Anti-erbB-2 (Her-2/neu) (Margetuximab) [Clone MGAH22]
Anti-erbB-2 (Her-2/neu) (Margetuximab) [Clone MGAH22]
Product No.: LT220
Product No.LT220 Clone MGAH22 Target erbB-2 Product Type Biosimilar Recombinant Human Monoclonal Antibody Alternate Names Anti erbB-2, erbB2, HER2, CD340 Isotype Human IgG1κ Applications ELISA , FA , FC , IP , WB |
Antibody DetailsProduct DetailsReactive Species Human Host Species Human Expression Host HEK-293 Cells FC Effector Activity Active Immunogen Human erbB2/EGFR2/CD340 Product Concentration ≥ 5.0 mg/ml Endotoxin Level < 1.0 EU/mg as determined by the LAL method Purity ≥95% by SDS Page ⋅ ≥98% 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. Pathogen Testing To protect mouse colonies from infection by pathogens and to assure that experimental preclinical data is not affected by such pathogens, all of Leinco’s recombinant biosimilar antibodies are tested and guaranteed to be negative for all pathogens in the IDEXX IMPACT I Mouse Profile. 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 ? FC, ELISA, WB, IP, FA 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 Margetuximab. This product is for research use only. Margetuximab activity is directed against Human erb-b2 receptor tyrosine kinase 2 (ERBB2; HER-2/neu). Background erbB-2 encodes a member of the epidermal growth factor (EGF) receptor family of receptor tyrosine kinases1. erbB-2 enhances kinase-mediated activation of downstream signaling pathways by forming a heterodimer with other ligand-bound EGF receptor family members. Dysregulation of erbB-2 contributes to tumorigenesis in breast, ovarian, gastric, and other cancers.
Margetuximab is a human/mouse chimeric anti-erbB-2 monoclonal IgG1 antibody derived from mouse clone 4D5, the precursor of trastuzumab2. Margetuximab has an Fc domain (MGFc0264) engineered for increased binding to both alleles of human activating Fcγ receptor IIIA (CD16A) and for reduced binding to CD32B. Compared with WT Fc domain, the optimized MGFc0264 domain demonstrates increased affinity for both alleles of human CD16A as well as human C1q but decreased binding to human CD32B (inhibitory FcγR) and the 131R allele of CD32A (human activating FcγR). Binding to the 131H allele is not substantially modified. The optimized Fc domain also confers improved antibody-dependent cell cytotoxicity against erbB-2-positive tumor cells, including low ERBB2 expressors, independent of the FcγR variant for the effector cells. The MGFc0264 Fc domain was generated by mutating five sites: L235V, F243L, R292P, Y300L, and P396L2. The L235V mutation was inserted to reduce CD32B binding. The Fc domain modifications do not influence antigen recognition or anti-proliferative activity in the absence of effector cells. In clinical trials, Margetuximab binds to erbB-2 with high affinity and produces direct growth suppression of erbB-2-expressing tumor cell lines3. Positive data from clinical trials led to US Food and Drug Administration approval for Margetuximab in the treatment of metastatic HER2-positive breast cancer in 20204. Antigen Distribution erbB-2 is an overexpressed cell-surface oncoprotein. Ligand/Receptor erbB-2/HER2/CD340, Receptor PubMed NCBI Gene Bank ID UniProt.org Research Area Biosimilars . Cancer . Immuno-Oncology . Immunology 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 Margetuximab biosimilars are commonly used as calibration standards or reference controls in PK bridging ELISA assays to ensure accurate quantification of drug concentration in serum samples. In these assays:
Supporting details:
Summary Table (for clarity):
This approach ensures that serum drug concentrations measured in clinical samples are reliable and comparable for both biosimilar and branded Margetuximab products, a critical requirement for biosimilar approval and postmarketing surveillance. In Vivo Models for Studying Anti-ErbB-2 Antibody Effects on Tumor Growth and TILsAnti-ErbB-2 monoclonal antibodies (mAbs) are a cornerstone of research and therapy for HER2-positive cancers, with both xenograft (humanized/immunocompetent) and syngeneic (mouse-derived, immunocompetent) models used to study direct tumor growth inhibition and immune-mediated effects, including characterization of tumor-infiltrating lymphocytes (TILs). Xenograft (Humanized) Models
Syngeneic (Immunocompetent) Models
Comparative Summary Table
Conclusions
In summary: Research-grade anti-ErbB-2 antibodies are most commonly studied in vivo using human tumor xenografts in immunodeficient mice for direct growth inhibition and mechanism studies, and in syngeneic, immune-competent models (notably NDLUCD) for comprehensive immune response and TIL characterization. Researchers are exploring the use of margetuximab, a novel anti-HER2 monoclonal antibody, in conjunction with other checkpoint inhibitors to study synergistic effects in immune-oncology models. While margetuximab itself is not a checkpoint inhibitor, its mechanism of enhancing antibody-dependent cellular cytotoxicity (ADCC) through engineered Fc domains can complement immunotherapies. Here’s how researchers might approach combining margetuximab with checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3: Combining Margetuximab with Checkpoint InhibitorsMechanism of Action
Synergistic EffectsCombining margetuximab with checkpoint inhibitors could potentially offer synergistic benefits by:
Research Approaches
Challenges and Opportunities
While there are no specific studies directly combining margetuximab with biosimilars of anti-CTLA-4 or anti-LAG-3, the concept of using such combinations to enhance immune-oncology treatments is an active area of research. The use of biosimilars could potentially make these therapies more accessible due to their lower cost compared to innovator molecules. Example CombinationA hypothetical approach could involve using margetuximab with a bispecific antibody like MGD013, which targets both PD-1 and LAG-3, to attack both the tumor and immune evasion mechanisms simultaneously. This combination could potentially increase the effectiveness of margetuximab by enhancing the immune system's ability to recognize and destroy cancer cells. Overall, while there are no direct studies mentioned on using a margetuximab biosimilar with checkpoint inhibitor biosimilars, the concept aligns with ongoing efforts to combine targeted therapies and immunotherapies to improve cancer treatment outcomes. In the context of immunogenicity testing, a Margetuximab biosimilar can be used as the capture or detection reagent in a bridging ADA (anti-drug antibody) ELISA to monitor a patient's immune response against the therapeutic drug through the following steps:
This approach allows for sensitive detection of ADAs, which is crucial for assessing the immunogenicity of therapeutic drugs and monitoring potential immune responses that could impact treatment efficacy and safety. General Steps in Bridging ADA ELISA:
Key Considerations:
References & Citations1. https://www.ncbi.nlm.nih.gov/gene/2064
2. Nordstrom JL, Gorlatov S, Zhang W, et al. Breast Cancer Res. 13(6):R123. 2011. 3. Bang YJ, Giaccone G, Im SA, et al. Ann Oncol. 28(4):855-861. 2017. 4. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761150s000lbl.pdf 5. Catenacci DVT, Kang YK, Park H, et al. Lancet Oncol. 21(8):1066-1076. 2020. Technical ProtocolsCertificate of Analysis |
Formats Available
Prod No. | Description |
---|---|
LT220 | |
LT225 |
