ERBB2 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

  • Receptor Blockade: Binds subdomain IV (e.g., trastuzumab biosimilars ) or subdomain II (e.g., pertuzumab ), preventing heterodimerization with EGFR/ErbB3 .

  • Immune Activation:

    • Antibody-dependent cellular cytotoxicity (ADCC)

    • Complement-dependent cytotoxicity (CDC)

  • Downstream Effects: Suppresses PI3K/Akt and MAPK/ERK pathways, inducing G1 cell cycle arrest .

Clinical Efficacy Data

Phase II Trial of MRG002 (HER2-targeted ADC) in Urothelial Carcinoma :

ParameterResult (n=43)
Objective Response Rate53% (95% CI: 38.9–67.5%)
Complete Response Rate6.9%
Median PFS7.0 months (95% CI: 5.4–NE)
Median OS14.9 months (95% CI: 11.9–NE)
Common Adverse EventsAnemia (51.2%), neutropenia (39.5%)

Preclinical Synergy: Combining trastuzumab, pertuzumab, and experimental antibody 3E10 eliminated BT-474 xenograft tumors in mice .

Diagnostic Use

ApplicationProtocol Example
IHC (Breast Tissue)4 µg/mL primary antibody, citrate buffer antigen retrieval
Flow CytometryDetects HER2 on MDA-MB-453 cells

Functional Assays

  • Proliferation Inhibition: ED₅₀ = 20–100 ng/mL in SK-BR-3 cells .

  • ADCC Enhancement: Anti-HER2 antibodies increase NK cell-mediated lysis by 40–60% .

Comparative Antibody Profiles

Antibody NameEpitope TargetKey FeatureSource
Trastuzumab BiosimilarSubdomain IVBlocks ligand-independent signaling R&D Systems
PertuzumabSubdomain IIInhibits dimerization PMC
3E10Domain IIIInduces conformational change Nature
29D8Intracellular domainWestern blot/IP applications CST

Emerging Directions

  • Antibody-Drug Conjugates (ADCs): MRG002 (anti-HER2-MMAE) shows 77.8% ORR in FISH+ urothelial cancer .

  • Combinatorial Immunotherapies: Co-administration with anti-PD-1/PD-L1 agents under investigation .

  • Biosimilars: Cost-effective alternatives with comparable efficacy to originators .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

The recombinant monoclonal antibody targeting ERBB2 is produced through a meticulous multi-step process. Firstly, ERBB2 antibody genes are precisely integrated into plasmid vectors. These engineered plasmid vectors are then introduced into suitable host cells, enabling the expression of the ERBB2 antibody using advanced exogenous protein expression techniques. Subsequently, the ERBB2 recombinant monoclonal antibody undergoes rigorous purification using affinity chromatography. Comprehensive validation has been performed on this antibody to ensure its suitability for specific applications, including ELISA and IHC. It is crucial to note that this antibody exhibits exclusive recognition of the human ERBB2 protein.

ERBB2 (HER2) plays a pivotal role in regulating cellular signaling pathways that control essential cellular processes such as growth, proliferation, differentiation, and survival. Dysregulation of ERBB2 signaling has been implicated in the initiation and progression of cancer.

Form
Liquid
Lead Time
Typically, we are able to ship your orders within 1-3 business days of receiving them. Delivery time may vary depending on the chosen purchasing method or location. For specific delivery time estimates, please consult with your local distributors.
Synonyms
Verb b2 erythroblastic leukemia viral oncogene homolog 2, neuro/glioblastoma derived oncogene homolog antibody; C erb B2/neu protein antibody; CD340 antibody; CD340 antigen antibody; Cerb B2/neu protein antibody; CerbB2 antibody; Erb b2 receptor tyrosine kinase 2 antibody; ErbB-2 proto-oncogene antibody; ERBB2 antibody; ERBB2_HUMAN antibody; HER 2 antibody; HER 2/NEU antibody; HER2 antibody; Herstatin antibody; Human epidermal growth factor receptor 2 antibody; Metastatic lymph node gene 19 protein antibody; MLN 19 antibody; MLN19 antibody; NEU antibody; NEU proto oncogene antibody; Neuro/glioblastoma derived oncogene homolog antibody; Neuroblastoma/glioblastoma derived oncogene homolog antibody; NGL antibody; p185erbB2 antibody; Proto-oncogene c-ErbB-2 antibody; Proto-oncogene Neu antibody; Receptor tyrosine-protein kinase erbB-2 antibody; TKR1 antibody; Tyrosine kinase type cell surface receptor HER2 antibody; Tyrosine kinase-type cell surface receptor HER2 antibody; V erb b2 avian erythroblastic leukemia viral oncogene homolog 2 (neuro/glioblastoma derived oncogene homolog) antibody; V erb b2 avian erythroblastic leukemia viral oncogene homolog 2 antibody; V erb b2 avian erythroblastic leukemia viral oncoprotein 2 antibody; V erb b2 erythroblastic leukemia viral oncogene homolog 2, neuro/glioblastoma derived oncogene homolog (avian) antibody; V erb b2 erythroblastic leukemia viral oncogene homolog 2, neuro/glioblastoma derived oncogene homolog antibody; Verb b2 erythroblastic leukemia viral oncogene homolog 2, neuro/glioblastoma derived oncogene homolog (avian) antibody
Target Names
Uniprot No.

Target Background

Function
ERBB2 is a protein tyrosine kinase that participates in multiple cell surface receptor complexes, but it appears to necessitate a coreceptor for ligand binding. It is an essential component of a neuregulin-receptor complex, although neuregulins alone do not interact with it. GP30 is a potential ligand for this receptor. ERBB2 regulates the outgrowth and stabilization of peripheral microtubules (MTs). Upon ERBB2 activation, the MEMO1-RHOA-DIAPH1 signaling pathway triggers the phosphorylation and subsequent inhibition of GSK3B at the cell membrane. This inhibition prevents the phosphorylation of APC and CLASP2, facilitating their association with the cell membrane. In turn, membrane-bound APC enables the localization of MACF1 to the cell membrane, which is crucial for microtubule capture and stabilization. In the nucleus, ERBB2 is involved in transcriptional regulation. It associates with the 5'-TCAAATTC-3' sequence in the PTGS2/COX-2 promoter and activates its transcription. ERBB2 is implicated in the transcriptional activation of CDKN1A, a process involving STAT3 and SRC. It participates in the transcription of rRNA genes by RNA Pol I, enhancing protein synthesis and cell growth.
Gene References Into Functions
  1. This abnormally sensitive electrochemical sensing performance resulting from anionic porphyrin for DNA sequences specific to HER2 gene will offer considerable promise for tumor diagnosis and treatment PMID: 30340409
  2. Authors demonstrated that mRNA and protein levels of COX2 and HER2 were upregulated in CRC compared with the adjacent tissues. COX2 protein levels and nuclear COX2 expression were correlated with a poor prognosis of CRC patients. COX2 expression was positively associated with HER2 expression. PMID: 29873317
  3. In patients with HER2-positive advanced breast cancer who have been extensively pretreated with anti-HER2 agents and cytotoxic chemotherapy, trastuzumab emtansine (T-DM1) has demonstrated favorable tolerability and provided a significant progression-free survival of 6 months. Notably, overall survival has not yet been reached. PMID: 29326401
  4. The expression of C-Met and HER2 protein in lung adenocarcinoma exhibits a strong correlation, and further investigation is warranted to determine whether their combined presence offers synergistic potential in targeted therapy for lung adenocarcinoma. PMID: 29400000
  5. While ST6GalI overexpression led to increased HER2 sialylation, corresponding to decreased HER2 phosphorylation, high alpha2,6sialylation unexpectedly enhanced Akt and ERK phosphorylation levels compared to those observed in the vector cell line. Conversely, ST6GalI knockdown had the opposite effects. Collectively, these findings suggest a functional role for ST6GalI in promoting tumor cell progression and resistance to trastuzumab. PMID: 30226606
  6. Studies indicate that miR-495 exerts stimulatory effects on GC chemosensitivity by inactivating the mTOR signaling pathway through suppression of ERBB2. This research provides compelling evidence supporting the potential use of miR-495 as a novel therapeutic target in GC chemotherapy. PMID: 30147110
  7. In early breast cancer, PIK3CA mutations appear to identify HER2+ patients who are less likely to achieve pCR. The clinical implications of PIK3CA mutations seem to vary between exon 9 and exon 20. Further research is necessary to elucidate this mechanism. PMID: 29575819
  8. HER2 and HER3 expression was detected in 22.2% and 86.1% of samples, respectively. The frequency of EGFR mutation was 45.7% and did not show a significant difference between stage 0 and IA1 (40.0% and 48.0%, respectively), suggesting that EGFR mutation may not be directly correlated with cancer progression from stage 0 to IA1. PMID: 29473311
  9. Research has demonstrated that heterogeneous HER2 expression accelerates the development of metastases, leading to poorer survival outcomes in mice with heterogeneous HER2 expression (HER2-60). PMID: 30042341
  10. Her-2/neu amplification increases with escalating grades of breast cancer. A high proportion of Her-2/neu gene amplified cases indicates aggressive disease in that area and warrants FISH testing on a large scale, which is the gold standard for equivocal cases on immunohistochemistry. PMID: 30060783
  11. Data indicate that the primary mechanism is the ability of p140Cap to interfere with ERBB2-dependent activation of Rac GTPase-controlled circuitries. PMID: 28300085
  12. Studies have shown that the expression levels of Gli1 and HER2 are significantly higher in gastric cancer and exhibit a positive relationship. HER2 may regulate Gli1 through the Akt-mTOR-p70S6K pathway. PMID: 29321573
  13. The combination of immunohistochemical expression of BRCA1, ER, PR, and HER-2/neu, coupled with clinicopathological details, may be useful in predicting individuals more likely to carry BRCA1 mutations. This information could facilitate targeted genetic screening for BRCA1 mutations in these individuals and their family members. PMID: 29567881
  14. In the current context, HER2/neu has not been established as a prognostic marker in head-and-neck cancers. PMID: 30004046
  15. These results suggest a possible link between HE4 expression and HER2/neu amplification. PMID: 30004048
  16. HER2 gene amplification in circulating tumor DNA has been shown to predict resistance to trastuzumab emtansine in HER2-positive breast neoplasms. PMID: 29700710
  17. Statistical analysis conducted in this study did not reveal a significant relationship between HER2 overexpression on tumor cells and microvessel density in the tumor stroma. PMID: 30334990
  18. Data highlight a high rate of discordance in matched pairs of primary tumors and metastases, suggesting the importance of accurate evaluation of proto-oncogene protein HER-2 (HER2) status before making therapeutic decisions. PMID: 30203148
  19. HER2 gene amplification has been observed to occur during the early stages of gastric cancer and has shown heterogeneity in several cases. HER2 gene amplification may play a role in tumor progression during the early stages of gastric cancer. PMID: 30120594
  20. Activating HER2 mutations are present in approximately 3% of bone metastases from breast cancers, with significantly higher rates observed in the pleomorphic subtype of lobular cancer. PMID: 30094493
  21. The results suggest a possible connection between tRNALeu overexpression and RSK1/MSK2 activation and ErbB2/ErbB3 signaling, particularly in breast cancer. PMID: 28816616
  22. High HER2 expression has been linked to metastasis in breast cancer. PMID: 29187405
  23. This study confirmed that biosimilar trastuzumab improves the overall response rate when combined with chemotherapy for HER2+ breast cancer. PMID: 30082554
  24. The study reveals a gender difference in the prognostic value of concomitant AIB1 and HER2 copy number gain (CNG) in glioma patients, which was previously largely unnoticed. These findings suggest that genetic alterations that interact with essential aspects of sex determination influence glioma biology and patient outcomes. PMID: 30153912
  25. The survival rates observed in this study are consistent with documented global rates. Notably, nodal disease burden emerged as the most significant prognostic factor. Additionally, in EBCs, a lack of hormone receptor expression and in LABC, Her2neu overexpression appear to negatively impact outcomes. PMID: 30147088
  26. Results indicate that HER2 and FGFR2 are regulated by DDX6 at the post-transcriptional level in gastric cancer. PMID: 29987267
  27. HER2 overexpression is associated with Gastric Cancer. PMID: 29938472
  28. The ERBB2 oncogene located at 17q12 is susceptible to palindromic gene amplification in HER2-positive breast tumors. PMID: 28211519
  29. Results demonstrate that mutations in ERBB2-exon17 were associated with poorer survival outcomes in patients with pancreatic neoplasm. [review] PMID: 30227250
  30. High HER2 expression and gene amplification have been linked to Upper Tract Urothelial Carcinomas. PMID: 28755093
  31. High HER2 expression has been associated with invasion and lymph node metastasis in gastric cancer. PMID: 29970682
  32. The basal HER2 phenotype exhibited poor DFS, but achieved an equivalent pCR rate after concurrent neo-adjuvant chemotherapy with trastuzumab. This finding suggests the need for a distinct treatment approach for the basal-HER2 type, even in cases that have shown adequate clinical response after neo-adjuvant chemotherapy. PMID: 29971625
  33. In the largest series reported to date, patients with HER2-amplified m17 cancers treated with trastuzumab have shown outcomes comparable to those observed in patients from large phase III adjuvant trastuzumab trials who were HER2-positive. This finding supports the critical role of HER2-directed therapy in this patient population. PMID: 28986743
  34. The interplay of dual MET/HER2 overexpression in the AKT and ERK pathways for esophageal cancer is described. This suggests that combination therapy could be a promising strategy for esophageal adenocarcinoma (EAC) characterized by amplification of both MET and HER2. PMID: 29223420
  35. Research provides evidence that the hostile environment developed in spheroids plays a key role in the acquisition of resistance to Trastuzumab. This resistance is associated with an increase in the number of breast cancer stem cells as well as a modulation in HER2 expression. PMID: 28722778
  36. A significant finding in this study is that one out of five (20%) patients with breast cancer BM exhibited a receptor discrepancy between the primary tumor and the subsequent BM. Loss of hormone receptors (ER and/or PR) expression and gain of HER2 overexpression were the most commonly observed changes. PMID: 28975433
  37. High HER2 expression is associated with Gastric Adenocarcinoma. PMID: 29802704
  38. Absence of HER2 expression in circulating tumor cells has been linked to non-metastatic esophageal cancer. PMID: 30275185
  39. HER2 positivity was found in a small proportion of rectal cancer patients and was not significantly associated with clinicopathologic and molecular characteristics. PMID: 30056472
  40. This study identified a novel enhancer, the HER2 gene body enhancer (HGE), located within the 3' gene body of HER2. The HGE activates promoters 1 and 2 in trans, contributing to the TFAP2C-mediated transcriptional induction of HER2 expression in breast cancer samples. PMID: 29035388
  41. ctDNA gene mutation profiles differed among HR/HER2 subtypes of metastatic breast cancer (MBC) patients. By identifying mutations associated with treatment resistance, the aim is to improve therapy selection for MBC patients who have received multiple lines of treatment. PMID: 29807833
  42. Research concluded that miR494 inhibited the cancer initiating cells phenotype and reversed resistance to lapatinib by inhibiting FGFR2 in HER2-positive gastric cancer. PMID: 29786108
  43. HER2 overexpression was evident in nearly 25% of Malaysian patients with locally advanced or metastatic gastric cancer. This overexpression correlated significantly with male gender and diffuse-type tumors. PMID: 28124769
  44. A statistically significant association was observed between positive p95-HER2 expression and negative hormonal receptors expression (p=0.004), high Ki-67 expression (p<0.001), and the development of visceral metastasis. PMID: 29779938
  45. The authors provide novel evidence demonstrating that the transcriptional repressor Blimp1 is a new mediator of p130Cas/ErbB2-mediated invasiveness. Notably, high Blimp1 expression levels are detected in invasive p130Cas/ErbB2 cells and correlate with metastatic status in human breast cancer patients. PMID: 28442738
  46. ERBB2 amplification is a driving force behind resistance to erlotinib in lung adenocarcinoma. PMID: 28870636
  47. Results demonstrate that combining the findings of IHC and FISH, following the HER2 testing algorithm, is a reliable method for accurately evaluating HER2-positive EMPD. PMID: 29744813
  48. Due to the lower concordance rates observed in HER2 IHC score 2/3+ cases compared to HER2 IHC score 0/1+ cases, further research is required to establish detailed analysis criteria for HER2 IHC score 2+ or 3+. PMID: 28478639
  49. HER2 interacts with Beclin 1 in breast cancer cells and inhibits autophagy. Mice genetically engineered with a mutation in Becn1, resulting in increased basal autophagy, are protected from human HER2-driven mammary tumorigenesis. This suggests that HER2-mediated inhibition of Beclin 1 and autophagy likely contributes to HER2-mediated tumorigenesis. PMID: 29610308
  50. These findings suggest that early-stage morphological alterations of HER2-positive BC cells during cancer progression can occur in a manner that is independent of physical interactions and signaling pathways. PMID: 27599456

Show More

Hide All

Database Links

HGNC: 3430

OMIM: 137800

KEGG: hsa:2064

STRING: 9606.ENSP00000269571

UniGene: Hs.446352

Involvement In Disease
Glioma (GLM); Ovarian cancer (OC); Lung cancer (LNCR); Gastric cancer (GASC)
Protein Families
Protein kinase superfamily, Tyr protein kinase family, EGF receptor subfamily
Subcellular Location
[Isoform 1]: Cell membrane; Single-pass type I membrane protein. Early endosome. Cytoplasm, perinuclear region. Nucleus.; [Isoform 2]: Cytoplasm. Nucleus.; [Isoform 3]: Cytoplasm. Nucleus.
Tissue Specificity
Expressed in a variety of tumor tissues including primary breast tumors and tumors from small bowel, esophagus, kidney and mouth.

Q&A

What is ERBB2/HER2 and why is it a significant target for monoclonal antibodies?

ERBB2 (also known as HER2) is a receptor tyrosine kinase that plays a crucial role in cell proliferation, differentiation, metastasis, and signal transduction. It is particularly significant as a research and therapeutic target because it is overexpressed in approximately 40% of human breast cancers as well as in various epithelial ovarian cancer tissues . The protein functions as a key mediator in cancer cell proliferation and survival pathways, making it an important oncogenic driver. Targeting ERBB2 with monoclonal antibodies has proven effective in both research contexts for understanding cancer biology and in therapeutic applications where antibodies like trastuzumab (Herceptin) are used for treating metastatic HER2-positive cancers . The receptor's prominence in multiple cancer types and its accessibility as a cell surface protein make it an ideal candidate for targeted antibody development and application.

How are recombinant monoclonal antibodies against ERBB2 produced?

Recombinant monoclonal antibodies against ERBB2 are produced using in vitro expression systems rather than traditional hybridoma technology. The process begins with cloning specific antibody DNA sequences from immunoreactive sources (such as rabbits) that have developed immune responses against ERBB2 . These DNA sequences, particularly those encoding the variable regions that determine antibody specificity, are then inserted into expression vectors. For humanized antibodies, the process often involves constructing a mammalian cell expression vector containing both light chain (LC) and heavy chain (HC) sequences .

The vectors are then transfected into production cell lines, commonly Human Embryonic Kidney 293 (HEK293) cells, using transient gene expression (TGE) technology or stable cell line development . In TGE systems, optimization of transfection conditions is crucial, including determining the optimal ratio of light chain to heavy chain plasmids (typically tested in ranges from 4:1 to 1:2) and the ratio of DNA to transfection reagents like polyethyleneimine (PEI) . After expression, the antibodies are purified from the cell culture supernatant using affinity chromatography methods such as rProtein A affinity chromatography . The final products undergo quality control testing for proper assembly, specificity, and biological activity.

What are the advantages of using recombinant monoclonal antibodies versus traditional monoclonal antibodies for ERBB2 research?

Recombinant monoclonal antibodies offer several significant advantages over traditional monoclonal antibodies for ERBB2 research:

  • Enhanced specificity and sensitivity: Recombinant antibodies typically demonstrate superior target recognition and binding characteristics, resulting in lower background and more reliable experimental results .

  • Lot-to-lot consistency: Unlike traditional hybridoma-derived antibodies that may show variation between production batches, recombinant antibodies are produced from defined genetic sequences, ensuring consistent performance across different lots .

  • Animal origin-free formulations: Many recombinant antibody production systems eliminate the need for animals in the manufacturing process, addressing ethical concerns and reducing potential contaminants .

  • Broader immunoreactivity: Particularly with rabbit-derived sequences, recombinant antibodies can capitalize on the larger immune repertoire of rabbits, potentially recognizing epitopes that might be missed by mouse-derived traditional antibodies .

  • Rapid production capability: Transient gene expression (TGE) systems allow researchers to quickly obtain significant quantities of antibodies compared to the time-consuming process of developing stable cell lines .

  • Customization potential: The recombinant approach allows for easier engineering of antibody properties, including humanization, isotype switching, or modification of Fc regions for specific research applications .

How do different epitope binding sites on ERBB2 affect the functional properties of recombinant monoclonal antibodies?

The epitope specificity of anti-ERBB2 recombinant monoclonal antibodies significantly influences their functional properties in both research and potential therapeutic applications. ERBB2 contains multiple domains, and antibodies targeting different regions can elicit distinct biological responses. For instance, antibodies like trastuzumab biosimilars target the extracellular domain of ERBB2, which results in specific inhibitory effects on receptor signaling . In contrast, antibodies targeting regions surrounding phosphorylation sites, such as tyrosine 1248, may provide insights into the activation status of the receptor .

The functional consequences of epitope specificity include:

  • Signaling pathway modulation: Antibodies binding to different domains can selectively inhibit specific downstream signaling cascades, allowing researchers to dissect complex regulatory networks.

  • Receptor internalization rates: Some epitopes, when bound by antibodies, trigger rapid receptor internalization and degradation, while others may stabilize surface expression.

  • Antibody-dependent cellular cytotoxicity (ADCC) potency: The orientation and positioning of the antibody binding can significantly affect its ability to recruit immune effector cells, with some epitopes providing optimal Fc presentation to immune receptors .

  • Combination effects: Antibodies targeting different epitopes may demonstrate synergistic or antagonistic effects when used together, providing valuable research tools for understanding receptor biology.

Researchers should carefully consider epitope specificity when selecting antibodies for experimental purposes, as this choice will directly impact the biological outcomes and interpretability of their results.

What methodological approaches can be used to optimize antibody-dependent cellular cytotoxicity (ADCC) in ERBB2-targeting recombinant antibodies?

Optimizing antibody-dependent cellular cytotoxicity (ADCC) in ERBB2-targeting recombinant antibodies involves several methodological approaches:

  • Fc region engineering: Modifications to the Fc region can enhance binding to Fcγ receptors on immune effector cells. Specific amino acid substitutions, glycoengineering, or isotype selection can significantly impact ADCC potency .

  • Expression system optimization: The choice of expression system affects glycosylation patterns, which in turn influences ADCC activity. HEK293 cells are commonly used for producing antibodies with human-compatible glycoforms that promote effective ADCC .

  • Light chain/Heavy chain ratio optimization: Adjusting the LC:HC ratio during antibody expression can impact proper folding and assembly, potentially enhancing ADCC functionality. Experimental determination of optimal ratios (typically ranging from 4:1 to 1:2) is recommended for each specific antibody construct .

  • Assay development for ADCC quantification: Lactate dehydrogenase (LDH) release assays provide a reliable method for quantifying ADCC activity. These assays measure the release of LDH from target cells as an indicator of immune-mediated cytotoxicity .

  • Target cell selection: ADCC potency assessment should utilize cell lines with clinically relevant ERBB2 expression levels. High-expressing cell lines like SK-BR-3 (breast cancer) provide robust models for evaluating ADCC potential .

  • Effector cell preparation: The source, activation status, and ratio of effector cells (typically NK cells or peripheral blood mononuclear cells) to target cells significantly impact ADCC assay sensitivity and reproducibility.

Researchers have demonstrated that optimized anti-ERBB2 humanized recombinant antibodies can achieve superior ADCC activity compared to reference antibodies like Herceptin, highlighting the importance of these methodological considerations in developing improved research tools .

How can recombinant anti-ERBB2 antibodies be leveraged for studying the relationship between receptor tyrosine kinase activity and cancer proliferation?

Recombinant anti-ERBB2 antibodies serve as sophisticated tools for investigating the complex relationship between ERBB2 receptor tyrosine kinase activity and cancer proliferation through multiple experimental approaches:

  • Phosphorylation site-specific antibodies: Antibodies targeting specific phosphorylation sites (such as Tyr1248, Tyr1221/1222, or Tyr877) allow researchers to monitor the activation status of ERBB2 and correlate it with downstream signaling events and cellular outcomes .

  • Proliferation inhibition assays: Quantitative assessment of antibody-mediated growth inhibition provides direct evidence of the relationship between ERBB2 blockade and cancer cell proliferation. For example, anti-ERBB2 antibodies have demonstrated concentration-dependent inhibition of proliferation in SK-BR-3 breast cancer cells, with ED50 values typically in the range of 20-100 ng/mL .

  • Combination with kinase inhibitors: Using recombinant antibodies in conjunction with small molecule kinase inhibitors allows researchers to distinguish between scaffolding functions and catalytic activities of ERBB2.

  • Degradation pathway analysis: Antibodies can be used to study how ERBB2 degradation affects proliferation. For instance, binding of c-Cbl ubiquitin ligase to ERBB2 at Tyr1112 leads to poly-ubiquitination and enhanced degradation, potentially reducing proliferative signaling .

  • In vivo xenograft models: Recombinant antibodies enable the establishment of correlations between receptor inhibition and tumor growth suppression in animal models, providing a system-level understanding of the ERBB2-proliferation relationship .

By systematically applying these approaches, researchers can delineate the signaling networks connecting ERBB2 activation to proliferative responses and identify potential vulnerabilities that could be exploited for therapeutic intervention.

What are the optimal validation techniques for confirming specificity of new recombinant anti-ERBB2 antibodies?

Validating the specificity of new recombinant anti-ERBB2 antibodies requires a multi-faceted approach that combines complementary techniques to establish confidence in antibody performance:

  • Western blotting: Essential for confirming the antibody recognizes a protein of the expected molecular weight (~185 kDa for full-length ERBB2) . This technique also verifies proper antibody assembly through non-reducing and reducing conditions, where intact IgG appears as ~150 kDa bands under non-reducing conditions and generates ~25 kDa light chain and ~50 kDa heavy chain bands under reducing conditions .

  • Flow cytometry with appropriate controls: Comparing staining patterns between known ERBB2-positive cell lines (e.g., SK-BR-3) and low-expressing cell lines (e.g., MCF-7) alongside isotype controls verifies surface recognition specificity . Quantitative assessment of cell surface binding provides evidence of functionality in recognizing native conformation.

  • Immunoprecipitation followed by mass spectrometry: This approach confirms that the antibody pulls down authentic ERBB2 rather than cross-reactive proteins by identifying unique peptide signatures.

  • Competitive binding assays: Demonstrating that the antibody competes with known anti-ERBB2 antibodies or natural ligands for binding provides evidence of epitope-specific recognition.

  • Genetic validation approaches: Testing antibody recognition in ERBB2-knockout or knockdown cell models provides definitive evidence of specificity by demonstrating loss of signal when the target is absent.

  • Cross-reactivity assessment: Testing against related family members (EGFR/ErbB1, ErbB3, ErbB4) confirms that the antibody specifically recognizes ERBB2 without binding to structurally similar proteins.

  • Immunohistochemistry correlation with established methods: Comparing staining patterns with clinically validated ERBB2 detection methods in tissue samples with known ERBB2 status validates applicability for tissue analysis .

A comprehensive validation approach incorporating multiple techniques provides the strongest evidence for antibody specificity and suitability for research applications.

What factors affect the reproducibility of experiments using ERBB2 recombinant monoclonal antibodies?

Several critical factors influence experimental reproducibility when working with ERBB2 recombinant monoclonal antibodies:

  • Antibody lot consistency: While recombinant antibodies generally demonstrate superior lot-to-lot consistency compared to traditional hybridoma-derived antibodies, variations can still occur . Researchers should record lot numbers and consider testing new lots against reference standards before use in critical experiments.

  • Storage and handling conditions: Antibody functionality can be compromised by improper storage, repeated freeze-thaw cycles, or exposure to unfavorable conditions. Following manufacturer recommendations for temperature, buffer conditions, and handling procedures is essential.

  • Cell line heterogeneity: ERBB2 expression levels can vary significantly across passages of the same cell line or between different sources of nominally identical cell lines. Regular validation of ERBB2 expression levels in experimental cell models is recommended.

  • Experimental protocol standardization: Variations in antibody concentration, incubation time, buffer composition, and detection methods can significantly impact results. Detailed protocol documentation and standardization are crucial for reproducibility.

  • Transfection efficiency in antibody production: When producing antibodies using transient gene expression systems, variations in transfection efficiency between experiments can affect antibody yield and quality . Standardizing transfection conditions, including DNA:PEI ratios and LC:HC plasmid ratios, improves consistency.

  • Appropriate controls: Inclusion of isotype controls, positive and negative cell line controls, and reference antibodies with established properties provides context for interpreting experimental outcomes .

  • Detection system sensitivity: The choice of secondary antibodies, fluorophores, or enzymatic detection systems affects signal-to-noise ratios and can influence apparent results, particularly in quantitative applications.

Addressing these factors through careful experimental design, thorough documentation, and consistent methodology significantly enhances the reproducibility of research using ERBB2 recombinant monoclonal antibodies.

How can researchers optimize flow cytometry protocols for detecting ERBB2 using recombinant monoclonal antibodies?

Optimizing flow cytometry protocols for ERBB2 detection requires attention to several key factors:

Table 1: Optimization Parameters for ERBB2 Flow Cytometry

ParameterRecommendationRationale
Antibody concentration1:200 - 1:400 dilutionProvides optimal signal-to-noise ratio while conserving antibody
Cell preparationSingle-cell suspension, viable cellsReduces aggregates and background from dead cells
Buffer compositionPBS with 1-2% BSA or FBSMinimizes non-specific binding
Incubation temperature4°CReduces internalization of receptor-antibody complexes
Incubation time30-60 minutesEnsures equilibrium binding without excessive background
Washing steps3x with cold bufferRemoves unbound antibody to reduce background
Secondary antibody (if needed)Anti-species IgG PE-conjugatedPE provides high sensitivity for detecting varying ERBB2 levels
ControlsIsotype control, blocking controlsEssential for distinguishing specific from non-specific binding

For detecting ERBB2 in clinical samples or cell lines with varying expression levels, researchers should consider these methodological approaches:

  • Titration of primary antibody: Determine the optimal concentration that provides maximum specific signal with minimal background by testing a dilution series.

  • Use of directly conjugated antibodies: When available, directly conjugated antibodies (such as Alexa Fluor 488-conjugated anti-ERBB2) can reduce protocol complexity and variability associated with secondary detection.

  • Gating strategy optimization: Implement hierarchical gating that first excludes debris and dead cells, then identifies single cells before analyzing ERBB2 expression.

  • Cell surface vs. total ERBB2 detection: For cell surface expression, maintain cells in non-permeabilized state; for total ERBB2, include a permeabilization step after fixation.

  • Standardization with calibration beads: Use calibration beads to establish a standard curve, allowing for quantitative comparison of ERBB2 expression levels across experiments.

When troubleshooting poor signal, researchers should systematically evaluate antibody quality, cell viability, receptor internalization, and detector sensitivity to identify and address the specific limiting factor.

What strategies can address common challenges in immunohistochemistry (IHC) applications of ERBB2 recombinant monoclonal antibodies?

Immunohistochemistry with ERBB2 recombinant monoclonal antibodies presents several common challenges that can be addressed through systematic optimization:

  • Epitope masking during fixation: Over-fixation with formalin can mask ERBB2 epitopes, reducing antibody accessibility.

    • Solution: Implement antigen retrieval protocols using citrate buffer (pH 6.0) or EDTA buffer (pH 8.0) with optimized heating conditions (95-100°C for 15-20 minutes) .

  • Variable expression levels across tissue specimens: ERBB2 expression can vary significantly within and between samples.

    • Solution: Include known positive and negative control tissues on each slide, and consider using a dilution series approach (1:200, 1:400, 1:800) to identify the optimal antibody concentration for each experiment .

  • Non-specific background staining: High background can complicate interpretation of true ERBB2 positivity.

    • Solution: Implement thorough blocking steps (3-5% BSA or serum matching the species of the secondary antibody), optimize antibody dilution, and include appropriate washing steps between each stage of the protocol.

  • Membranous vs. cytoplasmic staining interpretation: ERBB2 should primarily show membranous staining in overexpressing cells.

    • Solution: Clearly define scoring criteria that distinguish between membrane staining (relevant for ERBB2 status) and cytoplasmic staining (potentially non-specific).

  • Inconsistent tissue processing: Variations in fixation time and processing can affect staining quality.

    • Solution: Standardize tissue handling protocols, including fixation duration (12-24 hours in 10% neutral buffered formalin) and processing schedules.

  • Signal amplification considerations: Weak ERBB2 expression may require signal amplification.

    • Solution: Implement polymer-based detection systems or tyramide signal amplification while monitoring for increased background.

  • Digital quantification challenges: Objective quantification of ERBB2 staining can be difficult.

    • Solution: Establish consistent imaging parameters and utilize digital image analysis software with validated algorithms for membrane staining quantification.

By systematically addressing these challenges, researchers can achieve more consistent and interpretable results when using ERBB2 recombinant monoclonal antibodies for immunohistochemistry applications.

How should researchers design experiments to compare the efficacy of different anti-ERBB2 recombinant monoclonal antibodies?

Designing robust comparative experiments for evaluating different anti-ERBB2 recombinant monoclonal antibodies requires careful consideration of multiple parameters:

  • Standardized binding assessment:

    • Implement surface plasmon resonance (SPR) or bio-layer interferometry (BLI) to determine binding kinetics (kon, koff) and affinity (KD) under identical conditions

    • Use flow cytometry with quantitative beads to establish binding curves on cells expressing different levels of ERBB2

    • Maintain consistent experimental conditions including temperature, buffer composition, and instrument settings

  • Functional comparison framework:

    • Cell proliferation inhibition: Establish dose-response curves using multiple cell lines with varying ERBB2 expression levels, calculating IC50 values for each antibody

    • ADCC activity: Standardize effector:target ratios, effector cell preparation, and quantification methods (such as LDH release assays) to enable direct comparison

    • Receptor internalization rates: Measure the kinetics of antibody-induced ERBB2 internalization using pH-sensitive fluorophores or surface biotinylation approaches

  • In vivo efficacy comparison:

    • Use identical xenograft models with defined cell lines and consistent implantation protocols

    • Administer antibodies at equimolar concentrations rather than equal mass

    • Include pharmacokinetic assessment to account for potential differences in half-life

    • Measure multiple outcomes including tumor volume, downstream signaling inhibition, and receptor downregulation

  • Experimental design considerations:

    • Conduct experiments in a blinded fashion when possible

    • Include appropriate positive controls (e.g., commercial trastuzumab) and negative controls (non-targeting IgG)

    • Perform power calculations to determine adequate sample sizes for detecting meaningful differences

    • Replicate key experiments independently to confirm reproducibility

  • Comprehensive data analysis:

    • Utilize statistical approaches that account for multiple comparisons

    • Consider area under the curve (AUC) analyses for time-course experiments rather than single timepoint comparisons

    • Integrate multiple parameters into composite scoring systems when appropriate

By implementing these design principles, researchers can generate robust comparative data that accurately reflects the relative efficacy of different anti-ERBB2 recombinant monoclonal antibodies across relevant biological contexts.

How might next-generation anti-ERBB2 recombinant antibodies overcome current limitations in research applications?

Next-generation anti-ERBB2 recombinant antibodies are poised to address several limitations of current research tools through innovative engineering approaches:

  • Enhanced epitope specificity: Development of antibodies targeting novel epitopes or conformational states of ERBB2 will enable more precise investigation of receptor dynamics and heterogeneity. This includes antibodies that can distinguish between active/inactive conformations or those that recognize specific dimerization interfaces with other ErbB family members .

  • Multispecific antibody formats: Creating bispecific or multispecific antibodies that simultaneously target ERBB2 and other relevant molecules (such as immune checkpoints or other receptor tyrosine kinases) will facilitate investigation of complex signaling networks and receptor crosstalk mechanisms.

  • Intracellular delivery systems: Development of cell-penetrating antibodies or antibody-mimetic molecules capable of accessing intracellular ERBB2 pools would overcome the current limitation of antibodies being restricted to cell surface targets.

  • Photoswitchable antibodies: Integration of photoswitchable domains would allow temporal and spatial control over antibody activity, enabling more precise experimental manipulation of ERBB2 signaling in specific cellular compartments or at defined timepoints.

  • Signal-amplifying antibody conjugates: Conjugation with enzymatic reporters or proximity-based amplification systems could enhance detection sensitivity for low-expressing samples without increasing background.

  • Tunable affinity variants: Engineering antibodies with controllable binding properties would allow researchers to modulate residence time on the receptor, potentially revealing new insights into how binding kinetics influence biological outcomes.

  • Streamlined production approaches: Further refinement of transient gene expression systems could reduce production time and increase yield, making custom antibody generation more accessible to the research community .

These innovations will expand the experimental toolkit available for ERBB2 research, enabling more sophisticated investigations into receptor biology and potentially revealing new therapeutic opportunities.

What are the emerging applications of ERBB2 recombinant monoclonal antibodies beyond traditional research contexts?

ERBB2 recombinant monoclonal antibodies are expanding beyond traditional research applications into several innovative domains:

  • Single-cell analysis platforms: Integration with mass cytometry (CyTOF) and other single-cell technologies enables comprehensive profiling of ERBB2 expression and activation states at unprecedented resolution, revealing cellular heterogeneity within tumors .

  • Organoid and patient-derived xenograft (PDX) model development: Antibodies are being used to identify, isolate, and characterize ERBB2-positive cells for establishing more physiologically relevant ex vivo and in vivo models that better recapitulate patient tumor characteristics.

  • Antibody-based biosensors: Development of conformation-sensitive antibodies that change properties upon binding is enabling real-time monitoring of ERBB2 activation in living systems through FRET-based or other proximity-reporting approaches.

  • PROTAC (Proteolysis Targeting Chimera) development: Conjugation of ERBB2-targeting antibody fragments with E3 ligase recruiting moieties creates novel degradation-inducing research tools that offer advantages over traditional blocking antibodies for certain applications.

  • Synthetic immunology applications: Anti-ERBB2 antibodies are being incorporated into engineered immune cell therapies for research purposes, including CAR-T cell development and artificial antigen-presenting cell constructs.

  • Antibody-guided imaging probe development: Site-specific conjugation of imaging moieties to recombinant anti-ERBB2 antibodies is advancing molecular imaging capabilities for preclinical research.

  • Extracellular vesicle (EV) targeting: Antibodies against ERBB2 are enabling the capture and characterization of tumor-derived EVs, opening new avenues for studying intercellular communication and potential liquid biopsy applications.

These emerging applications highlight the versatility of recombinant anti-ERBB2 antibodies as adaptable research tools that continue to evolve alongside technological advances in biomedical research.

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.