PDCD1 Antibody

Programmed cell death protein 1, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Introduction to PDCD1 Antibody

PDCD1 antibodies are immunotherapeutic agents targeting programmed cell death protein 1 (PD-1/CD279), an immune checkpoint receptor expressed on activated T cells, B cells, and macrophages. These antibodies block PD-1 interactions with its ligands (PD-L1/PD-L2), reversing immune suppression and restoring anti-tumor T cell activity . Clinically, PDCD1 antibodies are pivotal in treating cancers such as melanoma, non-small cell lung carcinoma (NSCLC), and Hodgkin’s lymphoma .

Mechanism of Action

PDCD1 antibodies function by:

  • Blocking ligand binding: Preventing PD-1 engagement with PD-L1/PD-L2, thereby inhibiting downstream immunosuppressive signaling .

  • Restoring T cell effector function: Reversing T cell exhaustion (a state of functional impairment in chronic infections/cancer) .

  • Modulating phosphatase activity: Disrupting SHP-2 recruitment, which normally dephosphorylates TCR signaling molecules (e.g., ZAP70) .

Therapeutic Applications

PDCD1 antibodies are FDA-approved for:

Comparative Efficacy of PD-1 vs. PD-L1 Antibodies

ParameterPD-1 Antibodies (e.g., nivolumab)PD-L1 Antibodies (e.g., atezolizumab)
Blocking efficiencyModerateSuperior
Adverse event incidenceHigher (e.g., pneumonitis)Lower
Target ligandsPD-L1 + PD-L2PD-L1 only

Biomarkers for PDCD1 Antibody Response

BiomarkerRelevanceExample Cancer Types
PD-L1 expressionHigh expression correlates with better response (e.g., NSCLC) NSCLC, melanoma
Tumor mutational burdenTMB-H tumors respond better due to neoantigen load Colorectal, endometrial
MSI/dMMR statusDefective DNA repair enhances immunotherapy efficacy Colorectal, gastric

Preclinical Insights

  • HIV/Chronic infections: PD-1 blockade enhances antiviral T cell responses but risks immunopathology .

  • Alzheimer’s disease: Anti-PD-1 reduces amyloid-β plaques in mice via IFN-γ-mediated macrophage recruitment .

Notable Trials and Outcomes

AntibodyTrial PhaseCancer TypeKey OutcomeReference
PembrolizumabIIINSCLC30% reduced mortality vs. chemotherapy
NivolumabIIIMelanoma5-year survival: 34% (vs. 16% for chemo)
CemiplimabIICutaneous SCC47% objective response rate

Current Challenges and Limitations

  • Response variability: Only 20–40% of patients achieve durable responses .

  • Immune-related adverse events: Pneumonitis, colitis, and hepatitis occur in 10–20% of patients .

  • Biomarker limitations: PD-L1 expression alone is insufficient for predicting outcomes .

Future Directions

  1. Combination therapies: Pairing PDCD1 antibodies with anti-CTLA-4, chemotherapy, or radiotherapy to enhance efficacy .

  2. Neoadjuvant/adjuvant use: Early-stage trials show promise in reducing recurrence (e.g., NSCLC) .

  3. Next-gen biomarkers: Integrating TMB, gut microbiome, and multiplex imaging for personalized therapy .

Product Specs

Introduction
Programmed cell death protein 1 (PDCD1), also known as PD-1, is a transmembrane protein expressed on the surface of activated T cells, B cells, and macrophages. It plays a critical role in regulating immune responses and preventing autoimmune reactions. PDCD1 interacts with its ligands, PD-L1 and PD-L2, which are expressed on tumor cells and other cells in the tumor microenvironment. This interaction inhibits T cell activation, proliferation, and cytokine production, leading to immune suppression and tumor evasion. PDCD1 has emerged as a promising target for cancer immunotherapy, and monoclonal antibodies that block PD-1 signaling have shown remarkable clinical efficacy in various cancers.
Formulation
This antibody is supplied as a 1 mg/ml solution in phosphate-buffered saline (PBS) at pH 7.4, containing 0.1% sodium azide as a preservative.
Storage Procedures
For short-term storage (up to 1 month), store the antibody at 4°C. For long-term storage, store at -20°C. Avoid repeated freeze-thaw cycles to maintain antibody stability and activity.
Stability / Shelf Life
The antibody is stable for 12 months when stored at -20°C and for 1 month when stored at 4°C.
Applications
This PDCD1 antibody has been validated for use in ELISA and Western blot applications. The recommended dilution range for Western blot analysis is 1:500 to 1:1000, with an optimal starting dilution of 1:500. However, the optimal working dilution may vary depending on the specific application and experimental conditions. Users are advised to determine the optimal dilution for their experiments.
Synonyms
Programmed cell death protein 1, Protein PD-1, hPD-1, CD279, PDCD1, PD1, SLEB2.
Purification Method
PDCD1 antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
P4F12AT.
Immunogen
Anti-human PDCD1 mAb, is derived from hybridization of mouse FO myeloma cells with spleen cells from BALB/c mice immunized with recombinant human PDCD1 amino acids 21-167 purified from E. coli.
Ig Subclass
Mouse IgG2b heavy chain and κ light chain.

Q&A

What is the biological function of PDCD1/PD-1 and how does it regulate immune responses?

PDCD1/PD-1 is a transmembrane protein that functions as a T cell checkpoint and plays a central role in regulating T cell exhaustion. Following T-cell receptor (TCR) engagement, PD-1 associates with CD3-TCR in the immunological synapse and directly inhibits T-cell activation. It delivers inhibitory signals upon binding to its ligands CD274/PDCD1L1 (PD-L1) and CD273/PDCD1LG2 (PD-L2) .

The inhibitory mechanism involves a signaling cascade where PD-1 is phosphorylated within its ITSM motif after ligand binding. This leads to the recruitment of the protein tyrosine phosphatase PTPN11/SHP-2 that mediates dephosphorylation of key TCR proximal signaling molecules, such as ZAP70, PRKCQ/PKCtheta, and CD247/CD3zeta . This pathway is exploited by tumors to attenuate anti-tumor immunity and escape destruction by the immune system, thereby facilitating tumor survival .

The PD-1/PD-L1 pathway plays important roles across multiple contexts including autoimmune diseases, viral infections, transplantation immunology, and tumor immunity .

What are the main types of anti-PD-1 antibodies available for research and how do they differ?

Anti-PD-1 antibodies can be categorized based on several characteristics:

  • Species reactivity:

    • Anti-mouse PD-1 antibodies: Common clones include RMP1-14, 29F.1A12, and J43

    • Anti-human PD-1 antibodies: Various clones including those similar to therapeutic antibodies (nivolumab, pembrolizumab)

  • Isotype and origin:

    • Rat IgG2a, κ (RMP1-14)

    • Rat IgG2a (29F.1A12)

    • Armenian Hamster IgG (J43)

  • Application suitability:

    • Some antibodies are specialized for in vivo blocking (e.g., RMP1-14 has extensive publication records for this purpose)

    • Others have broader application profiles (e.g., 29F.1A12 and J43 can be used for in vitro neutralization, western blotting, immunohistochemistry, immunofluorescence, and flow cytometry)

  • Epitope recognition:

    • Antibodies target different epitopes on PD-1, with at least 10 distinct epitope bins identified

    • Some antibodies block PD-1/PD-L1 interaction while others do not

The differences between these antibodies have significant implications for research applications and experimental outcomes.

How do anti-PD-1 antibody binding properties relate to their functional effects?

Anti-PD-1 antibodies exhibit a remarkable diversity in their binding properties that directly impacts their functional effects:

These binding characteristics should be carefully considered when selecting antibodies for specific research applications.

What methodological approach should be used to select the optimal anti-PD-1 antibody for a specific research application?

Selecting the appropriate anti-PD-1 antibody requires a systematic approach considering multiple factors:

  • Species reactivity determination:

    • Identify which species you're studying (human, mouse, etc.)

    • Select antibodies validated for your target species (e.g., RMP1-14, 29F.1A12, or J43 for mouse studies)

  • Application-specific selection:

    • For in vivo PD-1 blocking: Choose antibodies with established in vivo efficacy records

    • For flow cytometry: Select antibodies validated for flow, potentially with appropriate conjugation

    • For multiple applications: Consider clones like 29F.1A12 or J43 that work across various techniques

  • Literature-based validation:

    • Search for published studies using your specific experimental model

    • Use specific search terms (e.g., "RMP1-14 MC38 BALB/c")

    • Review the methodology sections of relevant papers for dosing strategies and protocols

  • Epitope consideration:

    • Determine whether complete blockade of PD-1/PD-L1 interaction is required

    • For mechanistic studies, choose antibodies with well-characterized epitopes

  • Validation through preliminary experiments:

    • Test multiple candidates in small-scale pilot studies

    • Include appropriate controls, especially isotype controls matching the antibody being tested

This methodical approach helps ensure the selected antibody will perform optimally in your specific research context.

How can surface plasmon resonance (SPR) be optimized for characterizing anti-PD-1 antibody binding properties?

Based on published industry collaborations, optimal SPR methodology for anti-PD-1 antibody characterization involves:

  • Capture assay design:

    • Implement anti-human Fc capture assays for direct comparison between platforms

    • Capture each mAb onto multiple discrete spots at varying capacities to populate a comprehensive array

    • This enables parallel comparison of kinetic rate and affinity constants

  • Analyte concentration series:

    • Use a wide concentration range of PD-1 protein (e.g., 0.5 nM–1000 nM for higher affinity antibodies)

    • Adjust concentration ranges based on expected affinities (higher concentrations for lower-affinity interactions)

  • Replication strategy:

    • Incorporate replicate ligand measurements into the experimental design

    • This allows binding kinetics to be reported with statistical confidence

  • Epitope binning methodology:

    • Perform pairwise competition or "epitope binning" experiments via high-throughput SPR

    • Represent results as graphed network plots colored by bin

    • Combine with orthogonal data to categorize antibodies into sub-bins

  • Data analysis approach:

    • Apply global fitting methods to determine kon, koff, and KD values

    • Report complete binding parameters with statistical confidence intervals

    • Generate overlay plots of sensorgrams with global fits to visualize binding quality

This comprehensive SPR approach enables detailed characterization of anti-PD-1 antibodies, revealing important differences in binding properties that may impact their research applications.

What controls are essential when using anti-PD-1 antibodies in experimental systems?

Robust experimental design with anti-PD-1 antibodies requires several types of controls:

  • Isotype controls:

    • Match the isotype, species, and format of the experimental antibody

    • For RMP1-14 (Rat IgG2a, κ), use an irrelevant Rat IgG2a, κ antibody

    • For J43 (Armenian Hamster IgG), use an irrelevant Armenian Hamster IgG antibody

    • These control for non-specific effects related to the antibody's constant regions

  • Biological controls:

    • Positive expression controls: Samples known to express PD-1 (e.g., activated T cells)

    • Negative expression controls: Samples known not to express PD-1

    • PD-1 knockout or knockdown models where available

  • Technical controls for specific applications:

    • Flow cytometry: Include fluorescence-minus-one (FMO) controls

    • IHC: Include secondary-antibody-only controls

    • Western blot: Include size markers and loading controls

  • Experimental protocol controls:

    • For in vivo experiments: Include untreated groups and vehicle-treated groups

    • For blocking experiments: Include non-blocking antibody controls

  • Validation controls:

    • Cross-validation with different antibody clones targeting the same protein

    • Orthogonal methods to verify observations (e.g., correlating protein detection with mRNA expression)

Proper implementation of these controls enables accurate interpretation of results and identification of potential artifacts or non-specific effects.

How can I design experiments to distinguish between anti-PD-1 antibodies with different epitope specificities?

Differentiating anti-PD-1 antibodies based on epitope recognition requires a multi-faceted approach:

  • Epitope binning via competitive binding:

    • Perform pairwise competition binding experiments using high-throughput SPR

    • Generate network plots to visualize epitope relationships and groupings

    • This approach has revealed at least 10 distinct epitope bins among anti-PD-1 antibodies

  • Functional blocking analysis:

    • Test each antibody's ability to block PD-1/PD-L1 interaction

    • Categorize antibodies based on blocking capacity (e.g., complete blockers, partial blockers, non-blockers)

    • Some antibodies (like mAb05, mAb12, and mAb30) were unable to block binding to PD-L1, while others showed differential blocking capabilities

  • Displacement studies:

    • Analyze whether antibodies can displace one another from PD-1

    • Quantify displacement kinetics to determine spatial relationships

    • Several anti-PD-1 mAbs have been shown to displace one another, implying they target closely adjacent or minimally overlapping epitopes

  • Structural analysis:

    • Use X-ray crystallography or cryo-EM to determine binding interfaces

    • Compare binding orientations and angles of different antibodies against PD-1

    • Computational docking can provide preliminary insights when structural data is unavailable

  • Correlation with functional outcomes:

    • Test different epitope-binding antibodies in functional assays

    • Determine whether epitope specificity correlates with functional effects

    • Map critical functional epitopes based on these correlations

This systematic epitope mapping approach provides crucial insights for both basic research and therapeutic antibody development.

What methodological approach is optimal for using AI protein diffusion to discover novel anti-PD-1 antibodies?

Based on recent research, AI protein diffusion for anti-PD-1 antibody discovery follows this methodological framework:

  • Training data preparation:

    • Compile a large corpus of existing anti-PD-1 antibodies

    • Use this dataset to conditionally guide the AI system

  • Constraint implementation:

    • Constrain the binding site on PD-1 to target the normal interface between PD-1 and PD-L1

    • This interface is also the binding site for FDA-approved antibodies like pembrolizumab and nivolumab

  • Generation and screening pipeline:

    • Generate multiple antibody candidates through protein diffusion

    • Screen candidates in silico for desirable binding performance

    • Analyze binding orientation and angles against PD-1

  • Comparative assessment:

    • Compare AI-generated antibodies with existing therapeutic antibodies

    • Evaluate binding affinities and structural properties

    • Assess whether candidates bind similarly to other existing therapeutic antibodies

  • Selection criteria:

    • Identify candidates with favorable predicted binding performance

    • Select diverse binding modes to maximize discovery potential

    • Prioritize candidates for experimental validation

  • Experimental validation strategy:

    • Express selected antibody fragments

    • Confirm binding using biophysical methods (SPR, BLI)

    • Evaluate functional blocking in cell-based assays

This AI-driven approach represents a significant advancement in antibody discovery methodology, potentially accelerating the development of novel anti-PD-1 therapeutics with improved properties .

What are the methodological considerations for interpreting conflicting results from different anti-PD-1 antibody clones?

When faced with conflicting results from different anti-PD-1 antibody clones, consider these methodological approaches:

  • Epitope analysis:

    • Determine if antibodies bind to different epitopes on PD-1

    • High-throughput SPR has revealed at least 10 distinct epitope bins

    • Differences in epitope targeting can lead to different functional outcomes despite targeting the same protein

  • Binding property assessment:

    • Compare binding affinities (KD values) across antibodies

    • Anti-PD-1 antibodies can span a >4,000-fold range in binding affinities

    • Analyze binding kinetics (kon and koff rates) which may be more important than equilibrium affinity

  • Isotype effect evaluation:

    • Consider whether differences are due to antibody isotypes rather than target recognition

    • Different isotypes (e.g., Rat IgG2a vs. Armenian Hamster IgG) have different Fc-mediated effects

    • Always use proper isotype controls in comparative studies

  • Experimental model analysis:

    • Determine if model-dependent factors contribute to variability

    • Different tumor models may show variable responses to the same antibody

    • Factors like immune infiltration levels, PD-L1 expression, and tumor microenvironment contribute to variability

  • Technical standardization:

    • Ensure standardized conditions when comparing antibodies:

      • Equivalent concentrations and dosing schedules

      • Consistent experimental protocols

      • Identical readouts and endpoints

  • Direct head-to-head comparison:

    • Conduct side-by-side experiments under identical conditions

    • Test in multiple experimental models

    • Use multi-parameter analysis to comprehensively assess differences

This systematic approach allows researchers to determine whether conflicting results reflect true biological differences in antibody function or are artifacts of experimental design.

What are the emerging methodologies for optimizing anti-PD-1 antibody combinations with other immunotherapies?

Optimizing anti-PD-1 combination therapies requires sophisticated methodological approaches:

  • Mechanistic rationale-based selection:

    • Target complementary pathways to PD-1/PD-L1 (e.g., CTLA-4, LAG-3, TIM-3)

    • The PD-1/PD-L1 pathway plays roles in multiple immune contexts, providing numerous combination opportunities

    • Combinations should address distinct aspects of tumor immune evasion

  • Sequential vs. concurrent administration:

    • Test different timing strategies systematically:

      • Concurrent administration

      • Sequential administration (PD-1 blockade before or after partner therapy)

      • Intermittent scheduling to minimize toxicity

  • Dose optimization methodology:

    • Conduct dose-response studies for both single agents

    • Test multiple combination ratios rather than only maximum tolerated doses

    • Consider pharmacokinetic and pharmacodynamic interactions

  • Comprehensive endpoint analysis:

    • Multi-parameter assessment of tumor microenvironment:

      • Immune cell infiltration and phenotyping

      • Spatial relationships between tumor and immune cells

      • Functional status of tumor-infiltrating lymphocytes

    • Correlation of these parameters with therapeutic response

  • Predictive biomarker development:

    • Identify biomarkers that predict response to combination therapy

    • Develop companion diagnostics for patient selection

    • The blockage of the PD-1-mediated pathway requires personalized approaches

  • Resistance mechanism characterization:

    • Map adaptive resistance pathways that emerge during treatment

    • Design rational combinations to address these resistance mechanisms

    • Challenges like treatment resistance and variability in patient response persist despite PD-1 therapy success

These approaches aim to maximize the efficacy of anti-PD-1 therapy combinations while managing toxicity and addressing resistance mechanisms.

How can deep sequencing technologies enhance our understanding of anti-PD-1 antibody mechanisms of action?

Deep sequencing technologies offer powerful approaches for elucidating anti-PD-1 antibody mechanisms:

  • Single-cell RNA sequencing applications:

    • Profile transcriptional changes in individual immune cells following anti-PD-1 treatment

    • Identify cell populations responding to therapy

    • Map cellular trajectory changes induced by PD-1 blockade

    • These insights connect to the observation that PD-1 signaling affects T cell exhaustion states

  • Spatial transcriptomics methodology:

    • Analyze gene expression patterns while preserving spatial context

    • Map interactions between tumor cells and immune cells

    • Correlate spatial organization with response to anti-PD-1 therapy

    • This addresses the importance of the tumor microenvironment in PD-1/PD-L1 signaling

  • T cell receptor (TCR) repertoire analysis:

    • Sequence TCR repertoire before and after anti-PD-1 therapy

    • Track clonal expansion of specific T cell populations

    • Correlate TCR diversity with therapeutic response

    • This connects to PD-1's role in regulating T cell activation

  • Epigenetic profiling:

    • Map chromatin accessibility changes using ATAC-seq

    • Analyze histone modifications in responding vs. non-responding cells

    • Identify epigenetic signatures of T cell reinvigoration

  • Integration of multi-omics data:

    • Combine transcriptomic, epigenomic, and proteomic data

    • Develop computational methods to integrate different data types

    • Generate comprehensive models of anti-PD-1 response mechanisms

These sequencing approaches provide unprecedented resolution for understanding the complex cellular and molecular changes following anti-PD-1 therapy, potentially revealing new biomarkers and therapeutic targets.

What are the technical challenges in developing bispecific antibodies targeting PD-1 and complementary immune checkpoints?

Developing bispecific antibodies targeting PD-1 and other checkpoints involves several technical challenges:

  • Epitope selection complexity:

    • PD-1 has multiple epitope regions with different functional implications

    • At least 10 distinct epitope bins have been identified for anti-PD-1 antibodies

    • Selecting optimal epitopes for both targets that preserve blocking function is critical

  • Binding affinity optimization:

    • Balancing affinities for both targets is challenging

    • Anti-PD-1 antibodies show a >4,000-fold range in binding affinities (<100 pM to 424 nM)

    • Affinity for one target may influence binding to the second target

  • Structural engineering considerations:

    • Format selection (e.g., IgG-like, tandem scFv, diabody)

    • Ensuring proper folding and stability of both binding domains

    • Optimizing linker length and composition between binding domains

  • Functional assessment methodology:

    • Developing assays that can measure simultaneous blockade of both targets

    • Assessing the impact of bispecific binding on immune cell function

    • Comparing to combination therapy with individual antibodies

  • Manufacturing challenges:

    • Expression system optimization for complex bispecific formats

    • Purification strategies for heterodimeric molecules

    • Stability testing under various conditions

  • Preclinical evaluation approaches:

    • Selection of appropriate animal models that express both human targets

    • Pharmacokinetic and biodistribution studies

    • Toxicity assessment specific to dual checkpoint blockade

Addressing these challenges requires integrated expertise in antibody engineering, protein biochemistry, and immunology, combined with sophisticated screening and characterization methodologies.

Product Science Overview

Introduction

PD-1 was first identified in mice, where its expression is induced in the thymus when anti-CD3 antibodies are injected, leading to apoptosis of thymocytes . The human homolog of the PD-1 gene was identified in 1994, sharing 60% sequence homology with the mouse PD-1 protein . PD-1 is expressed in various types of tumors, including melanomas, and plays a significant role in anti-tumor immunity .

Preparation Methods

The preparation of mouse anti-human PD-1 antibodies involves immunizing mice with human PD-1 protein or peptides. The immune response generates antibodies specific to human PD-1, which can be harvested and purified from the mice. These antibodies are then characterized for their specificity and affinity to human PD-1.

Industrial Production Methods

Industrial production of mouse anti-human PD-1 antibodies typically involves the use of hybridoma technology. This process includes:

  1. Immunization: Mice are immunized with human PD-1 protein or peptides.
  2. Cell Fusion: Spleen cells from the immunized mice are fused with myeloma cells to create hybridoma cells.
  3. Screening: Hybridoma cells are screened for the production of antibodies specific to human PD-1.
  4. Cloning: Positive hybridoma cells are cloned to establish stable cell lines.
  5. Production: Large-scale production of antibodies is carried out using bioreactors.
  6. Purification: Antibodies are purified using techniques such as protein A/G affinity chromatography.
Chemical Reactions Analysis

The interaction between PD-1 and its ligands, PD-L1 and PD-L2, involves specific binding sites on the proteins. Upon ligand binding, PD-1 undergoes phosphorylation within its immunoreceptor tyrosine-based switch motif (ITSM), leading to the recruitment of protein tyrosine phosphatases such as PTPN11/SHP-2 . This recruitment results in the dephosphorylation of key signaling molecules, thereby inhibiting T-cell activation .

PD-1/PD-L1 blocking antibodies, such as those used in cancer immunotherapy, have shown profound clinical activity across diverse cancer types . These antibodies work by preventing the interaction between PD-1 and its ligands, thereby enhancing T-cell activation and promoting anti-tumor immunity .

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 2024 Thebiotek. All Rights Reserved.