The synthesis involves advanced recombinant technologies:
Immunogen: Recombinant human nectin-4 ectodomain fragments .
Hybridoma Generation: B-cells from immunized hosts (e.g., chickens, rabbits) are isolated, and antibody genes are cloned into phage display libraries .
Expression Systems: Produced in CHO cells or E. coli for consistent glycosylation and batch-to-batch reproducibility .
Purification: Protein A/G affinity chromatography, yielding >95% purity .
Immunization with nectin-4 ectodomain.
Phage display library construction and biopanning.
Clonal selection based on binding affinity (e.g., clones S21 and L4 in breast cancer models) .
Large-scale fermentation and purification.
NECTIN4 monoclonal antibodies are validated for multiple assays:
Prognostic biomarker in luminal-A breast cancer (membrane expression correlates with distant relapse ).
Predictive marker for antibody-drug conjugate (ADC) therapies .
NECTIN4 monoclonal antibodies serve as backbones for targeted therapies:
Mechanism: Internalization of the ADC-nectin-4 complex releases cytotoxic payloads intracellularly .
BT7480: Bicyclic peptide targeting nectin-4 + CD137 agonist to enhance antitumor immunity .
TLR8 Conjugates: Nectin-4 mAb linked to TLR8 agonists to activate myeloid cells in the tumor microenvironment .
Enfortumab vedotin:
ETx-22:
The NECTIN4 Recombinant Monoclonal Antibody is produced through a meticulous and controlled process to ensure its exceptional quality and specificity. The process starts with isolating B cells from an immunized animal, using recombinant human NECTIN4 protein as the immunogen. Total RNA is extracted from these B cells and transformed into cDNA through reverse transcription. The NECTIN4 antibody genes are then amplified using specific primers targeting the antibody constant regions and inserted into an expression vector. The vector is introduced into host cells through transfection, allowing for the production of the NECTIN4 Recombinant Monoclonal Antibody. After a period of cell culture, the antibody is harvested from the supernatant and purified using affinity chromatography, resulting in a highly purified form suitable for a wide range of applications. To confirm its reliability and functionality, the antibody undergoes ELISA to validate its specificity in detecting human NECTIN4 protein.
NECTIN4 appears to be involved in cell adhesion through trans-homophilic and -heterophilic interactions, the latter including specifically interactions with NECTIN1. It does not act as a receptor for alpha-herpesvirus entry into cells. In the context of microbial infections, it acts as a receptor for measles virus.
Structurally, Nectin-4 contains an extracellular domain that can be targeted by antibodies, with the V domain being particularly important for antibody binding. The extracellular domain of human Nectin-4 shares high homology (90% and 92%) with mouse and rat Nectin-4, respectively , which has implications for cross-species research applications.
NECTIN4 recombinant monoclonal antibodies can be utilized in various laboratory techniques and applications:
ELISA (Enzyme-Linked Immunosorbent Assay): For quantitative detection of Nectin-4 in solution or immobilized samples. For example, immobilized human NECTIN4-His at 2 μg/mL can bind to enfortumab (anti-Nectin-4 antibody) with an EC50 of 0.003846 μg/mL .
Flow Cytometry: For detecting Nectin-4 expression on cell surfaces. Human breast cancer cell lines such as MCF-7 and SK-BR-3 are commonly used for this application .
Western Blotting: For detecting Nectin-4 protein in cell lysates, typically observed at molecular weights of 62 and 75 kDa depending on glycosylation status .
Immunohistochemistry/Immunofluorescence: For evaluating Nectin-4 expression in tissue sections or fixed cells.
Functional Assays: For investigating the biological roles of Nectin-4 in cell adhesion, signaling, and other cellular processes .
Therapeutic Development: As the foundation for antibody-drug conjugates (ADCs) and other therapeutic modalities targeting Nectin-4-expressing tumors .
The most common expression system for NECTIN4 recombinant monoclonal antibodies is Chinese Hamster Ovary (CHO) cells . This mammalian expression system provides several advantages:
Proper post-translational modifications, particularly glycosylation patterns that are important for antibody function and stability
High protein yield and scalability for research and therapeutic applications
Consistent product quality across production batches
For example, enfortumab, a fully human IgG1 antibody targeting Nectin-4, is expressed from CHO cells with a huIgG1 heavy chain and hukappa light chain configuration, resulting in a predicted molecular weight of 145.44 kDa . For therapeutic applications, the transition from hybridoma-produced antibodies (such as AGS-22M6E) to CHO-produced antibodies (ASG-22CE) has been documented to maintain equivalent binding characteristics and potency .
Alternative expression systems, such as chicken-derived single-chain variable fragments (scFvs), have also been developed for research applications .
A comprehensive validation strategy for a new NECTIN4 recombinant monoclonal antibody should include:
Binding Specificity Assessment:
ELISA against recombinant Nectin-4 protein
Western blot analysis using positive control cell lines known to express Nectin-4 (e.g., MCF-7, SK-BR-3)
Comparison with established anti-Nectin-4 antibodies
Testing against related Nectin family members to confirm specificity
Functional Characterization:
Cross-reactivity Analysis:
Testing against Nectin-4 from different species if cross-reactivity is desired
Negative controls using cell lines that do not express Nectin-4
Application-specific Optimization:
For IHC/IF: Testing different fixation methods and antigen retrieval protocols
For flow cytometry: Optimizing antibody concentration and incubation conditions
For Western blotting: Determining optimal sample preparation and detection methods
When validating by flow cytometry, it's essential to include appropriate isotype controls, as demonstrated in studies with SK-BR-3 human breast cancer cells .
Proper storage and handling are critical for maintaining antibody functionality:
Storage Temperature:
Reconstitution Guidelines:
Formulation Considerations:
Freeze-Thaw Cycles:
Working Dilutions:
Prepare fresh working dilutions on the day of the experiment
Follow manufacturer's recommendations for application-specific dilutions
NECTIN4 antibodies have become valuable tools for investigating multiple aspects of cancer biology:
Expression Profiling and Biomarker Studies:
Functional Studies of Cancer Cell Behavior:
Mechanism of Action Studies:
Therapeutic Targeting Validation:
Developing therapeutic antibodies against NECTIN4 involves several key challenges:
Target Binding Optimization:
Antibody Format Selection:
ADC Design Considerations:
Resistance Mechanisms:
Understanding potential resistance mechanisms to antibody-based therapies
Developing strategies to overcome resistance through combination approaches or next-generation antibody designs
Translation from Pre-clinical to Clinical Studies:
Ensuring that pre-clinical efficacy in xenograft models translates to clinical benefit
Addressing potential immunogenicity concerns, particularly for novel antibody formats
The successful development of enfortumab vedotin, now approved for urothelial carcinoma, provides proof-of-concept for the clinical utility of Nectin-4-directed therapies .
Optimizing flow cytometry protocols for NECTIN4 detection requires attention to several parameters:
Antibody Selection and Concentration:
Cell Preparation Considerations:
Use single-cell suspensions with high viability (>90%)
For adherent cells like MCF-7 or SK-BR-3, ensure complete dissociation without damaging surface proteins
Include a viability dye to exclude dead cells from analysis
Staining Protocol:
Perform staining in buffers containing protein (1-2% BSA or FBS) to minimize non-specific binding
Incubate at 4°C for 30-60 minutes for optimal binding
Wash cells thoroughly to remove unbound antibody
Instrument Settings:
Set appropriate voltage for the fluorochrome used
Include single-stained controls for compensation if performing multicolor analysis
Use unstained and isotype controls to establish proper gating strategies
Analysis Considerations:
Present data as histograms comparing test samples with isotype controls
For quantitative analysis, calculate the mean or median fluorescence intensity ratio compared to control
For example, detection of Nectin-4 in MCF-7 human breast cancer cells has been successfully performed using PE-conjugated monoclonal antibodies with appropriate isotype controls .
Western blotting for NECTIN4 requires specific considerations:
Sample Preparation:
Use lysis buffers containing appropriate detergents to solubilize membrane proteins
Include protease inhibitors to prevent degradation
For glycosylated Nectin-4, samples may need to be treated with glycosidases to resolve band heterogeneity
Gel Electrophoresis Parameters:
Transfer Conditions:
Optimize transfer time and voltage for efficient transfer of higher molecular weight proteins
Consider semi-dry or wet transfer methods based on your specific setup
Blocking and Antibody Incubation:
Detection Methods:
Use appropriate HRP-conjugated secondary antibodies and ECL detection
For low abundance targets, consider more sensitive detection systems or signal amplification methods
Controls to Include:
Positive control: Lysate from cells known to express Nectin-4 (e.g., breast cancer cell lines)
Negative control: Lysate from cells not expressing Nectin-4
Loading control: Housekeeping protein (e.g., β-actin, GAPDH)
Several methodologies can be employed to characterize binding properties:
Surface Plasmon Resonance (SPR):
Enzyme-Linked Immunosorbent Assay (ELISA):
Epitope Mapping Techniques:
Peptide arrays covering the Nectin-4 sequence
Competition assays between different antibodies
Hydrogen-deuterium exchange mass spectrometry
X-ray crystallography of antibody-antigen complexes
Computational Approaches:
Binding to Truncated Protein Variants:
Express different domains of Nectin-4 to determine the binding region
Mutagenesis studies to identify critical binding residues
Development of antibody-drug conjugates (ADCs) targeting NECTIN4 involves several critical design parameters:
Antibody Selection:
Linker Chemistry:
Cytotoxic Payload Selection:
Drug-to-Antibody Ratio (DAR):
Pre-clinical Evaluation:
In vitro testing in Nectin-4-expressing cell lines
In vivo efficacy in xenograft models
Pharmacokinetic and biodistribution studies to confirm tumor targeting
The success of enfortumab vedotin in clinical trials validates the approach of targeting Nectin-4 with ADCs for cancer therapy, particularly in urothelial carcinoma .
Different antibody formats offer distinct advantages for targeting NECTIN4:
Antibody Format | Advantages | Limitations | Research/Therapeutic Applications |
---|---|---|---|
Full IgG (e.g., enfortumab) | - Long serum half-life - Fc-mediated effector functions - Well-established production platforms | - Larger size limits tumor penetration - Higher production costs - Potential immunogenicity | - ADC development (e.g., enfortumab vedotin) - Blocking Nectin-4 interactions - Flow cytometry, IHC, Western blotting |
scFv | - Smaller size improves tissue penetration - Less immunogenic - Amenable to phage display selection | - Short serum half-life - Lack of Fc-mediated functions - Potential stability issues | - Research tools for epitope mapping - Cell aggregation assays - Building blocks for next-generation therapeutics |
PE-conjugated antibodies | - Direct detection in flow cytometry - Higher sensitivity | - Limited to detection applications - Photosensitive | - Flow cytometry to detect Nectin-4 expression - Phenotypic characterization of cell lines |
The choice of format depends on the specific application. For example, scFv clones have demonstrated utility in diminishing attachment-free cell aggregation of Nectin-4-positive breast cancer cells , while full IgG formats are preferred for ADC development due to their longer circulation time and ability to carry multiple drug molecules .