may still respond) .

  • Tumor mutational burden (TMB) and MSI-high status are emerging predictive markers .

PD-1 vs. PD-L1 Inhibitors: Comparative Analysis

ParameterPD-1 InhibitorsPD-L1 Inhibitors
Binding TargetT-cell PD-1 receptorTumor/immune cell PD-L1 ligand
EC₅₀ (Signaling)Higher (e.g., nivolumab = 76.17 ng/ml)Lower (e.g., atezolizumab = 6.46 ng/ml)
Efficacy (NSCLC)Superior OS when combined with chemotherapySimilar ORR but variable OS

Clinical Implications:

  • PD-1 antibodies may offer broader immunomodulation, while PD-L1 antibodies target tumor-specific ligands .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery times.
Synonyms
CD279 antibody; CD279 antigen antibody; hPD 1 antibody; hPD l antibody; hPD-1 antibody; hSLE1 antibody; PD 1 antibody; PD-1 antibody; PD1 antibody; PDCD 1 antibody; PDCD1 antibody; PDCD1_HUMAN antibody; Programmed cell death 1 antibody; Programmed cell death 1 protein antibody; Programmed cell death protein 1 antibody; Protein PD 1 antibody; Protein PD-1 antibody; SLEB2 antibody; Systemic lupus erythematosus susceptibility 2 antibody
Target Names
Uniprot No.

Target Background

Function
PD-1 (Programmed Death-1) is an inhibitory receptor expressed on antigen-activated T cells. It plays a crucial role in establishing and maintaining immune tolerance to self-antigens. Upon binding to its ligands, CD274/PDCD1L1 (PD-L1) and CD273/PDCD1LG2 (PD-L2), PD-1 delivers inhibitory signals. Following T-cell receptor (TCR) engagement, PD-1 interacts with CD3-TCR within the immunological synapse, directly inhibiting T-cell activation. PD-1 suppresses T-cell activation through the recruitment of PTPN11/SHP-2. After ligand binding, PD-1 is phosphorylated within its ITSM motif, leading to the recruitment of the protein tyrosine phosphatase PTPN11/SHP-2, which mediates dephosphorylation of key TCR-proximal signaling molecules like ZAP70, PRKCQ/PKCtheta, and CD247/CD3zeta. This inhibitory pathway is exploited by tumors to dampen anti-tumor immunity and escape destruction by the immune system, thereby facilitating tumor survival. The interaction with CD274/PDCD1L1 inhibits cytotoxic T lymphocytes (CTLs) effector function. Blocking the PDCD1-mediated pathway reverses the exhausted T-cell phenotype and normalizes the anti-tumor response, providing a rationale for cancer immunotherapy.
Gene References Into Functions
  1. HLA-G, NRP1, and PD-1 may be involved in the immune response in psoriatic patients. PMID: 29790686
  2. In young melanoma patients, compared to age-matched controls, the proportions of naïve CD4+ T cells were lower, while percentages of memory CD4+ T cells expressing HLA-DR, Ki-67, and PD-1 were higher. This pattern was not observed in older patients. PMID: 29546435
  3. A meta-analysis indicates that the PD-1 rs36084323 A > G polymorphism is associated with a decreased risk of cancer in Asian populations. PMID: 30249505
  4. In a Southern Brazilian population, there is no significant association between PD1.3G/A - rs11568821 polymorphism and susceptibility to systemic lupus erythematosus or rheumatoid arthritis. PMID: 27914594
  5. SLC18A1 might complement other biomarkers currently under investigation regarding programmed cell death protein 1/programmed cell death protein ligand 1 inhibition. PMID: 30194079
  6. Structural and functional analyses unexpectedly revealed an N-terminal loop outside the IgV domain of PD-1. This loop is not involved in PD-L1 recognition but dominates binding to nivolumab, whereas N-glycosylation is not involved in binding at all. PMID: 28165004
  7. This finding suggests the potential application of PD-1 blockade in AML. The study demonstrated an excellent synergistic tumor therapeutic effect of PD-1 blockade and CTL therapy compared to either treatment alone. PMID: 29962321
  8. High PD-1 expression is associated with Mycobacterium avium complex-induced lung disease. PMID: 28169347
  9. PD-1/PD-L1 expression is a frequent occurrence in poorly differentiated neuroendocrine carcinomas of the digestive system. PMID: 29037958
  10. Results indicate that high-level PD1 expression may be a significant factor associated with the immune checkpoint pathway in liver cancer. PMID: 29620156
  11. These results suggest that the PD-1 genotype of the donor plays an important role in the development of acute GvHD after alloHSCT from HLA-identical sibling donors. PMID: 30019128
  12. A subgroup of advanced disease ovarian cancer patients with high-grade tumors expressing PD-L1 may be prime candidates for immunotherapy targeting PD-1 signaling. PMID: 29843813
  13. Both rs2227982 and rs2227981 polymorphisms were associated with T1 Diabetes (T1D) risk in East Asians, and rs2227982 also had a significant association with glycemic traits, suggesting that PDCD1 gene polymorphisms might contribute to T1D risk. [meta-analysis] PMID: 29774466
  14. PD.1 (-538G/A) gene polymorphism is associated with Colon Cancer Risk. PMID: 29580042
  15. The high-affinity PD-1 mutant could compete with the binding of antibodies specific to PD-L1 or PD-L2 on cancer cells. PMID: 29890018
  16. The G allele of rs36084323 of PDCD1 is associated with an increased risk of advanced TNM staging of colorectal cancer. PMID: 29652996
  17. Promoter methylation of CTLA4, PD-L1, PD-L2, and PD-1 in diffuse lower-grade gliomas (LGG) harboring isocitrate dehydrogenase (IDH) mutation is reported. PMID: 29396294
  18. Unlike other multiple autoimmune disease-associated genetic variants, there was no association between PDCD1 variants and Juvenile Idiopathic Arthritis. PMID: 28056736
  19. Alteration of the PD-1/PD-L1 pathway can modulate Treg/Th17 balance in asthmatic children. PMID: 29874664
  20. The expression of PD1 on T cells was elevated in patients with rheumatoid arthritis and was correlated with disease activity. PMID: 29257239
  21. High expression of programmed death-1 in sentinel lymph nodes is associated with breast cancer. PMID: 29193094
  22. This study presents three cases of patients with mRCC treated with anti-PD-1 antibody therapy in combination with targeted therapy (bevacizumab), anti-cytotoxic T lymphocyte antigen 4 therapy (ipilimumab), or radiotherapy. PMID: 29146617
  23. Anti-CTLA4/anti-PD-1/PD-L1 combinations versus anti-PD-1/PD-L1 monotherapy were selected as a factor independent of TMB for predicting better RR (77% vs. 21%; P = 0.004) and PFS (P = 0.024). Higher TMB predicts favorable outcome to PD-1/PD-L1 blockade across diverse tumors. PMID: 28835386
  24. The altered soluble (s)PD1 and sICOS serum levels in the different Hepatitis b (HBV) groups may reflect the dysregulation of T cell activation, and may be associated with the HBV pathological process. PMID: 28983583
  25. PD-L1 expression was upregulated following TGF-beta induction; in contrast, it was downregulated by TGF-beta receptor-kinase inhibitors and the MET process. Furthermore, chemo-treatment increased TGF-beta expression and enhances PD-L1 expression via autocrine TGF-beta induced EMT. Analysis of clinical samples revealed a significant relationship between PD-L1 expression and EMT status. PMID: 28849209
  26. This study demonstrated that PD-1 may be involved in lymph nodes metastasis and contributes to the understanding of the mechanism of immunotherapies in non-small cell lung cancer. PMID: 28799818
  27. PD-L1 expression in melanoma tumor cells is lower than NSCLC or renal cell carcinoma cells. The higher response rate in melanoma patients treated with PD-1 inhibitors is likely related to PD-L1 in tumor-associated inflammatory cells. Further studies are warranted to validate the predictive role of inflammatory cell PD-L1 expression in melanoma and determine its biological significance. PMID: 28223273
  28. Higher PRE PD-1(+) T cells in responders suggest active suppression of an engaged immune system that is disinhibited by anti-PD-1 therapies. Furthermore, immunoprofiling of EDT biopsies for increased PD-L1 expression and immune cell infiltration showed greater predictive utility than PRE biopsies and may allow better selection of patients most likely to benefit from anti-PD-1 therapies and warrants further evaluation. PMID: 28512174
  29. PD-1 was overexpressed on CD8+ T-cells from patients with Obstructive Sleep Apnoea in a severity-dependent manner. PMID: 29051270
  30. The results show that the distinctive pathological features of papillary thyroid carcinomas (PTCs), including tumor-infiltrating lymphocytes (TILs), background chronic lymphocytic thyroiditis (CLT), female gender, psammoma bodies, and stromal calcification, are useful parameters for predicting PD-L1 or PD-1 expression. PMID: 28974264
  31. Our results did not show any association between PDCD1 SNPs and the development of juvenile idiopathic arthritis in an Iranian population. PMID: 29307156
  32. High expressions of programmed cell death protein 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) were associated with poor prognosis after surgery. PMID: 29848685
  33. Programmed death-1 polymorphism is associated with the risk of esophagogastric junction adenocarcinoma. PMID: 28487496
  34. PD-1 was expressed in 26% of Ewing's sarcoma family of tumor cells and may have prognostic and therapeutic implications. PMID: 29445891
  35. This study shows that patients with low PD-L1 expression on myeloid cells have improved survival when treated with an antitumor vaccine, suggesting that PD-L1 expression on myeloid cells may be an important predictive biomarker in future clinical trials. Additionally, the combination of PD-1/PD-L1 inhibition and vaccination may increase the efficacy of this immunotherapeutic approach. PMID: 28193626
  36. Lower expression of PD-1 and PD-L1 was associated with better survival in patients who underwent surgery for the primary tumor and had multiple brain metastases. PMID: 28201746
  37. The PD-1/PD-L pathway inhibits Mycobacterium tuberculosis-specific CD4(+) T-cell functions and phagocytosis of macrophages in active tuberculosis. PMID: 27924827
  38. This study provides optimism that harmonization between assays may be possible, and that the three assays studied could potentially be used interchangeably to identify patients most likely to respond to anti-PD-1/PD-L1 immunotherapies, provided the appropriate clinically defined algorithm and agent are always linked. PMID: 28073845
  39. Results identified an overall low expression of PD-1 and PD-L1 in high-risk prostate cancer tissue. PMID: 28461179
  40. The biopsy tumor key protein measurements demonstrate substantial between-tumor variation in expression ratios of these proteins and suggest that programmed cell death 1 ligand 2 PD-L2 is present in some tumors at levels sufficient to contribute to programmed cell death-1 PD-1-dependent T-cell regulation and possibly to affect responses to PD-1- and programmed cell death 1 ligand 1 PD-L1-blocking drugs. PMID: 28546465
  41. Early phase clinical trials using PD-1 or PD-L1 inhibitors alone or in combination have shown objective tumor responses and durable long-term disease control, in heavily pre-treated patients, notably in the TN subtype. Blockade of PD-1 or PD-L1 shows impressive antitumor activity in some subsets of breast cancer patients. PMID: 28799073
  42. Differential expression of immunological markers relating to the PD-1/PD-L1 pathway in blood can be used as potential diagnostic and prognostic markers in ovarian cancers. These data have implications for the development and trial of anti-PD-1/PD-L1 therapy in ovarian cancer. PMID: 27986748
  43. LAG-3+ iTILs are enriched in estrogen receptor-negative breast cancers and represent an independent favorable prognostic factor. In addition, a high proportion of PD-1/PD-L1+ tumors are co-infiltrated with LAG-3+ TILs. PMID: 29045526
  44. An IDO1 inhibitor, epacadostat, also demonstrated promising activity in combination with the PD-1 checkpoint inhibitors in other solid tumors, including melanoma, urothelial carcinoma, renal cell carcinoma, and non-small cell lung cancer. PMID: 28760910
  45. PD-L1 was positive in tumor cells in 2/13 cases, weak positive in 7/13, and negative in 4/13 cases, respectively. PMID: 28807336
  46. This study reports the first case of immune microenvironment profiling and response to anti-PD-1 in a patient with Renal medullary carcinoma. This case suggests that anti-PD-1-based therapies may have clinical activity in Renal medullary carcinoma. PMID: 28105368
  47. This study presents two cases of metastatic melanoma treated with nivolumab and pembrolizumab (anti PD-1). Both patients developed acute interstitial nephritis during immune checkpoint therapy. PMID: 28105370
  48. The positive rate of PD-L2 did not show any differences between primary tumors and metastatic lymph nodes. In multivariate analysis, PD-L1 expression, PD-L2 expression, a low density of CD8(+) T cells in primary tumors, and PD-1 expression on CD8(+) T cells in primary tumors were associated with poor prognosis. PMID: 28754154
  49. Relative to controls, the expression of PD-1 and PD-L1 on peripheral blood and tumor infiltrating T cells increased with disease progression. Upregulation of expression promotes t-cells apoptosis in gastric adenocarcinoma. PMID: 29599324
  50. These data support the combinatorial approach of in situ suppression of the PD-L inhibitory checkpoints with DC-mediated IL15 transpresentation to promote antigen-specific T-cell responses and, ultimately, contribute to graft-versus-tumor immunity. PMID: 28637876

Show More

Hide All

Database Links

HGNC: 8760

OMIM: 109100

KEGG: hsa:5133

STRING: 9606.ENSP00000335062

UniGene: Hs.158297

Involvement In Disease
Systemic lupus erythematosus 2 (SLEB2)
Subcellular Location
Cell membrane; Single-pass type I membrane protein.

Q&A

What is the mechanism of action for PD-1 monoclonal antibodies in cancer immunotherapy?

When PD-1 monoclonal antibodies bind to the PD-1 receptor, they prevent its interaction with PD-L1/PD-L2, thereby removing the inhibitory signal. This blockade reinvigorates exhausted T cells, allowing them to recognize and attack tumor cells more effectively. The pathway plays critical roles in T cell coinhibition and exhaustion, with overexpression of PD-L1 and PD-1 on tumor cells and tumor-infiltrating lymphocytes correlating with poor disease outcomes in various cancers .

The expression of PD-L1 can be induced by inflammatory cytokines such as IFN-γ and TNF-α, which upregulate its expression on T cells, B cells, endothelial cells, and epithelial cells. Additionally, genetic alterations in cancer cells can trigger PD-L1 expression, though this varies by cancer type. For instance, PTEN dysfunction in human glioma cells induces PD-L1 expression through Akt activation, while human melanoma cells show no association between PTEN or Akt and PD-L1 induction .

For which cancer types have PD-1 antibodies demonstrated clinical efficacy?

PD-1 monoclonal antibodies have shown impressive response rates across several cancer types, with the most notable clinical success in:

  • Melanoma: One of the first and most responsive cancer types to PD-1 blockade

  • Non-small-cell lung cancer (NSCLC): Significant clinical benefit observed in multiple trials

  • Renal cell carcinoma: Demonstrated improved outcomes compared to standard therapies

  • Bladder cancer: Showing promising response rates in clinical studies

These variable responses highlight the need for biomarkers to predict which patients will benefit from PD-1 blockade therapy.

What are the major limitations of current PD-1 monoclonal antibody therapies?

Despite their clinical success, current PD-1 monoclonal antibodies face several key limitations:

  • Poor tissue/tumor penetrance: Antibodies are large molecules (approximately 150 kDa) that cannot efficiently penetrate solid tumors, particularly those with dense stroma or poor vascularization .

  • Detrimental Fc-effector functions: The Fc region of anti-PD-1 antibodies can engage Fcγ receptors (FcγRs), leading to depletion of activated CD8 T cells, thereby potentially counteracting the intended immunotherapeutic effect .

  • Tumor microenvironment influence: The efficacy of anti-PD-1 antibodies is significantly affected by the immune context of the tumor. "Cold" tumors with minimal immune infiltration show reduced response compared to "hot" immunologically active tumors .

  • Variable response rates: Clinical response to PD-1 blockade varies significantly among patients and cancer types, with many patients showing primary or acquired resistance .

  • Immune-related adverse events: While not detailed in the search results, these therapies can trigger immune-related toxicities due to enhanced immune activation.

Research has shown that engagement of activating FcγRs by anti-PD-1 mAbs leads to depletion of activated CD8 T cells both in vitro and in vivo, which can abrogate therapeutic activity . This effect is particularly pronounced in certain immune environments, highlighting the complex interplay between antibody design and the tumor microenvironment in determining therapeutic outcomes.

How can high-affinity PD-1 variants be engineered for optimized immunotherapy?

Engineering high-affinity PD-1 variants represents an innovative approach to overcome limitations of conventional antibody-based checkpoint inhibitors. Researchers have successfully used directed evolution with yeast-surface display to generate PD-1 variants with dramatically enhanced affinity for PD-L1.

The process involves a systematic two-library approach:

  • First-generation library: Identification of mutational "hotspots" that confer large affinity gains

    • Crystal structure analysis identified 22 residues at the PD-1:PD-L1 interface for randomization

    • Four rounds of selection using biotinylated human PD-L1 ectodomain

    • Resulting variants showed 400-500-fold increase in affinity but had poor biochemical properties

  • Second-generation library: Optimizing beneficial mutations while eliminating detrimental ones

    • Focused on positions showing clear selection preferences

    • Further selection yielded variants with improved affinity and biochemical characteristics

This approach yielded high-affinity PD-1 variants (HAC-PD-1) with approximately 35,000-fold enhanced affinity for PD-L1 (K<sub>D</sub> of 110 pM compared to 8.2 μM for wild-type PD-1). These engineered PD-1 variants demonstrated superior tumor penetration compared to anti-PD-L1 monoclonal antibodies without inducing depletion of peripheral effector T cells .

The HAC-PD-1 variant competitively antagonized PD-L1 on human SK-MEL-28 cells with an IC<sub>50</sub> of 210 pM, representing a 40,000-fold enhancement in potency compared to blockade with wild-type monomeric human PD-1 (IC<sub>50</sub> of 8.2 μM) . To improve interaction with mouse PD-L1 for in vivo studies, researchers created a high-affinity "microbody" (HACmb) by fusing HAC-PD-1 to a dimeric CH3 domain, which showed potent blocking of both human and mouse PD-L1 .

What role do Fc:FcγR interactions play in the efficacy of PD-1 monoclonal antibodies?

Fc:FcγR interactions significantly impact the therapeutic efficacy of PD-1 monoclonal antibodies, often in ways that can diminish their intended immunostimulatory effects. Research has revealed several critical aspects of these interactions:

  • Depletion of activated T cells: Engagement of activating FcγRs by anti-PD-1 mAbs leads to depletion of activated CD8 T cells both in vitro and in vivo, potentially abrogating therapeutic activity .

  • Immune environment dependence: The impact of Fc-mediated modulation is determined by the surrounding immune environment, with varying effects in different tumor contexts .

  • Isotype influence: Low FcγR-engaging mouse anti-PD-1 isotypes (often used as surrogates for human mAbs) show impaired ability to expand antigen-specific CD8 T cells compared to Fc-null mAbs .

  • Human FcγR interactions: In humanized mouse models expressing human FcγRs, clinically relevant human IgG4 anti-PD-1 antibodies showed reduced expansion of CD8 T cells compared to Fc-null counterparts .

  • Tumor model specificity: In "hot" immunologically active tumors, both low-engaging and Fc-null mAbs could induce long-term antitumor immunity, while in "cold" tumor models, the optimal anti-PD-1 isotype could delay tumor growth but not induce long-term protection .

These findings suggest that antibody engineering to minimize FcγR engagement (Fc-null designs) may be critical for maximizing the therapeutic efficacy of PD-1 blockade, particularly in immunologically "cold" tumors or early treatment settings where preserving activated T cells is crucial.

How can PD-1 variants be utilized as imaging tracers for PD-L1 expression?

High-affinity PD-1 variants can be repurposed as PET imaging tracers to non-invasively assess PD-L1 expression in tumors, providing an alternative to invasive biopsies and histological analysis. This approach, termed "Immuno-PET," enables simultaneous measurement of PD-L1 expression throughout an entire tumor.

The development process involves:

  • Engineered PD-1 modification: A mutated high-affinity PD-1 variant (HAC-N91C) is conjugated with a thiol-reactive bifunctional chelate DOTA-maleimide .

  • Radiolabeling: The conjugate is labeled with a radioactive isotope such as <sup>64</sup>Cu for PET imaging .

  • Validation: The radiolabeled tracer demonstrates the ability to distinguish between PD-L1-positive and PD-L1-negative tumors in living subjects .

Despite having slightly weaker apparent affinity for human PD-L1 than its parent sequence, the DOTA-conjugated HAC variant still antagonized human PD-L1 approximately 1,200-fold more potently than wild-type PD-1 .

This approach offers several advantages over conventional biopsy methods:

  • Non-invasive whole-tumor assessment

  • Ability to detect heterogeneous PD-L1 expression

  • Potential for longitudinal monitoring

  • Avoidance of sampling errors associated with biopsies

PD-L1 expression in tumors (by tumor cells or stroma) has been suggested as a potential biomarker to predict response to PD-1 or PD-L1-directed immunotherapies . The ability to assess this expression non-invasively could significantly improve patient selection and therapeutic monitoring.

What methodological approaches are used to assess anti-PD-1 monoclonal antibody efficacy in preclinical models?

Assessment of anti-PD-1 monoclonal antibody efficacy in preclinical models employs multiple complementary approaches:

  • Competitive binding assays:

    • Cell-based assays using PD-L1-expressing tumor cell lines (e.g., SK-MEL-28, B16-F10)

    • Measurement of IC<sub>50</sub> values for blockade of wild-type PD-1 binding to PD-L1

    • Yeast-display systems for evaluating binding to recombinant PD-L1/PD-L2

  • Functional T cell assays:

    • Assessment of T cell expansion following vaccination with model antigens (e.g., ovalbumin)

    • Flow cytometric analysis of T cell activation and proliferation markers

    • Measurement of cytokine production by activated T cells

  • Tumor models with varying immune profiles:

    • "Hot" immunologically active tumors (e.g., MC38 colon carcinoma)

    • "Cold" immune-excluded tumors (e.g., 9464D neuroblastoma)

    • Assessment of differential responses based on tumor immunological status

  • Pharmacokinetic and biodistribution studies:

    • Radiolabeling of antibodies or engineered PD-1 variants

    • PET imaging to assess tumor penetration and tissue distribution

    • Comparison between different antibody formats or engineered proteins

  • Therapeutic efficacy assessment:

    • Tumor growth inhibition in syngeneic mouse models

    • Survival analysis

    • Evaluation of durable responses and tumor recurrence

    • Analysis of tumor infiltrating lymphocytes by flow cytometry

In one systematic approach, researchers used high-affinity PD-1 variants in the CT26 tumor model and found they were effective in treating both small (50 mm<sup>3</sup>) and large tumors (150 mm<sup>3</sup>), whereas anti-PD-L1 antibodies showed completely abrogated activity against large tumors . This demonstrates the importance of using multiple tumor models and tumor sizes when assessing therapeutic efficacy.

What factors contribute to resistance to PD-1 monoclonal antibody therapy?

Resistance to PD-1 monoclonal antibody therapy remains a significant challenge, with both primary (innate) and acquired (developed during treatment) resistance observed. Several key factors contribute to therapeutic resistance:

  • Tumor Immunological Status:

    • "Cold" tumor microenvironments with limited T cell infiltration demonstrate reduced response to PD-1 blockade

    • The efficacy of even optimally-designed anti-PD-1 antibodies is significantly diminished in immunologically "cold" tumors like neuroblastoma compared to "hot" tumors

  • Fc-Mediated Effects:

    • Engagement of activating FcγRs by anti-PD-1 antibodies can deplete activated CD8 T cells

    • This depletion may counteract the intended immunostimulatory effects of PD-1 blockade

    • The magnitude of this effect varies with the immune environment and antibody isotype

  • Tumor Size and Penetration Limitations:

    • Large tumors show decreased response to conventional antibodies

    • Poor tumor penetration of large antibody molecules (~150 kDa) limits efficacy

    • In the CT26 tumor model, anti-PD-L1 antibodies showed completely abrogated activity against large tumors (150 mm<sup>3</sup>), while smaller engineered PD-1 variants maintained efficacy

  • MGMT Methylation Status:

    • In glioblastoma, neither methylated nor unmethylated MGMT status benefited from anti-PD-1 monoclonal antibody treatment

    • This suggests that certain molecular features may predict resistance to PD-1 blockade

  • PD-L1 Expression Heterogeneity:

    • Variable PD-L1 expression within tumors creates challenges for effective therapy

    • Spatial heterogeneity of PD-L1 expression complicates assessment by conventional biopsy

These resistance factors highlight the need for more sophisticated approaches, including combination therapies, biomarker-guided patient selection, and development of next-generation checkpoint inhibitors with improved properties like the engineered high-affinity PD-1 variants.

How should researchers design experiments to compare different anti-PD-1 monoclonal antibody formats?

When designing experiments to compare different anti-PD-1 monoclonal antibody formats, researchers should implement a comprehensive approach that addresses multiple aspects of antibody function:

  • Binding affinity and specificity assessment:

    • Surface plasmon resonance (SPR) to determine K<sub>D</sub> values

    • Competitive binding assays on cells expressing PD-L1

    • Cross-reactivity testing with related proteins (e.g., PD-L2)

  • Fc-mediated effects evaluation:

    • Compare antibodies with different Fc regions (IgG1, IgG4, Fc-null variants)

    • Use in vitro assays to assess antibody-dependent cellular cytotoxicity (ADCC)

    • Evaluate T cell depletion in both in vitro and in vivo systems

  • Multiple tumor models:

    • Include both "hot" (immunologically active) and "cold" (immune-excluded) tumor models

    • Test efficacy against tumors of different sizes (e.g., 50 mm<sup>3</sup> vs. 150 mm<sup>3</sup>)

    • Evaluate in models with variable PD-L1 expression levels

  • Comprehensive immune profiling:

    • Flow cytometry to analyze T cell activation, exhaustion, and proliferation

    • Assessment of tumor-infiltrating lymphocytes

    • Analysis of cytokine profiles in the tumor microenvironment

  • Humanized mouse models:

    • Use of mice expressing human FcγRs for testing clinically relevant antibody formats

    • Testing in models that better recapitulate human immune responses

A robust experimental design would include controls such as:

  • Isotype control antibodies

  • Wild-type PD-1 for comparison with engineered variants

  • Multiple dose levels to establish dose-response relationships

  • Longitudinal studies to assess durability of response

What analytical techniques are essential for characterizing engineered PD-1 variants?

Characterization of engineered PD-1 variants requires a diverse set of analytical techniques to fully understand their properties and therapeutic potential:

  • Biophysical characterization:

    • Surface plasmon resonance (SPR) for binding kinetics and affinity (K<sub>D</sub>)

    • Size-exclusion chromatography to assess aggregation tendency

    • Circular dichroism spectroscopy for secondary structure analysis

    • Thermal stability assessment (e.g., differential scanning calorimetry)

  • Functional assessment:

    • Competitive binding assays on PD-L1-expressing cells

    • IC<sub>50</sub> determination for blockade of PD-1:PD-L1 interaction

    • T cell activation assays to confirm biological activity

    • Cross-reactivity testing with related proteins (e.g., PD-L2)

  • Structural analysis:

    • X-ray crystallography or cryo-EM to determine molecular interactions

    • Epitope mapping to confirm binding interfaces

    • Computational modeling to predict and analyze mutations

  • In vivo biodistribution and pharmacokinetics:

    • Radiolabeling techniques (e.g., with <sup>64</sup>Cu)

    • PET imaging for tissue distribution assessment

    • Pharmacokinetic profiling of serum half-life and clearance

  • Production and yield evaluation:

    • Expression yield quantification in various production systems

    • Purification efficiency assessment

    • Stability testing under various storage conditions

For directed evolution approaches using yeast display, analytical techniques include:

  • Flow cytometry for sorting and selection of variants

  • Deep sequencing to identify mutation patterns and frequencies

  • Sequence-function correlation analysis

In the development of high-affinity PD-1 variants, researchers observed that first-generation libraries often contained variants with improved affinity but poor biochemical behavior (decreased expression yield, aggregation tendency). This highlighted the importance of comprehensive characterization beyond simple affinity measurements .

How should researchers interpret contradictory results from different tumor models in anti-PD-1 studies?

Interpreting contradictory results from different tumor models in anti-PD-1 studies requires careful consideration of multiple factors that influence treatment response:

  • Tumor immunological status assessment:

    • Characterize baseline immune infiltration in each model

    • Analyze "hot" versus "cold" tumor microenvironments

    • Quantify pre-existing antitumor immunity levels

    • Measure baseline PD-L1 expression on tumor and immune cells

  • Model-specific characteristics evaluation:

    • Consider tumor growth kinetics differences

    • Assess tumor location/tissue context

    • Evaluate vascularity and accessibility differences

    • Account for genetic backgrounds of tumor models

  • Therapeutic agent properties:

    • Analyze antibody isotype and Fc receptor engagement profiles

    • Consider molecular size and tumor penetration capabilities

    • Evaluate species cross-reactivity issues

    • Assess binding affinities to human vs. mouse targets

  • Experimental design variables:

    • Compare treatment timing (early vs. established tumors)

    • Analyze dosing regimens across studies

    • Consider route of administration differences

    • Evaluate concurrent treatments or interventions

When faced with contradictory results, researchers should:

  • Perform comprehensive immune profiling of responder vs. non-responder models

  • Test multiple antibody formats in the same models

  • Validate findings across multiple independent models

  • Consider tumor size effects, as demonstrated in the CT26 model where anti-PD-L1 antibodies showed completely abrogated activity against large tumors (150 mm<sup>3</sup>) while maintaining effectiveness against small tumors (50 mm<sup>3</sup>)

The research literature demonstrates significant model-dependent effects. For example, in "hot" immunologically active MC38 tumors, both low-FcγR-engaging and Fc-null antibodies induced long-term antitumor immunity, while in "cold" 9464D neuroblastoma models, even optimally designed antibodies could only delay tumor growth without inducing long-term protection .

What considerations are important when translating preclinical findings on PD-1 antibodies to clinical applications?

Translating preclinical findings on PD-1 antibodies to clinical applications requires addressing several critical considerations to maximize therapeutic success:

  • Species differences in immune systems:

    • Account for variations in PD-1/PD-L1 expression and regulation

    • Consider differences in Fc receptor distribution and function

    • Use humanized mouse models expressing human FcγRs when possible

    • Validate findings in multiple preclinical models

  • Antibody format optimization:

    • Carefully consider antibody isotype selection

    • Evaluate Fc-mediated effects on T cell populations

    • Consider engineered variants with modified Fc regions or Fc-null designs

    • Balance tumor penetration against serum half-life

  • Tumor heterogeneity assessment:

    • Account for differences between preclinical models and human tumors

    • Consider tumor size effects observed in preclinical models

    • Develop strategies for "cold" tumors with limited immune infiltration

    • Address PD-L1 expression heterogeneity

  • Biomarker development:

    • Identify predictive biomarkers of response from preclinical studies

    • Develop companion diagnostics (e.g., PD-L1 expression assessment)

    • Consider non-invasive imaging approaches for PD-L1 detection

    • Validate biomarkers across multiple models

  • Combination therapy strategies:

    • Identify synergistic combinations from preclinical studies

    • Consider sequencing of therapies

    • Address potential antagonistic effects

    • Evaluate safety profiles of combinations

Researchers should be aware that antibody formats that perform well in preclinical models may have unexpectedly different effects in humans. For example, the clinically relevant human IgG4 anti-PD-1 format showed reduced endogenous expansion of CD8 T cells compared with engineered Fc-null counterparts in mice expressing human FcγRs .

The development of high-affinity PD-1 variants demonstrates the potential for non-antibody biologics to overcome limitations of conventional antibodies, particularly in terms of tumor penetration and avoiding detrimental Fc-mediated effects .

What emerging approaches aim to overcome resistance to PD-1 monoclonal antibody therapy?

Several innovative approaches are being explored to overcome resistance to PD-1 monoclonal antibody therapy:

  • Engineered high-affinity PD-1 variants:

    • Non-antibody biologics based on the PD-1 ectodomain with dramatically enhanced affinity

    • Demonstrated superior tumor penetration compared to antibodies

    • Effective against large tumors where antibodies fail

    • Avoid detrimental Fc-mediated effects that can deplete T cells

  • Fc-engineered antibodies:

    • Development of Fc-null antibodies that minimize engagement with FcγRs

    • Optimization of Fc regions to avoid depletion of activated T cells

    • Isotype selection based on tumor microenvironment context

  • Combination therapy approaches:

    • Pairing PD-1 blockade with other checkpoint inhibitors

    • Combining with strategies to convert "cold" tumors to "hot" tumors

    • Integration with conventional therapies (radiation, chemotherapy)

    • Complementary targeting of multiple immune pathways

  • Biomarker-guided therapy:

    • Development of PET imaging tracers based on high-affinity PD-1 variants

    • Non-invasive assessment of PD-L1 expression throughout tumors

    • Identification of molecular signatures predicting response or resistance

  • Alternative formats and delivery systems:

    • Smaller antibody fragments with improved tumor penetration

    • Bispecific antibodies targeting PD-1 and other immune checkpoints

    • Local delivery approaches to increase intratumoral concentration

Research has demonstrated that high-affinity PD-1 variants can overcome key limitations of antibodies. In the CT26 tumor model, engineered PD-1 was effective in treating both small (50 mm<sup>3</sup>) and large tumors (150 mm<sup>3</sup>), whereas anti-PD-L1 antibodies showed completely abrogated activity against large tumors . This highlights the potential of novel protein formats to address resistance mechanisms related to tumor penetration.

Understanding the impact of Fc:FcγR interactions has led to the development of antibodies with optimized Fc regions, potentially preventing the depletion of activated CD8 T cells that can counteract therapeutic efficacy .

How might directed evolution approaches be optimized for next-generation PD-1 targeting therapeutics?

Directed evolution approaches for next-generation PD-1 targeting therapeutics can be optimized through several strategic enhancements:

  • Advanced library design strategies:

    • Structure-guided focused libraries targeting key interface residues

    • Computational prediction of beneficial mutations

    • Machine learning approaches to predict mutation impact

    • Combinatorial libraries exploring synergistic mutations

  • Multi-property optimization:

    • Simultaneous selection for affinity, stability, and expression

    • Negative selection steps to eliminate variants with poor biophysical properties

    • Sequential rounds with alternating selection pressures

    • Balance between affinity enhancement and maintenance of biological function

  • Alternative display technologies:

    • Combining yeast display with other platforms (phage, mammalian)

    • Cell-free display systems for larger library sizes

    • In vivo directed evolution approaches

    • Ribosome display for unbiased selection

  • Enhanced selection strategies:

    • Alternating positive and negative selection

    • Gradient selections with decreasing target concentrations

    • Competition-based selections mimicking physiological contexts

    • Multi-parameter FACS for simultaneous property optimization

  • Post-selection engineering:

    • Rational stabilization of evolved variants

    • Humanization of non-human scaffolds

    • PEGylation or half-life extension strategies

    • Multimerization approaches for avidity effects

The two-library approach used for engineering high-affinity PD-1 variants demonstrates the value of iterative optimization. The first-generation library identified mutational "hotspots" but yielded variants with poor biochemical behavior. The second-generation library then focused on positions showing clear selection preferences, resulting in variants with both improved affinity and biochemical characteristics .

Future approaches might include deep mutational scanning to comprehensively map the fitness landscape of PD-1, allowing more precise library design and potentially revealing non-obvious beneficial mutations that might be missed in conventional approaches.

What are the prospects for developing combination biomarkers to predict response to PD-1 monoclonal antibody therapy?

Developing combination biomarkers to predict response to PD-1 monoclonal antibody therapy represents a critical research direction with significant potential to improve patient selection and therapeutic outcomes:

  • PD-L1 expression assessment innovations:

    • Non-invasive PET imaging using radiolabeled high-affinity PD-1 variants

    • Comprehensive assessment of spatial heterogeneity throughout tumors

    • Dynamic monitoring of PD-L1 expression changes during treatment

    • Integration of PD-L1 expression on tumor cells versus immune cells

  • Tumor immune microenvironment characterization:

    • Multiplex immunohistochemistry to assess immune cell infiltration

    • Spatial analysis of "hot" versus "cold" tumor regions

    • Gene expression profiling of immune activation signatures

    • Assessment of tertiary lymphoid structure formation

  • Genetic and molecular tumor features:

    • Tumor mutational burden assessment

    • Microsatellite instability status

    • DNA damage repair pathway alterations

    • Oncogenic driver mutation analysis

    • MGMT methylation status in relevant tumors

  • Circulating biomarkers:

    • Peripheral immune cell phenotyping

    • Soluble checkpoint molecule quantification

    • Circulating tumor DNA analysis

    • Exosome profiling

  • Integrative multi-omics approaches:

    • Combination of genomics, transcriptomics, and proteomics

    • Machine learning algorithms to identify predictive signatures

    • Systems biology analysis of pathway interactions

    • Longitudinal assessment before and during treatment

The development of radiolabeled high-affinity PD-1 variants as PET imaging tracers offers a promising approach for non-invasive assessment of PD-L1 expression throughout entire tumors, potentially addressing the limitations of conventional biopsy-based testing which can miss spatial heterogeneity .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.