ErbB2 Human

ErbB-2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Protein Structure

ErbB2 is a 43.4 kDa transmembrane glycoprotein containing:

  • Extracellular domain: Ligand-binding region (though no direct ligand identified).

  • Transmembrane domain: Facilitates dimerization with other ErbB family members (EGFR, ErbB3, ErbB4).

  • Intracellular kinase domain: Mediates downstream signaling via phosphorylation .

FeatureDescriptionSource
Molecular Weight43.4 kDa (human recombinant Herstatin)
Amino Acids397 residues (Herstatin variant) + N-terminal methionine
DimerizationHeterodimerizes with EGFR/ErbB3/ErbB4; homodimers form under overexpression

Signal Transduction Pathways

ErbB2 activates key pathways that promote proliferation and survival:

  • MAPK/ERK: Regulates cell cycle progression.

  • PI3K/Akt: Inhibits apoptosis.

  • STAT: Modulates immune responses .

Role in Oncogenesis

  • Breast Cancer: Overexpression correlates with poor prognosis, reduced disease-free survival, and metastasis .

  • Other Cancers: Associated with ovarian, gastric, and lung carcinomas .

  • HPV-Associated Cancers: Regulates viral promoter activity, enhancing oncogene (E6/E7) expression .

Chemoresistance Mechanisms

ErbB2 overexpression confers resistance to:

DrugMechanism of ResistanceEvidence
Paclitaxel/DocetaxelReduced drug uptake; intrinsic resistance via ERBB2 amplification
AdriamycinDownregulation of pro-apoptotic pathways
TamoxifenEnhanced survival signaling via PI3K/Akt

Tissue Expression

ErbB2 expression varies across tissues:

TissueExpression LevelClinical RelevanceSource
BreastHigh (30% tumors)Predicts response to HER2-targeted therapies
Renal Clear Cell CarcinomaElevatedPotential diagnostic/prognostic biomarker
HeartModerateLinked to congenital heart defects (e.g., LVOTO)

Genetic Variants

VariantPathogenic EffectSource
R599CCardiomyocyte hypertrophy; left ventricular outflow tract obstruction (LVOTO)
L1157RConstitutive kinase activation; oncogenic in solid tumors

Targeted Therapies

DrugMechanismClinical ImpactSource
TrastuzumabMonoclonal antibody; inhibits dimerizationImproved survival in HER2+ breast cancer
TucatinibTyrosine kinase inhibitorEffective in CNS metastases
Afatinib/NeratinibIrreversible EGFR/ErbB2 inhibitorsActive against ERBB2-mutant tumors

Emerging Strategies

  • Autophagy Modulation: ERBB2 inhibition reduces autophagic flux in breast cancer cells, enhancing chemosensitivity .

  • HPV-Targeted Therapies: Silencing ErbB2 suppresses HPV oncogene expression, offering dual antiviral and anticancer effects .

Unresolved Mechanisms

  • Ligand Independence: No high-affinity ligand identified; signaling relies on heterodimerization .

  • Cardiac Toxicity: ERBB2 inhibitors (e.g., trastuzumab) may exacerbate cardiomyopathy .

Future Targets

  • ERBB2 Mutations: Developing inhibitors for activating mutations (e.g., L1157R) .

  • Immune Modulation: ERBB2’s role in immune cell infiltration (e.g., Th17, NK cells) warrants exploration .

Product Specs

Introduction
The HER-2/neu (erbB-2) gene encodes a 185-kDa orphan receptor tyrosine kinase that exhibits constitutive activity as a dimer and demonstrates potent oncogenic activity when overexpressed. Herstatin, generated from an alternative HER-2 transcript, retains intron 8. Herstatin mRNA is found in normal human fetal kidney and liver tissues, but its levels are lower compared to p185HER-2 mRNA in carcinoma cells characterized by HER-2 gene amplification. Herstatin is thought to act as an inhibitor of p185HER-2 by disrupting dimer formation, reducing tyrosine phosphorylation of p185, and suppressing the anchorage-independent growth of transformed cells that overexpress HER-2.
Description
Recombinant Human ErbB-2 is a 43.4 kDa protein comprising 397 amino acid residues of human Herstatin, with an additional Methionine residue at the N-terminus (indicated by underlining). It is produced in E. coli.
Physical Appearance
A clear, colorless solution after filtration.
Formulation
ErbB-2 is supplied as a 0.4 μm filtered solution in 0.05 M Acetate buffer at pH 4 containing 5% trehalose.
Stability
For short-term storage (up to 2-4 weeks), store the product at 4°C. For extended storage, freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 95.0% by SDS-PAGE analysis.
Synonyms
NEU, NGL, HER2, TKR1, HER-2, c-erb B2, HER-2/neu, Receptor tyrosine-protein kinase erbB-2, EC 2.7.10.1, p185erbB2, C-erbB-2, NEU proto-oncogene, Tyrosine kinase-type cell surface receptor HER2, MLN 19, CD340 antigen.
Source
Escherichia Coli.
Amino Acid Sequence
MTQVCTGTDM KLRLPASPET HLDMLRHLYQ GCQVVQGNLE LTYLPTNASL SFLQDIQEVQ GYVLIAHNQV RQVPLQRLRI VRGTQLFEDN YALAVLDNGD PLNNTTPVTG ASPGGLRELQ LRSLTEILKG GVLIQRNPQL CYQDTILWKD IFHKNNQLAL TLIDTNRSRA CHPCSPMCKG SRCWGESSED CQSLTRTVCA GGCARCKGPL PTDCCHEQCA AGCTGPKHSD CLACLHFNHS GICELHCPAL VTYNTDTFES MPNPEGRYTF GASCVTACPY NYLSTDVGSC TLVCPLHNQE VTAEDGTQRC EKCSKPCARG THSLPPRPAA VPVPLRMQPG PAHPVLSFLR PSWDLVSAFY SLPLAPLSPT SVPISPVSVG RGPDPDAHVA VDLSRYEG.

Q&A

What detection methods are most reliable for identifying ErbB2/HER2 expression in research samples?

Researchers commonly employ multiple complementary techniques to detect ErbB2/HER2 expression:

Flow cytometry provides quantitative assessment of cell surface ErbB2/HER2 expression with high sensitivity. For instance, research-grade trastuzumab biosimilar antibodies have been validated for flow cytometry applications in cancer cell lines like MCF-7 and MDA-MB-453 . This approach allows for single-cell analysis and can detect varying levels of ErbB2/HER2 expression across heterogeneous populations.

Direct ELISA represents another validated method for detecting human ErbB2/HER2, particularly when using antibodies that target the same epitope as therapeutic antibodies like trastuzumab . This provides consistency between research findings and potential clinical applications.

For comprehensive analysis, researchers should consider combining these methods with molecular approaches that detect ERBB2 gene amplification or mutation, as expression levels alone may not capture all clinically relevant alterations.

How do ErbB2/HER2 mutation profiles differ across cancer types?

ErbB2/HER2 mutations demonstrate remarkable heterogeneity across cancer types:

Comprehensive analysis of eleven datasets (n=211,726) revealed distinctive ERBB2 mutational hotspots across 25 different tumor types . Unlike amplification, which is well-characterized in breast and gastric cancers, activating mutations in ERBB2 appear across multiple solid tumor types and likely represent distinct molecular subtypes .

These mutations are not uniformly distributed - certain cancer types show predilection for specific mutations. For example, exon 20 insertion mutations (like Y772dupYVMA) are particularly relevant in non-small cell lung cancer (NSCLC), where targeted therapies have shown promising clinical responses .

Researchers should note that distinguishing driver mutations from passenger mutations remains challenging but essential for therapeutic development. The recent focus has shifted toward targeting these activating mutations rather than just amplification events, marking an important evolution in precision oncology .

What in vitro models best represent ErbB2/HER2 mutation effects for drug testing?

Ba/F3 cell systems have proven valuable for interrogating ErbB2/HER2 mutations:

These IL-3-dependent murine pro-B cells become growth factor-independent when transformed with constitutively active oncogenes, making them ideal for testing oncogenic ErbB2/HER2 mutations . Researchers can express specific mutations and measure sensitivity to various inhibitors using IC50 values.

When designing such experiments, it's crucial to include appropriate controls. For instance, Ba/F3 cells expressing wild-type EGFR supplemented with EGF (10 ng/ml) serve as useful comparators when evaluating inhibitor selectivity . This allows calculation of selectivity ratios (mutant/WT IC50) to identify compounds most likely to have favorable therapeutic windows.

The systematic testing of multiple compounds against panels of mutations in isogenic backgrounds provides powerful insights into structure-activity relationships. In one comprehensive analysis, poziotinib emerged as the most mutant-selective tyrosine kinase inhibitor (TKI) across all HER2 mutants (mutant/WT = 0.23) and specifically for exon 20 mutants (mutant/WT = 0.31) .

How can molecular dynamics simulations inform drug development for ErbB2/HER2 mutations?

Molecular dynamics simulations (MDS) provide crucial structural insights for rational drug design:

These computational approaches reveal how mutations affect protein conformation and drug binding. For example, MDS of HER2 mutants (L755S, L755P, Y772dupYVMA, and V777L) constructed from X-ray crystallography data (PDB 3PP0) demonstrated significant conformational differences that impact drug efficacy .

The technique uncovered that distinct mutations at the same residue can dramatically alter protein structure and dynamics. The L755P mutation, for instance, breaks a backbone hydrogen bond between L755 (β3 strand) and V790 (β5 strand), causing destabilization of the β-sheet and structural rearrangement in the kinase hinge region . This specific change reduces the binding pocket volume, explaining decreased sensitivity to larger TKIs.

For accurate simulations, researchers should employ accelerated molecular dynamics to adequately sample the conformational space of the protein. Analysis should focus on key structural elements like the αC-helix position ("in" active vs. "out" inactive conformations) and binding pocket volume changes that directly impact drug binding .

What is the relationship between ErbB2/HER2 status and tumor-infiltrating lymphocytes (TILs)?

The interaction between ErbB2/HER2 and TILs reveals important prognostic implications:

Research has demonstrated associations between ErbB2/HER2 status, TIL levels, and clinical outcomes. In clinical studies, TIL status is typically categorized as "low" (≤30%) or "high" (>30%), providing a practical threshold for stratification .

When planning studies examining this relationship, researchers should include interaction terms in their statistical models to account for the differential effects of ErbB2/HER2 and TILs within hormone receptor-positive versus hormone receptor-negative contexts .

How does ErbB2/HER2-low status correlate with demographic and clinical characteristics?

ErbB2/HER2-low tumors show distinct epidemiological patterns:

Recent cohort studies involving thousands of patients have identified significant differences in hormone receptor status, family history, and self-identified race and ethnicity associated with ErbB2/HER2-low tumors .

Table 1: Selected Clinical and Demographic Factors Associated with ErbB2/HER2 Status

FactorErbB2-LowErbB2-NegativeErbB2-PositiveStatistical Significance
Hormone receptor statusVariable distributionEnriched in triple-negativeOften HR-positivep < 0.05
Family historyDistinctive patternsDiffers from low/positiveDiffers from low/negativep < 0.05
Self-identified raceDemographic variationsDemographic variationsDemographic variationsp < 0.05

When designing studies to investigate these associations, researchers should stratify analyses by hormone receptor status and include comprehensive demographic data collection . The inclusion of self-identified race and ethnicity information is particularly important given the observed variations in ErbB2/HER2 distribution across different populations.

What mechanisms explain the differential response of ErbB2/HER2 mutations to tyrosine kinase inhibitors?

The structural consequences of ErbB2/HER2 mutations directly impact inhibitor efficacy:

Different mutations alter the kinase domain in distinct ways, affecting drug binding differentially. For example, mutations that reduce the binding pocket volume (like L755P) show decreased affinity for larger TKIs . This structural insight explains why smaller molecules like poziotinib demonstrate superior activity against certain mutations.

The position of the αC-helix (between "in"/active and "out"/inactive conformations) varies among mutants, further affecting inhibitor binding. These conformational differences must be considered when matching specific inhibitors to particular mutations .

When designing inhibitor studies, researchers should systematically evaluate structure-activity relationships across multiple mutations and compounds. The most comprehensive analyses have tested panels of TKIs against common HER2 mutants, revealing that poziotinib and pyrotinib were the most mutant-selective TKIs across all HER2 mutants tested .

How do clinical outcomes differ between ErbB2/HER2-targeted therapies for mutation-positive versus amplification-positive cancers?

Response patterns to ErbB2/HER2-targeted therapies vary by alteration type:

Clinical studies have demonstrated distinct efficacy profiles based on the underlying ErbB2/HER2 alteration. For instance, antibody-drug conjugates like T-DM1 showed a 54.5% objective response rate (ORR) in NSCLC patients with ERBB2 exon 20 insertion mutations, but no responses in patients with exon 19 mutations .

For poziotinib, phase II clinical testing in ERBB2 exon 20-mutant NSCLC yielded a confirmed ORR of 42% in the first twelve evaluable patients . These mutation-specific and cancer-specific differences highlight the importance of precise molecular characterization in therapeutic decision-making.

How can combination strategies enhance efficacy against ErbB2/HER2-mutant cancers?

Rational combinations leverage complementary mechanisms of action:

Preclinical research has demonstrated that certain TKIs can upregulate HER2 cell-surface expression, potentiating the activity of antibody-drug conjugates. For example, poziotinib increased HER2 surface expression and enhanced T-DM1 efficacy, resulting in complete tumor regressions with combination treatment .

This synergistic interaction represents a promising research direction, particularly for mutations that show partial resistance to either approach alone. Understanding the mechanisms underlying these combinatorial effects requires detailed investigation of receptor dynamics, internalization rates, and downstream signaling pathways.

When designing combination studies, researchers should consider sequence and timing effects, as the order of drug administration may significantly impact efficacy. Additionally, appropriate in vivo models are essential to capture the complex pharmacokinetic and pharmacodynamic interactions that occur in the tumor microenvironment.

What are the emerging approaches for addressing ERBB2 mutations as actionable biomarkers?

Precision medicine approaches for ERBB2 mutations continue to evolve:

The year 2018 marked an important milestone in establishing ERBB2 mutation as an actionable target across multiple cancer types . This shift from focusing solely on amplification to including mutations represents a paradigm change in how researchers approach ERBB2/HER2 targeting.

Current challenges include the low frequency of these mutations, incomplete understanding of their biological activity, and difficulty distinguishing driver from passenger mutations . Addressing these challenges requires integrated genomic, transcriptomic, and epigenomic analyses, as demonstrated in studies of HER2-low tumors in triple-negative breast cancer patients .

For researchers pursuing this direction, comprehensive molecular profiling that goes beyond simple mutation detection is essential. Integration of structural biology, functional validation, and clinical correlation will be necessary to fully exploit ERBB2 mutations as biomarkers and therapeutic targets.

Product Science Overview

Gene and Protein Structure

The ERBB2 gene, which encodes the ErbB-2 protein, is located on the long arm of human chromosome 17 (17q12) . The protein itself is composed of an extracellular ligand-binding domain, a transmembrane domain, and an intracellular domain that interacts with various signaling molecules . Unlike other members of the ErbB family, ErbB-2 does not have a ligand-binding domain of its own and cannot bind growth factors directly . Instead, it forms heterodimers with other ligand-bound ErbB family members, stabilizing ligand binding and enhancing kinase-mediated activation of downstream signaling pathways .

Function and Significance

ErbB-2 plays a crucial role in cell proliferation, differentiation, and development . It is particularly significant in the context of cancer biology. Amplification or over-expression of the ERBB2 gene has been associated with the development and progression of several aggressive types of cancer, including breast, prostate, ovarian, and lung cancers . Approximately 30% of breast cancer patients exhibit over-expression of HER2, making it an important biomarker and target for therapy .

Recombinant ErbB-2

Recombinant human ErbB-2 is produced using expression systems such as HEK293 cells . The recombinant protein is often tagged (e.g., with a His-tag) to facilitate purification and detection . It is used in various research applications, including studies on receptor-ligand interactions, signal transduction pathways, and the development of therapeutic antibodies .

Clinical Applications

The clinical significance of ErbB-2 is underscored by the development of targeted therapies such as Trastuzumab (Herceptin), which binds to the extracellular domain of HER2 and inhibits its activity . These therapies have significantly improved the prognosis for patients with HER2-positive cancers .

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.