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.
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 Amivantamab. Amivantamab binds with high
specificity to EGFR and MET, particularly targeting EGFR mutations, including exon 20
insertions, and MET alterations.
Background
Anti-Human EGFR x MET bispecific antibodies have shown promising results in cancer
therapy. These antibodies target both the epidermal growth factor receptor (EGFR) and the
MET receptor, which are crucial in cancer progression and resistance mechanisms. Studies
have demonstrated that bispecific antibodies can inhibit proliferation, migration, and invasive
growth of tumor cells more effectively than single-agent treatments. Additionally, these
antibodies have been found to enhance tumor growth inhibition and downregulate both
EGFR and MET receptors, highlighting their potential in combating therapeutic resistance in
patients with EGFR mutations. The combination of EGFR and MET inhibition through
bispecific antibodies presents a valuable strategy for overcoming resistance mechanisms
and improving treatment outcomes in cancer patients1-3.
Amivantamab (JNJ-611) is a human bispecific monoclonal antibody that targets both the
EGFR and the hepatocyte growth factor receptor (HGFR/cMet). This bispecific antibody is
engineered to simultaneously bind to wild-type and mutant forms of EGFR and MET, thereby
blocking their phosphorylation and subsequent signaling pathways essential for cancer cell
proliferation. Preclinical studies demonstrated the superior efficacy of Amivantamab in
models with EGFR exon 20 insertions compared to other therapies like cetuximab or
poziotinib. In clinical settings, Amivantamab has shown promise in treating patients with
NSCLC, particularly those with EGFR exon 20 insertion mutations who have progressed
after platinum-based chemotherapy1,3.
Antigen Distribution
EGFR and MET are widely expressed in various epithelial tissues,
including lung, skin, and digestive tract linings. In cancer cells, these receptors can be
overexpressed or mutated, driving tumorigenesis.
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 Amivantamab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs to enable accurate, quantitative measurement of drug concentrations in serum samples, ensuring comparability between biosimilar and reference (originator) products.
Essential context and application:
In the PK bridging ELISA, the typical approach is to use a single analytical standard—either the biosimilar or the reference Amivantamab—as the calibrator for quantification of both the biosimilar and reference products in test samples. This is considered best practice because it minimizes assay variability and obviates the need for multiple separate assays for each product.
Calibration standards are prepared using known concentrations of research-grade biosimilar Amivantamab, serially diluted to generate a standard curve in the relevant sample matrix (e.g., human serum). This curve is then used to interpolate the concentrations of Amivantamab in unknown samples.
Reference controls (quality control or QC samples) are typically prepared from both biosimilar and reference products at multiple concentrations, then spiked into serum or plasma. These QC samples are quantified against the standard curve to assess assay accuracy, precision, and to establish the bioanalytical comparability of the PK bridging ELISA for both product types.
The validation of the assay includes running independent sets of biosimilar and reference controls to demonstrate that the assay measures both forms with equivalent precision and accuracy, evaluating for any systematic bias between biosimilar and originator reagents.
If bioanalytical comparability is demonstrated—often by showing the 90% confidence interval of the ratio of measured concentrations falls within a predefined equivalence interval (e.g., 0.8–1.25)—the biosimilar Amivantamab can be used as the primary standard in all subsequent studies. This is documented and supported by regulatory expectations and industry guidelines.
Summary of use in practice:
Biosimilar Amivantamab serves as the calibrator standard: serial dilutions provide the reference for the ELISA curve.
Reference and biosimilar Amivantamab are each used to prepare QC samples to verify the assay's reliability and equivalence for both forms of the drug.
This setup enables accurate measurement of Amivantamab concentrations in serum during PK studies and bridging studies between biosimilar and reference products.
Regulatory and methodological best practices require thorough documentation of comparability and validation so the PK data produced by the ELISA is robust for both biosimilar and reference Amivantamab, ensuring scientific rigor throughout biosimilar development.
If you need information on a specific ELISA kit protocol (such as KRIBIOLISA), these kits are designed with biosimilar compatibility in mind and are validated for accuracy and precision in quantifying Amivantamab levels in clinical and research samples.
The primary in vivo models for studying the effects of anti-EGFR x MET (c-Met) bispecific antibodies on tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization are human tumor xenograft models and, less commonly, humanized or syngeneic mouse models, depending largely on the antibody species cross-reactivity and the specific goals of TIL analysis.
Key model types:
Human tumor xenograft models: Most published studies involving anti-EGFR x MET bispecific antibodies use human tumor cell lines implanted subcutaneously in immunodeficient mice (e.g., nude or SCID mice). An example is the non-small cell lung cancer xenograft model used to demonstrate the efficacy of a bispecific EGFR/MET antibody (MetHer1), resulting in significant tumor growth inhibition. These models allow direct assessment of antibody impact on tumor growth but have limited utility for TIL studies because of the mice's compromised immune system.
Syngeneic mouse models: These are murine tumor cell lines implanted into immunocompetent, genetically matched mice. Syngeneic models are critical for evaluating how immunotherapies alter TIL populations, since the mice have a fully functional immune system. However, development of true anti-human EGFR x MET antibodies that cross-react with mouse targets is uncommon, limiting the direct use of research-grade human reagents in these systems. Engineered murine surrogates of the antibody may be required to conduct such studies in this context.
Humanized mouse models: These are immunodeficient mice reconstituted with human immune cells, permitting the simultaneous study of human tumor–human antibody–human immune system interactions. These models are complex and less commonly reported in the anti-EGFR x MET space, but could enable both efficacy and TIL characterization relevant to human biology if the antibody is of human origin and targets human antigens.
Model selection:
Model Type
Tumor Growth Inhibition
TIL Characterization
Antibody Considerations
Human xenograft
Yes
Limited (mouse immune system compromised)
Recombinant human antibodies
Syngeneic (murine)
Yes
Yes (robust TIL analysis)
Antibody must be murine-reactive or a murine surrogate used
Humanized
Yes
Yes (human TILs possible)
Human-specific antibodies
Key details for anti-EGFR x MET bispecifics:
Most tumor growth inhibition studies referenced in the literature (e.g., using MetHer1, JNJ-61186372) employ human tumor xenograft models in immunocompromised mice.
TIL analysis is best performed in syngeneic or humanized systems, since xenograft models do not recapitulate the full immune response.
If the main goal is TIL characterization after antibody administration, the model must support both immune cell recruitment and antibody binding, which requires careful antibody engineering.
Summary:
Human tumor xenograft models are the most established system for anti-EGFR x MET bispecific antibody in vivo efficacy studies.
Syngeneic models are commonly used for immunotherapy and TIL research generally, but are rarely used directly with human-specific bispecific antibodies without murine-adapted versions.
Humanized mouse models offer the best surrogate for combined efficacy and TIL characterization with human reagents, though published data are less common.
Researchers should select their model based on the antibody’s species reactivity and whether tumor growth inhibition, immune modulation, or both are primary endpoints.
Researchers use the Amivantamab biosimilar in combination with other checkpoint inhibitors—most commonly with anti-PD-1/PD-L1 antibodies—to study synergistic immune effects in complex tumor models, focusing on how dual targeting can overcome resistance and remodel the tumor immune microenvironment. While specific preclinical studies involving anti-CTLA-4 or anti-LAG-3 biosimilars in combination with Amivantamab biosimilar have not been explicitly documented in available sources, the methodology and rationale for such combinations are informed by approaches taken with anti-PD-1 agents.
Essential context and supporting details:
Combination Rationale: Amivantamab is a bispecific antibody targeting both EGFR and MET, exhibiting immune cell–directing activity and receptor downmodulation. When combined with checkpoint inhibitors, the goal is to simultaneously block tumor-intrinsic growth signals (EGFR/MET pathways) and release immune suppression mediated by pathways like PD-1, CTLA-4, or LAG-3.
Synergy and Immune Remodeling: In humanized patient-derived xenograft (PDX) models of squamous cell carcinoma, combining Amivantamab with pembrolizumab (anti-PD-1) led to:
Significant reduction in tumor growth, where neither agent alone was effective.
Increased infiltration and activation of granzyme B-producing CD8+ T cells and central memory CD8+ T cells in both the tumor microenvironment and circulation, resulting in enhanced tumor killing.
Suppression of glycolytic and lactate production markers in tumor cells, which normally promote immunosuppression and resistance to immunotherapy.
Immune Mechanism Analysis: Single-cell RNA sequencing and immunophenotyping allow researchers to assess changes in T cell subsets, activation status, and other immune cell populations after combination therapy.
Use of Biosimilars: The Amivantamab biosimilar (research-grade antibody) is employed in preclinical studies to mechanistically probe receptor blockade, receptor internalization, and effector function activation in EGFR/MET-driven tumor models, making it suitable for combinatorial studies with other checkpoint inhibitors.
Potential for Expansion:
Although most published studies focus on Amivantamab with anti-PD-1/PD-L1 agents, the same methodologies—tumor modeling, immune profiling, and functional assays—are applicable for combinations with anti-CTLA-4 and anti-LAG-3 biosimilars, enabling analysis of synergistic effects in models where T cell activation, exhaustion, and regulatory cell infiltration are of interest.
Summary Table: Example Mechanisms Studied in Amivantamab Biosimilar/Checkpoint Inhibitor Combination Models
Combination Partner
Main Synergistic Effects Studied
Model Types Used
Anti-PD-1/PD-L1 (e.g. pembrolizumab)
Enhanced CD8+ cytotoxic T cell activation, tumor growth inhibition, overcoming resistance via TME remodeling
Humanized PDX, syngeneic tumor models
Anti-CTLA-4 biosimilar
(Potential) Expanded CD4/CD8 T cell priming, regulatory T cell suppression
(Method inferred, not documented)
Anti-LAG-3 biosimilar
(Potential) Modulation of T cell exhaustion, additive effects with other inhibitors
(Method inferred, not documented)
Preclinical research using Amivantamab biosimilar in combination with checkpoint inhibitors leverages advanced immunogenomic and immune profiling technologies to unravel synergistic mechanisms—primarily focusing on enhanced T cell activity and overcoming immunosuppressive tumor adaptations.
An Amivantamab biosimilar can be used as the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response against Amivantamab by leveraging its identical antigen-binding domains to the therapeutic drug.
Essential context and supporting details:
A bridging ADA ELISA is designed to detect antibodies (ADAs) generated by a patient against a therapeutic antibody—here, Amivantamab—by using the drug (or biosimilar) as both the capture and detection reagent.
Biosimilars of Amivantamab share the same variable (antigen-binding) regions as the clinical antibody, making them suitable for immunoassay applications without using clinical-grade material.
Protocol overview (based on general ADA bridging ELISA principles):
Plate coating (capture reagent): The Amivantamab biosimilar is immobilized onto the ELISA plate (often directly adsorbed or via a biotin-streptavidin system). This reagent will capture any anti-Amivantamab antibodies present in a patient’s serum.
Incubation with patient samples: Serum samples from patients are incubated on the coated plate, allowing any ADAs specific to Amivantamab to bind.
Detection reagent: The same Amivantamab biosimilar—now labeled (e.g., with HRP enzyme or biotin)—is added. If ADAs are present, they form a "bridge" between the immobilized and labeled biosimilar reagent via their two antigen-binding sites.
Detection: A substrate for the label (such as TMB for HRP) is added, and the signal generated corresponds to the presence and amount of ADAs against Amivantamab.
Key scientific considerations:
The Amivantamab biosimilar’s identical antigen-binding specificity ensures the assay reliably detects immune responses against the therapeutic drug. Using a biosimilar rather than the clinical drug reduces cost and avoids depletion of clinical material.
By serving as both capture and detection reagent, the biosimilar enables the bridging format, which is highly sensitive for bivalent antibodies present in patients’ serum.
The assay can be customized for detection of different ADA classes (IgG, IgM, or IgE), facilitated by specific secondary reagents or detection formats.
Additional relevant details:
This approach ensures the measured immune response is specific to the therapeutic domains of Amivantamab, crucial for safety and efficacy monitoring in treated patients.
Bridging ADA ELISAs are widely used for immunogenicity testing of biotherapeutics and routinely adapted for bispecific monoclonal antibodies like Amivantamab.
In summary, an Amivantamab biosimilar is an effective, highly-specific reagent for both capturing and detecting anti-Amivantamab antibodies in patient samples using bridging ADA ELISA methodologies.
References & Citations
1. Petrini I, Giaccone G. Onco Targets Ther. 2022;15:1197-1210.
2. Neijssen J, Cardoso RMF, Chevalier KM, et al. J Biol Chem. 2021;296:100641.
3. Zhou C, Tang KJ, Cho BC, et al. N Engl J Med. 2023;389(22):2039-2051.
4. Amivantamab (JNJ-61186372) | EGFR-MET Dual Antibody | MedChemExpress. MedchemExpress.com. Accessed September 30, 2024.
https://www.medchemexpress.com/amivantamab.html