Anti-Human EGFR (Panitumumab) – Fc Muted™

Anti-Human EGFR (Panitumumab) – Fc Muted™

Product No.: LT625

- -
- -
Product No.LT625
Clone
ABX-EGF
Target
EGFR
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Epidermal growth factor receptor, ErbB1, Anti-Human EGFR, ABX-EGF
Isotype
Human IgG2κ
Applications
ELISA
,
FA
,
FC
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
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 ?
IP,
FC
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 Panitumumab. This product is for research use only. Panitumumab 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. 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 is associated with neurological diseases (e.g. Parkinson’s2, Alzheimer’s1, 2, and amyotrophic lateral sclerosis2) and multiple cancers (lung, glioblastoma, brain, breast, colorectal, ovarian)10. Additionally, in cancer, binding of ligands to EGFR is associated with aberrant cell proliferation, invasion, metastasis, angiogenesis, and decreased apoptosis11. As such, EGFR is the target of multiple cancer therapies, including monoclonal humanized antibodies, such as panitumumab, as well as selective small molecule inhibitors.

Panitumumab was generated in a XenoMouse IgG2 strain immunized with the human cervical epidermal carcinoma cell line A43112. Panitumumab binds specifically to EGFR and inhibits the growth and survival of selected human tumor cell lines over-expressing EGFR in vitro and in vivo13. Panitumumab binds EGFR with high affinity, blocking the binding of both EGF and TGF-α, and preventing EGF-activated EGFR tyrosine autophosphorylation and downstream activation of receptor-associated kinases12. Panitumumab inhibits cell growth, tumor cell activation, in vitro tumor cell proliferation12, and metastasis13. Panitumumab also induces apoptosis and decreases proinflammatory cytokine and vascular growth factor production13. Additionally, upon binding, panitumumab causes EGFR internalization in tumor cells12.

Panitumumab was approved in the United States for the treatment of some patients with EGFR-expressing metastatic colorectal cancer14, 15.
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 panitumumab biosimilars can be used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs by serving as the quantifiable reference material against which unknown serum sample concentrations are measured. This approach enables robust, direct comparison of drug levels for both the biosimilar and reference product within the same analytical method, critical for bioequivalence assessments and PK bridging studies.

Context and Supporting Details:

  • In a PK bridging ELISA, calibration standards (also referred to as calibrators or analytical standards) are prepared using a known concentration of the test article, which can be the biosimilar, the reference product, or both.
  • The assay generally employs a single, well-characterized biosimilar preparation as the analytical standard for constructing the standard curve, covering a defined concentration range relevant for quantifying therapeutic levels in serum. For panitumumab, calibrator concentrations often span from low ng/mL to tens of mg/L depending on the assay's sensitivity and therapeutic range requirements.
  • Reference controls or quality controls (QCs) are typically prepared separately using either the biosimilar or the reference product, at multiple concentrations relevant to the expected clinical range. These evaluate assay precision and accuracy for both drug forms.

Why biosimilars can be used as calibrators/standards:

  • Regulatory and scientific consensus recommends using a single PK assay and potentially a single analytical standard (usually the biosimilar intended for clinical use) to minimize analytical variability and ensure comparable quantification of both the biosimilar and the originator (reference) product.
  • Before using a biosimilar as the reference standard, bioanalytical comparability between the biosimilar and the reference product must be demonstrated, typically by spiking both into serum and showing the ELISA quantifies both with equivalent precision and accuracy.
  • Once equivalence is confirmed, the biosimilar is selected as the analytical standard for all sample and QC measurements.

How are these standards and controls used practically?

  • Serum samples from PK studies are diluted and compared against the standard curve generated with the biosimilar calibrator.
  • QC samples prepared with both biosimilar and reference-panatumumab at low, medium, and high concentrations are run on each plate to ensure assay validity, precision, and accuracy across the expected clinical range.
  • This method allows consistent and robust comparison across batches, studies, and between the biosimilar and originator, forming the basis for bioequivalence and bridging assessments.

Example:

  • In validated panitumumab PK ELISAs, six calibrators (0.1–20 mg/L) plus an anchor point (up to 50 mg/L) are defined using reference material, with QCs at representative concentrations throughout the range to monitor accuracy and precision. Kits on the market sometimes note that their standards are "calibrated against commercially sourced Vectibix Injection," which means either the biosimilar or reference product is used for curve generation.

Summary Table:

Role in ELISASource MaterialPurpose
Calibration StandardsPanitumumab biosimilar (or reference)Generate standard curve for quantifying unknowns
Quality Control SamplesBoth biosimilar and referenceConfirm precision and accuracy of quantitation

This approach is essential to ensure confidence in measured drug concentrations and support regulatory submissions for biosimilars.

The primary in vivo models to study anti-EGFR antibody effects on tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are:

  • Human tumor xenograft models in immunodeficient mice (such as athymic nude or NOD/SCID mice), using human cancer cell lines that overexpress human EGFR and administering research-grade anti-EGFR antibodies.
  • Syngeneic mouse tumor models genetically engineered to express human EGFR (or with knock-in human antigens), enabling study in immunocompetent mice and robust TIL characterization.

Context and Supporting Details:

  • Xenograft Models: Standard models involve implanting human cancer cells (e.g., A431 cells, which are EGFR-overexpressing) into immunodeficient mice and treating with anti-EGFR antibodies, including classical and next-generation formats such as nanobodies. While tumor growth inhibition and some immune correlates can be observed, these models lack a functional mouse immune system, so detailed TIL profiling is limited.

  • Syngeneic/Transgenic Humanized Models: To assess both tumor response and immune cell (including TIL) dynamics, syngeneic mouse models are used, where mouse tumor cell lines are engineered to express human antigens such as EGFR. This approach leverages fully immunocompetent hosts, enabling comprehensive analysis of TIL recruitment and function upon antibody treatment. These models permit in vivo immunoprofiling, including the study of CD8+, CD4+, and myeloid populations infiltrating the tumor after antibody therapy.

Key Points:

  • Syngeneic tumor models (e.g., MC38, TC-1 with engineered human EGFR expression) are preferred for in-depth TIL studies, as they offer a more physiologic tumor-immune interaction.
  • Humanized mouse models (mice with human immune system engraftment or with human EGFR knock-in) are essential when the anti-EGFR antibody is human-specific and requires human target/immune context for activity.
  • Immunocompetent syngeneic models are widely used for preclinical immunotherapy evaluation due to their predictive value for human clinical outcomes and the ability to analyze TILs using flow cytometry and multiplex immunohistochemistry.

In summary, the choice between xenograft and syngeneic/humanized models depends on the specificity of the anti-EGFR antibody and the requirement to profile immune responses, with syngeneic and humanized models being essential when TIL characterization is a primary endpoint.

Researchers utilize panitumumab biosimilars in combination with checkpoint inhibitors like anti-CTLA-4 and anti-LAG-3 to investigate complex mechanistic interactions and potential synergistic effects in immune-oncology research. This approach leverages the distinct but complementary mechanisms of action of these therapeutic classes to create more comprehensive preclinical models.

Mechanistic Rationale for Combination Studies

The strategic combination of panitumumab biosimilars with checkpoint inhibitors is based on their different yet potentially synergistic mechanisms of action. Panitumumab is a fully human IgG2 monoclonal antibody that binds with high affinity to the ligand-binding region of EGFR, completely blocking its association with ligands and activation of downstream kinase cascades. This EGFR blockade can prevent xenograft formation and completely eradicate established tumors, with sustained inhibition lasting up to 8 months after discontinuation of therapy.

Meanwhile, checkpoint inhibitors operate through distinct immune system modulation pathways. The combination of multiple checkpoint inhibitors targets different mechanisms, with anti-CTLA-4 primarily acting in the lymph node compartment to restore induction and proliferation of activated T cells, while anti-PD-1 mainly functions at the tumor periphery, preventing neutralization of cytotoxic T cells by PD-L1 expressing tumor cells.

Differential Immune Cell Activation Patterns

Recent research has revealed that different checkpoint inhibitor combinations activate distinct immune cell subsets, providing researchers with opportunities to study how panitumumab biosimilars might interact with these varied immune responses. Anti-PD-1/LAG-3 combinations require the presence of CD4 T-cells for their anticancer effects in both cutaneous melanoma and brain metastases, decreasing Treg cell activity and increasing CD4 helper T cell activity that leads to CD8 T-cell activation. In contrast, anti-PD-1/CTLA-4 combinations do not require CD4 T-cell presence and result in direct accumulation and activation of more cytotoxic CD8 T cells.

Research Applications and Model Systems

Researchers employ these combination strategies in sophisticated preclinical models to study synergistic effects. The panitumumab biosimilar, which uses the same variable regions as the therapeutic antibody, provides an ideal research tool for investigating these interactions. When combined with chemotherapeutic agents, panitumumab has demonstrated enhanced inhibition of tumor growth and eradication of carcinoma cells in both in vitro and in vivo models.

Patient Selection and Biomarker Integration

A critical aspect of these combination studies involves understanding how EGFR targeting interacts with immune checkpoint modulation based on specific biomarkers. Research has shown that K-ras mutation status significantly impacts panitumumab efficacy, with wild-type K-ras patients showing median progression-free survival of 12.3 weeks compared to 7.3 weeks in controls, while mutated K-ras patients showed no benefit. This biomarker-driven approach allows researchers to study how checkpoint inhibitor combinations might overcome resistance mechanisms in different genetic contexts.

The integration of panitumumab biosimilars with checkpoint inhibitors in research models enables scientists to investigate whether the substantial survival benefits observed with individual therapies can be enhanced through combination approaches, potentially transferring the 40% survival benefit seen in wild-type RAS patients into earlier treatment settings and exploring whether such combinations can rival the survival advantages observed with other therapeutic approaches.

A Panitumumab biosimilar is typically used as both the capture and detection reagent in a bridging ADA (anti-drug antibody) ELISA to monitor a patient’s immune response against the therapeutic drug. In this assay format, the biosimilar serves as a surrogate for the reference drug and enables detection of antibodies directed against Panitumumab.

Mechanism in Bridging ADA ELISA:

  • Capture Reagent: The Panitumumab biosimilar (often identical in structure to the originator) is immobilized on a microtiter plate. When patient serum is added, any ADAs specific to Panitumumab present in the serum will bind to the immobilized biosimilar.
  • Detection Reagent: A labeled (e.g., HRP- or biotin-conjugated) Panitumumab biosimilar is then added. If anti-Panitumumab antibodies from the patient's serum are present, they will have bound to plate-coated biosimilar at one paratope, leaving the other paratope free. The labeled biosimilar binds to this free paratope, thus forming a "bridge" between the capture and detection reagents via the ADA.

Readout: After washing away unbound detection reagent, a substrate is added for enzymatic detection, generating a measurable signal proportional to the amount of ADA "bridging" the biosimilar reagents.

Why Use a Biosimilar as Reagent?

  • Biosimilars are structurally and functionally equivalent to the originator drug, so they present the same epitopes for immune recognition.
  • They are used interchangeably with originator in ADA testing to ensure that the assay detects antibodies against both reference and biosimilar forms, which is crucial for pharmacovigilance when switching between products.

Additional notes:

  • The bridging ADA ELISA can detect all isotypes of antibodies that can bridge two identical antigens, most commonly IgG and IgM.
  • If a biosimilar is used in clinical practice, it is appropriate to use the biosimilar as both the capture and detection reagent to accurately monitor anti-biosimilar responses.
  • The assay must be properly validated to ensure sensitivity and specificity for both the reference and biosimilar forms, since minor sequence or glycosylation differences could affect binding in rare cases.

Summary Table: Bridging ADA ELISA Using Biosimilar Panitumumab

StepReagentPurpose
Plate coatingBiosimilar PanitumumabCapture ADA from patient serum
Serum additionPatient sampleSource of potential anti-Panitumumab ADA
Detection reagent additionLabeled biosimilar PanitumumabBinds to ADA to complete the "bridge"
Signal generationSubstrate for label (e.g., TMB for HRP)Quantifies ADA presence

This approach is a standard in immunogenicity testing, allowing detection and monitoring of patient immune responses to monoclonal antibody therapies like Panitumumab.

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. Sigismund S, Avanzato D, Lanzetti L. Mol Oncol. 12(1):3-20. 2018.
11. Garnock-Jones KP. Drugs. 76(2):283-289. 2016.
12. Yang XD, Jia XC, Corvalan JR, et al. Crit Rev Oncol Hematol. Apr;38(1):17-23. 2001.
13. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/125147s080lbl.pdf
14. Dubois EA, Cohen AF. Br J Clin Pharmacol. 68(4):482-483. 2009.
15. Saltz L, Easley C, Kirkpatrick P. Nat Rev Drug Discov. 5(12):987-988. 2006.
16. Giusti RM, Shastri KA, Cohen MH, et al. Oncologist. 12(5):577-583. 2007.
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.