EGF Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Structure and Production

EGF Recombinant Monoclonal Antibodies are typically IgG-class immunoglobulins generated through hybridoma or phage display technologies. Key structural features include:

  • Antigen-binding regions (Fab): Engineered to recognize specific epitopes on EGF or EGFR .

  • Fc region: Mediates immune effector functions such as antibody-dependent cellular cytotoxicity (ADCC) .

  • Recombinant modifications: Humanization (e.g., cetuximab) or chimerization (e.g., panitumumab) to reduce immunogenicity .

Mechanism of Action

EGF mAbs inhibit ligand-receptor interactions or downstream signaling pathways:

MechanismExample AntibodiesEffect
Ligand binding blockadeMGR1 , E7.6.3 Prevents EGF/TGF-α from activating EGFR, blocking autocrine growth signals
Receptor dimerization inhibitionCetuximab , Panitumumab Disrupts EGFR homo-/heterodimerization, impairing kinase activation
Immune-mediated cytotoxicityIgE anti-EGFR Triggers degranulation of effector cells (e.g., mast cells, eosinophils)

In cancer cells, these antibodies suppress proliferation and induce apoptosis by blocking RAS-RAF-MEK-ERK and PI3K-AKT pathways .

Research Applications

EGF mAbs are widely used in experimental and diagnostic settings:

  • Western blot/Immunofluorescence: Detects EGF/EGFR expression in tissues (e.g., MAB236 in human skin keratinocytes) .

  • Neutralization assays: Inhibits EGF-induced cell proliferation (IC₅₀: 0.05–0.1 µg/mL for MAB236 in Balb/3T3 fibroblasts) .

  • Immunohistochemistry: Identifies EGFR overexpression in tumors (e.g., meningiomas, head and neck cancers) .

Cancer Treatment

  • Colorectal/Head and Neck Cancers: Cetuximab (chimeric) and panitumumab (human) improve survival in KRAS wild-type tumors .

  • Xenograft eradication: Fully human E7.6.3 antibody eliminated A431 epidermoid carcinomas in mice at doses as low as 0.6 mg .

  • Combination therapies: Synergizes with radiation/chemotherapy to enhance tumor sensitivity .

Autoimmune and Inflammatory Diseases

  • Asthma: Omalizumab (anti-IgE) reduces allergic responses .

  • Rheumatoid arthritis: TNF-α inhibitors like adalimumab .

Clinical and Preclinical Findings

AntibodyTargetKey OutcomeStudy
E7.6.3 (human IgG)EGFRAchieved 100% A431 xenograft eradication in mice; no recurrence after 8 monthsPhase I trials
MGR1 (mouse IgG)EGFRInhibited tumor growth in cells with >5×10⁴ receptors/cellPreclinical
CetuximabEGFRImproved median survival in SCCHN by 19.7 months with radiotherapyClinical

Challenges and Future Directions

  • Resistance mechanisms: Tumors develop EGFR polymorphisms, nuclear internalization, or exosome-mediated evasion .

  • Novel formats: Bispecific antibodies and CAR-T cells incorporating EGFR-specific scFv fragments show promise in glioma models .

  • Safety: Soluble EGFR (sEGFR) may trigger off-target degranulation, necessitating epitope-specific engineering .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

CUSABIO's approach to developing a recombinant monoclonal antibody targeting EGF commenced with the immunization of a rabbit using a synthesized peptide derived from human EGF protein. Subsequent steps involved isolating B cells from the immunized rabbit and extracting RNA from these cells. The extracted RNA was reverse-transcribed into cDNA, which served as a template for extending EGF antibody genes using degenerate primers. These engineered EGF antibody genes were then integrated into a plasmid vector and introduced into host cells for expression. The resulting EGF recombinant monoclonal antibody was purified from the cell culture supernatant via affinity chromatography and evaluated for its suitability in ELISA, IHC, and FC applications, demonstrating specific recognition of human EGF protein.

EGF is a versatile and essential growth factor that plays a pivotal role in regulating various cellular processes, including cell proliferation, differentiation, and tissue repair. Its signaling through the EGFR is tightly controlled to ensure proper tissue development and maintenance.

Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary based on different purchasing methods or location. Please consult your local distributors for specific delivery timelines.
Synonyms
Pro-epidermal growth factor (EGF) [Cleaved into: Epidermal growth factor (Urogastrone)], EGF
Target Names
EGF
Uniprot No.

Target Background

Function
EGF stimulates the growth of various epidermal and epithelial tissues both in vivo and in vitro, as well as certain fibroblasts in cell culture. As a magnesiotropic hormone, it promotes magnesium reabsorption in the renal distal convoluted tubule by engaging EGFR and activating the magnesium channel TRPM6. Notably, EGF can induce neurite outgrowth in motoneurons of the pond snail Lymnaea stagnalis in vitro.
Gene References Into Functions
  1. Our research indicates that the chimeric EGFETA toxin demonstrates exceptional effectiveness against EGFR-positive cancers, suggesting its potential for further development as a therapeutic agent for targeting EGFR-positive tumors resistant to monoclonal antibodies. PMID: 30226622
  2. These findings highlight the potential role of EGF in promoting hepatocellular carcinoma (HCC) metastasis, revealing a novel pathway for regulating FN expression and identifying potential targets for HCC prevention and treatment. PMID: 29315755
  3. The abnormally elevated expression of EGF and TGF-alpha is closely associated with the occurrence and development of chronic pancreatitis and pancreatic cancer. PMID: 29125273
  4. ERRa positively regulated the cell proliferation, migration, and invasion of colon cancer cells, and the suppression of ERRa completely reduced the EGF treatment-induced proliferation of colon cancer cells. PMID: 30185207
  5. EGF significantly upregulated RFPL3 and hTERT protein levels in the nonsmall cell lung cancer cells. RFPL3 and hTERT proteins upregulation by EGF were attenuated by pretreatment with AG1478 and erlotinib. EGF promoted proliferation and inhibited apoptosis; PD98059 decreased RFPL3 and hTERT protein expression; and RFPL3 overexpression increased the expression of hTERT and related MEKpathway proteins. PMID: 29749533
  6. We have identified the novel N-72, and it was found to be critical for EGF-induced migration by targeting MMP2 in Human amnion mesenchymal stem cells (hAMSCs). PMID: 29734654
  7. The spleen can regulate the functions of hematopoietic stem cells in cirrhotic hypersplenism by modulating EGF signaling. PMID: 29721775
  8. After HIP1 expression was blocked by siRNAs, EGFR endocytosis was accelerated, and this effect was dependent on the EGF concentration. This endocytosis was colocalized with clathrin expression. These findings indicate that the inhibition of HIP1 can accelerate the endocytosis and degradation of EGFR. PMID: 29039605
  9. The present study demonstrated that EGF induced aggressiveness of gastric cancer cells by activating epithelial to mesenchymal transition, which involved the activation of the ERK1/2 pathway and, subsequently, uPAR expression. PMID: 28849196
  10. The EGF system acts as a mechanosensitizer in bone marrow stromal cells. PMID: 28843157
  11. EGF counteracts Tat modulation of human endogenous retroviruses of the W family in astrocytes. PMID: 28474333
  12. FTIR spectra of EGF, unconjugated, post treatment with alpha-lipoic acid, attached to gold nanoparticle, and bound to the bifunctional nanoprobe, showed decreasing disordered structures and turns, and increasing loops, as the synthesis process progressed. There was an overall increase in beta-sheets in the final product compared to pure EGF, but this increase was not linear and fluctuated. PMID: 29122663
  13. EGF-mediated lysosome trafficking, protease secretion, and invasion are regulated by the activity of p38 mitogen activated protein kinase (MAPK) and sodium hydrogen exchangers (NHEs). Interestingly, EGF stimulates anterograde lysosome trafficking through a different mechanism than previously reported for HGF, suggesting that there are redundant signaling pathways that control lysosome positioning. PMID: 28978320
  14. Although the diabetic chronic wounds microenvironment is hostile for local GFs bioavailability, EGF local infiltration circumvented the limitations of its topical application, thus expanding its therapeutic prospect. Our clinical pharmacovigilance and basic studies attest to the significance of the GF local infiltration for chronic wounds healing. PMID: 28904952
  15. These results provide the first evidence for an association between the EGF rs2298999 C/T polymorphism and gout. PMID: 27506295
  16. The increased EGFR expression observed in patients with seborrheic keratomas (SK) and concomitant type 2 diabetes mellitus (DM2) is caused by insulin resistance and hyperinsulinemia, where the dysregulation of insulin signal transmission into the cell leads to changes in EGF synthesis and signaling pathway that regulates cell proliferation and growth. PMID: 28791994
  17. A novel EGFR-NF-kappaB-FOXC1 signaling axis that is critical for BLBC cell function has been identified. PMID: 28629477
  18. EGFR pathway gene expression analysis indicated that DeltaNp63 alters EGFR-regulated genes involved in cell adhesion, migration, and angiogenesis. Addition of EGF or neutralizing EGFR antibodies demonstrated that EGFR activation is responsible for DeltaNp63-mediated loss of cellular adhesion. PMID: 28349272
  19. EGF up-regulated CCL2 expression in HNSCC cells, which recruited monocytes and turned them into M2-like macrophages, thus forming a positive feedback paracrine loop. PMID: 27888616
  20. This study demonstrates that EGF induces epithelial-mesenchymal transition through phospho-Smad2/3-Snail signaling pathway in breast cancer cells. PMID: 27829223
  21. EGF and TNFalpha cooperatively promoted the motility of HCC cells mainly through NF-kappaB/p65 mediated synergistic induction of FN in vitro. These findings highlight the crosstalk between EGF and TNFalpha in promoting HCC, and provide potential targets for HCC prevention and treatment. PMID: 28844984
  22. Data suggest that EGF induces colorectal cancer cells to undergo epithelial-mesenchymal transition, enhances their ability to invade/migrate, and promotes phosphorylation of Ezrin at Tyr353. (EGF = epidermal growth factor) PMID: 28535417
  23. Simulation results indicate that human epidermal growth factor receptor (hEGFR) soluble soluble extracellular domains (sECD):EGF show different dynamic properties between the two pHs, and the complex may have a higher tendency of activation at pH 8.5. PMID: 27179806
  24. EGF and IP-10 were significantly elevated and GRO levels were lower in the tear profile of HIV patients with dry eye disease (DED) compared to immunocompetent patients with DED. PMID: 27585367
  25. Data (including data from studies using transgenic/knockout mice) suggest that surfactant protein A1 (SPA1) interferes with EGF binding to EGFR in pulmonary alveoli cell lines; SPA1 directly binds the extracellular domain of EGFR; binding of SPA1 to EGFR appears to be different from binding of SPD to EGFR; binding of SPA1 to EGFR does not suppress EGF-induced phosphorylation of EGFR or cell proliferation. PMID: 28972165
  26. EGF-AREG interplay in airway basal cell stem/progenitor cells is one of the mechanisms that mediates the interconnected pathogenesis of all major smoking-induced lesions in the human airway epithelium. PMID: 27709733
  27. Caspase-3 inhibitors also suppressed the attenuation of cell adhesion and phosphorylation of p38 MAPK by EGF-F9. Our data indicated that EGF-F9 activated signals for apoptosis and induced de-adhesion in a caspase-3 dependent manner. PMID: 27129300
  28. Evidence suggests that CDK1/2 participate in the regulation of constitutive pre-mRNA splicing by EGF stimulation in MDA-MB-468 cells. PMID: 27109354
  29. The EGF rs4444903 GG genotype is associated with higher susceptibility to HCV-related liver cirrhosis and hepatocellular carcinoma in the Chinese Han population. PMID: 28397482
  30. TGF-beta opposes EGF-mediated sensitization to TRAIL-induced caspase-8 activation and apoptosis in non-transformed breast epithelial cells. EGF and TGF-beta finely regulate the sensitivity of human breast epithelial cells to TRAIL, which may be relevant during morphogenesis. PMID: 27208428
  31. Amplification of the EGFR gene can be maintained and modulated by variation of EGF concentrations in in vitro models of glioblastoma multiforme. PMID: 28934307
  32. Our study showed that the EGF61 rs4444903GA genotype had a decreased risk of non-syndromic cleft lip with or without cleft palate. Our data provides further evidence regarding the role of EGF61 variations in the development of non-syndromic cleft lip with or without cleft palate in families of the studied populations. PMID: 28906376
  33. Interestingly, EGF rapidly downregulates LINC01089 (here renamed LncRNA Inhibiting Metastasis; LIMT) expression by enhancing histone deacetylation at the respective promoter. PMID: 27485121
  34. EGF-induced, calpain-mediated proteolysis contributes to the rapid destruction of cyclin G2 and that the PEST domain is critical for EGF/calpain actions. PMID: 28640887
  35. The salivary levels of EGF are significantly increased during the acute phase of natural rotavirus infection. PMID: 28558652
  36. Findings have identified a role for members of these signaling pathways in the regulation of EGF-induced vimentin expression in the MDA-MB-468 breast cancer cell line. PMID: 27163529
  37. miR-223 downregulated the local expression of epidermal growth factor (EGF), leading to decreased activation of EGF receptor (EGFR) on target cells and, eventually, dampening a positive EGF-EGFR autocrine/paracrine stimulation loop induced by the post-surgical wound-healing response. PMID: 26876200
  38. EGFR and EGF expression showed no significant difference between placentas from normal pregnancies and those complicated with preeclampsia. PMID: 27657362
  39. Atomistic molecular dynamics simulations show that N-glycosylation of the EGFR extracellular domain plays critical roles in the binding of growth factors, monoclonal antibodies, and the dimeric partners to the monomeric EGFR extracellular domain. PMID: 28486782
  40. CMTM3 decreases EGFR expression, facilitates EGFR degradation, and inhibits the EGF-mediated tumorigenicity of gastric cancer cells by enhancing Rab5 activity. PMID: 27867015
  41. Findings suggest that EGF not only promotes the proliferation of adipose stem cells and delays their senescence, but also maintains the differentiation potency of adipose stem cells, which are related to the EGF-induced activation of the STAT signal pathway. PMID: 28746211
  42. The results show that the interaction between STS-1 and ShcA is regulated in response to EGF receptor activation. PMID: 28690151
  43. Insulin treatment caused sustained Akt activity, whereas EGF or PDGF-AA promoted transient signaling; PDGF-BB produced sustained responses at higher concentrations. Transient responses to EGF were caused by negative feedback at the receptor level, as a second treatment yielded minimal responses, whereas parallel exposure to IGF-I caused full Akt activation. PMID: 27044757
  44. Our results indicate that different concentrations of bFGF and EGF supplemented during propagation of neural rosettes are involved in altering the identity of the resultant neural cells. PMID: 27321088
  45. F25P preproinsulin effectively reduced the concentrations of EGF, VEGF, and MMP-9 in the blood of tumor-bearing mice with EGFR-mutant glioblastoma. PMID: 27317648
  46. Conformational stability of the EGFR as influenced by glycosylation, dimerization, and EGF hormone binding has been described. PMID: 28019699
  47. Differential expression patterns of EGF, EGFR, and ERBB4 are essential in epithelial restitution and remodeling in nasal epithelium. PMID: 27285994
  48. Phosphorylation and immunohistochemical assays on the EGF receptor in HeLa cells indicate that the EGF protein produced in soybean seed is bioactive and comparable to commercially available human EGF. This work demonstrates the feasibility of using soybean seeds as a biofactory to produce therapeutic agents in a soymilk delivery platform. PMID: 27314851
  49. Data suggest that activated platelets release ADAMDEC1, which hydrolyzes pro-EGF (epidermal growth factor) to soluble, active HMW-EGF; proteolytic cleavage of pro-EGF first occurs at the C-terminal arginyl residue of the EGF domain; proteolysis is the regulated, rate-limiting step in generating soluble EGF from activated platelets. PMID: 28455445
  50. Subgroup analysis in a Slovak population by gender showed that the genotype EGF G61G and allele G was associated with a non-significantly increased risk of MDD. PMID: 27755861

Show More

Hide All

Database Links

HGNC: 3229

OMIM: 131530

KEGG: hsa:1950

STRING: 9606.ENSP00000265171

UniGene: Hs.419815

Involvement In Disease
Hypomagnesemia 4 (HOMG4)
Subcellular Location
Membrane; Single-pass type I membrane protein.
Tissue Specificity
Expressed in kidney, salivary gland, cerebrum and prostate.

Q&A

What is an EGF Recombinant Monoclonal Antibody and how is it produced?

Epidermal Growth Factor (EGF) Recombinant Monoclonal Antibodies are laboratory-engineered antibodies produced using in vitro expression systems. These systems function by cloning specific antibody DNA sequences from immunoreactive organisms (commonly rabbits) and then screening individual clones to select optimal candidates for production. Unlike traditional hybridoma-derived antibodies, recombinant antibodies offer superior consistency between production lots and can be designed with precise binding characteristics. The recombinant technology enables production of antibodies with identical amino acid sequences to established therapeutics like cetuximab, while maintaining consistent structure and function.

What are the key advantages of using recombinant monoclonal antibodies over traditional antibodies?

Recombinant monoclonal antibodies provide several significant advantages over traditional hybridoma-derived antibodies in research applications:

  • Enhanced specificity and sensitivity in target recognition

  • Superior lot-to-lot consistency due to defined genetic sequences

  • Animal origin-free formulations that reduce biosafety concerns

  • Broader immunoreactivity to diverse targets due to larger immune repertoire

  • Improved reproducibility in experimental results

  • Ability to engineer specific binding domains for specialized applications

These advantages make recombinant antibodies particularly valuable for longitudinal studies where consistent antibody performance is critical for reliable data comparison across experiments.

What is the biological significance of EGF and its receptor in cellular processes?

EGF (Epidermal Growth Factor) functions primarily by binding to the EGF Receptor (EGFR), a 170 kDa protein kinase. This interaction triggers receptor dimerization and initiates diverse cellular pathways critical for both normal and pathological processes. EGFR activation initiates at least four major downstream signaling cascades:

  • RAS-RAF-MEK-ERK pathway

  • PI3 kinase-AKT pathway

  • PLCgamma-PKC module

  • STAT signaling module

EGF is widely expressed in tissues including kidney, cerebrum, prostate, and salivary glands, where it acts as a potent mitogenic factor. The phosphorylated receptor recruits adapter proteins like GRB2 that activate complex downstream signaling cascades. Research indicates EGF may also play an important role in activating the NF-kappa-B signaling cascade. Dysregulation of EGF signaling has been associated with the growth and progression of certain cancers.

How should researchers design validation protocols for EGF recombinant antibodies in their specific experimental systems?

When validating EGF recombinant antibodies for experimental use, researchers should implement a multi-technique validation approach:

  • Initial specificity assessment: Perform ELISA and Western blot analyses using both positive controls (recombinant human EGF) and potential cross-reactive proteins (HB-EGF, TGF-alpha) to confirm antibody specificity.

  • Cross-species reactivity testing: Evaluate antibody performance across relevant species (human, mouse, rat) if cross-species experiments are planned. For example, anti-human EGF antibodies may recognize rat EGF but not mouse EGF in Western blots.

  • Application-specific validation: For each intended application (Western blot, IHC, IF, FACS), perform positive and negative control experiments using:

    • Cell lines with confirmed EGF expression

    • Knockout/knockdown models

    • Competing peptide blocking to confirm binding specificity

  • Functional validation: For neutralizing antibodies, establish dose-response curves using cell proliferation assays (e.g., with Balb/3T3 mouse embryonic fibroblast cells) to determine the neutralization dose (ND50).

  • Reproducibility assessment: Perform technical and biological replicates to ensure consistent antibody performance across different experimental conditions.

Documentation of all validation steps creates a comprehensive antibody validation profile that ensures experimental reliability and facilitates troubleshooting.

What are the optimal methodologies for determining antibody affinity and specificity for EGF versus related growth factors?

To determine antibody affinity and specificity for EGF versus related growth factors, researchers should employ a systematic approach combining multiple complementary techniques:

Affinity Determination Methods:

  • ELISA-based affinity measurement: Establish saturation binding curves to determine Kd values. For EGF antibodies, affinity constants in the range of 10^7 M^-1 are considered good for research applications.

  • Surface Plasmon Resonance (SPR): Provides real-time kinetic analysis of antibody-antigen interactions, yielding association (kon) and dissociation (koff) rate constants in addition to equilibrium dissociation constants (KD).

  • Bio-Layer Interferometry (BLI): Offers similar kinetic information to SPR but with different technical advantages for certain applications.

Specificity Assessment Methods:

  • Competitive binding assays: Compare binding of the antibody to EGF in the presence of increasing concentrations of related growth factors (TGF-α, HB-EGF).

  • Western blot analysis: Test antibody recognition against a panel of recombinant proteins including human EGF, mouse EGF, rat EGF, human HB-EGF, and human TGF-alpha. Document molecular weights and band intensities.

  • Immunoprecipitation followed by mass spectrometry: To identify potential cross-reactive proteins in complex biological samples.

TechniquePrimary InformationTypical Values for High-Quality Anti-EGF Antibodies
ELISAEquilibrium binding constantKd = 10^-8 to 10^-10 M
SPRAssociation/dissociation kineticskon = 10^4 to 10^6 M^-1s^-1, koff = 10^-4 to 10^-2 s^-1
Western BlotCross-reactivity profileSpecific detection of 6-10 kDa EGF band
Cell-based assayNeutralization dose (ND50)0.05-0.1 μg/mL for cell proliferation inhibition

What are the recommended storage and handling procedures to maintain antibody stability and activity?

Proper storage and handling of EGF recombinant monoclonal antibodies is crucial for maintaining their stability and activity throughout the research process:

Storage Recommendations:

  • Temperature: Store lyophilized antibody formulations at -20°C to -80°C for long-term stability. For reconstituted antibodies, follow manufacturer guidelines, typically recommending -20°C for aliquots.

  • Avoid freeze-thaw cycles: Minimize repeated freeze-thaw cycles by preparing appropriately sized single-use aliquots upon initial reconstitution.

  • Buffer conditions: For reconstitution, sterile PBS is typically recommended for research-grade antibodies. Reconstitute at appropriate concentrations (e.g., 500 μg/mL) as specified in product documentation.

Handling Procedures:

  • Thawing: Thaw frozen antibody aliquots at room temperature or at 4°C rather than using heat to prevent protein denaturation.

  • Working dilutions: Prepare working dilutions freshly on the day of use whenever possible, especially for sensitive applications like cell-based assays.

  • Sterility: Maintain aseptic technique when handling antibody solutions, particularly for cell culture applications.

  • Centrifugation: Briefly centrifuge antibody vials before opening to collect liquid at the bottom of the tube and reduce loss.

  • Transport: When shipping or transporting antibodies between locations, use cold packs or dry ice as appropriate for the antibody formulation.

Proper documentation of storage conditions, reconstitution dates, and freeze-thaw cycles for each antibody lot can help troubleshoot unexpected experimental results and maintain consistent antibody performance over time.

How can researchers effectively use EGF recombinant antibodies in cancer research and therapeutic development?

EGF recombinant antibodies serve as powerful tools in cancer research and therapeutic development through multiple strategic applications:

Diagnostic Applications:

Therapeutic Research Applications:

  • Mechanism of action studies: Neutralizing anti-EGF antibodies can block ligand-receptor interactions in experimental models to elucidate the role of EGF signaling in tumor progression.

  • Combination therapy investigation: Researchers can use these antibodies to study potential synergies between EGF pathway inhibition and other therapeutic approaches (chemotherapy, radiotherapy, immunotherapy).

  • Resistance mechanism studies: As tumors frequently develop resistance to EGFR-targeted therapies, these antibodies help investigate alternative signaling pathways and resistance mechanisms.

Therapeutic Development:

  • Antibody optimization: The recombinant platform allows for engineering of enhanced antibody properties including:

    • Increased affinity for target

    • Modified Fc functions

    • Altered pharmacokinetic profiles

    • Reduced immunogenicity

  • Target validation: Neutralizing antibodies provide proof-of-concept data for EGF pathway inhibition before committing to costly drug development programs.

Anti-EGFR monoclonal antibody therapies (e.g., cetuximab) have demonstrated clinical efficacy in EGFR-positive epithelial tumors including common malignancies of the head and neck, lung, and colon, establishing EGF/EGFR as a validated therapeutic target pathway.

What strategies can be employed to develop highly specific antibodies against EGF receptor variants such as EGFRvIII?

Developing highly specific antibodies against EGFR variants like EGFRvIII requires sophisticated strategies that leverage recombinant antibody technology:

Immunization and Selection Strategies:

  • Peptide-based immunization: Design immunogens that specifically highlight the unique junction epitope created by the EGFRvIII deletion mutation (deletion of exons 2-7). This junction creates a novel glycine residue not present in wild-type EGFR.

  • Negative selection: Implement screening protocols that eliminate antibody clones showing cross-reactivity with wild-type EGFR while preserving binding to EGFRvIII.

Antibody Engineering Approaches:

  • CDR modification: Strategic mutation of complementarity-determining regions (CDRs) can enhance specificity. For example, targeted mutation of tyrosine residues in the CDRH2 and CDRH3 domains to phenylalanine, as demonstrated in the development of RAb DMvIII antibody, can dramatically improve specificity for EGFRvIII over wild-type EGFR.

  • Bispecific antibody design: Creating antibodies that simultaneously recognize the unique EGFRvIII junction epitope and another EGFRvIII-specific epitope can provide enhanced specificity through avidity effects.

  • Affinity maturation: Directed evolution techniques can be employed to enhance binding affinity while maintaining specificity, targeting affinity constants in the range of 10^7 M^-1 or higher.

Validation Methodology:

  • Comprehensive cross-reactivity testing: Validate specificity using multiple techniques (Western blot, immunohistochemistry, immunofluorescence, FACS) against cell lines expressing:

    • EGFRvIII only

    • Wild-type EGFR only

    • Both EGFRvIII and wild-type EGFR

    • Neither receptor (negative control)

  • Primary tissue validation: Confirm specificity using GBM primary tissue samples with known EGFRvIII expression status.

These strategies have successfully produced antibodies like RAb DMvIII that specifically detect EGFRvIII with minimal cross-reactivity to wild-type EGFR, making them valuable tools for both research and potential clinical applications in cancers harboring this variant.

How do the pharmacokinetic profiles of EGF recombinant monoclonal antibodies influence their research and clinical applications?

The pharmacokinetic (PK) profiles of EGF recombinant monoclonal antibodies significantly impact their utility in both research and clinical applications through several critical parameters:

Key Pharmacokinetic Parameters and Their Research Implications:

  • Half-life (T1/2):

    • Recombinant anti-EGFR antibodies typically demonstrate extended half-lives ranging from 77-86 hours after single infusion to 102-119 hours after multiple infusions.

    • Research implication: Longer half-lives enable less frequent dosing in animal models and potentially reduced experimental variability.

  • Distribution volume (Vd):

    • Monoclonal antibodies generally have limited tissue distribution due to their large molecular size.

    • Research implication: Affects experimental design when targeting tissues with restricted antibody penetration (e.g., brain tumors).

  • Clearance mechanisms:

    • Target-mediated drug disposition (TMDD) often occurs when target expression levels are high.

    • Research implication: Requires consideration of target expression levels in experimental models when designing dosing regimens.

  • Dose-exposure relationship:

    • Anti-EGFR antibodies commonly exhibit non-linear PK profiles across dose ranges (e.g., 100 to 400 mg/m²).

    • Research implication: Necessitates careful dose optimization in experimental models to achieve desired target occupancy.

Clinical Translation Considerations:

  • Immunogenicity assessment:

    • The development of anti-drug antibodies (ADAs) can significantly alter PK profiles and efficacy.

    • Recombinant antibodies specifically engineered to reduce immunogenicity show advantages in this regard, with studies showing no detectable neutralizing anti-antibody antibodies in clinical trials.

  • PK/PD modeling:

    • Integration of pharmacokinetic data with pharmacodynamic biomarkers (e.g., receptor occupancy, downstream signaling inhibition) provides crucial information for translational research.

    • Mean Cmax often increases in roughly dose-proportional manner while AUC₀-∞ can show greater than dose-proportionate increases across dose ranges.

The comprehensive understanding of these PK parameters enables researchers to design more effective experimental protocols and facilitates more accurate translation of research findings to clinical applications, particularly in oncology where precise dosing is critical for balancing efficacy and toxicity.

What are the common technical challenges when using EGF antibodies in various experimental techniques and how can they be addressed?

Researchers frequently encounter technical challenges when working with EGF antibodies across different experimental platforms. Here are the most common issues and their solutions:

Western Blot Challenges:

  • Problem: Multiple bands or unexpected molecular weight detection
    Solution: Verify sample preparation conditions (reducing vs. non-reducing), as EGF has multiple forms including precursor (130 kDa) and mature forms (6.7 kDa). Implement positive controls using recombinant EGF protein alongside experimental samples.

  • Problem: Weak signal despite confirmed EGF expression
    Solution: Optimize protein extraction methods specifically for growth factors, which may require specialized lysis buffers. Consider concentration steps for secreted EGF in culture media samples.

Immunohistochemistry Challenges:

  • Problem: Inconsistent staining between tissue sections
    Solution: Standardize antigen retrieval methods (pH, temperature, duration) and optimize antibody concentration through titration experiments. For EGF detection in skin, optimal concentrations around 25 μg/mL have been reported for certain antibodies.

  • Problem: High background staining
    Solution: Implement additional blocking steps using bovine serum albumin or normal serum matching the secondary antibody host species. Add washing steps with detergent (0.1% Tween-20) to reduce non-specific binding.

Neutralization Assay Challenges:

  • Problem: Variable neutralization potency between experiments
    Solution: Standardize cell culture conditions including passage number, seeding density, and assay duration. Establish dose-response curves for each antibody lot using reference cell lines (e.g., Balb/3T3 fibroblasts) and calculate ND₅₀ values (typically 0.05-0.1 μg/mL for quality anti-EGF antibodies).

Flow Cytometry Challenges:

  • Problem: Poor discrimination between positive and negative populations
    Solution: Optimize fixation and permeabilization protocols specifically for EGF/EGFR detection. Include appropriate isotype controls and implement fluorescence-minus-one (FMO) controls for accurate gating.

General Quality Control Measures:

  • Implement routine validation of each new antibody lot using standard positive controls

  • Maintain detailed records of antibody performance across applications

  • Store validation data (images, graphs) alongside antibody information for future reference

How can researchers distinguish between experimental artifacts and true biological effects when using EGF recombinant antibodies?

Distinguishing between experimental artifacts and true biological effects requires systematic experimental design and rigorous controls when using EGF recombinant antibodies:

Essential Control Experiments:

  • Antibody Specificity Controls:

    • Peptide competition assays: Pre-incubation of the antibody with excess recombinant EGF should eliminate specific signals.

    • Genetic controls: Utilize EGF knockout/knockdown systems alongside wild-type controls to verify signal specificity.

    • Cross-reactivity testing: Test against related proteins (HB-EGF, TGF-alpha) to confirm signals are not due to off-target binding.

  • Technical Controls:

    • Concentration gradients: Test multiple antibody concentrations to identify optimal signal-to-noise ratios.

    • Multiple detection methods: Confirm findings using orthogonal techniques (e.g., verify Western blot results with ELISA).

    • Secondary antibody-only controls: Exclude signals arising from non-specific secondary antibody binding.

  • Biological Variability Assessment:

    • Biological replicates: Test samples from multiple independent sources to distinguish reproducible effects from anomalies.

    • Temporal analyses: Examine EGF/EGFR expression and activation at multiple time points to identify consistent patterns versus transient artifacts.

Data Interpretation Framework:

  • Quantitative analysis: Implement appropriate statistical methods to distinguish significant differences from experimental noise.

  • Correlation with functional outcomes: Link antibody-detected signals to functional readouts (e.g., cell proliferation, migration, or signaling pathway activation).

  • Dose-response relationships: True biological effects typically show consistent dose-dependent responses, while artifacts often display random or inconsistent patterns.

  • Mechanistic validation: Confirm antibody-detected effects through mechanistic studies (e.g., if an anti-EGF antibody detects increased EGF expression, verify this leads to expected downstream signaling events).

What are the latest advances in recombinant antibody technology that enhance reproducibility and performance in EGF/EGFR research?

Recent technological advances have significantly improved the reproducibility and performance of recombinant antibodies in EGF/EGFR research:

Structural and Functional Enhancements:

  • Antibody humanization and de-immunization: Advanced computational algorithms now guide the redesign of antibody frameworks to reduce immunogenicity while preserving binding characteristics, extending the utility of these reagents in in vivo models and potentially clinical applications.

  • Affinity maturation technologies: Directed evolution techniques including phage, yeast, and mammalian display platforms enable the systematic enhancement of binding affinity and specificity through iterative selection processes, leading to antibodies with exquisite specificity for targets like EGFRvIII over wild-type EGFR.

  • Site-specific conjugation: Precision engineering of conjugation sites for fluorophores, enzymes, or other detection moieties minimizes batch-to-batch variation and ensures consistent performance in imaging and detection applications.

Production and Quality Control Innovations:

  • Serum-free, chemically defined expression systems: Advanced production platforms eliminate animal-derived components, reducing batch variation and potential contaminants that can affect experimental outcomes.

  • Automated clone selection technologies: High-throughput screening platforms enable more comprehensive evaluation of antibody clones, identifying those with optimal specificity, affinity, and stability characteristics.

  • Standardized validation metrics: Implementation of industry-wide validation standards enhances comparability between different antibody reagents and improves experimental reproducibility across laboratories.

Application-Specific Optimizations:

  • Single-domain antibodies (nanobodies): These smaller antibody fragments offer improved tissue penetration and stability for certain applications, particularly in imaging dense tissues.

  • Bispecific antibody platforms: Advanced formats enable simultaneous targeting of EGF/EGFR and secondary targets (immune effector cells, other tumor markers) for enhanced specificity and functionality.

  • Intrabodies: Engineered variants optimized for intracellular expression and targeting specific protein conformations or post-translational modifications of EGFR, expanding the research toolkit beyond traditional applications.

The integration of these technologies has substantially enhanced the reliability and utility of recombinant antibodies in EGF/EGFR research, addressing historical challenges related to specificity, reproducibility, and functional versatility.

How are EGF recombinant antibodies being utilized in developing organoid models and personalized medicine approaches?

EGF recombinant antibodies play increasingly important roles in organoid technology and personalized medicine, functioning both as essential research tools and potential therapeutic agents:

Applications in Organoid Development:

  • Growth factor modulation: EGF is a critical component in organoid culture media, where recombinant antibodies can be used to precisely control EGF signaling through neutralization at specific developmental stages. This enables researchers to model pathway-dependent developmental processes in tissues like intestine, where EGF works synergistically with other factors including Noggin, R-Spondin 1, and Wnt-3a.

  • Pathway dissection: Selective inhibition of EGF/EGFR signaling using specific monoclonal antibodies allows researchers to distinguish EGF-dependent versus EGF-independent cellular processes in complex 3D tissue models.

  • Differentiation induction: Manipulating EGF signaling through timed application of neutralizing antibodies can drive organoid differentiation toward specific lineages, particularly in epithelial tissues where EGF regulates stem cell maintenance versus differentiation decisions.

  • Disease modeling: Anti-EGFR antibodies enable the modeling of therapeutic responses in patient-derived organoids, particularly for EGFR-dependent cancers of the colon, lung, and head and neck.

Personalized Medicine Applications:

  • Predictive biomarker development: Patient-derived organoids treated with anti-EGFR antibodies can predict individual patient responses to EGFR-targeted therapies, allowing for personalized treatment selection.

  • Resistance mechanism investigation: By exposing patient-derived organoids to increasing concentrations of therapeutic anti-EGFR antibodies, researchers can identify and characterize resistance mechanisms that may emerge during treatment.

  • Combination therapy optimization: Testing anti-EGFR antibodies alongside other targeted agents or conventional chemotherapies in patient-derived organoids enables personalized optimization of combination regimens.

  • Patient-specific dosing strategies: Pharmacokinetic and pharmacodynamic studies in organoid models can inform individualized dosing strategies based on patient-specific antibody metabolism and target expression levels.

These applications represent significant advances toward precision medicine approaches that account for individual patient differences in disease pathophysiology and treatment response.

What are the latest developments in using EGF recombinant antibodies for targeted drug delivery systems?

Recent innovations have expanded the utility of EGF recombinant antibodies in targeted drug delivery systems, leveraging their specificity to improve therapeutic efficacy while reducing off-target effects:

Advanced Antibody-Drug Conjugate (ADC) Platforms:

  • Site-specific conjugation technologies: Newer methods enable precise attachment of cytotoxic payloads at defined antibody positions, resulting in homogeneous ADCs with improved stability and pharmacokinetic profiles compared to earlier-generation random conjugation approaches.

  • Novel linker chemistry: Stimuli-responsive linkers designed to release payloads selectively in tumor microenvironments (responding to pH, proteases, or redox conditions) enhance the therapeutic index of anti-EGFR antibody conjugates.

  • Payload diversification: Beyond traditional cytotoxic agents, anti-EGF/EGFR antibodies are being conjugated to:

    • Immunomodulatory molecules

    • Radionuclides for theranostic applications

    • Photosensitizers for photodynamic therapy

    • siRNA/mRNA for gene modulation

Nanoparticle-Antibody Hybrid Systems:

  • Antibody-decorated nanoparticles: Recombinant anti-EGFR antibodies and fragments serve as targeting moieties on various nanocarriers (liposomes, polymeric nanoparticles, dendrimers) to enhance tumor-specific delivery of encapsulated therapeutics.

  • Multi-targeting approaches: Systems incorporating both anti-EGFR antibodies and secondary targeting ligands achieve more precise delivery to tumor subpopulations or specific intracellular compartments.

  • Triggered release mechanisms: Smart delivery systems using anti-EGFR antibodies for targeting incorporate release triggers (light, ultrasound, magnetic fields) to control payload release with spatial and temporal precision.

Biological Barriers Navigation:

  • Blood-brain barrier crossing: Modified anti-EGFR antibody fragments with enhanced BBB penetration capabilities facilitate delivery to brain tumors expressing EGFR/EGFRvIII.

  • Tumor microenvironment modulation: Bifunctional antibodies that simultaneously target EGFR and components of the tumor stroma improve penetration of therapeutic payloads into solid tumors.

These technological advances significantly enhance the potential of EGF/EGFR-targeted delivery systems, addressing historical limitations related to payload delivery, specificity, and biological barrier penetration, with several candidates advancing through preclinical and early clinical development.

How can researchers effectively integrate computational approaches with experimental studies to optimize EGF recombinant antibody design and application?

The integration of computational approaches with experimental studies creates powerful synergies that accelerate and enhance EGF recombinant antibody development:

Antibody Structure Optimization:

  • In silico affinity maturation: Computational methods including molecular dynamics simulations and machine learning algorithms predict beneficial mutations in complementarity-determining regions (CDRs) to enhance binding affinity and specificity for EGF or specific EGFR variants like EGFRvIII.

  • Stability engineering: Algorithms identify destabilizing residues and suggest modifications to improve thermal and colloidal stability without compromising binding characteristics, extending shelf-life and experimental reliability.

  • Humanization optimization: Computational tools guide the humanization process by identifying minimum essential murine residues to maintain binding properties while maximizing human content to reduce immunogenicity for in vivo applications.

Target Interaction Analysis:

  • Epitope mapping and optimization: Molecular docking and simulation techniques predict antibody-antigen interactions at atomic resolution, guiding experimental design of antibodies targeting specific EGF/EGFR epitopes.

  • Cross-reactivity prediction: Computational screening against proteome databases identifies potential off-target interactions, prioritizing antibody candidates with minimal cross-reactivity profiles for experimental validation.

  • Dynamic binding analysis: Advanced simulation techniques model conformational changes during antibody-antigen binding, revealing insights into binding kinetics that inform experimental optimization.

Integrated Workflow Implementation:

  • Iterative design-build-test cycles: Effective integration follows a workflow where:

    • Computational design generates multiple antibody candidates

    • High-throughput experimental screening validates predictions

    • Experimental data feeds back to refine computational models

    • Improved models guide the next design iteration

  • Experimental validation prioritization: Computational approaches help prioritize the most promising antibody candidates for resource-intensive experimental validation, optimizing research efficiency.

  • Data integration platforms: Custom bioinformatics pipelines integrate structural, functional, and experimental data to continuously refine antibody design parameters.

Computational ApproachExperimental ValidationIntegrated Outcome
Binding affinity predictionSurface plasmon resonanceOptimized antibody variants with enhanced EGF/EGFR binding
Stability simulationDifferential scanning calorimetryAntibodies with improved thermal tolerance for diverse applications
Epitope predictionHydrogen-deuterium exchange mass spectrometryAntibodies targeting specific functional epitopes on EGF/EGFR
Cross-reactivity screeningProtein microarray testingAntibodies with minimal off-target binding

This integrated approach significantly accelerates development timelines while producing antibodies with superior performance characteristics for both research and potential therapeutic applications.

What criteria should researchers use to select the optimal EGF recombinant antibody for their specific experimental needs?

Selecting the optimal EGF recombinant antibody requires systematic evaluation of multiple parameters aligned with specific experimental requirements:

Application-Specific Selection Criteria:

  • Intended application compatibility: Different applications require antibodies with distinct properties:

    • Western blot applications benefit from antibodies recognizing denatured epitopes

    • Immunohistochemistry requires antibodies effective with fixed tissues

    • Neutralization assays demand antibodies targeting functional epitopes

    • Flow cytometry applications often require higher affinity antibodies

  • Validated performance metrics: Evaluate published validation data including:

    • Affinity constants (optimal ranges vary by application)

    • Specificity profiles against related proteins (HB-EGF, TGF-alpha)

    • Neutralization potency (ND₅₀) for functional studies

    • Cross-species reactivity if using non-human models

  • Technical specifications compatibility:

    • Clonality (monoclonal for consistent epitope targeting)

    • Host species (avoiding same species as experimental samples)

    • Isotype (relevant for certain detection systems)

    • Conjugation status (directly labeled vs. requiring secondary detection)

Experimental System Considerations:

  • Model system validation: Prioritize antibodies with demonstrated effectiveness in your specific:

    • Species of interest (human, mouse, rat)

    • Tissue or cell type

    • Disease model context

  • Signal-to-noise requirements: Projects requiring detection of low-abundance targets benefit from antibodies with higher affinity and specificity, even at premium cost.

  • Reproducibility needs: Long-term projects should prioritize recombinant antibodies with defined sequences that ensure consistent performance across production lots.

By systematically evaluating these criteria against experimental requirements, researchers can select antibodies that optimize results while minimizing troubleshooting time and resource expenditure.

What future directions are emerging in EGF/EGFR-targeted antibody research and therapeutic applications?

The field of EGF/EGFR-targeted antibody research is rapidly evolving, with several promising directions emerging at the intersection of biological understanding, technological innovation, and clinical need:

Next-Generation Therapeutic Approaches:

  • Bispecific and multispecific antibodies: Emerging formats simultaneously target EGFR and complementary pathways (HER2, HER3, c-MET) or immune cells (T cells, NK cells), addressing resistance mechanisms and enhancing therapeutic efficacy beyond traditional monospecific antibodies.

  • Conditional activation antibodies: Smart antibody designs that become activated only in the tumor microenvironment reduce off-target effects in normal EGFR-expressing tissues.

  • Intracellular antibody delivery: Novel delivery technologies enabling antibodies to reach intracellular EGFR signaling components expand the therapeutic landscape beyond surface receptor targeting.

Precision Medicine Integration:

  • Companion diagnostics development: Advanced antibody-based diagnostic tools more precisely identify patients likely to benefit from EGFR-targeted therapies by detecting specific mutations, expression levels, or activation states.

  • Combination therapy optimization: Systematic evaluation of EGFR antibodies with emerging immunotherapies, targeted agents, and conventional treatments using patient-derived models enables more personalized treatment strategies.

  • Resistance mechanism targeting: Antibodies specifically designed to address known resistance mechanisms to first-generation EGFR-targeted therapies show promise in extending treatment durability.

Technological Frontiers:

  • AI-designed antibodies: Machine learning approaches integrating structural, functional, and clinical data accelerate the development of antibodies with optimized properties for specific applications.

  • Antibody-enabled imaging: Novel EGF/EGFR antibody-based imaging agents provide real-time visualization of receptor expression, localization, and occupancy, enhancing both research and clinical applications.

  • PROTAC-antibody conjugates: Emerging proteolysis-targeting chimera technology combined with antibody targeting enables selective degradation of EGFR, potentially addressing resistance mechanisms resistant to traditional inhibition approaches.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.