Anti-RSV F Protein (Nirsevimab) [Clone MEDI8897] — Fc Muted™
Anti-RSV F Protein (Nirsevimab) [Clone MEDI8897] — Fc Muted™
Product No.: R215
Product No.R215 Clone MEDI8897 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 |
Antibody DetailsProduct DetailsReactive Species Human Host Species Human Expression Host HEK-293 Cells FC Effector Activity Muted Immunogen Prefusion RSV F protein 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 ? FA, ELISA Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change. DescriptionDescriptionSpecificity This non-therapeutic biosimilar antibody uses the same variable region sequence as the therapeutic antibody Nirsevimab. Nirsevimab binds the F1 and F2 subunits of the prefusion RSV F protein at a highly conserved epitope in antigenic site Ø. 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 postfusion form. Approximately 25% of isolate specific variability for F protein is found within an antigenic site at the apex of the prefusion trimer (antigenic site Ø), composed of an α-helix from F1 (aa 196–210) and a strand from F2 (aa 62–69). Nirsevimab is a long-acting, neutralizing recombinant human monoclonal antibody that binds the F1 and F2 subunits of F protein at a highly conserved epitope in antigenic site Ø and locks the RSV F protein in the prefusion conformation, blocking viral entry into the host cell1, 3, 4. In vitro, nirsevimab binds to immobilized human FcγRs (FcγRI, FcγRIIA, FcγRIIB and FcγRIII)3. Protection from infection is thought to be dependent on neutralization activity rather than Fc-mediated effector function based on data from a cotton rat model of RSV infection3. Nirsevimab has been modified with a triple amino acid substitution (YTE) in the Fc region to extend the serum half-life3. Nirsevimab originates from the D25 antibody developed by AIMM Therapeutics and was jointly developed and commercialized by AstraZeneca and Sanofi for the prevention of RSV infection in neonates and infants. 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 AdvisorPowered 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 Nirsevimab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as the definitive material against which drug concentrations in serum samples are quantitatively measured. In this application:
Essential details:
In summary, in PK bridging ELISAs for Nirsevimab, research-grade biosimilars serve dual roles—as calibration standards to generate standard curves for quantitative drug measurement and as reference controls for validation and equivalence testing—ensuring precise, accurate, and comparable PK data between biosimilar and originator drug samples. The primary models used to study the effects of anti-Respiratory Syncytial Virus (RSV) therapeutics—including antibodies—on tumor growth inhibition and characterization of tumor-infiltrating lymphocytes (TILs) are predominantly murine syngeneic models and, to a lesser extent, human xenograft models in immunodeficient mice. Key Model Types:
Essential Details:
Summary Table:
Conclusion: Currently, there is no specific evidence or research on using Nirsevimab biosimilars in conjunction with other checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 biosimilars to study synergistic effects in complex immune-oncology models. Nirsevimab is primarily used as an anti-RSV antibody, targeting the fusion (F) protein of the respiratory syncytial virus to prevent infections in high-risk populations, such as infants and older adults. However, researchers often explore combination strategies in immunotherapy, particularly in cancer models, where multiple checkpoint inhibitors are combined to enhance therapeutic efficacy. This includes combining CTLA-4 inhibitors with PD-1/PD-L1 inhibitors to target different aspects of the immune response. If researchers were to explore Nirsevimab in immune-oncology models, it would likely involve studying its effects in complex systems where viral infections influence cancer outcomes or immune function, rather than as a direct checkpoint inhibitor. Hypothetical ApplicationIn a hypothetical scenario where researchers might study the effects of Nirsevimab alongside checkpoint inhibitors, the focus would not be on direct synergistic effects for cancer treatment but possibly on understanding how viral infections and their treatments influence cancer immunity.
However, such applications are speculative and not directly related to the current use of Nirsevimab or its biosimilars. Current research focuses on its role in preventing RSV infections rather than exploring synergies with checkpoint inhibitors in cancer models. Future Research DirectionsFor future research, it would be intriguing to investigate how reducing the burden of viral infections could indirectly affect cancer outcomes, potentially by improving immune system function or reducing chronic inflammation. This could involve studying the impact of viral infections on the tumor microenvironment and how treatments like Nirsevimab might influence these dynamics. However, this would require a significant shift in how Nirsevimab is used and studied, from a primary focus on RSV prevention to broader immunological effects. Nirsevimab biosimilars play a crucial role in bridging ADA ELISA assays for monitoring patient immune responses against the therapeutic drug. These assays are specifically designed to detect anti-drug antibodies that can form in patients receiving nirsevimab treatment, which could lead to loss of efficacy or adverse reactions. Mechanism of Bridging ADA ELISA with Nirsevimab BiosimilarIn a bridging ADA ELISA format, the nirsevimab biosimilar serves dual roles as both capture and detection reagents. The biotinylated nirsevimab biosimilar is first captured on streptavidin-coated plates, creating the capture surface. Patient serum samples are then added, and any anti-nirsevimab antibodies present will bind to the captured drug on the plate. For detection of bivalent anti-drug antibodies, a second nirsevimab biosimilar conjugated with a detection label (such as HRP or a fluorescent dye) is added. This labeled biosimilar binds to the other binding site of the anti-drug antibody, forming a "bridge" between the capture and detection reagents - hence the name "bridging ELISA". Use of Non-Therapeutic BiosimilarsThe nirsevimab biosimilar used in these assays is specifically designed as a non-therapeutic research reagent that uses the same variable regions as the therapeutic antibody nirsevimab. This ensures that the biosimilar maintains the same binding specificity and epitope recognition as the therapeutic drug, making it ideal for detecting antibodies that would interfere with the actual treatment. Since nirsevimab is a human IgG1 monoclonal antibody that targets the fusion (F) glycoprotein of respiratory syncytial virus (RSV), specifically binding to a highly conserved epitope on the prefusion conformation of the RSV F protein, the biosimilar maintains these same binding characteristics for accurate immunogenicity assessment. Clinical Significance and InterpretationThe formation of anti-drug antibodies against nirsevimab has been associated with loss of response, hypersensitivity reactions, and severe therapy-limiting side effects. The bridging ELISA format offers high sensitivity and allows for high-throughput screening of patient samples, making it particularly valuable for monitoring large patient populations receiving nirsevimab therapy. However, the results must be interpreted carefully in the context of simultaneously measured drug levels. The presence of anti-drug antibodies does not necessarily correlate directly with treatment failure if sufficient drug levels remain in circulation. Low-level antibodies detected by highly sensitive assays like bridging ELISA may not significantly influence treatment response if adequate nirsevimab concentrations are maintained. Assay ConsiderationsThe specificity of bridging ELISA assays can be challenged by matrix components in human serum, soluble target molecules, or drug components. Therefore, the use of high-quality biosimilar reagents and appropriate blocking solutions is crucial for obtaining meaningful results. Each laboratory must customize and validate the protocol according to their specific requirements while maintaining the sensitivity needed to detect clinically relevant anti-drug antibodies. References & Citations1. Hammitt LL, Dagan R, Yuan Y, et al. N Engl J Med. 386(9):837-846. 2022. 2. McLellan JS, Ray WC, Peeples ME. Curr Top Microbiol Immunol. 372:383-104. 2013. 3. Keam SJ. Drugs. 83(2):181-187. 2023. 4. Zhu Q, McLellan JS, Kallewaard NL, et al. Sci Transl Med. 9(388):eaaj1928. 2017. 5. Domachowske JB, Khan AA, Esser MT, et al. Pediatr Infect Dis J. 37(9):886-892. 2018. 6. Zhu Q, Lu B, McTamney P, et al. J Infect Dis. 218(4):572-580. 2018. 7. Griffin MP, Yuan Y, Takas T, et al. N Engl J Med. 383(5):415-425. 2020. Technical ProtocolsCertificate of Analysis |
Formats Available
Prod No. | Description |
---|---|
R210 | |
R215 |
