跳转至内容
Merck
CN

Epidermal growth factor receptor (EGFR), its family members Her-2/ErbB-2, Her-3, Her-4 and their ligands, are involved in over 70% of all cancers. EGFR itself has been implicated in ~30% of all solid human tumors. EGFR is associated not only with the proliferation of tumor cells, but also with enhanced tumor cell survival, angiogenesis and metastatic spread. The enhanced activity of the EGFR due to over-expression, co-expression of the receptor and its ligands, as well as activating mutations is the hallmark of many human carcinomas.

The co-expression of the EGFR and its ligands, especially TGFα and EGF, plays a key role in EGFR-mediated tumorigenesis. EGFR expression is a prognostic indicator, predicting poor survival and indicating an advanced state of the disease. When EGFR is co-expressed with other members of the Her family, the various combinations of Her dimers confer different degrees of malignancy. It has been noted that the co-expression of EGFR with Her-2 and Her-3 is associated with more aggressive clinical behavior. In many types of tumor, including lung, breast, prostate, ovary, gastrointestinal tract and brain, the EGFR receptor is expressed approximately 100 times the normal number of EGF receptors found on the surface of normal cells. Furthermore, expression of high levels of these two receptors in nonmalignant cell lines, either alone or in combination, leads to a transformed phenotype.

Due to all these observations, it is no surprise that EGFR and Her-2/Erb-2 were identified early on as important targets for drug development. Indeed, the first signal transduction therapeutic agent introduced into the clinic was Herceptin, an anti-Her-2 antibody, followed closely by the protein tyrosine kinase inhibitors Iressa (ZD 1839) and Tarceva (OSI-774) and the anti-EGFR antibody Erbitux (mAb 225).

The enhanced activity of the EGFR is due to a number of molecular events. Most common is the overexpression of the receptor along with the expression of the EGFR receptor ligands like TGFα, EGF, amphiregulin and HB-EGF, leading to persistent autocrine stimulation. Another common occurrence is an activating mutation resulting from deletion of exons 2 through 7, leading to a persistently active receptor Δ (2-7)EGFR (also known as EFFRvIII) in the absence of a ligand. The emergence of this mutation occurs in the most aggressive forms of EGFR overexpressing tumors.

Activation of the EGFR pathway is not limited to members of the EGFR family, but frequently occurs due to the transactivation by other signaling pathways such as mitogenic G protein-coupled receptors and the PDGF receptor. Furthermore, the EGFR pathway cooperates in a synergistic manner with pp60c-Src, and the deletion of PTEN, the negative regulator of PKB/Akt. The frequent involvement of EGFR in human tumors has identified it as a target for novel therapies. The first breakthrough was the development of selective EGFR kinase inhibitors (tyrphostins) like tyrphostins, AG 1478 and ZD 1839 (Iressa). Iressa is one of the two kinase inhibitors (the other being Gleevec) to receive approval for clinical application. It is important to note that it was recently found that the response of patients suffering from non-small-cell lung carcinoma to Iressa, is limited to the 7-10% harboring mutations in the kinase domain of the receptor. Other inhibitors, similar to Iressa, like Tarceva (OSI-774), the pan-Her reversible inhibitor GW 2016, the irreversible inhibitor CI-1033, which targets both RGFR and Her-2, are in the pipeline. Antibodies to the EGFR, like Erbitux and TGFα fused to a mutated form of pseudomonas exotoxin, TP-38, have also undergone clinical development.

The Table below contains accepted modulators and additional information. 

Materials
Loading

References

1.
Alroy I, Yarden Y. 1997. The ErbB signaling network in embryogenesis and oncogenesis: signal diversification through combinatorial ligand-receptor interactions. 410(1):83-86. https://doi.org/10.1016/s0014-5793(97)00412-2
2.
Bartolotti M, Franceschi E, Brandes AA. 2012. EGF receptor tyrosine kinase inhibitors in the treatment of brain metastases from non-small-cell lung cancer. Expert Review of Anticancer Therapy. 12(11):1429-1435. https://doi.org/10.1586/era.12.121
3.
Blagosklonny MV. 2004. Gefitinib (Iressa) in Oncogene-Addictive Cancers and Therapy for Common Cancers. Cancer Biology & Therapy. 3(5):436-440. https://doi.org/10.4161/cbt.3.5.984
4.
Grünwald V, Hidalgo M. 2003. Development of The Epidermal Growth Factor Receptor Inhibitor Tarcevatm(Osi-774).235-246. https://doi.org/10.1007/978-1-4615-0081-0_19
5.
He H, Levitzki A, Zhu H, Walker F, Burgess A, Maruta H. 2001. Platelet-derived Growth Factor Requires Epidermal Growth Factor Receptor to Activate p21-activated Kinase Family Kinases. J. Biol. Chem.. 276(29):26741-26744. https://doi.org/10.1074/jbc.c100229200
6.
Huang HS, Nagane M, Klingbeil CK, Lin H, Nishikawa R, Ji X, Huang C, Gill GN, Wiley HS, Cavenee WK. 1997. The Enhanced Tumorigenic Activity of a Mutant Epidermal Growth Factor Receptor Common in Human Cancers Is Mediated by Threshold Levels of Constitutive Tyrosine Phosphorylation and Unattenuated Signaling. J. Biol. Chem.. 272(5):2927-2935. https://doi.org/10.1074/jbc.272.5.2927
7.
Jamal-Hanjani M, Spicer J. 2012. Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in the Treatment of Epidermal Growth Factor Receptor-Mutant Non-Small Cell Lung Cancer Metastatic to the Brain. Clinical Cancer Research. 18(4):938-944. https://doi.org/10.1158/1078-0432.ccr-11-2529
8.
Kim T, Murren J. 2002. Erlotinib OSI/Roche/Genentech. Curr. Opin. Investig.. Drugs(3):1385-1395.
9.
Leserer, Andreas Gschwind, Axel Ull M. 2000. Epidermal Growth Factor Receptor Signal Transactivation. IUBMB Life (International Union of Biochemistry and Molecular Biology: Life). 49(5):405-409. https://doi.org/10.1080/152165400410254
10.
Levitzki A, Gazit A. 1995. Tyrosine kinase inhibition: an approach to drug development. Science. 267(5205):1782-1788. https://doi.org/10.1126/science.7892601
11.
Mendelsohn J. 2001. The epidermal growth factor receptor as a target for cancer therapy..3-9. https://doi.org/10.1677/erc.0.0080003
12.
Nakata A, Gotoh N. 2012. Recent understanding of the molecular mechanisms for the efficacy and resistance of EGF receptor-specific tyrosine kinase inhibitors in non-small cell lung cancer. Expert Opinion on Therapeutic Targets. 16(8):771-781. https://doi.org/10.1517/14728222.2012.697155
13.
Norman P. 2002. ZD-1839 (AstraZeneca). Curr. Opin. Investig. Drugs. 2428-434.
14.
Okines A, Cunningham D, Chau I. 2011. Targeting the human EGFR family in esophagogastric cancer. Nat Rev Clin Oncol. 8(8):492-503. https://doi.org/10.1038/nrclinonc.2011.45
15.
Osherov N, Levitzki A. 1994. Epidermal-Growth-Factor-Dependent Activation of the Src-Family Kinases. Eur J Biochem. 225(3):1047-1053. https://doi.org/10.1111/j.1432-1033.1994.1047b.x
16.
Settleman J. 2004. Inhibition of Mutant EGF Receptors by Gefitinib: Targeting an Achilles? Heel of Lung Cancer. Cell Cycle. 3(12):1496-1497. https://doi.org/10.4161/cc.3.12.1325
17.
Sgambato A, Casaluce F, Maione P, Rossi A, Rossi E, Napolitano A, Palazzolo G, A. Bareschino M, Schettino C, C. Sacco P, et al. 2012. The Role of EGFR Tyrosine Kinase Inhibitors in the First-Line Treatment of Advanced Non Small Cell Lung Cancer Patients Harboring EGFR Mutation. curr med chem. 19(20):3337-3352. https://doi.org/10.2174/092986712801215973
18.
Uitdehaag JC, Verkaar F, Alwan H, de Man J, Buijsman RC, Zaman GJ. 2012. A guide to picking the most selective kinase inhibitor tool compounds for pharmacological validation of drug targets. 166(3):858-876. https://doi.org/10.1111/j.1476-5381.2012.01859.x
登录以继续。

如要继续阅读,请登录或创建帐户。

暂无帐户?