Anti-Human EGFR (Necitumumab)

Anti-Human EGFR (Necitumumab)

Product No.: LT610

- -
- -
Product No.LT610
Clone
IMC-11F8
Target
EGFR
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Epidermal growth factor receptor, ErbB1, Anti-Human EGFR, 906805-06-9, IMC-11F8
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC
,
IP
,
WB

- -
- -
Select Product Size

Data

Necitumumab-EGFR Direct Binding Data Leinco Prod. No.: LT610Direct binding of Human Recombinant EGFR (Leinco Prod. No.: E309) to anti-Human EGFR Necitumumab (Leinco Prod. No.: LT610)
Binding was measured by ELISA. Recombinant Human EGFR was immobilized at 1 µg/mL. Necitumumab antibody was titrated.
Necitumumab-EGFR Western Data Leinco Prod. No.: LT610WESTERN
Purified recombinant human EGFR (Leinco Prod. No.: E309) was separated on SDS-PAGE under non-reducing conditions and probed with Necitumumab (Leinco Prod. No.: LT610).
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Human EGFR/ErbB1
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.
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
Applications and Recommended Usage?
Quality Tested by Leinco
FA,
ELISA,
WB
Additional Applications Reported In Literature ?
FC,
IP,
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 Necitumumab. This product is for research use only. Necitumumab activity is directed against human EGFR.
Background
Epidermal growth factor receptor (EGFR, also known as ErbB1 or HER-1) belongs to the receptor tyrosine kinase superfamily and is a transmembrane glycoprotein that activates various signaling pathways fundamental to cellular proliferation, differentiation, and survival1, 2. EGFR plays important roles during embryogenesis, organogenesis, and in the growth, differentiation, maintenance, and repair of adult tissues2, including autophagy3. EGFR is also a host factor that facilitates viral entry for hepatitis B4, hepatitis C5, and gastroenteritis6 and plays a role in SARS-CoV-2 infection7, 8, 9. Dysregulation, somatic mutation, and/or altered signaling of EGFR are associated with disease (Parkinson’s2, Alzheimer’s1,2, and amyotrophic lateral sclerosis2) and various cancers (lung, glioblastoma, brain, breast, colorectal, ovarian)3. Additionally, in cancer, aberrant activation of EGFR is associated with increased cell proliferation, invasion, metastasis, angiogenesis, and decreased apoptosis10. As such, EGFR is the target of multiple cancer therapies, including monoclonal humanized antibodies, such as necitumumab, as well as selective small molecule inhibitors.

Necitumumab inhibits EGFR-dependent tumor cell proliferation and metastasis11 by acting as an EGFR antagonist that binds specifically and with high affinity to human EGFR, thereby blocking ligand binding and neutralizing ligand-induced EGFR phosphorylation and downstream signaling pathways10, 11, 12. Anti-tumor activity has been demonstrated in vitro and in vivo 10, and necitumumab is FDA approved for treatment of adult patients with locally advanced or metastatic EGFR-expressing squamous non-small cell lung cancer11. In vitro studies have shown that necitumumab induces internationalization and degradation of EGFR, leading to antibody-dependent cellular cytotoxicity in EGFR-expressing cells10, 11. Necitumumab was generated from the Dyax Corp proprietary phage display library by ImClone Systems, a wholly owned subsidiary of Eli Lilly10.
Antigen Distribution
EGFR is overexpressed on the cell surfaces of various tumor cell types and is also found in the plasma membranes, cytoplasm, and cell junctions of many healthy tissues, including those associated with the Skin – Epidermis development cluster of The Human Protein Atlas. EGFR is also found in the blood secretome.
Ligand/Receptor
Epidermal growth factor receptor
PubMed
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Cell Biology
.
Immuno-Oncology
.
Immunology

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 Necitumumab biosimilars can be used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to quantitatively measure drug concentrations in serum samples.

In a PK bridging ELISA for biosimilar development:

  • A single analytical standard (often the biosimilar itself) is selected as the assay calibrator for both the biosimilar and reference product, after demonstrating bioanalytical equivalence through method qualification studies.
  • Calibration is achieved by preparing serial dilutions of the research-grade Necitumumab biosimilar to establish a standard curve on each ELISA plate. The measured absorbance (O.D.) at each concentration is fit to a mathematical model, such as a 4-parameter logistic (4PL) curve, to interpolate unknown sample concentrations.
  • Reference controls (quality controls), typically samples prepared with either the biosimilar or reference product at various known concentrations, are included in each assay to monitor assay performance, precision, and accuracy across plates and runs.
  • Bioanalytical comparability between the biosimilar and the reference product is statistically assessed—typically using methods like Bland-Altman analysis—to ensure that the biosimilar calibrator accurately measures both products in test samples.

Essential context and key steps:

  • The selected research-grade Necitumumab biosimilar standard must be well-characterized and traceable, ensuring reliable quantitative comparison.
  • Method validation ensures that the ELISA assay is robust, precise, and accurate for measuring both biosimilar and reference Necitumumab in serum matrix.
  • Negative and positive controls are run on each plate to control for background and assay sensitivity, improving inter-assay reproducibility and reliability.
  • Use of a single ELISA method and standard minimizes variability and simplifies assay logistics in blinded clinical and nonclinical studies.

In summary, research-grade Necitumumab biosimilars serve as highly characterized calibration standards and reference controls in PK bridging ELISA assays, enabling precise and equivalent quantification of drug concentrations for clinical and nonclinical bioequivalence studies.

Primary Models for Studying Anti-EGFR Antibodies and Tumor-Infiltrating Lymphocytes (TILs)

To study the effects of anti-EGFR antibodies on tumor growth inhibition and characterize TILs, researchers often employ syngeneic or humanized mouse models. Here are the primary models used:

1. Murine Syngeneic Models

  • Description: These models involve using mouse-derived tumor cells implanted into immunocompetent mice. This approach allows for the study of tumor growth and immune interactions in a genetically identical host, which is beneficial for understanding the role of the immune system in cancer progression and therapy.
  • Use in Anti-EGFR Studies: Syngeneic models are commonly used to evaluate the efficacy of anti-cancer therapies, including anti-EGFR antibodies, by monitoring tumor growth and immune responses. However, they may not directly reflect human immune responses due to species differences.

2. Humanized Mouse Models

  • Description: Humanized mice are genetically modified to express human antigens or immune systems, allowing for a more accurate representation of human immune responses. These models are particularly useful when studying human-specific antigens or therapies.
  • Use in Anti-EGFR Studies: Humanized models, such as those with human EGFR expression, can be used to study the effects of anti-EGFR antibodies in a setting that more closely mimics human biology. This can help in understanding how these antibodies interact with TILs and other components of the human immune system.

3. Specific Example: Athymic (Nude) Mice with Xenografts

  • Description: Athymic mice lack a functional immune system and are used for xenografting human tumor cells. This model is particularly useful for studying the direct effects of anti-EGFR antibodies on tumor growth without the confounding effects of an intact immune system.
  • Use in Anti-EGFR Studies: As seen in studies involving anti-EGFR Nanobodies, athymic mice with human tumor xenografts (e.g., A431 cells) are used to assess the efficacy of these antibodies in inhibiting tumor growth in vivo without the influence of a host immune response.

Characterizing TILs

In both syngeneic and humanized models, researchers can analyze TILs to understand how anti-EGFR therapies affect the tumor microenvironment. This includes examining the types and functions of immune cells present in the tumor, such as T cells and macrophages, which can provide insights into how these therapies modulate immune responses against tumors.

Based on the available search results, there is limited specific information about researchers using necitumumab biosimilars in conjunction with other checkpoint inhibitors to study synergistic effects. However, I can provide insights into the current research landscape and methodological approaches being explored in this area.

Current State of Necitumumab Research

Necitumumab is an anti-EGFR monoclonal antibody that has been established as a standard therapy component for advanced lung squamous cell carcinoma when combined with gemcitabine and cisplatin. The drug works by binding to EGFR with high affinity and blocking signal transmission, inhibiting tumor growth. Clinical studies like the international SQUIRE trial and the Japanese JFCM study have demonstrated significant improvements in overall survival when necitumumab is added to chemotherapy regimens.

Combination Immunotherapy Strategies

The field of immune-oncology is actively exploring combination strategies that target multiple immune checkpoints simultaneously. The rationale behind combining different checkpoint inhibitors stems from their distinct mechanisms of action - for example, anti-CTLA-4 agents primarily act in lymph node compartments to restore T cell activation and proliferation, while anti-PD-1/PD-L1 agents work at the tumor periphery to prevent cytotoxic T cell neutralization.

Research Methodologies for Studying Synergistic Effects

Researchers are employing several approaches to study combination effects:

Preclinical Models: Multiple preclinical studies are investigating combinations of immune checkpoint inhibitors with various therapeutic agents. These models help establish the mechanistic basis for beneficial therapeutic interactions before advancing to clinical trials.

Clinical Trial Designs: Early-phase clinical studies are being conducted to evaluate combination strategies. For instance, there are ongoing phase Ib/II trials examining necitumumab in combination with other targeted therapies like trastuzumab and osimertinib for EGFR-mutated non-small cell lung cancer.

Real-World Evidence Studies: Researchers are conducting multicenter retrospective studies to assess the efficacy and tolerability of combination regimens in real-world settings, particularly examining how treatments perform after patients have received prior immune checkpoint inhibitor therapy.

Challenges and Considerations

The major limitation of combination immunotherapy approaches is the increased risk of grade 3 or grade 4 toxicities. This necessitates careful dose optimization and patient selection strategies. Additionally, the efficacy of combination approaches may vary based on biomarker status - for example, patients with PD-L1-negative tumors may benefit more from combination therapy than those with PD-L1-positive tumors.

While the search results do not provide specific details about necitumumab biosimilar combinations with anti-CTLA-4 or anti-LAG-3 inhibitors, the general approach to studying such combinations would likely involve preclinical immune-oncology models followed by carefully designed early-phase clinical trials with appropriate safety monitoring and biomarker-driven patient selection strategies.

A Necitumumab biosimilar can be used as a reagent in a bridging anti-drug antibody (ADA) ELISA to detect patient immune responses (ADAs) against Necitumumab or its biosimilar by exploiting the antibody’s ability to "bridge" two forms of the drug: one immobilized on the plate (capture) and one labeled for detection.

Context and Explanation:

  • Bridging ADA ELISA Principle: In this assay, patient serum is added to a plate coated with Necitumumab (or its biosimilar). If anti-drug antibodies (ADA) are present in the serum, they will bind to this immobilized drug. Then, a labeled Necitumumab biosimilar (e.g., biotinylated or HRP-conjugated) is added; ADAs that recognize Necitumumab will also bind the labeled drug, forming a "bridge" between the capture and detection reagents.

  • Use of Biosimilar as Reagent:

    • The biosimilar can be used for either or both of the following roles:
      • As the capture reagent (coated onto the ELISA plate)
      • As the detection reagent (labeled with a tag such as HRP or biotin for detection)
    • This dual use leverages the similarity of the biosimilar to the reference product (Necitumumab), allowing detection of ADAs that cross-react with both the reference drug and its biosimilar.
  • Assay Workflow Overview:

    • Patient serum is incubated on a plate coated with Necitumumab biosimilar.
    • ADAs in the serum bind to the immobilized drug.
    • After washing, a labeled Necitumumab biosimilar is added, which binds to a different epitope on the same ADA.
    • The formation of a "bridge" (Drug–ADA–Drug) is detected via a colorimetric or chemiluminescent signal after adding a substrate (if HRP or similar is used).
  • Advantages:

    • Sensitivity to all isotypes: The bridging format can detect a wide range of ADA isotypes (e.g., IgG, IgM) as long as they are multivalent.
    • Detection of cross-reactivity: The use of a biosimilar for capture or detection allows the assay to pick up ADAs that can bind to both the reference drug and biosimilar product, which is important for immunogenicity assessments.
  • Relevance for Monitoring: Such assays are used in clinical trials and post-market surveillance to monitor for the development of ADA responses, which can impact efficacy and safety of the therapeutic, including altered pharmacokinetics or loss of drug effect.

Summary Table: Necitumumab Biosimilar in Bridging ADA ELISA

Assay ComponentRoleReagent UsedFunction
Plate coatingCapture reagentNecitumumab biosimilarImmobilizes drug for ADA binding
DetectionDetection reagentLabeled Necitumumab biosimilar (HRP/Biotin)Binds to a separate epitope of ADA, allowing signal generation
Patient serumAnalyte (ADA)Antibodies (if present)Bridges between capture and detection drug molecules

Key Point:
Using a Necitumumab biosimilar as either the capture or detection reagent in a bridging ADA ELISA enables sensitive and specific monitoring of the patient's immune response against the therapeutic drug, detecting both biosimilar- and reference drug-reactive ADAs.

If you need specific labeling or procedural details for a particular platform or regulatory guidance, please clarify, as protocols may vary depending on assay design and regulatory requirements.

References & Citations

1. Jayaswamy PK, Vijaykrishnaraj M, Patil P, et al. Ageing Res Rev. 83:101791. 2023.
2. Romano R, Bucci C. Cells. 9(8):1887. 2020.
3. Sigismund S, Avanzato D, Lanzetti L. Mol Oncol. 12(1):3-20. 2018.
4. Iwamoto M, Saso W, Sugiyama R, et al. Proc Natl Acad Sci U S A. 116(17):8487-8492. 2019.
5. Lupberger J, Zeisel MB, Xiao F, et al. Nat Med. 17(5):589-595. 2011.
6. Hu W, Zhang S, Shen Y, et al. Virology. 521:33-43. 2018.
7. Klann K, Bojkova D, Tascher G, et al. Mol Cell. 80(1):164-174.e4. 2020.
8. Xu G, Li Y, Zhang S, et al. Cell Res. 31(12):1230-1243. 2021.
9. Wang S, Qiu Z, Hou Y, et al. Cell Res. 31(2):126-140. 2021.
10. Garnock-Jones KP. Drugs. 76(2):283-289. 2016.
11. Fala L. Am Health Drug Benefits. 9(Spec Feature):119-122. 2016.
12. https://www.cancer.gov/publications/dictionaries/cancer-drug/def/necitumumab?redirect=true
Indirect Elisa Protocol
FA
Flow Cytometry
Immunoprecipitation Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

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