EGFR Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Mechanisms of Action

EGFR mAbs exert antitumor effects through multiple complementary pathways:

MechanismDescriptionKey AntibodiesReferences
Ligand BlockadeCompetitively inhibit EGFR binding to EGF, TGF-α, or other ligands.Cetuximab, Panitumumab
Receptor Dimerization InhibitionSteric hindrance prevents EGFR-EGFR or HER family member (HER2/3/4) dimerization.Cetuximab, Necitumumab
Internalization and DegradationPromote receptor endocytosis, reducing surface EGFR levels and signaling.All EGFR mAbs
Immune ActivationADCC: Engage NK cells/macrophages via Fcγ receptors. CDC: Complement-mediated lysis.Cetuximab (IgG1)
Signaling InhibitionSuppress MAPK/PI3K/AKT pathways, inducing G1 arrest, apoptosis, and anti-angiogenesis.Cetuximab, Panitumumab

Approved EGFR Monoclonal Antibodies

Four EGFR mAbs are currently approved, with others in clinical trials:

DrugTargetIndicationsApproval StatusReferences
CetuximabEGFR (Domain III)mCRC (RAS WT), HNSCC, NSCLCFDA/EMA Approved
PanitumumabEGFR (Domain III)mCRC (RAS WT)FDA/EMA Approved
NimotuzumabEGFR (Domain III)HNSCC, Glioblastoma, Esophageal cancerApproved (Regional)
NecitumumabEGFR (Domain III)Squamous NSCLCFDA Approved (2015)

Pipeline Therapies:

  • Duligotuzumab: Dual EGFR/HER3 inhibitor (e.g., MEHD7945A).

  • Futuximab: Targets EGFR Domain III (Sym004).

  • GC1118: Humanized IgG1 for colorectal/gastric cancers .

Clinical Efficacy and Outcomes

EGFR mAbs demonstrate variable efficacy across cancer types and histologies:

Colorectal Cancer (mCRC)

  • RAS Wild-Type: Cetuximab/Panitumumab + chemotherapy improves PFS (HR 0.74) and OS (HR 0.83) in first-line treatment .

  • Maintenance Therapy: Combining anti-EGFR mAbs with 5-FU/LV extends PFS (16.0 vs. 10.1 months in non-maintenance) .

Head and Neck Squamous Cell Carcinoma (HNSCC)

  • Combination with RT/CT: Cetuximab improves locoregional control and survival .

Non-Small Cell Lung Cancer (NSCLC)

  • Squamous Subtype: Necitumumab + chemotherapy improves OS (HR 0.83) .

  • EGFR-Positive NSCLC: High EGFR protein expression (H-score ≥200) or gene amplification predicts better response .

Biomarkers for Response Prediction

BiomarkerThresholdClinical RelevanceReferences
EGFR Protein ExpressionH-score ≥200Strong predictor of response in NSCLC
EGFR Gene Amplification≥40% cells with ≥4 copiesCorrelates with improved outcomes
KRAS Mutation StatusWild-TypeMandatory for mCRC eligibility

Resistance Mechanisms

Resistance to EGFR mAbs arises through:

  1. Secondary EGFR Mutations: Rare in mAb-treated patients .

  2. KRAS/NRAS/BRAF Mutations: Pre-existing or acquired .

  3. HER2/HER3 Compensatory Signaling: Upregulation of alternate ErbB family members .

  4. Tumor Microenvironment: Exosome-mediated drug resistance, non-coding RNA modulation .

Strategies to Overcome Resistance:

  • Combination Therapies: EGFR mAbs + TKIs (e.g., afatinib) or immunotherapy (e.g., avelumab + irinotecan) .

  • Bispecific Antibodies: Target EGFR and HER3 (e.g., duligotuzumab) .

Emerging Therapies and Future Directions

  1. Bispecific T-cell Engagers (BiTEs): Redirect T-cells to EGFR-expressing tumors.

  2. Engineered scFv Fragments: Enhanced tumor penetration in CAR-T or oncolytic viruses .

  3. Dual Inhibition: EGFR + VEGF blockade (e.g., ziv-aflibercept + cetuximab).

Product Specs

Buffer
Phosphate-buffered saline (PBS), pH 7.4, containing 0.02% sodium azide as a preservative and 50% glycerol.
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery times.
Synonyms
Avian erythroblastic leukemia viral (v erb b) oncogene homolog antibody; Cell growth inhibiting protein 40 antibody; Cell proliferation inducing protein 61 antibody; EGF R antibody; EGFR antibody; EGFR_HUMAN antibody; Epidermal growth factor receptor (avian erythroblastic leukemia viral (v erb b) oncogene homolog) antibody; Epidermal growth factor receptor (erythroblastic leukemia viral (v erb b) oncogene homolog avian) antibody; Epidermal growth factor receptor antibody; erb-b2 receptor tyrosine kinase 1 antibody; ERBB antibody; ERBB1 antibody; Errp antibody; HER1 antibody; mENA antibody; NISBD2 antibody; Oncogen ERBB antibody; PIG61 antibody; Proto-oncogene c-ErbB-1 antibody; Receptor tyrosine protein kinase ErbB 1 antibody; Receptor tyrosine-protein kinase ErbB-1 antibody; SA7 antibody; Species antigen 7 antibody; Urogastrone antibody; v-erb-b Avian erythroblastic leukemia viral oncogen homolog antibody; wa2 antibody; Wa5 antibody
Target Names
Uniprot No.

Target Background

Function
EGFR (Epidermal Growth Factor Receptor) is a receptor tyrosine kinase that binds to ligands from the EGF family, initiating various signaling cascades. These cascades convert external stimuli into appropriate cellular responses. Known ligands include EGF, TGFA/TGF-alpha, AREG, epigen/EPGN, BTC/betacellulin, epiregulin/EREG, and HBEGF/heparin-binding EGF. Ligand binding triggers receptor homo- and/or heterodimerization, followed by autophosphorylation on key cytoplasmic residues. The phosphorylated receptor then recruits adapter proteins like GRB2, ultimately activating complex downstream signaling cascades. EGFR activates at least four major downstream signaling pathways, including the RAS-RAF-MEK-ERK, PI3 kinase-AKT, PLCgamma-PKC, and STATs modules. It may also activate the NF-kappa-B signaling cascade. Furthermore, EGFR directly phosphorylates other proteins such as RGS16, boosting its GTPase activity and potentially linking EGFR signaling to G protein-coupled receptor signaling. EGFR also phosphorylates MUC1, enhancing its interaction with SRC and CTNNB1/beta-catenin. EGFR positively regulates cell migration by interacting with CCDC88A/GIV, which retains EGFR at the cell membrane after ligand stimulation. This promotes EGFR signaling, ultimately triggering cell migration. EGFR plays a role in enhancing learning and memory performance. Isoform 2 of EGFR might act as an antagonist of EGF action. EGFR acts as a receptor for hepatitis C virus (HCV) in hepatocytes, facilitating its cell entry. It mediates HCV entry by promoting the formation of the CD81-CLDN1 receptor complexes, essential for HCV entry, and by enhancing membrane fusion of cells expressing HCV envelope glycoproteins.
Gene References Into Functions
  1. Amphiregulin, contained in non-small-cell lung carcinoma-derived exosomes, induces osteoclast differentiation through the activation of the EGFR pathway. PMID: 28600504
  2. Combining vorinostat with an EGFRTKI can reverse EGFRTKI resistance in NSCLC. PMID: 30365122
  3. The feasibility of using the radiocobalt labeled antiEGFR affibody conjugate ZEGFR:2377 as an imaging agent is being investigated. PMID: 30320363
  4. Among all transfection complexes, 454 lipopolyplexes modified with the bidentate PEG-GE11 agent demonstrate the best, EGFR-dependent uptake, as well as luciferase and NIS gene expression into PMID: 28877405
  5. EGFR amplification was observed to be higher in the OSCC group compared to the control group (P=0.018) and was associated with advanced clinical stage (P=0.013), independent of age. Patients with EGFR overexpression exhibited worse survival rates, as did patients with T3-T4 tumors and positive margins. EGFR overexpression has a negative impact on disease progression. PMID: 29395668
  6. Clonal analysis revealed that the dominant JAK2 V617F-positive clone in Polycythemia Vera harbors EGFR C329R substitution, suggesting this mutation may contribute to clonal expansion. PMID: 28550306
  7. Baseline Circulating tumor cell count could be a predictive biomarker for EGFR-mutated and ALK-rearranged non-small cell lung cancer, offering improved guidance and monitoring of patients during molecular targeted therapies. PMID: 29582563
  8. High EGFR expression is associated with cystic fibrosis. PMID: 29351448
  9. These findings suggest a mechanism for EGFR inhibition to suppress respiratory syncytial virus by activating endogenous epithelial antiviral defenses. PMID: 29411775
  10. This study detected the emergence of the T790M mutation within the EGFR cDNA in a subset of erlotinib-resistant PC9 cell models through Sanger sequencing and droplet digital PCR-based methods, demonstrating that the T790M mutation can emerge via de novo events following treatment with erlotinib. PMID: 29909007
  11. The current study demonstrated that miR145 regulates the EGFR/PI3K/AKT signaling pathway in patients with nonsmall cell lung cancer. PMID: 30226581
  12. Among NSCLC patients treated with EGFR-TKI, those with T790M mutations were found to frequently also exhibit 19 dels, in contrast to T790M-negative patients. Additionally, T790M-positive patients exhibited a longer PFS. Therefore, screening these patients for T790M mutations may contribute to improving survival. PMID: 30150444
  13. High EGFR expression is associated with Breast Carcinoma. PMID: 30139236
  14. Results indicate that CAV-1 could promote anchorage-independent growth and anoikis resistance in detached SGC-7901 cells. This was associated with the activation of Src-dependent epidermal growth factor receptor-integrin beta signaling, as well as the phosphorylation of PI3K/Akt and MEK/ERK signaling pathways. PMID: 30088837
  15. Our findings suggest that FOXK2 inhibits the malignant phenotype of clear-cell renal cell carcinoma and functions as a tumor suppressor, possibly through the inhibition of EGFR. PMID: 29368368
  16. EGFR mutation status in advanced non-small cell lung cancer (NSCLC) patients altered significantly. PMID: 30454543
  17. Different Signaling Pathways in Regulating PD-L1 Expression in EGFR Mutated Lung Adenocarcinoma. PMID: 30454551
  18. Internal tandem duplication of the kinase domain delineates a genetic subgroup of congenital mesoblastic nephroma transcending histological subtypes. PMID: 29915264
  19. The expression level of EGFR increased alongside higher stages and pathologic grades of BTCC, and the significantly increased expression of HER-2 was statistically associated with clinical stages and tumor recurrence. Additionally, the expression level of HER-2 increased with higher clinical stages of BTCC. EGFR expression and HER-2 levels exhibited a positive association in BTCC samples. PMID: 30296252
  20. Results show that GGA2 interacts with the EGFR cytoplasmic domain to stabilize its expression and reduce its lysosomal degradation. PMID: 29358589
  21. Combination therapy of apatinib with icotinib for primary acquired resistance to icotinib might be an option for patients with advanced pulmonary adenocarcinoma with EGFR mutations. However, physicians must also be aware of the potential side effects associated with this therapy. PMID: 29575765
  22. We report a rare case presenting as multiple lung adenocarcinomas with four different EGFR gene mutations detected in three lung tumors. PMID: 29577613
  23. The study supports the involvement of EGFR, HER2, and HER3 in BCC aggressiveness and tumor differentiation towards distinct histological subtypes. PMID: 30173251
  24. The ratio of sFlt-1/sEGFR could serve as a novel candidate biochemical marker for monitoring the severity of preterm preeclampsia. sEndoglin and sEGFR might be involved in the pathogenesis of small for gestational age in preterm preelampsia. PMID: 30177039
  25. The study confirmed the prognostic effect of EGFR and VEGFR2 for recurrent disease and survival rates in patients with epithelial ovarian cancer. PMID: 30066848
  26. The data suggest that diagnostic or therapeutic chest radiation may predispose patients with decreased stromal PTEN expression to secondary breast cancer. Prophylactic EGFR inhibition might mitigate this risk. PMID: 30018330
  27. This study suggests a unique regulatory feature of PHLDA1 to inhibit the ErbB receptor oligomerization process, thereby controlling the activity of the receptor signaling network. PMID: 29233889
  28. The study observed the occurrence of not only EGFR C797S mutation but also L792F/Y/H in three NSCLC clinical subjects with acquired resistance to osimertinib treatment. PMID: 28093244
  29. Data indicate that the expression level of epidermal growth factor-like domain 7 (EGFL7) and epidermal growth factor receptor (EGFR) in invasive growth hormone-producing pituitary adenomas (GHPA) was considerably higher than that of non-invasive GHPA. PMID: 29951953
  30. Concurrent mutations in genes such as CDKN2B or RB1 were associated with a poorer clinical outcome in lung adenocarcinoma patients with EGFR active mutations. PMID: 29343775
  31. The ER-alpha36/EGFR signaling loop promotes the growth of hepatocellular carcinoma cells. PMID: 29481815
  32. High EGFR expression is associated with colorectal cancer. PMID: 30106444
  33. High EGFR expression is associated with gefitinib resistance in lung cancer. PMID: 30106446
  34. High EGFR expression is associated with tumor-node-metastasis in nonsmall cell lung cancer. PMID: 30106450
  35. Data suggest that Thr264 in TRPV3 is a key ERK1 phosphorylation site mediating EGFR-induced sensitization of TRPV3 to stimulate signaling pathways involved in regulating skin homeostasis. (TRPV3 = transient receptor potential cation channel subfamily V member-3; ERK1 = extracellular signal-regulated kinase-1; EGFR = epidermal growth factor receptor). PMID: 29084846
  36. The EGFR mutation frequency in Middle East and African patients is higher than that observed in white populations but still lower than the frequency reported in Asian populations. PMID: 30217176
  37. EGFR-containing exosomes derived from cancer cells could favor the development of a liver-like microenvironment, promoting liver-specific metastasis. PMID: 28393839
  38. The results reveal that the EGF-STAT3 signaling pathway promotes and sustains colorectal cancer (CRC) stemness. Additionally, a crosstalk between STAT3 and Wnt activates the Wnt/beta-catenin signaling pathway, which is also responsible for cancer stemness. Therefore, STAT3 represents a putative therapeutic target for CRC treatment. PMID: 30068339
  39. This result indicated that the T790M mutation is not only associated with EGFR-TKI resistance but may also play a functional role in the malignant progression of lung adenocarcinoma. PMID: 29887244
  40. LOX regulates EGFR cell surface retention to drive tumor progression. PMID: 28416796
  41. In a Han Chinese population, EGFR gene polymorphisms, rs730437 and rs1468727, and haplotype A-C-C were shown to be potential protective factors for the development of Alzheimer's Disease. PMID: 30026459
  42. EGFR proteins at different cellular locations in lung adenocarcinoma could influence the biology of cancer cells and serve as an independent indicator of a more favorable prognosis and treatment response. PMID: 29950164
  43. We report the crystal structure of EGFR T790M/C797S/V948R in complex with EAI045, a new type of EGFR TKI that binds to EGFR reversibly and does not rely on Cys 797. PMID: 29802850
  44. Overexpression of miR-452-3p promoted cell proliferation and mobility while suppressing apoptosis. MiR-452-3p enhanced EGFR and phosphorylated AKT (pAKT) expression but inhibited p21 expression levels. MiR-452-3p promoted hepatocellular carcinoma (HCC) cell proliferation and mobility by directly targeting the CPEB3/EGFR axis. PMID: 29332449
  45. This study demonstrates that the D2A sequence of the UPAR induces cell growth through alphaVbeta3 integrin and EGFR. PMID: 29184982
  46. BRAF and EGFR inhibitors can synergize to increase cytotoxic effects and decrease stem cell capacities in BRAF(V600E)-mutant colorectal cancer cells. PMID: 29534162
  47. This study confirms a direct correlation between MSI1 and EGFR, potentially supporting the significant role of MSI1 in EGFR activation via NOTCH/WNT pathways in esophageal squamous cell carcinoma. PMID: 30202417
  48. Three lines of tyrosine kinase inhibitors (TKIs) therapy can prolong survival in non-small cell lung cancer (NSCLC) patients. Elderly patients can benefit from TKI therapy. EGFR mutation-positive patients can benefit from second-line or third-line TKI therapy. PMID: 29266865
  49. EGFR 19Del and L858R mutations are valuable biomarkers for predicting the clinical response to EGFR-TKIs. 19Del mutations might yield a better clinical outcome. PMID: 29222872
  50. HMGA2-EGFR constitutively induced a higher level of phosphorylated STAT5B compared to EGFRvIII. PMID: 29193056

Show More

Hide All

Database Links

HGNC: 3236

OMIM: 131550

KEGG: hsa:1956

STRING: 9606.ENSP00000275493

UniGene: Hs.488293

Involvement In Disease
Lung cancer (LNCR); Inflammatory skin and bowel disease, neonatal, 2 (NISBD2)
Protein Families
Protein kinase superfamily, Tyr protein kinase family, EGF receptor subfamily
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Endoplasmic reticulum membrane; Single-pass type I membrane protein. Golgi apparatus membrane; Single-pass type I membrane protein. Nucleus membrane; Single-pass type I membrane protein. Endosome. Endosome membrane. Nucleus.; [Isoform 2]: Secreted.
Tissue Specificity
Ubiquitously expressed. Isoform 2 is also expressed in ovarian cancers.

Q&A

What is the primary mechanism of action for anti-EGFR monoclonal antibodies?

Anti-EGFR monoclonal antibodies exert their antitumor effects through multiple mechanisms. Primarily, they competitively bind to specific extracellular regions of EGFR, blocking ligand binding and inhibiting downstream signaling pathways that control cell proliferation and survival. Additionally, they sterically hinder EGFR heterodimerization with other HER family members, promote receptor internalization and degradation, and increase cell cycle inhibitor p27kip1 while inhibiting proliferating cell nuclear antigen (PCNA), leading to G1 cell cycle arrest . The variable region fragment (FV) of these antibodies is responsible for the specific recognition of EGFR, while the constant region (Fc) mediates immune-related mechanisms .

How do anti-EGFR antibodies activate immune responses against tumor cells?

Anti-EGFR monoclonal antibodies, particularly IgG1 antibodies like cetuximab, engage immune mechanisms through their Fc region. Two primary immune-mediated mechanisms are:

  • Complement-dependent cytotoxicity (CDC): The antibody Fc region activates the complement cascade, leading to formation of the membrane attack complex and tumor cell lysis .

  • Antibody-dependent cellular cytotoxicity (ADCC): The Fc segment binds to FcR receptors on natural killer (NK) cells or macrophages, triggering immune cell activation and tumor cell killing .

Different anti-EGFR antibodies demonstrate varying capacities to engage these immune mechanisms. For example, comparative studies have shown that cetuximab is more effective at mediating anti-tumor immune responses than panitumumab, despite similar EGFR signaling inhibition .

What are the structural characteristics of EGFR that enable targeting by monoclonal antibodies?

EGFR, as one of the 60 receptor protein tyrosine kinases (RTKs) in the human genome, has a characteristic structure that facilitates antibody targeting. It consists of:

  • Extracellular domain: Divided into four sub-structures where domains I and III have a β-helical fold and bind ligands, while domains II and IV are cysteine-rich regions responsible for exposing the dimerization interface .

  • Transmembrane domain: Contains an alpha helix transmembrane peptide that anchors the receptor to the cell membrane .

  • Intracellular domain: Comprises a 250-amino acid conserved protein tyrosine kinase core and 229 C-terminal residues with regulatory tyrosine residues .

Different anti-EGFR antibodies target distinct epitopes on the extracellular domain. For example, cetuximab and panitumumab compete with ligand binding to domain III, while depatuxizumab targets domain II to prevent receptor dimerization .

What biomarkers predict response to anti-EGFR monoclonal antibody therapy?

RAS mutation status is the primary predictive biomarker for anti-EGFR monoclonal antibody therapy. In colorectal cancer, KRAS mutations occur in 30-50% of cases and predict poor sensitivity to cetuximab or panitumumab . Only patients with RAS wild-type tumors typically respond to these therapies, though even among this population, the final response rate is only about 15% .

Additional biomarkers with predictive potential include:

  • EGFR expression levels and polymorphisms

  • Circulating tumor DNA (ctDNA) mutational status

  • BRAF mutation status

  • Other downstream pathway alterations in PI3K/AKT signaling

A methodological approach to biomarker assessment should include both tissue and liquid biopsy-based testing, with sequential monitoring to detect emerging resistance mechanisms .

What are the primary resistance mechanisms to anti-EGFR monoclonal antibodies and how can they be overcome?

Resistance mechanisms to anti-EGFR monoclonal antibodies can be classified into four categories:

  • Pre-target resistance:

    • KRAS mutations (occurring in 30-50% of colorectal cancers)

    • Tumor microenvironment factors, such as hyaluronic acid accumulation creating a barrier to antibody and NK cell penetration

  • On-target resistance:

    • High expression of EGFR or its ligands

    • Mutations or deletions in the extracellular domain binding sites

  • Post-target resistance:

    • Alterations in EGFR downstream signaling pathways

    • Bypass pathway activation

  • Off-target resistance:

    • Upregulation of non-EGFR receptors (FGFR1, PDGFRA, VEGFR1)

    • Increased heterodimerization with other receptors (HER2, HER3, C-MET)

    • Expression of factors like milk-derived growth inhibitor (MDGI)

    • Exosome-mediated resistance through the PTEN/Akt pathway

Strategies to overcome resistance include:

  • Combining anti-EGFR antibodies with inhibitors of downstream or parallel pathways

  • Developing bispecific antibodies targeting multiple receptors

  • Using antibody-drug conjugates

  • Exploring immune checkpoint inhibitor combinations

  • Engineering novel formats like single-chain variable fragments (scFv) for CAR-T or oncolytic virus applications

How should maintenance therapy with anti-EGFR monoclonal antibodies be designed to optimize clinical outcomes?

The optimal design of maintenance therapy with anti-EGFR monoclonal antibodies remains an area of active investigation. Based on recent studies, methodological approaches include:

  • Patient selection: Identify patients with RAS wild-type mCRC who have achieved complete or partial response to first-line chemotherapy combined with anti-EGFR therapy .

  • Maintenance regimen options:

    • Anti-EGFR monotherapy (e.g., panitumumab alone)

    • Anti-EGFR antibody combined with fluoropyrimidine (e.g., panitumumab + 5-FU/LV)

    • De-escalated chemotherapy regimens

  • Monitoring strategies:

    • Regular assessment of circulating tumor DNA (ctDNA) for early detection of resistance mutations

    • Patients with RAS/BRAF wild-type ctDNA show superior outcomes (mOS of 17.3 months) compared to those with mutated ctDNA (mOS of 10.4 months)

  • Duration considerations:

    • Continue until disease progression or unacceptable toxicity

    • Evaluate intermittent schedules to manage toxicity while maintaining efficacy

The Valentino study compared panitumumab monotherapy with panitumumab + 5-FU/LV for maintenance therapy in 229 patients with RAS wild-type mCRC after 8 cycles of panitumumab + FOLFOX4, providing important data on comparative efficacy of these approaches .

What are the latest developments in engineering anti-EGFR antibodies to enhance efficacy and overcome resistance?

Recent advances in antibody engineering have produced several innovative anti-EGFR therapeutic candidates:

  • Bispecific antibodies:

    • Duligotuzumab (MEHD7945A): Dual EGFR/HER3 inhibitory antibody designed to overcome resistance mediated by HER3 signaling

    • Other bispecific formats targeting EGFR and immune effector cells

  • Antibody mixtures:

    • Sym004: A 1:1 mixture of two recombinant antibodies (futuximab and modotuximab) that bind to non-overlapping epitopes in the EGFR extracellular domain, enhancing receptor internalization and degradation

  • Antibody-drug conjugates:

    • Depatuxizumab mafodotin (depatux-m): An anti-EGFR antibody conjugated to a cytotoxic payload, showing promise in recurrent glioblastoma with a 14.3% objective response rate, 25.2% PFS, and 69.1% OS

  • Single-chain variable fragments (scFv):

    • EGFR-targeted CAR-T cells: Incorporating anti-EGFR scFv in the extracellular domain of chimeric antigen receptors

    • EGFR-retargeted oncolytic viruses: Armed with anti-EGFR scFv, showing efficacy in orthotopic mouse models of primary human glioma

  • Pan-ErbB targeting:

    • PanErbB-CAR: Currently in clinical trials for head and neck squamous cell carcinoma (HNSCC)

These engineered formats aim to enhance tumor targeting, improve immune cell engagement, increase cytotoxic payload delivery, and overcome established resistance mechanisms .

How do the immune effects of different anti-EGFR monoclonal antibodies compare in experimental models?

Comparative studies of anti-EGFR monoclonal antibodies have revealed significant differences in their immune effects, despite targeting the same receptor. Methodologically, these studies involve:

  • Side-by-side comparisons in specific cancer models:

    • In head and neck cancer models, cetuximab and panitumumab were compared, revealing that panitumumab, despite similar EGFR signaling inhibition, was less effective in mediating anti-tumor immune mechanisms .

  • Evaluation of multiple immune parameters:

    • ADCC potency

    • Complement activation

    • NK cell recruitment and activation

    • Macrophage phagocytosis

    • T-cell response stimulation

  • Relationship to antibody structure:

    • IgG subclass determines Fc receptor binding (cetuximab is IgG1)

    • Glycosylation patterns affect immune cell interaction

    • Epitope binding location may influence receptor clustering and immune recognition

  • Comparison across different antibodies:

    • Cetuximab, panitumumab, and nimotuzumab were directly compared by Mazora et al., demonstrating different immune effect profiles despite targeting the same receptor

These comparative studies inform combination strategies, as researchers can better pair drugs based on their complementary immune mechanisms .

What experimental models best predict clinical response to anti-EGFR monoclonal antibodies?

Selecting appropriate experimental models to predict clinical responses to anti-EGFR monoclonal antibodies requires a methodical approach:

  • Cell line models:

    • Panels of cancer cell lines with defined genetic backgrounds (RAS/RAF status)

    • 3D organoid cultures that better recapitulate tumor architecture and heterogeneity

    • Cell line-derived xenografts (CDX) for in vivo assessment

  • Patient-derived models:

    • Patient-derived xenografts (PDX) that maintain tumor heterogeneity and microenvironment

    • Patient-derived organoids allowing high-throughput drug screening

    • Ex vivo tumor slice cultures for short-term drug response assays

  • Immune-competent models:

    • Humanized mouse models with reconstituted human immune systems to study ADCC and other immune mechanisms

    • Syngeneic mouse models with murine tumors and intact mouse immunity (requires murine-specific or cross-reactive antibodies)

  • Co-clinical trials:

    • Parallel testing in patient-matched models and the corresponding patients

    • Real-time model refinement based on clinical outcomes

  • Liquid biopsy integration:

    • Models incorporating circulating tumor DNA analysis to monitor evolving resistance

    • Ex vivo testing of circulating tumor cells

The predictive value of these models should be systematically evaluated by comparing preclinical findings with clinical outcomes, with particular attention to resistance mechanisms and biomarkers of response .

How should combination therapy with anti-EGFR antibodies and immune checkpoint inhibitors be designed and evaluated?

Designing combination therapies with anti-EGFR antibodies and immune checkpoint inhibitors requires systematic consideration of multiple factors:

  • Preclinical validation methodology:

    • Assess synergistic potential using immune-competent models

    • Evaluate changes in tumor immune microenvironment

    • Determine optimal dosing and sequencing through factorial design experiments

  • Patient selection strategy:

    • Primary focus on microsatellite stable (MSS) tumors that typically don't respond to checkpoint inhibitors alone

    • Biomarker-guided selection based on EGFR expression, PD-L1 status, and tumor mutational burden

    • RAS mutation status assessment, as RAS wild-type status predicts better response to anti-EGFR therapy

  • Clinical trial design considerations:

    • Phase IIa proof-of-concept studies with careful monitoring of immune-related adverse events

    • Incorporate pharmacodynamic biomarkers to confirm target engagement

    • Include translational endpoints to understand mechanisms of response/resistance

What are the optimal pharmacokinetic/pharmacodynamic models for dosing anti-EGFR monoclonal antibodies?

Determining optimal PK/PD models for anti-EGFR monoclonal antibodies requires understanding their complex pharmacological properties:

  • Pharmacokinetic considerations:

    • Anti-EGFR antibodies typically exhibit non-linear pharmacokinetics, as observed with CMAB009 across the 100-400 mg/m² dose range

    • Target-mediated drug disposition (TMDD) models should account for:

      • Saturable binding to EGFR

      • Receptor-mediated internalization and degradation

      • Influence of tumor burden on drug clearance

      • Impact of soluble EGFR on pharmacokinetics

  • Exposure-response relationships:

    • Determine minimal effective concentration for EGFR pathway inhibition

    • Establish exposure thresholds for maximal ADCC activity

    • Correlate trough concentrations with biomarkers of target engagement

    • Assess exposure correlation with toxicity, particularly skin reactions

  • Dosing strategy design:

    • Loading dose followed by maintenance dosing to rapidly achieve and maintain therapeutic concentrations

    • Individualized dosing based on patient factors (body weight, tumor burden)

    • Toxicity-adjusted dosing protocols

    • Investigation of extended interval dosing regimens

  • Special populations:

    • Adjust models for hepatic impairment (many anti-EGFR mAbs cause transaminase elevation )

    • Consider impact of anti-drug antibodies on clearance

    • Account for ethnic differences in drug metabolism and response

These models should integrate clinical data from multiple phases of drug development, with continuous refinement based on emerging evidence about resistance mechanisms and combination therapies .

How might anti-EGFR single-chain variable fragments (scFv) be utilized in next-generation immunotherapies?

Anti-EGFR scFv fragments represent a versatile platform for developing next-generation immunotherapies through several methodological approaches:

  • CAR-T cell therapy development:

    • Anti-EGFR scFv can serve as the antigen recognition domain in chimeric antigen receptors

    • CAR design considerations include:

      • Optimal scFv affinity to balance efficacy and on-target/off-tumor toxicity

      • Co-stimulatory domains selection (CD28, 4-1BB) to enhance T cell persistence

      • Inclusion of safety switches (suicide genes) to manage toxicity

    • PanErbB-CAR targeting multiple ErbB family members is currently in clinical trials for head and neck squamous cell carcinoma

  • Bispecific T-cell engagers (BiTEs):

    • Anti-EGFR scFv linked to anti-CD3 scFv to redirect T cells to tumors

    • Format optimization for:

      • Serum half-life extension

      • Tissue penetration

      • Manufacturability

  • Oncolytic virus armament:

    • EGFR-retargeted oncolytic viruses incorporating anti-EGFR scFv

    • Demonstrated efficacy in orthotopic mouse models of primary human glioma

    • Design considerations include:

      • Virus selection (adenovirus, herpes simplex virus, vaccinia)

      • scFv display strategy

      • Additional transgene payload selection

  • Nanoparticle targeting:

    • Anti-EGFR scFv-decorated nanoparticles for targeted drug delivery

    • Integration with imaging agents for theranostic applications

The smaller size of scFv compared to full antibodies (approximately 25 kDa versus 150 kDa) offers advantages in tissue penetration and versatility for engineering applications, while still retaining the variable region functions of monoclonal antibodies .

What are the emerging strategies to overcome acquired resistance to anti-EGFR monoclonal antibodies?

Emerging strategies to address acquired resistance to anti-EGFR monoclonal antibodies encompass several methodological approaches:

Each strategy requires methodical evaluation through well-designed preclinical models and clinical trials with integrated biomarker analyses to identify the most promising approaches for specific resistance mechanisms .

What are the optimal endpoints for evaluating efficacy of anti-EGFR monoclonal antibodies in clinical trials?

Selecting appropriate endpoints for anti-EGFR monoclonal antibody clinical trials requires careful consideration of multiple factors:

The selection of endpoints should align with the specific research question, stage of development, and patient population, with increased attention to patient-reported outcomes and quality of life measures in later-phase studies .

How should toxicity management protocols be designed for anti-EGFR monoclonal antibody therapy?

Designing effective toxicity management protocols for anti-EGFR monoclonal antibody therapy requires a systematic approach based on the known adverse event profile:

  • Common toxicities requiring management:

    • Dermatologic toxicities (skin rash, paronychia)

    • Gastrointestinal effects (diarrhea, nausea)

    • Electrolyte disturbances (magnesium, potassium)

    • Hepatic effects (transaminase elevation)

    • Infusion-related reactions (fever, asthenia)

  • Prophylactic measures:

    • Proactive skin care regimens starting before therapy

    • Premedication protocols to prevent infusion reactions

    • Electrolyte monitoring and replacement guidelines

    • Antibiotic prophylaxis for skin toxicity when indicated

  • Grading systems:

    • Standardized adverse event grading using CTCAE (Common Terminology Criteria for Adverse Events)

    • Specific grading adaptations for EGFR inhibitor-related skin toxicity

  • Dose modification algorithms:

    • Clear guidelines for dose reductions, delays, or discontinuations based on toxicity grade

    • Specific thresholds for intervention (e.g., hold therapy for Grade 3 skin toxicity until improvement to Grade ≤1)

    • Rechallenge protocols after toxicity resolution

  • Multidisciplinary management:

    • Dermatology consultation for severe skin reactions

    • Pharmacist involvement for supportive medication management

    • Nursing education for toxicity assessment and patient counseling

  • Patient education materials:

    • Visual guides for skin toxicity self-assessment

    • Detailed home care instructions

    • Clear guidance on when to contact healthcare providers

  • Quality of life considerations:

    • Regular assessment of toxicity impact on quality of life

    • Psychological support when needed

    • Cosmetic interventions for visible skin changes

These protocols should be evidence-based, regularly updated with emerging data, and include special considerations for combination therapies where toxicity profiles may overlap or interact .

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 2025 TheBiotek. All Rights Reserved.