PVR antibodies modulate immune-tumor interactions through:
NK Cell Activation: Binding PVR on tumor cells triggers NK cell adhesion, lytic granule secretion, and IFN-γ production via interactions with CD226 .
Checkpoint Inhibition: Blocking PVR-TIGIT/CD96 interactions reverses immunosuppression in tumors .
Viral Entry Interference: Neutralizes poliovirus entry by competing for the D1 domain .
NK Cell Cytotoxicity: Antibodies like BLR074G enhance CD107a expression (a degranulation marker) by 240% over controls in NK cell assays .
Tumor Growth Inhibition: PVR knockdown reduces tumor volume by 50% in colon cancer models .
Immunoassays:
Immunohistochemistry (IHC): Localizes PVR overexpression in tumor tissues (e.g., pancreatic adenocarcinoma) .
Checkpoint Therapy: Humanized antibodies (e.g., Patent WO2021070181A1) block TIGIT-PVR interactions, restoring CD8+ T cell activity .
Oncolytic Virotherapy: Recombinant polioviruses targeting PVR-overexpressing tumors are in clinical trials .
Overexpression: PVR is upregulated in glioblastoma, colorectal carcinoma, and lung cancer .
Prognostic Marker: High PVR levels correlate with poor survival (HR = 2.1, p < 0.01) .
Humanization: CDR grafting onto human frameworks reduces immunogenicity (e.g., N56E variant improves cross-reactivity with monkey PVR by 35%) .
Affinity Optimization: Mutations in CDR2 (e.g., E→N) enhance binding affinity by 50% .
The PVR recombinant monoclonal antibody undergoes a rigorous production process, beginning with in vitro cloning. During this stage, genes encoding the PVR antibody's heavy and light chains are incorporated into expression vectors. These vectors are subsequently transfected into host cells, enabling recombinant antibody expression within a controlled cell culture system. Following expression, the PVR recombinant monoclonal antibody is purified from the supernatant of transfected host cell lines utilizing an affinity-chromatography purification technique. A key attribute of this antibody is its highly specific binding capability with the human PVR protein. Notably, its versatility makes it suitable for a range of applications, including ELISA, Western blot analysis (WB), and immunohistochemistry (IHC).
The poliovirus receptor (PVR), also known as the CD155 receptor, primarily functions as a cell surface receptor for poliovirus and related enteroviruses. It plays a pivotal role in the initial attachment and entry of the virus into host cells.
PVR serves as a mediator of NK cell adhesion and triggers NK cell effector functions. It binds to two distinct NK cell receptors: CD96 and CD226. These interactions accumulate at the cell-cell contact site, leading to the formation of a mature immunological synapse between the NK cell and target cell. This process may trigger adhesion and the secretion of lytic granules and IFN-gamma, ultimately activating the cytotoxic activity of activated NK cells. PVR may also promote NK cell-target cell modular exchange and PVR transfer to the NK cell. This transfer is particularly significant in certain tumor cells expressing high levels of PVR and can trigger fratricide NK cell activation, providing tumors with a potential mechanism for immune evasion.
PVR plays a role in mediating tumor cell invasion and migration. In the context of microbial infection, PVR acts as a receptor for poliovirus. It may contribute to the axonal transport of poliovirus by targeting virion-PVR-containing endocytic vesicles to the microtubular network through interaction with DYNLT1. This interaction drives the virus-containing vesicle towards axonal retrograde transport. PVR also acts as a receptor for Pseudorabies virus. Furthermore, PVR is prevented from reaching the cell surface upon infection by Human cytomegalovirus /HHV-5, likely to avoid immune recognition of the infected cell by NK cells.
PVR/CD155 is a transmembrane glycoprotein belonging to the nectin family with multiple isoforms. The predominant isoforms include membrane-bound variants (α) and soluble or secreted variants (β and γ), which are present in various body fluids including blood, cerebrospinal fluid, and urine . The protein has a molecular weight ranging from 60-80 kDa when detected by Western blotting .
Most recombinant monoclonal antibodies target the extracellular domain of human PVR/CD155. For example, some antibodies are developed against a synthetic peptide corresponding to residues surrounding Asn188 of human CD155 protein , while others target the extracellular domain spanning Met1-Asn343 (UniProt #P15151-1) . This region contains the immunoglobulin-like domains that mediate PVR's interactions with its binding partners.
In normal immune homeostasis, PVR/CD155 plays crucial roles in:
Mediating NK cell adhesion and triggering NK cell effector functions
Binding to receptors CD96 and CD226 (DNAM-1), accumulating at cell-cell contact sites and forming mature immunological synapses
Triggering adhesion, secretion of lytic granules, IFN-gamma production, and activating cytotoxicity in NK cells
Maintaining immune tolerance and preventing autoimmunity through T-cell inhibitory signaling
In pathological conditions, particularly cancer:
PVR is dramatically overexpressed in multiple malignancies while maintaining low or absent expression in most healthy tissues
Overexpression promotes tumor cell invasion, migration, and proliferation
Associated with poor prognosis and enhanced tumor progression
Contributes to immune escape mechanisms through immunoregulatory functions
The dual role of PVR in immune activation (via DNAM-1) and inhibition (via TIGIT and CD96) creates a complex regulatory network that cancer cells exploit through PVR overexpression.
PVR recombinant monoclonal antibodies have been validated for multiple applications, each with specific optimization considerations:
When selecting the appropriate clone, researchers should consider:
Target epitope - different clones recognize distinct regions which may affect functionality detection
Species cross-reactivity - most validated for human samples only
Conjugation needs - whether unconjugated or directly labeled (e.g., HRP-conjugated) antibodies are required
Validation status - preference for clones with published validation for your specific application
Detection of PVR/CD155 in patient samples requires careful optimization:
For immunohistochemistry:
Antigen retrieval is critical - most protocols use citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Background reduction techniques are essential as PVR can be expressed at varying levels on stromal cells
Comparison with healthy tissue controls is necessary due to the differential expression pattern
Quantification systems should be established (H-score or percentage of positive cells) for consistent assessment
For liquid biopsies detecting soluble PVR:
Soluble PVR isoforms (β and γ) are present in patient sera and represent potential biomarkers of cancer development and progression
ELISA-based detection should include calibration standards
Pre-analytical variables (collection, processing, storage) must be standardized
Validation against tissue expression is recommended for correlation studies
When analyzing patient samples, researchers should consider that PVR is expressed at low levels on specific healthy cells including vascular endothelial cells, spinal cord motor neurons, and some immune cell subsets, which may serve as internal controls for antibody specificity .
PVR/CD155 orchestrates complex immunoregulatory functions through its interactions with multiple receptors on immune cells:
DNAM-1 (CD226) Interaction - Activating pathway:
Promotes NK and T cell activation against tumor cells
Enhances cytotoxic functions and cytokine production
Facilitates immune surveillance
TIGIT Interaction - Inhibitory pathway:
Functions as an immune checkpoint receptor with an ITIM domain
Suppresses NK and T cell functions upon binding to PVR
Counterbalances DNAM-1-mediated activation
Often upregulated in the tumor microenvironment
CD96 Interaction - Context-dependent inhibitory role:
Tumors exploit this system by:
Overexpressing PVR to engage inhibitory receptors (TIGIT, CD96)
Shifting the balance from immune activation toward suppression
Promoting NK cell fratricide through PVR transfer mechanisms
Utilizing soluble PVR to potentially compete for receptor binding
These interactions represent the molecular basis for current immunotherapeutic approaches targeting the PVR-receptor axis.
Researchers investigating PVR-targeted immunotherapies should consider these experimental models:
In vitro models:
Co-culture systems using PVR-expressing tumor cells with immune effectors (NK cells, T cells)
CRISPR/Cas9-mediated PVR knockout or overexpression cell lines
Reporter assays measuring activation/inhibition through PVR-receptor interactions
3D organoid models incorporating immune components
In vivo models:
Syngeneic mouse models with orthotopic tumors expressing human PVR
Humanized mouse models for evaluating human-specific PVR interactions
Genetically engineered mouse models with tissue-specific PVR expression
Patient-derived xenograft models for testing PVR-targeting approaches
Readouts and endpoints to assess:
Tumor volume and weight measurements (demonstrated in colon cancer models)
Metastatic burden quantification (validated in multiple mouse tumor models)
Immune infiltration analysis by flow cytometry and multiplex IHC
Results from these models have validated several therapeutic approaches currently in clinical trials, including checkpoint inhibitors targeting TIGIT and adoptive cell therapies recognizing PVR-overexpressing tumors.
Ensuring antibody specificity in complex tissue samples requires systematic validation:
Positive and negative controls:
Use cell lines with known high PVR expression (many tumor cell lines) as positive controls
Include PVR-knockout cell lines generated by CRISPR/Cas9 as negative controls
Compare with tissues known to express low levels of PVR (most healthy tissues)
Epitope validation techniques:
Peptide competition assays to confirm epitope specificity
Western blotting to verify molecular weight (60-80 kDa)
Cross-validation with multiple antibody clones targeting different epitopes
Signal-to-noise optimization:
Titrate antibody concentrations (typical starting dilutions 1:100-1:400 for IHC)
Compare signal patterns with known PVR distribution (e.g., vascular endothelial cells serve as internal positive controls)
Use isotype controls to identify non-specific binding
Technical considerations:
If inconsistencies persist, consider epitope masking by protein-protein interactions or post-translational modifications affecting antibody recognition.
PVR detection requires strategies that account for its complexity:
For transmembrane isoform (α) detection:
Use antibodies targeting extracellular domains when studying surface expression
Employ membrane extraction protocols for Western blotting
Consider fixation conditions that preserve membrane integrity
Account for potential internalization during procedures
For soluble isoform (β/γ) detection:
Optimize sample preparation techniques for bodily fluids
Use sandwich ELISA approaches with capture/detection antibody pairs
Consider concentration steps for dilute samples
Recognize that soluble PVR levels are increased in cancer patient sera
For dynamic expression analysis:
Implement time-course experiments following relevant stimuli
Consider stress conditions that induce PVR (PVR is a stress-induced ligand)
Analyze sorted cell populations when examining heterogeneous tissues
Use single-cell techniques to capture expression heterogeneity
For multiparametric analysis:
Combine PVR detection with its binding partners (DNAM-1, TIGIT, CD96)
Correlate with functional assays (cytotoxicity, cytokine production)
Integrate with analysis of downstream signaling pathways
These strategies will provide a more comprehensive understanding of PVR biology in experimental systems.
Several promising therapeutic approaches targeting the PVR/CD155 axis warrant investigation:
Direct PVR targeting:
Development of neutralizing antibodies against PVR
Small molecule inhibitors disrupting PVR interactions
Evaluation of soluble receptor decoys competing for PVR binding
Validation methods should include receptor-binding competition assays and functional immune readouts
Checkpoint inhibition strategies:
Anti-TIGIT antibodies blocking the PVR-TIGIT inhibitory axis
Dual targeting of TIGIT and other checkpoints (PD-1, CTLA-4)
Bispecific antibodies engaging both inhibitory pathways
Validation requires immune activation assessment in PVR-expressing tumor models
Adoptive cell therapy approaches:
Engineering NK or T cells with enhanced DNAM-1 signaling
CAR-T cells targeting PVR-overexpressing tumors
Genetic modification to render effector cells resistant to PVR-mediated inhibition
Validation through cytotoxicity assays against PVR-positive tumor panels
Oncolytic virotherapy:
These approaches require rigorous preclinical validation before clinical translation, with particular attention to potential on-target/off-tumor effects given PVR expression on some healthy tissues.
Emerging research areas exploring non-canonical PVR functions include:
Metabolic regulation:
Investigating PVR's influence on cellular metabolism in tumors
Examining potential interactions with metabolic checkpoints
Exploring metabolic consequences of PVR signaling in immune cells
Experimental approaches include metabolomics and bioenergetic analyses
Extracellular vesicle biology:
Analyzing PVR incorporation into exosomes and microvesicles
Studying intercellular transfer of PVR through vesicular mechanisms
Investigating the immunomodulatory roles of PVR-containing vesicles
Methods include vesicle isolation, characterization, and functional studies
PVR in tissue homeostasis:
Exploring functions beyond immune regulation and viral entry
Investigating roles in tissue repair and regeneration
Examining developmental functions in organogenesis
Approaches include tissue-specific knockout models and developmental studies
Post-translational regulation:
Understanding these non-canonical functions may reveal new therapeutic vulnerabilities and explain context-dependent PVR activities observed in different pathological states.