Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-r6qrq Total loading time: 0 Render date: 2024-04-28T21:33:18.703Z Has data issue: false hasContentIssue false

18 - PI3K

from Part 2.1 - Molecular pathways underlying carcinogenesis: signal transduction

Published online by Cambridge University Press:  05 February 2015

Kevin D. Courtney
Affiliation:
Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX, USA
Lewis C. Cantley
Affiliation:
Weill Cornell Cancer Center, New York – Presbyterian Hospital, Weill Cornell Medical College, New York, NY, 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

Introduction

Phosphatidylinositol 3-kinase (PI3K) was first discovered in the 1980s through the association of its enzymatic activity with viral oncoproteins (1–5). Since that time, a firm connection between PI3K and cancer has been established. Components of the PI3K signaling pathway are among the most frequently altered in human cancer, leading to deregulation of a signaling cascade that is central to normal cell metabolism, proliferation, motility, and survival (6,7). Intact PI3K signaling is also critical for the processes of angiogenesis and anti-tumor immune surveillance that support and combat tumor growth, respectively (8–10). The enzymes involved in this pathway consequently have become attractive targets for cancer therapy. It is therefore crucial that we examine the diverse roles that components of PI3K signaling play if we are to optimally target these enzymes for therapeutic gain.

PI3K family members

Mammalian PI3K is comprised of three classes of lipid kinases (11). These evolved from a single enzyme that is conserved in all eukaryotes and was first described in yeast as vacuolar protein-sorting defective 34 (Vps34), corresponding to class III PI3K in mammals (6,12). Class III PI3K catalyzes the phosphorylation of phosphatidylinositol (PI) to phophatidylinositol-3-phosphate (PI-3-P; 6,12). Three genes encode isoforms of class II PI3K, which converts PI to PI-3-P and PI-4-P to PI-3,4-P2. Class II PI3Ks have been proposed to be involved in membrane trafficking (6). Class I PI3Ks include both a catalytic and a separate regulatory subunit and catalyze the phosphorylation of PI-4,5-P2 to PI-3,4,5-P3. Class I PI3Ks are further categorized into class IA and class IB enzymes. For class IA PI3K, the genes PIK3R1, PIK3R2, and PIK3R3 encode the regulatory subunits p85α (p85α, p55α, and p50α isoforms), p85β, and p55γ, respectively, which are referred to collectively as p85 (6,7,12). The catalytic isoforms of class IA PI3K, p110α, p110β, and p110δ, are the products of PIK3CA, PIK3CB, and PIK3CD genes, respectively. The p110α and -β isoforms are ubiquitously expressed in mammals, while p110δ expression is predominantly leukocyte-restricted (13). The catalytic subunit p110γ, which is also leukocyte-restricted, and the regulatory subunits p101, p84, or p87PIKAP, constitute Class IB PI3K (6,13). Unlike the catalytic subunits of class IA PI3Ks, p110γ can be active without binding to its regulatory subunit (14). Class IA PI3Ks are the most widely studied in mammalian systems and have been directly linked to cancer.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 218 - 230
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

Kaplan, DR, Whitman, M, Schaffhausen, B, et al. Common elements in growth factor stimulation and oncogenic transformation: 85 kd phosphoprotein and phosphatidylinositol kinase activity. Cell. 1987;50:1021–9.CrossRef
Kaplan, DR, Whitman, M, Schaffhausen, B, et al. Phosphatidylinositol metabolism and polyoma-mediated transformation. Proceedings of the National Academy of Sciences USA 1986;83:3624–8.CrossRef
Sugimoto, Y, Whitman, M, Cantley, LC, Erikson, RL. Evidence that the Rous sarcoma virus transforming gene product phosphorylates phosphatidylinositol and diacylglycerol. Proceedings of the National Academy of Sciences USA 1984;81:2117–21.CrossRef
Whitman, M, Downes, CP, Keeler, M, Keller, T, Cantley, L. Type I phosphatidylinositol kinase makes a novel inositol phospholipid, phosphatidylinositol-3-phosphate. Nature. 1988;332:644–6.CrossRef
Whitman, M, Kaplan, DR, Schaffhausen, B, Cantley, L, Roberts, TM. Association of phosphatidylinositol kinase activity with polyoma middle-T competent for transformation. Nature. 1985;315:239–42.CrossRef
Engelman, JA, Luo, J, Cantley, LC. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nature Reviews Genetics 2006;7:606–19.CrossRef
Yuan, TL, Cantley, LC. PI3K pathway alterations in cancer: variations on a theme. Oncogene. 2008;27:5497–510.CrossRef
Graupera, M, Guillermet-Guibert, J, Foukas, LC, et al. Angiogenesis selectively requires the p110alpha isoform of PI3K to control endothelial cell migration. Nature. 2008;453:662–6.CrossRef
Yuan, TL, Choi, HS, Matsui, A, et al. Class 1A PI3K regulates vessel integrity during development and tumorigenesis. Proceedings of the National Academy of Sciences USA 2008;105:9739–44.CrossRef
Zebedin, E, Simma, O, Schuster, C, et al. Leukemic challenge unmasks a requirement for PI3Kdelta in NK cell-mediated tumor surveillance. Blood. 2008;112:4655–64.CrossRef
Walker, EH, Perisic, O, Ried, C, Stephens, L, Williams, RL. Structural insights into phosphoinositide 3-kinase catalysis and signalling. Nature. 1999;402:313–20.CrossRef
Katso, R, Okkenhaug, K, Ahmadi, K, et al. Cellular function of phosphoinositide 3-kinases: implications for development, homeostasis, and cancer. Annual Review of Cell and Developmental Biology 2001;17:615–75.CrossRef
Kang, S, Denley, A, Vanhaesebroeck, B, Vogt, PK. Oncogenic transformation induced by the p110beta, -gamma, and -delta isoforms of class I phosphoinositide 3-kinase. Proceedings of the National Academy of Sciences USA 2006;103:1289–94.CrossRef
Huang, CH, Mandelker, D, Schmidt-Kittler, O, et al. The structure of a human p110alpha/p85alpha complex elucidates the effects of oncogenic PI3Kalpha mutations. Science 2007;318:1744–8.CrossRef
Miled, N, Yan, Y, Hon, WC, et al. Mechanism of two classes of cancer mutations in the phosphoinositide 3-kinase catalytic subunit. Science. 2007;317:239–42.CrossRef
Vanhaesebroeck, B, Waterfield, MD. Signaling by distinct classes of phosphoinositide 3-kinases. Experimental Cell Research 1999;253:239–54.CrossRef
Luo, J, Cantley, LC. The negative regulation of phosphoinositide 3-kinase signaling by p85 and its implication in cancer. Cell Cycle 2005;4:1309–12.CrossRef
Yu, J, Zhang, Y, McIlroy, J, et al. Regulation of the p85/p110 phosphatidylinositol 3’-kinase: stabilization and inhibition of the p110alpha catalytic subunit by the p85 regulatory subunit. Molecular and Cellular Biology 1998;18:1379–87.CrossRef
Carpenter, CL, Auger, KR, Chanudhuri, M, et al. Phosphoinositide 3-kinase is activated by phosphopeptides that bind to the SH2 domains of the 85-kDa subunit. Journal of Biological Chemistry 1993;268:9478–83.Google ScholarPubMed
Songyang, Z, Shoelson, SE, Chaudhuri, M, et al. SH2 domains recognize specific phosphopeptide sequences. Cell1993;72:767–78.
Ueki, K, Fruman, DA, Brachmann, SM, et al. Molecular balance between the regulatory and catalytic subunits of phosphoinositide 3-kinase regulates cell signaling and survival. Molecular and Cellular Biology 2002;22:965–77.CrossRef
Ueki, K, Fruman, DA, Yballe, CM, et al. Positive and negative roles of p85 alpha and p85 beta regulatory subunits of phosphoinositide 3-kinase in insulin signaling. Journal of Biological Chemistry 2003;278:48 453–66.CrossRefGoogle ScholarPubMed
Pacold, ME, Suire, S, Perisic, O, et al. Crystal structure and functional analysis of Ras binding to its effector phosphoinositide 3-kinase gamma. Cell 2000;103:931–43.CrossRef
Nolte, RT, Eck, MJ, Schlessinger, J, Shoelson, SE, Harrison, SC. Crystal structure of the PI 3-kinase p85 amino-terminal SH2 domain and its phosphopeptide complexes. Natural Structural Biology 1996;3:364–74.CrossRef
Escobedo, JA, Navankasattusas, S, Kavanaugh, WM, et al. cDNA cloning of a novel 85 kd protein that has SH2 domains and regulates binding of PI3-kinase to the PDGF beta-receptor. Cell 1991;65:75–82.CrossRef
Levy-Toledano, R, Blaettler, DH, LaRochelle, WJ, Taylor, SI. Insulin-induced activation of phosphatidylinositol (PI) 3-kinase. Insulin-induced phosphorylation of insulin receptors and insulin receptor substrate-1 displaces phosphorylated platelet-derived growth factor receptors from binding sites on PI 3-kinase. Journal of Biological Chemistry 1995;270:30 018–22.Google ScholarPubMed
Sun, XJ, Rothenberg, P, Kahn, CR, et al. Structure of the insulin receptor substrate IRS-1 defines a unique signal transduction protein. Nature 1991;352:73–7.CrossRef
Engelman, JA, Janne, PA, Mermel, C, et al. ERBB-3 mediates phosphoinositide 3-kinase activity in gefitinib-sensitive non-small cell lung cancer cell lines. Proceedings of the National Academy of Sciences USA 2005;102:3788–93.CrossRef
Sithanandam, G, Smith, GT, Fields, JR, Fornwald, LW, Anderson, LM. Alternate paths from epidermal growth factor receptor to Akt in malignant versus nontransformed lung epithelial cells: ERBB3 versus Gab1. American Journal of Respiratory Cell and Molecular Biology 2005;33:490–9.CrossRefGoogle ScholarPubMed
Soltoff, SP, Carraway, KL, Prigent, SA, Gullick, WG, Cantley, LC. ERBB3 is involved in activation of phosphatidylinositol 3-kinase by epidermal growth factor. Molecular and Cellular Biology 1994;14:3550–8.CrossRef
Engelman, JA, Cantley, LC. The role of the ERBB family members in non-small cell lung cancers sensitive to epidermal growth factor receptor kinase inhibitors. Clinical Cancer Research 2006;12:4372s-6s.
Junttila, TT, Akita, RW, Parsons, K, et al. Ligand-independent HER2/HER3/PI3K complex is disrupted by trastuzumab and is effectively inhibited by the PI3K inhibitor GDC-0941. Cancer Cell 2009;15:429–40.CrossRef
Shaw, RJ, Cantley, LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature 2006;441:424–30.CrossRef
Zhao, L, Vogt, PK. Class I PI3K in oncogenic cellular transformation. Oncogene 2008;27:5486–96.CrossRef
Gewinner, C, Wang, ZC, Richardson, A, et al. Evidence that inositol polyphosphate 4-phosphatase type II is a tumor suppressor that inhibits PI3K signaling. Cancer Cell 2009;16:115–25.CrossRef
Cantley, LC. The phosphoinositide 3-kinase pathway. Science 2002;296:1655–7.CrossRef
Alessi, DR, James, SR, Downes, CP, et al. Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha. Current Biology 1997;7:261–9.CrossRef
Currie, RA, Walker, KS, Gray, A, et al. Role of phosphatidylinositol 3,4,5-trisphosphate in regulating the activity and localization of 3-phosphoinositide-dependent protein kinase-1. Biochemical Journal 1999;337 (Pt ):575–83.
Majumder, PK, Sellers, WR. Akt-regulated pathways in prostate cancer. Oncogene 2005;24:7465–74.CrossRef
Milburn, CC, Deak, M, Kelly, SM, et al. Binding of phosphatidylinositol 3,4,5-trisphosphate to the pleckstrin homology domain of protein kinase B induces a conformational change. Biochemical Journal 2003;375:531–8.CrossRef
Huang, J, Manning, BD. The TSC1-TSC2 complex: a molecular switchboard controlling cell growth. Biochemical Journal 2008;412:179–90.CrossRef
Duronio, V. The life of a cell: apoptosis regulation by the PI3K/PKB pathway. Biochemical Journal 2008;415:333–44.CrossRef
Hresko, RC, Mueckler, M. mTOR.RICTOR is the Ser473 kinase for Akt/protein kinase B in 3T3-L1 adipocytes. Journal of Biological Chemistry 2005;280:40 406–16.CrossRefGoogle ScholarPubMed
Sarbassov, DD, Guertin, DA, Ali, SM, Sabatini, DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005;307:1098–101.CrossRef
Carracedo, A, Pandolfi, PP. The PTEN-PI3K pathway: of feedbacks and cross-talks. Oncogene 2008;27:5527–41.CrossRef
O’Reilly, KE, Rojo, F, She, QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Research 2006;66:1500–8.CrossRef
Salmena, L, Carracedo, A, Pandolfi, PP. Tenets of PTEN tumor suppression. Cell 2008;133:403–14.CrossRef
Samuels, Y, Velculescu, VE. Oncogenic mutations of PIK3CA in human cancers. Cell Cycle 2004;3:1221–4.CrossRef
Bader, AG, Kang, S, Vogt, PK. Cancer-specific mutations in PIK3CA are oncogenic in vivo. Proceedings of the National Academy of Sciences USA 2006;103:1475–9.CrossRef
Carpten, JD, Faber, AL, Horn, C, et al. A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature 2007;448:439–44.CrossRef
Cheng, JQ, Godwin, AK, Bellacosa, A, et al. AKT2, a putative oncogene encoding a member of a subfamily of protein-serine/threonine kinases, is amplified in human ovarian carcinomas. Proceedings of the National Academy of Sciences USA 1992;89:9267–71.CrossRef
Cheng, JQ, Ruggeri, B, Klein, WM, et al. Amplification of AKT2 in human pancreatic cells and inhibition of AKT2 expression and tumorigenicity by antisense RNA. Proceedings of the National Academy of Sciences USA 1996;93:3636–41.CrossRef
Ikenoue, T, Kanai, F, Hikiba, Y, et al. Functional analysis of PIK3CA gene mutations in human colorectal cancer. Cancer Research 2005;65:4562–7.CrossRef
Lim, KH, Counter, CM. Reduction in the requirement of oncogenic Ras signaling to activation of PI3K/AKT pathway during tumor maintenance. Cancer Cell 2005;8:381–92.CrossRef
Mizoguchi, M, Nutt, CL, Mohapatra, G, Louis, DN. Genetic alterations of phosphoinositide 3-kinase subunit genes in human glioblastomas. Brain Pathology 2004;14:372–7.CrossRef
Parsons, DW, Wang, TL, Samuels, Y, et al. Colorectal cancer: mutations in a signalling pathway. Nature 2005;436:792.CrossRef
Philp, AJ, Campbell, IG, Leet, C, et al. The phosphatidylinositol 3’-kinase p85alpha gene is an oncogene in human ovarian and colon tumors. Cancer Research 2001;61:7426–9.
Samuels, Y, Wang, Z, Bardelli, A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004;304:554.CrossRef
Shayesteh, L, Lu, Y, Kuo, WL, et al. PIK3CA is implicated as an oncogene in ovarian cancer. Nature Genetics 1999;21:99–102.CrossRef
Shoji, K, Oda, K, Nakagawa, S, et al. The oncogenic mutation in the pleckstrin homology domain of AKT1 in endometrial carcinomas. British Journal of Cancer 2009;101:145–8.CrossRefGoogle ScholarPubMed
Stommel, JM, Kimmelman, AC, Ying, H, et al. Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science 2007;318:287–90.CrossRef
Keniry, M, Parsons, R. The role of PTEN signaling perturbations in cancer and in targeted therapy. Oncogene 2008;27:5477–85.CrossRef
Li, J, Yen, C, Liaw, D, et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 1997;275:1943–7.CrossRef
Steck, PA, Pershouse, MA, Jasser, SA, et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nature Genetics 1997;15:356–62.CrossRef
Hickey, FB, Cotter, TG. BCR-ABL regulates phosphatidylinositol 3-kinase-p110gamma transcription and activation and is required for proliferation and drug resistance. Journal of Biological Chemistry 2006;281:2441–50.CrossRefGoogle ScholarPubMed
Zhao, L, Vogt, PK. Helical domain and kinase domain mutations in p110alpha of phosphatidylinositol 3-kinase induce gain of function by different mechanisms. Proceedings of the National Academy of Sciences USA 2008;105:2652–7.CrossRef
Mandelker, D, Gabelli, SB, Schmidt-Kittler, O, et al. A frequent kinase domain mutation that changes the interaction between PI3Kalpha and the membrane. Proceedings of the National Academy of Sciences USA 2009;106:16 996–7001.
Astanehe, A, Arenillas, D, Wasserman, WW, et al. Mechanisms underlying p53 regulation of PIK3CA transcription in ovarian surface epithelium and in ovarian cancer. Journal of Cell Science 2008;121:664–74.CrossRefGoogle ScholarPubMed
Hui, RC, Gomes, AR, Constantinidou, D, et al. The forkhead transcription factor FOXO3a increases phosphoinositide-3 kinase/Akt activity in drug-resistant leukemic cells through induction of PIK3CA expression. Molecular and Cellular Biology 2008;28:5886–98.CrossRef
Astanehe, A, Finkbeiner, MR, Hojabrpour, P, et al. The transcriptional induction of PIK3CA in tumor cells is dependent on the oncoprotein Y-box binding protein-1. Oncogene 2009;28:2406–18.CrossRef
Haas-Kogan, D, Shalev, N, Wong, M, et al. Protein kinase B (PKB/Akt) activity is elevated in glioblastoma cells due to mutation of the tumor suppressor PTEN/MMAC. Current Biology 1998;8:1195–8.CrossRef
Myers, MP, Pass, I, Batty, IH, et al. The lipid phosphatase activity of PTEN is critical for its tumor suppressor function. Proceedings of the National Academy of Sciences USA 1998;95:13 513–8.
Sun, X, Huang, J,Homma, T, et al. Genetic alterations in the PI3K pathway in prostate cancer. Anticancer Research 2009;29:1739–43.
Ali, IU, Schriml, LM, Dean, M. Mutational spectra of PTEN/MMAC1 gene: a tumor suppressor with lipid phosphatase activity. Journal of the National Cancer Institute 1999;91:1922–32.CrossRefGoogle ScholarPubMed
Han, SY, Kato, H, Kato, S, et al. Functional evaluation of PTEN missense mutations using in vitro phosphoinositide phosphatase assay. Cancer Research 2000;60:3147–51.
Garcia, JM, Silva, J, Pena, C, et al. Promoter methylation of the PTEN gene is a common molecular change in breast cancer. Genes Chromosomes and Cancer 2004;41:117–24.CrossRef
Goel, A, Arnold, CN, Niedzwiecki, D, et al. Frequent inactivation of PTEN by promoter hypermethylation in microsatellite instability-high sporadic colorectal cancers. Cancer Research 2004;64:3014–21.CrossRef
Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008;455:1061–8.CrossRef
Davies, MA, Stemke-Hale, K, Tellez, C, et al. A novel AKT3 mutation in melanoma tumours and cell lines. British Journal of Cancer 2008;99:1265–8.CrossRefGoogle ScholarPubMed
Moasser, MM, Basso, A, Averbuch, SD, Rosen, N. The tyrosine kinase inhibitor ZD1839 (“Iressa”) inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing tumor cells. Cancer Research 2001;61:7184–8.
Mellinghoff, IK, Wang, MY, Vivanco, I, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. New England Journal of Medicine 2005;353:2012–24.CrossRefGoogle ScholarPubMed
Zhao, JJ, Liu, Z, Wang, L, et al. The oncogenic properties of mutant p110alpha and p110beta phosphatidylinositol 3-kinases in human mammary epithelial cells. Proceedings of the National Academy of Sciences USA 2005;102:18 443–8.
Isakoff, SJ, Engelman, JA, Irie, HY, et al. Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells. Cancer Research 2005;65:10 992–1000.
Engelman, JA, Chen, L, Tan, X, et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nature Medicine 2008;14:1351–6.CrossRef
Zhao, JJ, Cheng, H, Jia, S, et al. The p110alpha isoform of PI3K is essential for proper growth factor signaling and oncogenic transformation. Proceedings of the National Academy of Sciences USA 2006;103:16 296–300.
Rodriguez-Viciana, P, Warne, PH, Khwaja, A, et al. Role of phosphoinositide 3-OH kinase in cell transformation and control of the actin cytoskeleton by Ras. Cell 1997;89:457–67.CrossRef
Gupta, S, Ramjaun, AR, Haiko, P, et al. Binding of ras to phosphoinositide 3-kinase p110alpha is required for ras-driven tumorigenesis in mice. Cell 2007;129:957–68.CrossRef
Jia, S, Liu, Z, Zhang, S, et al. Essential roles of PI(3)K-p110beta in cell growth, metabolism and tumorigenesis. Nature 2008;454:776–9.CrossRef
Wee, S, Wiederschain, D, Maira, SM, et al. PTEN-deficient cancers depend on PIK3CB. Proceedings of the National Academy of Sciences USA 2008;105:13 057–62.
Carmeliet, P, Lampugnani, MG, Moons, L, et al. Targeted deficiency or cytosolic truncation of the VE-cadherin gene in mice impairs VEGF-mediated endothelial survival and angiogenesis. Cell 1999;98:147–57.CrossRef
Hamada, K, Sasaki, T, Koni, PA, et al. The PTEN/PI3K pathway governs normal vascular development and tumor angiogenesis. Genes and Development 2005;19:2054–65.CrossRef
Xia, C, Meng, Q, Cao, Z, Shi, X, Jiang, BH. Regulation of angiogenesis and tumor growth by p110 alpha and AKT1 via VEGF expression. Journal of Cell Physiology 2006;209:56–66.CrossRefGoogle ScholarPubMed
Luo, J, Sobkiw, CL, Hirshman, MF, et al. Loss of class IA PI3K signaling in muscle leads to impaired muscle growth, insulin response, and hyperlipidemia. Cell Metabolism 2006;3:355–66.CrossRef
Cho, H, Mu, J, Kim, JK, et al. Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKB beta). Science 2001;292:1728–31.CrossRef
Taniguchi, CM, Tran, TT, Kondo, T, et al. Phosphoinositide 3-kinase regulatory subunit p85alpha suppresses insulin action via positive regulation of PTEN. Proceedings of the National Academy of Sciences USA 2006;103:12 093–7.
Ueki, K, Yballe, CM, Brachmann, SM, et al. Increased insulin sensitivity in mice lacking p85beta subunit of phosphoinositide 3-kinase. Proceedings of the National Academy of Sciences USA 2002;99:419–24.CrossRef
Wijesekara, N, Konrad, D, Eweida, M, et al. Muscle-specific Pten deletion protects against insulin resistance and diabetes. Molecular Cell Biology 2005;25:1135–45.CrossRef
Kalaany, NY, Sabatini, DM. Tumours with PI3K activation are resistant to dietary restriction. Nature 2009;458:725–31.CrossRef
Courtney, KD, Corcoran, RB, Engelman, JA. The PI3K pathway as drug target in human cancer. Journal of Clinical Oncology 2010;28:1075–83.CrossRefGoogle ScholarPubMed
Garcia-Echeverria, C, Sellers, WR. Drug discovery approaches targeting the PI3K/Akt pathway in cancer. Oncogene 2008;27:5511–26.CrossRef
Ma, WW, Adjei, AA. Novel agents on the horizon for cancer therapy. CA: A Cancer Journal for Clinicians 2009;59:111–37.
Hudes, G, Carducci, M, Tomczak, P, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. New England Journal of Medicine 2007;356:2271–81.CrossRefGoogle ScholarPubMed
Motzer, RJ, Escudier, B, Oudard, S, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled Phase III trial. Lancet 2008;372:449–56.CrossRef
Carracedo, A, Ma, L, Teruya-Feldstein, J, et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. Journal of Clinical Investigation 2008;118:3065–74.Google ScholarPubMed

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.

  • PI3K
    • By Kevin D. Courtney, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX, USA, Lewis C. Cantley, Weill Cornell Cancer Center, New York – Presbyterian Hospital, Weill Cornell Medical College, New York, NY, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.019
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.

  • PI3K
    • By Kevin D. Courtney, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX, USA, Lewis C. Cantley, Weill Cornell Cancer Center, New York – Presbyterian Hospital, Weill Cornell Medical College, New York, NY, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.019
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.

  • PI3K
    • By Kevin D. Courtney, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX, USA, Lewis C. Cantley, Weill Cornell Cancer Center, New York – Presbyterian Hospital, Weill Cornell Medical College, New York, NY, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.019
Available formats
×