InVivoMab anti-human/rat HER2 (neu)

Clone 7.16.4
Catalog # BE0277
Category InVivoMab Antibodies
Price
Size Regular Price
1 mg $ 150.00
5 mg $ 550.00
25 mg $ 1,840.00
50 mg $ 2,770.00
100 mg $ 3,920.00
About InVivoMab anti-human/rat HER2 (neu)

The 7.16.4 monoclonal antibody reacts with human and rat HER2 (human epidermal growth factor receptor 2) also known as neu peptide, CD340, ErbB-2, and p185. HER2 is a 185 kDa transmembrane, receptor-like glycoprotein with intrinsic tyrosine kinase activity that is part of several cell surface receptor complexes. HER2 lacks an identified ligand however, the kinase can be activated in the absence of a ligand when overexpressed. HER2 is a proto-oncoprotein that is commonly overexpressed on a variety of different tumors. Approximately 40% of human breast cancers overexpress HER2. HER2 overexpression is associated with poorer overall survival rates, shorter times to disease progression, and increased resistance to chemotherapy. Because of these clinical characteristics anti-HER2 monoclonal antibody therapy is now a standard for the treatment of advanced breast cancers that overexpress HER2. The 7.16.4 antibody has been shown to inhibit the growth of HER2-overexpressing tumors both in vitro and in vivo.

InVivoMab anti-human/rat HER2 (neu) Specifications
IsotypeMouse IgG2a, κ
Immunogenneu-transfected NIH 3T3 cells
Reported Applications
  • in vivo HER2/neu inhibition
  • in vitro HER2/neu inhibition
  • Immunoprecipitation
  • Immunofluorescence
  • Flow cytometry
Formulation
  • PBS, pH 7.0
  • Contains no stabilizers or preservatives
Endotoxin
  • <2EU/mg (<0.002EU/μg)
  • Determined by LAL gel clotting assay
Purity
  • >95%
  • Determined by SDS-PAGE
Sterility0.2 μM filtered
ProductionPurified from tissue culture supernatant in an animal free facility
PurificationProtein G
RRIDAB_2687800
Molecular Weight150 kDa
StorageThe antibody solution should be stored at the stock concentration at 4°C. Do not freeze.
Application References

INVIVOMAB ANTI-HUMAN/RAT HER2 (NEU) (CLONE: 7.16.4)

 

Park, S., et al. (2010). “The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity.” Cancer Cell 18(2): 160-170. PubMed

Anti-HER2/neu antibody therapy is reported to mediate tumor regression by interrupting oncogenic signals and/or inducing FcR-mediated cytotoxicity. Here, we demonstrate that the mechanisms of tumor regression by this therapy also require the adaptive immune response. Activation of innate immunity and T cells, initiated by antibody treatment, was necessary. Intriguingly, the addition of chemotherapeutic drugs, although capable of enhancing the reduction of tumor burden, could abrogate antibody-initiated immunity leading to decreased resistance to rechallenge or earlier relapse. Increased influx of both innate and adaptive immune cells into the tumor microenvironment by a selected immunotherapy further enhanced subsequent antibody-induced immunity, leading to increased tumor eradication and resistance to rechallenge. This study proposes a model and strategy for anti-HER2/neu antibody-mediated tumor clearance.

Knutson, K. L., et al. (2004). “Neu antigen-negative variants can be generated after neu-specific antibody therapy in neu transgenic mice.” Cancer Res 64(3): 1146-1151. PubMed

Prolonged administration of HER-2/neu-specific monoclonal antibody therapy is now widely used for the treatment of HER-2/neu-overexpressing tumors in advanced-stage breast cancer patients. Monoclonal antibody therapy has the potential to promote reduced tumor expression of HER-2/neu by receptor down-modulation and/or the generation of antigen-negative variants. Loss of antigen by either mechanism could potentially impact subsequent therapeutic strategies targeting HER-2/neu. In this study, the effects of chronic neu-specific monoclonal antibody therapy on tumor growth and neu protein expression were examined in a murine model of neu-overexpressing breast cancer. Treatment of neu-overexpressing tumors with neu-specific antibody, in vitro or in vivo, resulted in significant tumor growth inhibition. When neu antibody was used to treat neu-overexpressing tumor cells both in vitro and in vivo in tumor-bearing mice, neu receptor expression was not diminished after cessation of therapy. However, in the setting of clinically undetectable disease in a fraction of animals, antigen-negative variants were generated. An understanding of the effects of monoclonal antibodies on target antigen expression is critical for the future design and testing of novel HER-2/neu-targeted therapies administered in combination with or after HER-2/neu-specific monoclonal antibody therapy.

Zhang, H., et al. (1999). “Shared antigenic epitopes and pathobiological functions of anti-p185(her2/neu) monoclonal antibodies.” Exp Mol Pathol 67(1): 15-25. PubMed

We have studied two anti-p185 antibodies: the monoclonal antibody 7. 16.4 and rhuMAb 4D5, which were raised against the the ectodomain of rat (p185(neu)), and the human (p185(her2/neu)) homolog, respectively. Studies on the structure of these two antibodies indicate that they share structural similarity in the variable region, especially the CDR3 region, which determines the antibody-antigen interaction. Further studies by flow cytometry revealed that 7.16.4 can compete with rhuMAb4D5 for binding to the cell surface p185(her2/neu), suggesting that these two antibodies share an epitope on the p185 receptor. Furthermore, 7.16.4 can also inhibit proliferation and transformation caused by p185(her2/neu). Moreover the rhuMAb 4D5 binds to the rat p185(neu). With the observation that 7.16.4 positively stains human breast cancer tissues that overexpress p185(her2/neu), 7.16.4 may be useful for the pathological diagnosis and therapy of human tumors.

Drebin, J. A., et al. (1986). “Inhibition of tumor growth by a monoclonal antibody reactive with an oncogene-encoded tumor antigen.” Proc Natl Acad Sci U S A 83(23): 9129-9133. PubMed

The neu oncogene encodes a 185-kDa transmembrane glycoprotein tumor antigen, termed p185. We have recently described a monoclonal antibody reactive with a cell surface domain of the p185 molecule. In vivo treatment with this anti-p185 monoclonal antibody was able to significantly inhibit the tumorigenic growth of neu-transformed NIH 3T3 cells implanted into nude mice. Such treatment had no effect on the tumorigenic growth of Ha-ras-transformed NIH 3T3 cells. Furthermore, anti-p185 antibody treatment was able to inhibit the growth of the rat neuroblastoma cells from which the neu oncogene was initially isolated. These results demonstrate that a monoclonal antibody reactive with the extracellular domain of an oncogene-encoded protein can exert a significant antitumor effect; such antibodies may prove useful in the therapy of certain malignancies.

Drebin, J. A., et al. (1984). “Monoclonal antibodies identify a cell-surface antigen associated with an activated cellular oncogene.” Nature 312(5994): 545-548. PubMed

A variety of antigens have been identified on the surface of the malignant cell. However, identical antigens are often found on non-malignant cells of the same or different histological origin, or of a different stage of embryonic development. Many of these tumour-associated antigens appear to be only incidentally expressed on neoplastic cells. Clearly, it would be of great interest to identify cell-surface antigens whose expression is associated specifically with the transformed state and linked directly with the mechanisms responsible for transformation. The detection of activated cellular oncogenes in human and animal cancer cells by the technique of DNA transfection has allowed the isolation of genetic elements which are thought to have a critical role in malignancy. Here, in an effort to identify cell-surface antigens associated with the neoplastic process, we have generated hybridomas which secrete monoclonal antibodies that react specifically with cell-surface determinants found on NIH 3T3 cells transformed by transfection with a group of rat neuroblastoma oncogenes. These antibodies bind to and immunoprecipitate a phosphoprotein of relative molecular mass 185,000 (185 K) from a DNA donor rat neuroblastoma and 13 independent rat neuroblastoma DNA transfectants. There was no antibody reactivity with normal NIH 3T3 cells or with NIH 3T3 cells transformed by various other agents.