Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-wq2xx Total loading time: 0 Render date: 2024-04-16T12:19:58.482Z Has data issue: false hasContentIssue false

79 - Targeting the EGFR family of receptor tyrosine kinases

from Part 4 - Pharmacologic targeting of oncogenic pathways

Published online by Cambridge University Press:  05 February 2015

Siyuan Zhang
Affiliation:
University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
Dihua Yu
Affiliation:
University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

General structure

Epidermal growth-factor receptor (EGFR) proteins or erythroblastic leukemia viral oncogene homolog (ERBB) proteins are a group of transmembrane receptors with intrinsic tyrosine kinase activity. The EGFR family is one of the most important groups of transmembrane cell surface receptors that integrate extra-cellular signals (e.g. growth factors, cytokines, and hormones) to drive multiple critical cellular processes, including cell proliferation, differentiation, and survival, via localized paracrine signals (1). EGFR family proteins are classified as subclass I receptor tyrosine kinases (RTKs; Figure 79.1), containing four structurally related RTKs (2): EGFR (also known as ERBB-1/HER1), ERBB-2 (HER2 in humans and Neu in rodents), ERBB-3 (HER3), and ERBB-4 (HER4). Each EGFR family receptor has an extra-cellular ligand-binding domain, a single α-helix transmembrane domain, an intra-cellular tyrosine kinase domain (with the exception of ERBB-3/HER3) and a cytoplasmic tail with tyrosine autophosphorylation sites (3; Figure 79.2). Although all four of these members share a similar domain structure, each has unique properties. EGFR (ERBB1) and ERBB4 have several known extra-cellular ligands and tyrosine-kinase activity on their cytoplasmic tail. ERBB2 has a similar active tyrosine-kinase domain. However, no direct ligand for ERBB2 has been identified. The other member, ERBB3 is characterized by a lack of tyrosine-kinase activity, although it binds to its extra-cellular ligands (4,5).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 843 - 853
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Hynes, NE, Lane, HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nature Reviews Cancer 2005;5:341–54.CrossRef
Yarden, Y, Ullrich, A. Growth factor receptor tyrosine kinases. Annual Review of Biochemistry 1988;57:443–78.CrossRef
Olayioye, MA, Neve, RM, Lane, HA, Hynes, NE. The ERBB signaling network: receptor heterodimerization in development and cancer. EMBO Journal 2000;19:3159–67.CrossRef
Yarden, Y, Sliwkowski, MX. Untangling the ERBB signalling network. Nature Reviews Molecular and Cellular Biology 2001;2:127–37.CrossRef
Pinkas-Kramarski, R, Soussan, L, Waterman, H, et al. Diversification of Neu differentiation factor and epidermal growth factor signaling by combinatorial receptor interactions. EMBO Journal 1996;15:2452–67.
Jones, JT, Akita, RW, Sliwkowski, MX. Binding specificities and affinities of egf domains for ERBB receptors. FEBS Letters 1999;447:227–31.CrossRef
Tzahar, E, Pinkas-Kramarski, R, Moyer, JD, et al. Bivalence of EGF-like ligands drives the ERBB signaling network. EMBO Journal 1997;16:4938–50.CrossRef
Graus-Porta, D, Beerli, RR, Daly, JM, Hynes, NE. ERBB-2, the preferred heterodimerization partner of all ERBB receptors, is a mediator of lateral signaling. EMBO Journal 1997;16:1647–55.CrossRef
Olayioye, MA, Graus-Porta, D, Beerli, RR, et al. ERBB-1 and ERBB-2 acquire distinct signaling properties dependent upon their dimerization partner. Molecular and Cellular Biology 1998;18:5042–51.CrossRef
Wong, AJ, Ruppert, JM, Bigner, SH, et al. Structural alterations of the epidermal growth factor receptor gene in human gliomas. Proceedings of the National Academy of Sciences USA 1992;89:2965–9.CrossRef
Lee, JW, Soung, YH, Kim, SY, et al. Somatic mutations of EGFR gene in squamous cell carcinoma of the head and neck. Clinical Cancer Research 2005;11:2879–82.CrossRef
Douglas, DA, Zhong, H, Ro, JY, et al. Novel mutations of epidermal growth factor receptor in localized prostate cancer. Frontiers in Bioscience 2006;11:2518–25.CrossRef
Batra, SK, Castelino-Prabhu, S, Wikstrand, CJ, et al. Epidermal growth factor ligand-independent, unregulated, cell-transforming potential of a naturally occurring human mutant EGFRvIII gene. Cell Growth and Differentiation 1995;6:1251–9.
Slamon, DJ, Clark, GM, Wong, SG, et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987;235:177–82.CrossRef
Meert, AP, Martin, B, Paesmans, M, et al. The role of HER-2/neu expression on the survival of patients with lung cancer: a systematic review of the literature. British Journal of Cancer 2003;89:959–65.CrossRefGoogle ScholarPubMed
Lee, JW, Soung, YH, Kim, SY, et al. ERBB2 kinase domain mutation in the lung squamous cell carcinoma. Cancer Letters 2006;237:89–94.CrossRef
Jaehne, J, Urmacher, C, Thaler, HT, et al. Expression of Her2/neu oncogene product p185 in correlation to clinicopathological and prognostic factors of gastric carcinoma. Journal of Cancer Research Clinical Oncology 1992;118:474–9.CrossRefGoogle ScholarPubMed
Di Lorenzo, G, Autorino, R, De Laurentiis, M, et al. HER-2/neu receptor in prostate cancer development and progression to androgen independence. Tumori 2004;90:163–70.CrossRef
Chen, H, Hung, MC.Involvement of co-activator p300 in the transcriptional regulation of the HER-2/neu gene. Journal of Biological Chemistry 1997;272:6101–4.CrossRefGoogle ScholarPubMed
Muraoka-Cook, RS, Sandahl, M, Husted, C, et al. The intracellular domain of ERBB4 induces differentiation of mammary epithelial cells. Molecular Biology of the Cell 2006;17:4118–29.CrossRef
Feng, SM, Sartor, CI, Hunter, D, et al. The HER4 cytoplasmic domain, but not its C terminus, inhibits mammary cell proliferation. Molecular Endocrinology 2007;21:1861–76.CrossRef
Arnould, L, Gelly, M, Penault-Llorca, F, et al. Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism?British Journal of Cancer 2006;94:259–67.CrossRefGoogle Scholar
Bier, H, Hoffmann, T, Haas, I, van Lierop, A. Anti-(epidermal growth factor) receptor monoclonal antibodies for the induction of antibody-dependent cell-mediated cytotoxicity against squamous cell carcinoma lines of the head and neck. Cancer Immunology, Immunotherapy 1998;46:167–73.CrossRef
Sato, JD, Kawamoto, T, Le, AD, et al. Biological effects in vitro of monoclonal antibodies to human epidermal growth factor receptors. Molecular Biology and Medicine 1983;1:511–29.
Goldstein, NI, Prewett, M, Zuklys, K, Rockwell, P, Mendelsohn, J. Biological efficacy of a chimeric antibody to the epidermal growth factor receptor in a human tumor xenograft model. Clinical Cancer Research 1995;1:1311–18.
Bonner, JA, Harari, PM, Giralt, J, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. New England Journal of Medicine 2006;354:567–78.CrossRefGoogle ScholarPubMed
Vermorken, JB, Mesia, R, Rivera, F, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. New England Journal of Medicine 2008;359:1116–27.CrossRefGoogle ScholarPubMed
Pirker, R, Pereira, JR, Szczesna, A, et al. Cetuximab plus chemotherapy in patients with advanced non-small-cell lung cancer (FLEX): an open-label randomised Phase III trial. Lancet 2009;373:1525–31.CrossRef
Drebin, JA, Link, VC, Greene, MI. Monoclonal antibodies reactive with distinct domains of the neu oncogene-encoded p185 molecule exert synergistic anti-tumor effects in vivo. Oncogene 1988;2:273–7.
Hudziak, RM, Lewis, GD, Winget, M, et al. p185HER2 monoclonal antibody has antiproliferative effects in vitro and sensitizes human breast tumor cells to tumor necrosis factor. Molecular and Cellular Biology 1989;9:1165–72.CrossRef
Shalaby, MR, Shepard, HM, Presta, L, et al. Development of humanized bispecific antibodies reactive with cytotoxic lymphocytes and tumor cells overexpressing the HER2 protooncogene. Journal of Experimental Medicine 1992;175:217–25.CrossRefGoogle ScholarPubMed
Pegram, MD, Lipton, A, Hayes, DF, et al. Phase II study of receptor-enhanced chemosensitivity using recombinant humanized anti-p185HER2/neu monoclonal antibody plus cisplatin in patients with HER2/neu-overexpressing metastatic breast cancer refractory to chemotherapy treatment. Journal of Clinical Oncology 1998;16:2659–71.CrossRefGoogle ScholarPubMed
Slamon, DJ, Leyland-Jones, B, Shak, S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. New England Journal of Medicine 2001;344:783–92.CrossRefGoogle Scholar
Cho, HS, Mason, K, Ramyar, KX, et al. Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature 2003;421:756–60.CrossRef
Nagata, Y, Lan, KH, Zhou, X, et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell 2004;6:117–27.CrossRef
Lane, HA, Beuvink, I, Motoyama, AB, et al. ERBB2 potentiates breast tumor proliferation through modulation of p27(Kip1)-Cdk2 complex formation: receptor overexpression does not determine growth dependency. Molecular and Cellular Biology 2000;20:3210–23.CrossRef
Le, XF, Claret, FX, Lammayot, A, et al. The role of cyclin-dependent kinase inhibitor p27Kip1 in anti-HER2 antibody-induced G1 cell cycle arrest and tumor growth inhibition. Journal of Biological Chemistry 2003;278:23 441–50.CrossRefGoogle ScholarPubMed
Wehrman, TS, Raab, WJ, Casipit, CL, et al. A system for quantifying dynamic protein interactions defines a role for Herceptin in modulating ERBB2 interactions. Proceedings of the National Academy of Sciences USA 2006;103:19 063–8.
Clynes, RA, Towers, TL, Presta, LG, Ravetch, JV. Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nature Medicine 2000;6:443–6.CrossRef
Spicer, J. Technology evaluation: pertuzumab, Roche/Genentech/Chugai. Current Opinion in Molecular Therapy 2004;6:337–43.
Yao, E, Zhou, W, Lee-Hoeflich, ST, et al. Suppression of HER2/HER3-mediated growth of breast cancer cells with combinations of GDC-0941 PI3K inhibitor, trastuzumab, and pertuzumab. Clinical Cancer Research 2009;15:4147–56.CrossRef
Pohl, M, Stricker, I, Schoeneck, A, et al. Antitumor activity of the HER2 dimerization inhibitor pertuzumab on human colon cancer cells in vitro and in vivo. Journal of Cancer Research Clinical Oncology 2009;135:1377–86.CrossRefGoogle ScholarPubMed
Hughes, JB, Berger, C, Rodland, MS, et al. Pertuzumab increases epidermal growth factor receptor down-regulation by counteracting epidermal growth factor receptor-ERBB2 heterodimerization. Molecular Cancer Therapeutics 2009;8:1885–92.CrossRef
Lewis, PGD, Li, G, Dugger, DL, et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Research 2008;68:9280–90.CrossRef
Mandler, R, Kobayashi, H, Hinson, ER, Brechbiel, MW, Waldmann, TA. Herceptin-geldanamycin immunoconjugates: pharmacokinetics, biodistribution, and enhanced antitumor activity. Cancer Research 2004;64:1460–7.CrossRef
Senter, PD. Potent antibody drug conjugates for cancer therapy. Current Opinion in Chemical Biology 2009;13:235–44.CrossRef
Kiewe, P, Hasmuller, S, Kahlert, S, et al. Phase I trial of the trifunctional anti-HER2 x anti-CD3 antibody ertumaxomab in metastatic breast cancer. Clinical Cancer Research 2006;12:3085–91.CrossRef
Jager, M, Schoberth, A, Ruf, P, Hess, J, Lindhofer, H. The trifunctional antibody ertumaxomab destroys tumor cells that express low levels of human epidermal growth factor receptor 2. Cancer Research 2009;69:4270–6.CrossRef
Bostrom, J, Yu, SF, Kan, D, et al. Variants of the antibody herceptin that interact with HER2 and VEGF at the antigen binding site. Science 2009;323:1610–14.CrossRef
Schaefer, G, Haber, L, Crocker, LM, et al. A two-in-one antibody against HER3 and EGFR has superior inhibitory activity compared with monospecific antibodies. Cancer Cell 2011;20:472–86.CrossRef
Zhang, J, Yang, PL, Gray, NS. Targeting cancer with small molecule kinase inhibitors. Nature Reviews Cancer 2009;9:28–39.CrossRef
Traxler, P, Furet, P. Strategies toward the design of novel and selective protein tyrosine kinase inhibitors. Pharmacology and Therapeutics 1999;82:195–206.CrossRef
Janne, PA, Gray, N, Settleman, J. Factors underlying sensitivity of cancers to small-molecule kinase inhibitors. Nature Reviews Drug Discovery 2009;8:709–23.CrossRef
Reck, M. Gefitinib in the treatment of advanced non-small-cell lung cancer. Expert Review of Anticancer Therapy 2009;9:401–12.CrossRef
Weinstein, IB. Cancer. Addiction to oncogenes: the Achilles heal of cancer. Science 2002;297:63–4.CrossRef
Sharma, SV, Settleman, J. Oncogene addiction: setting the stage for molecularly targeted cancer therapy. Genes and Development 2007;21:3214–31.CrossRef
Lynch, TJ, Bell, DW, Sordella, R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. New England Journal of Medicine 2004;350:2129–39.CrossRefGoogle ScholarPubMed
Sequist, LV, Bell, DW, Lynch, TJ, Haber, DA.Molecular predictors of response to epidermal growth factor receptor antagonists in non-small-cell lung cancer. Journal of Clinical Oncology 2007;25:587–95.CrossRefGoogle ScholarPubMed
Paez, JG, Janne, PA, Lee, JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497–500.CrossRef
Montemurro, F, Valabrega, G, Aglietta, M. Lapatinib: a dual inhibitor of EGFR and HER2 tyrosine kinase activity. Expert Opinion on Biological Therapy 2007;7:257–68.CrossRef
Wood, ER, Truesdale, AT, McDonald, OB, et al. A unique structure for epidermal growth factor receptor bound to GW572016 (Lapatinib): relationships among protein conformation, inhibitor off-rate, and receptor activity in tumor cells. Cancer Research 2004;64:6652–9.CrossRef
Geyer, CE, Forster, J, Lindquist, D, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. New England Journal of Medicine 2006;355:2733–43.CrossRefGoogle ScholarPubMed
Scaltriti, M, Rojo, F, Ocana, A, et al. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. Journal of the National Cancer Institute 2007;99:628–38.CrossRefGoogle ScholarPubMed
Miettinen, PJ, Berger, JE, Meneses, J, et al. Epithelial immaturity and multiorgan failure in mice lacking epidermal growth factor receptor. Nature 1995;376:337–41.CrossRef
Sibilia, M, Wagner, EF. Strain-dependent epithelial defects in mice lacking the EGF receptor. Science 1995;269:234–8.CrossRef
Sipples, R. Common side effects of anti-EGFR therapy: acneform rash. Seminars in Oncology Nursing 2006;22:28–34.CrossRef
Agero, AL, Dusza, SW, Benvenuto-Andrade, C, et al. Dermatologic side effects associated with the epidermal growth factor receptor inhibitors. Journal of the American Academy of Dermatology 2006;55:657–70.CrossRefGoogle ScholarPubMed
Camus, P, Kudoh, S, Ebina, M.Interstitial lung disease associated with drug therapy. British Journal of Cancer 2004;91:S18–23.CrossRefGoogle ScholarPubMed
Lee, KF, Simon, H, Chen, H, et al. Requirement for neuregulin receptor erbB2 in neural and cardiac development. Nature 1995;378:394–8.CrossRef
Crone, SA, Zhao, YY, Fan, L, et al. ERBB2 is essential in the prevention of dilated cardiomyopathy. Nature Medicine 2002;8:459–65.CrossRef
Schneider, JW, Chang, AY, Garratt, A. Trastuzumab cardiotoxicity: speculations regarding pathophysiology and targets for further study. Seminars in Oncology 2002;29:22–8.CrossRef
Azim, H, Azim, HA, Escudier, B. Trastuzumab versus lapatinib: The cardiac side of the story. Cancer Treatment Reviews 2009;35:633–8.CrossRef
Blencke, S, Zech, B, Engkvist, O, et al. Characterization of a conserved structural determinant controlling protein kinase sensitivity to selective inhibitors. Chemical Biology 2004;11:691–701.CrossRef
Cappuzzo, F, Varella-Garcia, M, Finocchiaro, G, et al. Primary resistance to cetuximab therapy in EGFR FISH-positive colorectal cancer patients. British Journal of Cancer 2008;99:83–9.CrossRefGoogle ScholarPubMed
Normanno, N, Tejpar, S, Morgillo, F, et al. Implications for KRAS status and EGFR-targeted therapies in metastatic CRC. Nature Reviews Clinical Oncology 2009;6:519–27.CrossRef
Pao, W, Miller, VA, Politi, KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Medicine 2005;2:e73.
Kobayashi, S, Ji, H, Yuza, Y, et al. An alternative inhibitor overcomes resistance caused by a mutation of the epidermal growth factor receptor. Cancer Research 2005;65:7096–101.CrossRef
Kuang, Y, Rogers, A, Yeap, BY, et al. Noninvasive detection of EGFR T790M in gefitinib or erlotinib resistant non-small cell lung cancer. Clinical Cancer Research 2009;15:2630–6.CrossRef
Wheeler, DL, Huang, S, Kruser, TJ, et al. Mechanisms of acquired resistance to cetuximab: role of HER (ERBB) family members. Oncogene 2008;27:3944–56.CrossRef
Engelman, JA, Zejnullahu, K, Mitsudomi, T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007;316:1039–43.CrossRef
Guix, M, Faber, AC, Wang, SE, et al. Acquired resistance to EGFR tyrosine kinase inhibitors in cancer cells is mediated by loss of IGF-binding proteins. Journal of Clinical Investigation 2008;118:2609–19.Google ScholarPubMed
Lan, KH, Lu, CH, Yu, D. Mechanisms of trastuzumab resistance and their clinical implications. Annals of the New York Academy of Science 2005;1059:70–5.CrossRef
Berns, K, Horlings, HM, Hennessy, BT, et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell 2007;12:395–402.CrossRef
Esteva, FJ, Guo, H, Zhang, S, et al. PTEN, PIK3CA, p-AKT, and p-p70S6K status: association with trastuzumab response and survival in patients with HER2-positive metastatic breast cancer. American Journal of Pathology 2010;177:1647–56.CrossRefGoogle ScholarPubMed
Moulder, SL, Yakes, FM, Muthuswamy, SK, et al. Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer Research 2001;61:8887–95.
Lu, Y, Zi, X, Zhao, Y, Mascarenhas, D, Pollak, M.Insulin-like growth factor-I receptor signaling and resistance to trastuzumab (Herceptin). Journal of the National Cancer Institute 2001;93:1852–7.CrossRefGoogle Scholar
Shattuck, DL, Miller, JK, Carraway, KL, Sweeney, C. Met receptor contributes to trastuzumab resistance of Her2-overexpressing breast cancer cells. Cancer Research 2008;68:1471–7.CrossRef
Nahta, R, Yuan, LX, Zhang, B, Kobayashi, R, Esteva, FJ. Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Research 2005;65:11 118–28.
Gong, C, Yao, Y, Wang, Y, et al. Up-regulation of miR-21 mediates resistance to trastuzumab therapy for breast cancer. Journal of Biological Chemistry 2011;286:19 127–37.CrossRefGoogle ScholarPubMed
Scaltriti, M, Eichhorn, PJ, Cortes, J, et al. Cyclin E amplification/overexpression is a mechanism of trastuzumab resistance in HER2+ breast cancer patients. Proceedings of the National Academy of Sciences USA 2011;108:3761–6.CrossRef
Zhang, S, Huang, WC, Li, P, et al. Combating trastuzumab resistance by targeting SRC, a common node downstream of multiple resistance pathways. Nature Medicine 2011;17:461–9.CrossRef
Liang, K, Esteva, FJ, Albarracin, C, et al. Recombinant human erythropoietin antagonizes trastuzumab treatment of breast cancer cells via Jak2-mediated Src activation and PTEN inactivation. Cancer Cell 2010;18:423–35.CrossRef
Eichhorn, PJ, Gili, M, Scaltriti, M, et al. Phosphatidylinositol 3-kinase hyperactivation results in lapatinib resistance that is reversed by the mTOR/phosphatidylinositol 3-kinase inhibitor NVP-BEZ235. Cancer Research 2008;68:9221–30.CrossRef
Xia, W, Bacus, S, Hegde, P, et al. A model of acquired autoresistance to a potent ERBB2 tyrosine kinase inhibitor and a therapeutic strategy to prevent its onset in breast cancer. Proceedings of the National Academy of Sciences USA 2006;103:7795–800.CrossRef
Narayan, M, Wilken, JA, Harris, LN, et al. Trastuzumab-induced HER reprogramming in “resistant” breast carcinoma cells. Cancer Research 2009;69:2191–4.CrossRef
Piccart, M. Circumventing de novo and acquired resistance to trastuzumab: new hope for the care of ERBB2-positive breast cancer. Clinical Breast Cancer 2008;8:S100–13.
Rexer, BN, Ham, AJ, Rinehart, C, et al. Phosphoproteomic mass spectrometry profiling links Src family kinases to escape from HER2 tyrosine kinase inhibition. Oncogene 2011;30:4163–74.CrossRef
Lu, CH, Wyszomierski, SL, Tseng, LM, et al. Preclinical testing of clinically applicable strategies for overcoming trastuzumab resistance caused by PTEN deficiency. Clinical Cancer Research 2007;13:5883–8.CrossRef
Morrow, PK, Wulf, GM, Ensor, J, et al. Phase I/II study of trastuzumab in combination with everolimus (RAD001) in patients with HER2-overexpressing metastatic breast cancer who progressed on trastuzumab-based therapy. Journal of Clinical Oncology 2011;29:3126–32.CrossRefGoogle ScholarPubMed
Serra, V, Markman, B, Scaltriti, M, et al. NVP-BEZ235, a dual PI3K/mTOR inhibitor, prevents PI3K signaling and inhibits the growth of cancer cells with activating PI3K mutations. Cancer Research 2008;68:8022–30.CrossRef
Engelman, JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nature Reviews Cancer 2009;9:550–62.CrossRef
Ueno, NT, Yu, D, Hung, MC. E1A: tumor suppressor or oncogene? Preclinical and clinical investigations of E1A gene therapy. Breast Cancer 2001;8:285–93.CrossRef
Renard, V, Leach, DR. Perspectives on the development of a therapeutic HER-2 cancer vaccine. Vaccine 2007;25:B17–23.
Satyanarayanajois, S, Villalba, S, Jianchao, L, Lin, GM. Design, synthesis, and docking studies of peptidomimetics based on HER2-herceptin binding site with potential antiproliferative activity against breast cancer cell lines. Chemical Biology and Drug Design 2009;74:246–57.CrossRef
Neckers, L, Neckers, K. Heat-shock protein 90 inhibitors as novel cancer chemotherapeutics: an update. Expert Opinion on Emerging Drugs 2005;10:137–49.CrossRef
Munster, PN, Marchion, DC, Basso, AD, Rosen, N. Degradation of HER2 by ansamycins induces growth arrest and apoptosis in cells with HER2 overexpression via a HER3, phosphatidylinositol 3ʹ-kinase-AKT-dependent pathway. Cancer Research 2002;62:3132–7.

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×