Anti-RSV F protein (Palivizumab) [Clone MEDI493]

Anti-RSV F protein (Palivizumab) [Clone MEDI493]

Product No.: R190

- -
- -
Product No.R190
Clone
MEDI493
Target
Respiratory Syncytial Virus
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Human Respiratory Syncytial Virus (hRSV), Respiratory Syncytial Virus (RSV)
Isotype
Human IgG1κ
Applications
ELISA
,
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Human RSV strain A2
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
< 1.0 EU/mg as determined by the LAL method
Purity
≥95% by SDS Page
≥95% monomer by analytical SEC
Formulation
This biosimilar antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
State of Matter
Liquid
Product Preparation
Recombinant biosimilar antibodies are manufactured in an animal free facility using only in vitro protein free cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Additional Applications Reported In Literature ?
ELISA,
FA
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.

Description

Description

Specificity
This non-therapeutic biosimilar antibody uses the same variable region sequence as the therapeutic antibody Palivizumab. This product is for research use only. Palivizumab binds to an epitope present in Site II, on the linear region of the F1 subunit, of both the prefusion and postfusion forms of RSV F protein.
Background
Respiratory syncytial virus (RSV) is a major cause of acute lower respiratory tract infection and hospitalization in infants1. RSV F protein is a type I integral membrane protein essential for viral membrane fusion that is highly conserved among isolates of RSV A and B subgroups2. F protein has been investigated as a target for neutralizing antibodies, small molecular antiviral drug development, as a vaccine antigen, and as an antibody target for passive prophylaxis.

F protein is synthesized as an inactive, palmitoylated precursor (F0) and is decorated with N-linked glycans2. Three F0 monomers form a trimer and become activated by a furin-like host protease as they pass through the Golgi. The protease cleaves twice, generating three polypeptides: F2 and F1, which are covalently linked, and pep27, an intervening peptide that dissociates after cleavage. When functional F protein trimer in the virion membrane is triggered, it undergoes a major conformational change from a prefusion to a postfusion form.

Palivizumab is a humanized monoclonal antibody developed for the prevention of serious RSV in high risk infants3 and is the first monoclonal antibody introduced into clinical practice for the prevention of an infectious disease4. Palivizumab was generated by immunizing BALB/c mouse with human RSV strain A2 and fusing the lymphocytes with murine myeloma cell line NS0 to produce a hybridoma4, 5. The murine monoclonal antibody Mab 1129 was then humanized by grafting the antigen binding site to gene segments coding for an intact human IgG1 molecule. The resulting antibody sequence is 95% human, with a small number of murine residues retained to ensure the structural integrity of the binding site. Palivizumab effectively neutralizes over 500 clinical isolates of RSV subtypes A and B3. Its binding epitope is present in both prefusion and postfusion forms of RSV F6 and binding to the postfusion F ectodomain has been experimentally confirmed7, 8.
Antigen Distribution
F protein is found in RSV virion membranes in either an inactive prefusion conformation or an active postfusion conformation.
Ligand/Receptor
site A of the RSV-F glycoprotein
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Immunology
.
Seasonal and Respiratory Infections
.
Viral

Leinco Antibody Advisor

Powered by AI: AI is experimental and still learning how to provide the best assistance. It may occasionally generate incorrect or incomplete responses. Please do not rely solely on its recommendations when making purchasing decisions or designing experiments.

Research-grade Palivizumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs to quantify drug concentrations in serum samples by establishing a standard curve against which unknown serum levels are measured.

Context and Essential Details:

  • In a PK bridging ELISA for palivizumab, the assay relies on a precisely characterized standard (reference material) to generate a calibration curve, allowing accurate measurement of palivizumab concentrations in test samples, typically serum or plasma.
  • Research-grade biosimilars (which are analytically similar versions of palivizumab) are often used as standards because they are more readily available and cost-effective than commercial (clinical-grade) palivizumab. As biosimilars, they are expected to match the reference product in structure, activity, and immunoreactivity—attributes essential for reliable standardization.
  • The biosimilar standard is serially diluted to produce a range of known concentrations. These are added to the plate alongside the test (serum) samples. The ELISA is configured so that the biosimilar and the drug in patient samples are detected equivalently by the assay antibodies.
  • The measured optical densities (ODs) for each standard concentration are used to construct a standard curve. Drug concentrations in unknown serum samples are interpolated from this curve, assuming comparable behavior between the standard and native drug in matrix.

Additional Technical Points:

  • To ensure comparability, bridging ELISAs validated for palivizumab biosimilars must demonstrate that the biosimilar and the reference drug behave identically in the assay, particularly with respect to capture and detection antibodies.
  • Calibration with a validated biosimilar standard maintains accuracy and reproducibility in quantitation, which is crucial for PK analysis, bioequivalence studies, and therapeutic drug monitoring.
  • According to guidelines and commercial ELISA kit documentation, both free drug and anti-drug antibody ELISAs may use biosimilar calibrators; these must be well-characterized and representative of in vivo palivizumab.
  • Use of low- or high-affinity biosimilars as standards could introduce bias, so analytical comparability is typically evaluated prior to large-scale use in regulated assays.

Summary Table: Use of Biosimilar Standards in PK ELISA

StepRole of Biosimilar Palivizumab Standard
PreparationSerially diluted to create calibration curve
Addition to plateIncluded on each ELISA plate with test samples
DetectionMust be recognized identically to reference drug
AnalysisStandard curve used to interpolate unknowns

Key Point:
The bridging ELISA for palivizumab PK relies fundamentally on using either research-grade reference palivizumab or a biosimilar, provided the two are shown to be analytically equivalent within the assay system.

Based on the available research, there appears to be some confusion in your query regarding the use of anti-RSV antibodies for tumor growth inhibition studies. The current scientific literature does not describe studies where anti-RSV antibodies are used to inhibit tumor growth or characterize tumor-infiltrating lymphocytes.

Current RSV Cancer Research Focus

The research actually demonstrates the opposite approach - using RSV itself as an oncolytic virus therapy rather than blocking it with antibodies. RSV has been shown to have anticancer oncolytic activity, where the virus directly infects and destroys cancer cells.

Established RSV Oncolytic Models

Xenograft Models

Human prostate tumor xenograft models in nude mice have been successfully used to demonstrate RSV's oncolytic effects. In these studies:

  • PC-3 human prostate tumor cells were implanted subcutaneously
  • RSV was administered both intratumorally and intraperitoneally
  • Significant tumor regression was observed compared to untreated controls
  • The virus showed selective toxicity toward cancer cells while sparing normal tissues

Cancer Cell Line Studies

RSV has been tested against various cancer cell lines in vitro, showing differential susceptibility:

  • Hepatocellular carcinoma cells: BNL-HCC, Hep3B, Huh-7, and SNU-739 showed significant growth inhibition
  • Colon cancer cells: CT-26, HCT-116, HT-29, and LoVo showed minimal response to RSV infection

Syngeneic Model Characteristics for Immunotherapy

While not specifically for anti-RSV antibody studies, several well-characterized syngeneic models are available for tumor immunotherapy research:

ModelImmune InfiltrationKey Characteristics
RENCAHighHighly immune infiltrated, responsive to immunotherapy
CT26ModerateImmune infiltrated with unique features
EMT6ModerateImmune infiltrated
B16F10LowPoorly infiltrated, less responsive to immunotherapy

Research Gap

The specific application you're asking about - using anti-RSV antibodies to study tumor growth inhibition and TIL characterization - does not appear to exist in the current literature. This could represent a novel research opportunity, as the existing work focuses on RSV as a therapeutic agent rather than a target for antibody-mediated tumor therapy.

If you're interested in developing such studies, you might consider adapting the established xenograft or syngeneic models mentioned above, but this would represent pioneering work in an unexplored research direction.

Researchers can use research-grade palivizumab biosimilars in preclinical studies, but there are no clinically approved palivizumab biosimilars for patient use as of now. In immune-oncology models, the combination of checkpoint inhibitors—such as anti-CTLA-4 or anti-LAG-3 antibodies—has been frequently explored to study synergistic immune activation, especially in solid tumors. However, palivizumab biosimilars are targeted against respiratory syncytial virus (RSV) and are not standard agents in oncology or immune checkpoint research.

Researchers investigating synergistic effects in complex immune-oncology models typically:

  • Use research-grade biosimilars or monoclonal antibodies as tools for mechanistic studies of the immune response.
  • Combine checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) to examine enhanced T cell activation and tumor response, often leveraging established combinations like nivolumab (anti-PD-1) plus relatlimab (anti-LAG-3), which have shown clinical benefit in melanoma.

Key practices:

  • Biosimilars used in combination experiments must have demonstrated analytical and functional comparability to reference antibodies in nonclinical studies.
  • For checkpoint inhibitors, researchers combine agents in murine tumor models, humanized mouse models, or ex vivo human immune cell assays to study additive or synergistic activation of anti-tumor immunity, changes in the tumor microenvironment, and toxicity profiles.
  • Immune profiling (e.g., flow cytometry, cytokine assays) assesses T cell activation, regulatory T cell depletion, and reversal of immune exhaustion.

Important context:

  • To date, there are no published studies on using palivizumab biosimilars in direct combination with checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3) for immune-oncology purposes, since palivizumab is specific for RSV and not for tumor-associated immune checkpoints.
  • Approved checkpoint inhibitors in synergy research are primarily CTLA-4, PD-1/PD-L1, and LAG-3 antibodies, which are directly involved in cancer immunotherapy.

In summary, while research-grade palivizumab biosimilars exist and can be used in investigational combination studies, there is no recognized synergistic application with immune checkpoint inhibitors in oncology models, given their distinct mechanisms and disease contexts. Studies of checkpoint inhibitor synergy are instead focused on combinations within established immune regulatory pathways.

A Palivizumab biosimilar is commonly used as both the capture reagent and the detection reagent in a bridging ADA ELISA to detect anti-drug antibodies (ADAs) generated in response to Palivizumab therapy.

In a typical bridging ADA ELISA:

  • The biosimilar (identical or highly similar to the reference drug, e.g., Palivizumab) is immobilized on the plate as the capture reagent.
  • Patient serum is added; if ADAs are present against Palivizumab, they will bind to the immobilized biosimilar.
  • The same biosimilar is then used—this time labeled (e.g., with biotin, HRP, or a dye)—as the detection reagent. The ADA, having two antigen-binding sites, bridges between the plate-bound and labeled biosimilar.
  • The detection signal (colorimetric or chemiluminescent) indicates the presence of ADAs bound to both biosimilar reagents.

This approach specifically measures bivalent ADA capable of binding two identical epitopes, leveraging the fact that patient ADAs can crosslink two molecules of the biosimilar, forming a "bridge".

Advantages

  • Specificity: Using the biosimilar as both the capture and detection reagent ensures that detected antibodies are specific to the therapeutic drug.
  • Sensitivity: The format is highly sensitive, able to detect low levels of ADA in patient serum.

Considerations

  • The biosimilar used must be sufficiently similar to the reference product (structurally and functionally) to ensure accurate ADA recognition, which is validated through physicochemical and structural characterization.
  • The assay may need optimization to minimize interference from drug in the sample and other serum factors.

Context for Palivizumab

  • Since Palivizumab is a monoclonal antibody, patient immune responses are monitored for anti-drug antibodies to assess immunogenicity.
  • Comparative immunogenicity studies utilize both reference Palivizumab and its biosimilar in these bridging assays to ensure biosimilarity in immune response profiles.

Summary Table: Bridging ADA ELISA with Palivizumab Biosimilar

StepReagent UsedRole
Plate coatingPalivizumab biosimilarCapture ADA from patient sample
Sample incubationPatient serumADA binds to biosimilar
DetectionLabeled biosimilarBridges ADA, detected via label

Using the Palivizumab biosimilar in this capacity is a standard method for monitoring immunogenicity against the therapeutic, reflecting the patient’s immune response and helping ensure the biosimilar’s safety and efficacy profile.

References & Citations

Indirect Elisa Protocol
FA

Certificate of Analysis

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.