메뉴 건너뛰기




Volumn 1835, Issue 2, 2013, Pages 194-210

Targeting LKB1 signaling in cancer

Author keywords

LKB1; Metformin; MTOR; Peutz Jeghers syndrome; Rapamycin; Sporadic cancer

Indexed keywords

1 (1 CYANO 1 METHYLETHYL) 3 METHYL 8 (3 QUINOLINYL)IMIDAZO[4,5 C]QUINOLIN 2(1H,3H) ONE; 1 [4 [1 (1,4 DIOXASPIRO[4.5]DEC 8 YL) 4 (8 OXA 3 AZABICYCLO[3.2.1]OCT 3 YL) 1H PYRAZOLO[3,4 D]PYRIMIDIN 6 YL]PHENYL] 3 METHYLUREA; 2 (4 AMINO 1 ISOPROPYL 1H PYRAZOLO[3,4 D]PYRIMIDIN 3 YL) 1H INDOL 5 OL; 2 (4 HYDROXYPHENYL) 4 MORPHOLINOPYRIDO[3',2':4,5]FURO[3,2 D]PYRIMIDINE; 4 (4 AMINO 5 (7 METHOXY 1H INDOL 2 YL)IMIDAZO[5,1 F][1,2,4]TRIAZIN 7 YL)CYCLOHEXANECARBOXYLIC ACID; 6,7 DIHYDRO 4 HYDROXY 3 (2' HYDROXY 1,1' BIPHENYL 4 YL) 6 OXOTHIENO[2,3 B]PYRIDINE 5 CARBONITRILE; ADENYLATE KINASE; ALPHA INTERFERON; AROMATASE INHIBITOR; AZD 8055; BEVACIZUMAB; CISPLATIN; DOXORUBICIN; EVEROLIMUS; GEMCITABINE; GLUCOSE; INSULIN; MAMMALIAN TARGET OF RAPAMYCIN; METFORMIN; OCTREOTIDE; PACLITAXEL; PLACEBO; PROTEIN KINASE LKB1; RAPAMYCIN; RIDAFOROLIMUS; SOMATOMEDIN C; SORAFENIB; SUNITINIB; TEMSIROLIMUS; UNINDEXED DRUG;

EID: 84873167692     PISSN: 0304419X     EISSN: 18792561     Source Type: Journal    
DOI: 10.1016/j.bbcan.2012.12.006     Document Type: Review
Times cited : (97)

References (339)
  • 1
    • 0000043603 scopus 로고
    • Over een zeer merkwaardige, gecombineerde familiaire polyposis van de slijmvliezen van den tractus intestinalis met die van de neuskeelholte en gepaard met eigenaardige pigmentaties van huid en slijmvliezen
    • Peutz J.L.A. Over een zeer merkwaardige, gecombineerde familiaire polyposis van de slijmvliezen van den tractus intestinalis met die van de neuskeelholte en gepaard met eigenaardige pigmentaties van huid en slijmvliezen. Ned. Maandschr. v. Geneesk. 1921, 10:134-146.
    • (1921) Ned. Maandschr. v. Geneesk. , vol.10 , pp. 134-146
    • Peutz, J.L.A.1
  • 2
    • 0000152018 scopus 로고
    • Generalized intestinal polyposis and melanin spots of the oral mucosa, lips and digits; a syndrome of diagnostic significance
    • Jeghers H., Mc K.V., Katz K.H. Generalized intestinal polyposis and melanin spots of the oral mucosa, lips and digits; a syndrome of diagnostic significance. N. Engl. J. Med. 1949, 241:1031-1036.
    • (1949) N. Engl. J. Med. , vol.241 , pp. 1031-1036
    • Jeghers, H.1    Mc, K.V.2    Katz, K.H.3
  • 5
    • 84855174861 scopus 로고    scopus 로고
    • Peutz-Jeghers syndrome: intriguing suggestion of gastrointestinal cancer prevention from surveillance
    • Latchford A.R., Neale K., Phillips R.K., Clark S.K. Peutz-Jeghers syndrome: intriguing suggestion of gastrointestinal cancer prevention from surveillance. Dis. Colon Rectum 2011, 54:1547-1551.
    • (2011) Dis. Colon Rectum , vol.54 , pp. 1547-1551
    • Latchford, A.R.1    Neale, K.2    Phillips, R.K.3    Clark, S.K.4
  • 9
    • 79955598460 scopus 로고    scopus 로고
    • High cumulative risk of intussusception in patients with Peutz-Jeghers syndrome: time to update surveillance guidelines?
    • van Lier M.G., Mathus-Vliegen E.M., Wagner A., van Leerdam M.E., Kuipers E.J. High cumulative risk of intussusception in patients with Peutz-Jeghers syndrome: time to update surveillance guidelines?. Am. J. Gastroenterol. 2011, 106:940-945.
    • (2011) Am. J. Gastroenterol. , vol.106 , pp. 940-945
    • van Lier, M.G.1    Mathus-Vliegen, E.M.2    Wagner, A.3    van Leerdam, M.E.4    Kuipers, E.J.5
  • 28
    • 78650938892 scopus 로고    scopus 로고
    • Negative regulation of TGFbeta signaling by the kinase LKB1 and the scaffolding protein LIP1
    • Moren A., Raja E., Heldin C.H., Moustakas A. Negative regulation of TGFbeta signaling by the kinase LKB1 and the scaffolding protein LIP1. J. Biol. Chem. 2011, 286:341-353.
    • (2011) J. Biol. Chem. , vol.286 , pp. 341-353
    • Moren, A.1    Raja, E.2    Heldin, C.H.3    Moustakas, A.4
  • 29
    • 79955478010 scopus 로고    scopus 로고
    • Liver kinase B1 (LKB1) in the pathogenesis of epithelial cancers
    • Herrmann J.L., Byekova Y., Elmets C.A., Athar M. Liver kinase B1 (LKB1) in the pathogenesis of epithelial cancers. Cancer Lett. 2011, 306:1-9.
    • (2011) Cancer Lett. , vol.306 , pp. 1-9
    • Herrmann, J.L.1    Byekova, Y.2    Elmets, C.A.3    Athar, M.4
  • 30
    • 37149042642 scopus 로고    scopus 로고
    • A role for LKB1 gene in human cancer beyond the Peutz-Jeghers syndrome
    • Sanchez-Cespedes M. A role for LKB1 gene in human cancer beyond the Peutz-Jeghers syndrome. Oncogene 2007, 26:7825-7832.
    • (2007) Oncogene , vol.26 , pp. 7825-7832
    • Sanchez-Cespedes, M.1
  • 34
    • 0042355248 scopus 로고    scopus 로고
    • Restoration of silenced Peutz-Jeghers syndrome gene, LKB1, induces apoptosis in pancreatic carcinoma cells
    • Qanungo S., Haldar S., Basu A. Restoration of silenced Peutz-Jeghers syndrome gene, LKB1, induces apoptosis in pancreatic carcinoma cells. Neoplasia 2003, 5:367-374.
    • (2003) Neoplasia , vol.5 , pp. 367-374
    • Qanungo, S.1    Haldar, S.2    Basu, A.3
  • 37
    • 0035992276 scopus 로고    scopus 로고
    • The tumor suppressor gene LKB1 is associated with prognosis in human breast carcinoma
    • Shen Z., Wen X.F., Lan F., Shen Z.Z., Shao Z.M. The tumor suppressor gene LKB1 is associated with prognosis in human breast carcinoma. Clin. Cancer Res. 2002, 8:2085-2090.
    • (2002) Clin. Cancer Res. , vol.8 , pp. 2085-2090
    • Shen, Z.1    Wen, X.F.2    Lan, F.3    Shen, Z.Z.4    Shao, Z.M.5
  • 38
    • 4644309913 scopus 로고    scopus 로고
    • High-resolution 19p13.2-13.3 allelotyping of breast carcinomas demonstrates frequent loss of heterozygosity
    • Yang T.L., Su Y.R., Huang C.S., Yu J.C., Lo Y.L., Wu P.E., Shen C.Y. High-resolution 19p13.2-13.3 allelotyping of breast carcinomas demonstrates frequent loss of heterozygosity. Genes Chromosom. Cancer 2004, 41:250-256.
    • (2004) Genes Chromosom. Cancer , vol.41 , pp. 250-256
    • Yang, T.L.1    Su, Y.R.2    Huang, C.S.3    Yu, J.C.4    Lo, Y.L.5    Wu, P.E.6    Shen, C.Y.7
  • 39
    • 68049137624 scopus 로고    scopus 로고
    • LKB1 is required for adiponectin-mediated modulation of AMPK-S6K axis and inhibition of migration and invasion of breast cancer cells
    • Taliaferro-Smith L., Nagalingam A., Zhong D., Zhou W., Saxena N.K., Sharma D. LKB1 is required for adiponectin-mediated modulation of AMPK-S6K axis and inhibition of migration and invasion of breast cancer cells. Oncogene 2009, 28:2621-2633.
    • (2009) Oncogene , vol.28 , pp. 2621-2633
    • Taliaferro-Smith, L.1    Nagalingam, A.2    Zhong, D.3    Zhou, W.4    Saxena, N.K.5    Sharma, D.6
  • 40
    • 33845332753 scopus 로고    scopus 로고
    • Enhanced expression of LKB1 in breast cancer cells attenuates angiogenesis, invasion, and metastatic potential
    • Zhuang Z.G., Di G.H., Shen Z.Z., Ding J., Shao Z.M. Enhanced expression of LKB1 in breast cancer cells attenuates angiogenesis, invasion, and metastatic potential. Mol. Cancer Res. 2006, 4:843-849.
    • (2006) Mol. Cancer Res. , vol.4 , pp. 843-849
    • Zhuang, Z.G.1    Di, G.H.2    Shen, Z.Z.3    Ding, J.4    Shao, Z.M.5
  • 41
    • 25444524851 scopus 로고    scopus 로고
    • Mutations in the human LKB1/STK11 gene
    • Launonen V. Mutations in the human LKB1/STK11 gene. Hum. Mutat. 2005, 26:291-297.
    • (2005) Hum. Mutat. , vol.26 , pp. 291-297
    • Launonen, V.1
  • 42
    • 79953232702 scopus 로고    scopus 로고
    • LKB1 in lung cancerigenesis: a serine/threonine kinase as tumor suppressor
    • Gao Y., Ge G., Ji H. LKB1 in lung cancerigenesis: a serine/threonine kinase as tumor suppressor. Protein Cell 2011, 2:99-107.
    • (2011) Protein Cell , vol.2 , pp. 99-107
    • Gao, Y.1    Ge, G.2    Ji, H.3
  • 47
    • 0031827021 scopus 로고    scopus 로고
    • Genetic pathways of colorectal carcinogenesis rarely involve the PTEN and LKB1 genes outside the inherited hamartoma syndromes
    • Wang Z.J., Taylor F., Churchman M., Norbury G., Tomlinson I. Genetic pathways of colorectal carcinogenesis rarely involve the PTEN and LKB1 genes outside the inherited hamartoma syndromes. Am. J. Pathol. 1998, 153:363-366.
    • (1998) Am. J. Pathol. , vol.153 , pp. 363-366
    • Wang, Z.J.1    Taylor, F.2    Churchman, M.3    Norbury, G.4    Tomlinson, I.5
  • 49
  • 50
    • 0033915012 scopus 로고    scopus 로고
    • 5'-CpG island methylation of the LKB1/STK11 promoter and allelic loss at chromosome 19p13.3 in sporadic colorectal cancer
    • Trojan J., Brieger A., Raedle J., Esteller M., Zeuzem S. 5'-CpG island methylation of the LKB1/STK11 promoter and allelic loss at chromosome 19p13.3 in sporadic colorectal cancer. Gut 2000, 47:272-276.
    • (2000) Gut , vol.47 , pp. 272-276
    • Trojan, J.1    Brieger, A.2    Raedle, J.3    Esteller, M.4    Zeuzem, S.5
  • 51
    • 0033775035 scopus 로고    scopus 로고
    • An investigation of the Peutz-Jeghers gene (LKB1) in sporadic breast and colon cancers
    • Forster L.F., Defres S., Goudie D.R., Baty D.U., Carey F.A. An investigation of the Peutz-Jeghers gene (LKB1) in sporadic breast and colon cancers. J. Clin. Pathol. 2000, 53:791-793.
    • (2000) J. Clin. Pathol. , vol.53 , pp. 791-793
    • Forster, L.F.1    Defres, S.2    Goudie, D.R.3    Baty, D.U.4    Carey, F.A.5
  • 52
    • 0034490014 scopus 로고    scopus 로고
    • Frequent loss of heterozygosity at the 19p13.3 locus without LKB1/STK11 mutations in human carcinoma metastases to the brain
    • Sobottka S.B., Haase M., Fitze G., Hahn M., Schackert H.K., Schackert G. Frequent loss of heterozygosity at the 19p13.3 locus without LKB1/STK11 mutations in human carcinoma metastases to the brain. J. Neurooncol. 2000, 49:187-195.
    • (2000) J. Neurooncol. , vol.49 , pp. 187-195
    • Sobottka, S.B.1    Haase, M.2    Fitze, G.3    Hahn, M.4    Schackert, H.K.5    Schackert, G.6
  • 58
    • 0032054770 scopus 로고    scopus 로고
    • Low frequency of somatic mutations in the LKB1/Peutz-Jeghers syndrome gene in sporadic breast cancer
    • Bignell G.R., Barfoot R., Seal S., Collins N., Warren W., Stratton M.R. Low frequency of somatic mutations in the LKB1/Peutz-Jeghers syndrome gene in sporadic breast cancer. Cancer Res. 1998, 58:1384-1386.
    • (1998) Cancer Res. , vol.58 , pp. 1384-1386
    • Bignell, G.R.1    Barfoot, R.2    Seal, S.3    Collins, N.4    Warren, W.5    Stratton, M.R.6
  • 60
    • 0033876205 scopus 로고    scopus 로고
    • Somatic mutations in the STK11/LKB1 gene are uncommon in rare gynecological tumor types associated with Peutz-Jegher's syndrome
    • Connolly D.C., Katabuchi H., Cliby W.A., Cho K.R. Somatic mutations in the STK11/LKB1 gene are uncommon in rare gynecological tumor types associated with Peutz-Jegher's syndrome. Am. J. Pathol. 2000, 156:339-345.
    • (2000) Am. J. Pathol. , vol.156 , pp. 339-345
    • Connolly, D.C.1    Katabuchi, H.2    Cliby, W.A.3    Cho, K.R.4
  • 61
    • 4444353724 scopus 로고    scopus 로고
    • The STK11/LKB1 Peutz-Jegher gene is not involved in the pathogenesis of sporadic sex cord-stromal tumors, although loss of heterozygosity at 19p13.3 indicates other gene alteration in these tumors
    • Kato N., Romero M., Catasus L., Prat J. The STK11/LKB1 Peutz-Jegher gene is not involved in the pathogenesis of sporadic sex cord-stromal tumors, although loss of heterozygosity at 19p13.3 indicates other gene alteration in these tumors. Hum. Pathol. 2004, 35:1101-1104.
    • (2004) Hum. Pathol. , vol.35 , pp. 1101-1104
    • Kato, N.1    Romero, M.2    Catasus, L.3    Prat, J.4
  • 64
    • 65249093808 scopus 로고    scopus 로고
    • Somatic alterations of the serine/threonine kinase LKB1 gene in squamous cell (SCC) and large cell (LCC) lung carcinoma
    • Strazisar M., Mlakar V., Rott T., Glavac D. Somatic alterations of the serine/threonine kinase LKB1 gene in squamous cell (SCC) and large cell (LCC) lung carcinoma. Cancer Investig. 2009, 27:407-416.
    • (2009) Cancer Investig. , vol.27 , pp. 407-416
    • Strazisar, M.1    Mlakar, V.2    Rott, T.3    Glavac, D.4
  • 67
    • 0033522144 scopus 로고    scopus 로고
    • Somatic mutation of the Peutz-Jeghers syndrome gene, LKB1/STK11, in malignant melanoma
    • Guldberg P., thor Straten P., Ahrenkiel V., Seremet T., Kirkin A.F., Zeuthen J. Somatic mutation of the Peutz-Jeghers syndrome gene, LKB1/STK11, in malignant melanoma. Oncogene 1999, 18:1777-1780.
    • (1999) Oncogene , vol.18 , pp. 1777-1780
    • Guldberg, P.1    thor Straten, P.2    Ahrenkiel, V.3    Seremet, T.4    Kirkin, A.F.5    Zeuthen, J.6
  • 71
    • 21144470441 scopus 로고    scopus 로고
    • Functional analysis of Peutz-Jeghers mutations reveals that the LKB1 C-terminal region exerts a crucial role in regulating both the AMPK pathway and the cell polarity
    • Forcet C., Etienne-Manneville S., Gaude H., Fournier L., Debilly S., Salmi M., Baas A., Olschwang S., Clevers H., Billaud M. Functional analysis of Peutz-Jeghers mutations reveals that the LKB1 C-terminal region exerts a crucial role in regulating both the AMPK pathway and the cell polarity. Hum. Mol. Genet. 2005, 14:1283-1292.
    • (2005) Hum. Mol. Genet. , vol.14 , pp. 1283-1292
    • Forcet, C.1    Etienne-Manneville, S.2    Gaude, H.3    Fournier, L.4    Debilly, S.5    Salmi, M.6    Baas, A.7    Olschwang, S.8    Clevers, H.9    Billaud, M.10
  • 73
    • 0036499467 scopus 로고    scopus 로고
    • Identification and characterization of four novel phosphorylation sites (Ser31, Ser325, Thr336 and Thr366) on LKB1/STK11, the protein kinase mutated in Peutz-Jeghers cancer syndrome
    • Sapkota G.P., Boudeau J., Deak M., Kieloch A., Morrice N., Alessi D.R. Identification and characterization of four novel phosphorylation sites (Ser31, Ser325, Thr336 and Thr366) on LKB1/STK11, the protein kinase mutated in Peutz-Jeghers cancer syndrome. Biochem. J. 2002, 362:481-490.
    • (2002) Biochem. J. , vol.362 , pp. 481-490
    • Sapkota, G.P.1    Boudeau, J.2    Deak, M.3    Kieloch, A.4    Morrice, N.5    Alessi, D.R.6
  • 74
    • 0035375261 scopus 로고    scopus 로고
    • Phosphorylation of the protein kinase mutated in Peutz-Jeghers cancer syndrome, LKB1/STK11, at Ser431 by p90(RSK) and cAMP-dependent protein kinase, but not its farnesylation at Cys(433), is essential for LKB1 to suppress cell growth
    • Sapkota G.P., Kieloch A., Lizcano J.M., Lain S., Arthur J.S., Williams M.R., Morrice N., Deak M., Alessi D.R. Phosphorylation of the protein kinase mutated in Peutz-Jeghers cancer syndrome, LKB1/STK11, at Ser431 by p90(RSK) and cAMP-dependent protein kinase, but not its farnesylation at Cys(433), is essential for LKB1 to suppress cell growth. J. Biol. Chem. 2001, 276:19469-19482.
    • (2001) J. Biol. Chem. , vol.276 , pp. 19469-19482
    • Sapkota, G.P.1    Kieloch, A.2    Lizcano, J.M.3    Lain, S.4    Arthur, J.S.5    Williams, M.R.6    Morrice, N.7    Deak, M.8    Alessi, D.R.9
  • 75
    • 0034141636 scopus 로고    scopus 로고
    • LKB1, a novel serine/threonine protein kinase and potential tumour suppressor, is phosphorylated by cAMP-dependent protein kinase (PKA) and prenylated in vivo
    • Collins S.P., Reoma J.L., Gamm D.M., Uhler M.D. LKB1, a novel serine/threonine protein kinase and potential tumour suppressor, is phosphorylated by cAMP-dependent protein kinase (PKA) and prenylated in vivo. Biochem. J. 2000, 345(Pt 3):673-680.
    • (2000) Biochem. J. , vol.345 , Issue.PART 3 , pp. 673-680
    • Collins, S.P.1    Reoma, J.L.2    Gamm, D.M.3    Uhler, M.D.4
  • 77
    • 45549094869 scopus 로고    scopus 로고
    • Protein kinase Czeta-dependent LKB1 serine 428 phosphorylation increases LKB1 nucleus export and apoptosis in endothelial cells
    • Song P., Xie Z., Wu Y., Xu J., Dong Y., Zou M.H. Protein kinase Czeta-dependent LKB1 serine 428 phosphorylation increases LKB1 nucleus export and apoptosis in endothelial cells. J. Biol. Chem. 2008, 283:12446-12455.
    • (2008) J. Biol. Chem. , vol.283 , pp. 12446-12455
    • Song, P.1    Xie, Z.2    Wu, Y.3    Xu, J.4    Dong, Y.5    Zou, M.H.6
  • 78
    • 0032815880 scopus 로고    scopus 로고
    • The mouse Peutz-Jeghers syndrome gene Lkb1 encodes a nuclear protein kinase
    • Smith D.P., Spicer J., Smith A., Swift S., Ashworth A. The mouse Peutz-Jeghers syndrome gene Lkb1 encodes a nuclear protein kinase. Hum. Mol. Genet. 1999, 8:1479-1485.
    • (1999) Hum. Mol. Genet. , vol.8 , pp. 1479-1485
    • Smith, D.P.1    Spicer, J.2    Smith, A.3    Swift, S.4    Ashworth, A.5
  • 79
    • 0345107247 scopus 로고    scopus 로고
    • Complexes between the LKB1 tumor suppressor, STRAD alpha/beta and MO25 alpha/beta are upstream kinases in the AMP-activated protein kinase cascade
    • Hawley S.A., Boudeau J., Reid J.L., Mustard K.J., Udd L., Makela T.P., Alessi D.R., Hardie D.G. Complexes between the LKB1 tumor suppressor, STRAD alpha/beta and MO25 alpha/beta are upstream kinases in the AMP-activated protein kinase cascade. J. Biol. 2003, 2:28.
    • (2003) J. Biol. , vol.2 , pp. 28
    • Hawley, S.A.1    Boudeau, J.2    Reid, J.L.3    Mustard, K.J.4    Udd, L.5    Makela, T.P.6    Alessi, D.R.7    Hardie, D.G.8
  • 80
    • 14244251499 scopus 로고    scopus 로고
    • Identification of the sucrose non-fermenting related kinase SNRK, as a novel LKB1 substrate
    • Jaleel M., McBride A., Lizcano J.M., Deak M., Toth R., Morrice N.A., Alessi D.R. Identification of the sucrose non-fermenting related kinase SNRK, as a novel LKB1 substrate. FEBS Lett. 2005, 579:1417-1423.
    • (2005) FEBS Lett. , vol.579 , pp. 1417-1423
    • Jaleel, M.1    McBride, A.2    Lizcano, J.M.3    Deak, M.4    Toth, R.5    Morrice, N.A.6    Alessi, D.R.7
  • 82
    • 0037461735 scopus 로고    scopus 로고
    • A role for Drosophila LKB1 in anterior-posterior axis formation and epithelial polarity
    • Martin S.G., St Johnston D. A role for Drosophila LKB1 in anterior-posterior axis formation and epithelial polarity. Nature 2003, 421:379-384.
    • (2003) Nature , vol.421 , pp. 379-384
    • Martin, S.G.1    St Johnston, D.2
  • 83
    • 0034090975 scopus 로고    scopus 로고
    • The C. elegans par-4 gene encodes a putative serine-threonine kinase required for establishing embryonic asymmetry
    • Watts J.L., Morton D.G., Bestman J., Kemphues K.J. The C. elegans par-4 gene encodes a putative serine-threonine kinase required for establishing embryonic asymmetry. Development 2000, 127:1467-1475.
    • (2000) Development , vol.127 , pp. 1467-1475
    • Watts, J.L.1    Morton, D.G.2    Bestman, J.3    Kemphues, K.J.4
  • 85
    • 34247338972 scopus 로고    scopus 로고
    • Acto-myosin reorganization and PAR polarity in C. elegans
    • Cowan C.R., Hyman A.A. Acto-myosin reorganization and PAR polarity in C. elegans. Development 2007, 134:1035-1043.
    • (2007) Development , vol.134 , pp. 1035-1043
    • Cowan, C.R.1    Hyman, A.A.2
  • 87
    • 33751204464 scopus 로고    scopus 로고
    • AMP-activated protein kinase regulates the assembly of epithelial tight junctions
    • Zhang L., Li J., Young L.H., Caplan M.J. AMP-activated protein kinase regulates the assembly of epithelial tight junctions. Proc. Natl. Acad. Sci. U. S. A. 2006, 103:17272-17277.
    • (2006) Proc. Natl. Acad. Sci. U. S. A. , vol.103 , pp. 17272-17277
    • Zhang, L.1    Li, J.2    Young, L.H.3    Caplan, M.J.4
  • 88
    • 38849204971 scopus 로고    scopus 로고
    • The tumor suppressor LKB1 regulates lung cancer cell polarity by mediating cdc42 recruitment and activity
    • Zhang S., Schafer-Hales K., Khuri F.R., Zhou W., Vertino P.M., Marcus A.I. The tumor suppressor LKB1 regulates lung cancer cell polarity by mediating cdc42 recruitment and activity. Cancer Res. 2008, 68:740-748.
    • (2008) Cancer Res. , vol.68 , pp. 740-748
    • Zhang, S.1    Schafer-Hales, K.2    Khuri, F.R.3    Zhou, W.4    Vertino, P.M.5    Marcus, A.I.6
  • 89
    • 33846521353 scopus 로고    scopus 로고
    • Regulation of epithelial tight junction assembly and disassembly by AMP-activated protein kinase
    • Zheng B., Cantley L.C. Regulation of epithelial tight junction assembly and disassembly by AMP-activated protein kinase. Proc. Natl. Acad. Sci. U. S. A. 2007, 104:819-822.
    • (2007) Proc. Natl. Acad. Sci. U. S. A. , vol.104 , pp. 819-822
    • Zheng, B.1    Cantley, L.C.2
  • 91
    • 34548179000 scopus 로고    scopus 로고
    • Suppression of tubulin polymerization by the LKB1-microtubule-associated protein/microtubule affinity-regulating kinase signaling
    • Kojima Y., Miyoshi H., Clevers H.C., Oshima M., Aoki M., Taketo M.M. Suppression of tubulin polymerization by the LKB1-microtubule-associated protein/microtubule affinity-regulating kinase signaling. J. Biol. Chem. 2007, 282:23532-23540.
    • (2007) J. Biol. Chem. , vol.282 , pp. 23532-23540
    • Kojima, Y.1    Miyoshi, H.2    Clevers, H.C.3    Oshima, M.4    Aoki, M.5    Taketo, M.M.6
  • 93
    • 13644267037 scopus 로고    scopus 로고
    • Mammalian SAD kinases are required for neuronal polarization
    • Kishi M., Pan Y.A., Crump J.G., Sanes J.R. Mammalian SAD kinases are required for neuronal polarization. Science 2005, 307:929-932.
    • (2005) Science , vol.307 , pp. 929-932
    • Kishi, M.1    Pan, Y.A.2    Crump, J.G.3    Sanes, J.R.4
  • 94
    • 4344648897 scopus 로고    scopus 로고
    • Dare to be different: asymmetric cell division in Drosophila, C. elegans and vertebrates
    • Betschinger J., Knoblich J.A. Dare to be different: asymmetric cell division in Drosophila, C. elegans and vertebrates. Curr. Biol. 2004, 14:R674-R685.
    • (2004) Curr. Biol. , vol.14
    • Betschinger, J.1    Knoblich, J.A.2
  • 97
    • 84869098775 scopus 로고    scopus 로고
    • Cell confinement controls centrosome positioning and lumen initiation during epithelial morphogenesis
    • Rodriguez-Fraticelli A.E., Auzan M., Alonso M.A., Bornens M., Martin-Belmonte F. Cell confinement controls centrosome positioning and lumen initiation during epithelial morphogenesis. J. Cell Biol. 2012, 198:1011-1023.
    • (2012) J. Cell Biol. , vol.198 , pp. 1011-1023
    • Rodriguez-Fraticelli, A.E.1    Auzan, M.2    Alonso, M.A.3    Bornens, M.4    Martin-Belmonte, F.5
  • 98
    • 0141616620 scopus 로고    scopus 로고
    • LKB1 (XEEK1) regulates Wnt signalling in vertebrate development
    • Ossipova O., Bardeesy N., DePinho R.A., Green J.B. LKB1 (XEEK1) regulates Wnt signalling in vertebrate development. Nat. Cell Biol. 2003, 5:889-894.
    • (2003) Nat. Cell Biol. , vol.5 , pp. 889-894
    • Ossipova, O.1    Bardeesy, N.2    DePinho, R.A.3    Green, J.B.4
  • 99
  • 100
    • 67650898228 scopus 로고    scopus 로고
    • LKB1 and AMPK family signaling: the intimate link between cell polarity and energy metabolism
    • Jansen M., Ten Klooster J.P., Offerhaus G.J., Clevers H. LKB1 and AMPK family signaling: the intimate link between cell polarity and energy metabolism. Physiol. Rev. 2009, 89:777-798.
    • (2009) Physiol. Rev. , vol.89 , pp. 777-798
    • Jansen, M.1    Ten Klooster, J.P.2    Offerhaus, G.J.3    Clevers, H.4
  • 101
    • 80555131518 scopus 로고    scopus 로고
    • Epithelial organization, cell polarity and tumorigenesis
    • McCaffrey L.M., Macara I.G. Epithelial organization, cell polarity and tumorigenesis. Trends Cell Biol. 2011, 21:727-735.
    • (2011) Trends Cell Biol. , vol.21 , pp. 727-735
    • McCaffrey, L.M.1    Macara, I.G.2
  • 102
    • 0028406897 scopus 로고
    • Role of the AMP-activated protein kinase in the cellular stress response
    • Corton J.M., Gillespie J.G., Hardie D.G. Role of the AMP-activated protein kinase in the cellular stress response. Curr. Biol. 1994, 4:315-324.
    • (1994) Curr. Biol. , vol.4 , pp. 315-324
    • Corton, J.M.1    Gillespie, J.G.2    Hardie, D.G.3
  • 103
    • 0344081177 scopus 로고    scopus 로고
    • Minireview: the AMP-activated protein kinase cascade: the key sensor of cellular energy status
    • Hardie D.G. Minireview: the AMP-activated protein kinase cascade: the key sensor of cellular energy status. Endocrinology 2003, 144:5179-5183.
    • (2003) Endocrinology , vol.144 , pp. 5179-5183
    • Hardie, D.G.1
  • 104
    • 0029925785 scopus 로고    scopus 로고
    • Characterization of AMP-activated protein kinase beta and gamma subunits. Assembly of the heterotrimeric complex in vitro
    • Woods A., Cheung P.C., Smith F.C., Davison M.D., Scott J., Beri R.K., Carling D. Characterization of AMP-activated protein kinase beta and gamma subunits. Assembly of the heterotrimeric complex in vitro. J. Biol. Chem. 1996, 271:10282-10290.
    • (1996) J. Biol. Chem. , vol.271 , pp. 10282-10290
    • Woods, A.1    Cheung, P.C.2    Smith, F.C.3    Davison, M.D.4    Scott, J.5    Beri, R.K.6    Carling, D.7
  • 105
    • 0029910018 scopus 로고    scopus 로고
    • Characterization of the AMP-activated protein kinase from rat liver and identification of threonine 172 as the major site at which it phosphorylates AMP-activated protein kinase
    • Hawley S.A., Davison M., Woods A., Davies S.P., Beri R.K., Carling D., Hardie D.G. Characterization of the AMP-activated protein kinase from rat liver and identification of threonine 172 as the major site at which it phosphorylates AMP-activated protein kinase. J. Biol. Chem. 1996, 271:27879-27887.
    • (1996) J. Biol. Chem. , vol.271 , pp. 27879-27887
    • Hawley, S.A.1    Davison, M.2    Woods, A.3    Davies, S.P.4    Beri, R.K.5    Carling, D.6    Hardie, D.G.7
  • 106
  • 108
    • 0024786438 scopus 로고
    • Purification and characterization of the AMP-activated protein kinase. Copurification of acetyl-CoA carboxylase kinase and 3-hydroxy-3-methylglutaryl-CoA reductase kinase activities
    • Carling D., Clarke P.R., Zammit V.A., Hardie D.G. Purification and characterization of the AMP-activated protein kinase. Copurification of acetyl-CoA carboxylase kinase and 3-hydroxy-3-methylglutaryl-CoA reductase kinase activities. Eur. J. Biochem. 1989, 186:129-136.
    • (1989) Eur. J. Biochem. , vol.186 , pp. 129-136
    • Carling, D.1    Clarke, P.R.2    Zammit, V.A.3    Hardie, D.G.4
  • 109
    • 0024335432 scopus 로고
    • The substrate and sequence specificity of the AMP-activated protein kinase. Phosphorylation of glycogen synthase and phosphorylase kinase
    • Carling D., Hardie D.G. The substrate and sequence specificity of the AMP-activated protein kinase. Phosphorylation of glycogen synthase and phosphorylase kinase. Biochim. Biophys. Acta 1989, 1012:81-86.
    • (1989) Biochim. Biophys. Acta , vol.1012 , pp. 81-86
    • Carling, D.1    Hardie, D.G.2
  • 110
    • 0023896220 scopus 로고
    • The low activity of acetyl-CoA carboxylase in basal and glucagon-stimulated hepatocytes is due to phosphorylation by the AMP-activated protein kinase and not cyclic AMP-dependent protein kinase
    • Sim A.T., Hardie D.G. The low activity of acetyl-CoA carboxylase in basal and glucagon-stimulated hepatocytes is due to phosphorylation by the AMP-activated protein kinase and not cyclic AMP-dependent protein kinase. FEBS Lett. 1988, 233:294-298.
    • (1988) FEBS Lett. , vol.233 , pp. 294-298
    • Sim, A.T.1    Hardie, D.G.2
  • 112
    • 0345167800 scopus 로고    scopus 로고
    • TSC2 mediates cellular energy response to control cell growth and survival
    • Inoki K., Zhu T., Guan K.L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 2003, 115:577-590.
    • (2003) Cell , vol.115 , pp. 577-590
    • Inoki, K.1    Zhu, T.2    Guan, K.L.3
  • 113
    • 0038141979 scopus 로고    scopus 로고
    • Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins
    • Zhang Y., Gao X., Saucedo L.J., Ru B., Edgar B.A., Pan D. Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins. Nat. Cell Biol. 2003, 5:578-581.
    • (2003) Nat. Cell Biol. , vol.5 , pp. 578-581
    • Zhang, Y.1    Gao, X.2    Saucedo, L.J.3    Ru, B.4    Edgar, B.A.5    Pan, D.6
  • 121
    • 78649811793 scopus 로고    scopus 로고
    • Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells
    • Nakada D., Saunders T.L., Morrison S.J. Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells. Nature 2010, 468:653-658.
    • (2010) Nature , vol.468 , pp. 653-658
    • Nakada, D.1    Saunders, T.L.2    Morrison, S.J.3
  • 122
    • 33845345621 scopus 로고    scopus 로고
    • LKB1 is recruited to the p21/WAF1 promoter by p53 to mediate transcriptional activation
    • Zeng P.Y., Berger S.L. LKB1 is recruited to the p21/WAF1 promoter by p53 to mediate transcriptional activation. Cancer Res. 2006, 66:10701-10708.
    • (2006) Cancer Res. , vol.66 , pp. 10701-10708
    • Zeng, P.Y.1    Berger, S.L.2
  • 123
    • 0037096833 scopus 로고    scopus 로고
    • Growth arrest by the LKB1 tumor suppressor: induction of p21(WAF1/CIP1)
    • Tiainen M., Vaahtomeri K., Ylikorkala A., Makela T.P. Growth arrest by the LKB1 tumor suppressor: induction of p21(WAF1/CIP1). Hum. Mol. Genet. 2002, 11:1497-1504.
    • (2002) Hum. Mol. Genet. , vol.11 , pp. 1497-1504
    • Tiainen, M.1    Vaahtomeri, K.2    Ylikorkala, A.3    Makela, T.P.4
  • 124
    • 0037444404 scopus 로고    scopus 로고
    • Growth and molecular profile of lung cancer cells expressing ectopic LKB1: down-regulation of the phosphatidylinositol 3'-phosphate kinase/PTEN pathway
    • Jimenez A.I., Fernandez P., Dominguez O., Dopazo A., Sanchez-Cespedes M. Growth and molecular profile of lung cancer cells expressing ectopic LKB1: down-regulation of the phosphatidylinositol 3'-phosphate kinase/PTEN pathway. Cancer Res. 2003, 63:1382-1388.
    • (2003) Cancer Res. , vol.63 , pp. 1382-1388
    • Jimenez, A.I.1    Fernandez, P.2    Dominguez, O.3    Dopazo, A.4    Sanchez-Cespedes, M.5
  • 125
    • 34250170149 scopus 로고    scopus 로고
    • Association of LKB1 with a WD-repeat protein WDR6 is implicated in cell growth arrest and p27(Kip1) induction
    • Xie X., Wang Z., Chen Y. Association of LKB1 with a WD-repeat protein WDR6 is implicated in cell growth arrest and p27(Kip1) induction. Mol. Cell. Biochem. 2007, 301:115-122.
    • (2007) Mol. Cell. Biochem. , vol.301 , pp. 115-122
    • Xie, X.1    Wang, Z.2    Chen, Y.3
  • 127
    • 34250793435 scopus 로고    scopus 로고
    • LKB1 catalytically deficient mutants enhance cyclin D1 expression
    • Scott K.D., Nath-Sain S., Agnew M.D., Marignani P.A. LKB1 catalytically deficient mutants enhance cyclin D1 expression. Cancer Res. 2007, 67:5622-5627.
    • (2007) Cancer Res. , vol.67 , pp. 5622-5627
    • Scott, K.D.1    Nath-Sain, S.2    Agnew, M.D.3    Marignani, P.A.4
  • 128
    • 75749096671 scopus 로고    scopus 로고
    • Endogenous LKB1 knockdown accelerates G(1)/S transition through p53 and p16 pathways
    • Liang X., Wang P., Gao Q., Xiang T., Tao X. Endogenous LKB1 knockdown accelerates G(1)/S transition through p53 and p16 pathways. Cancer Biol. Ther. 2010, 9:156-160.
    • (2010) Cancer Biol. Ther. , vol.9 , pp. 156-160
    • Liang, X.1    Wang, P.2    Gao, Q.3    Xiang, T.4    Tao, X.5
  • 133
    • 33745212865 scopus 로고    scopus 로고
    • JNK pathway mediates apoptotic cell death induced by tumor suppressor LKB1 in Drosophila
    • Lee J.H., Koh H., Kim M., Park J., Lee S.Y., Lee S., Chung J. JNK pathway mediates apoptotic cell death induced by tumor suppressor LKB1 in Drosophila. Cell Death Differ. 2006, 13:1110-1122.
    • (2006) Cell Death Differ. , vol.13 , pp. 1110-1122
    • Lee, J.H.1    Koh, H.2    Kim, M.3    Park, J.4    Lee, S.Y.5    Lee, S.6    Chung, J.7
  • 135
    • 70350238781 scopus 로고    scopus 로고
    • PTEN hamartoma tumor syndrome: an overview
    • Hobert J.A., Eng C. PTEN hamartoma tumor syndrome: an overview. Genet. Med. 2009, 11:687-694.
    • (2009) Genet. Med. , vol.11 , pp. 687-694
    • Hobert, J.A.1    Eng, C.2
  • 136
    • 26444610333 scopus 로고    scopus 로고
    • LKB1 interacts with and phosphorylates PTEN: a functional link between two proteins involved in cancer predisposing syndromes
    • Mehenni H., Lin-Marq N., Buchet-Poyau K., Reymond A., Collart M.A., Picard D., Antonarakis S.E. LKB1 interacts with and phosphorylates PTEN: a functional link between two proteins involved in cancer predisposing syndromes. Hum. Mol. Genet. 2005, 14:2209-2219.
    • (2005) Hum. Mol. Genet. , vol.14 , pp. 2209-2219
    • Mehenni, H.1    Lin-Marq, N.2    Buchet-Poyau, K.3    Reymond, A.4    Collart, M.A.5    Picard, D.6    Antonarakis, S.E.7
  • 137
    • 34848892187 scopus 로고    scopus 로고
    • Reactive nitrogen species induced by hyperglycemia suppresses Akt signaling and triggers apoptosis by upregulating phosphatase PTEN (phosphatase and tensin homologue deleted on chromosome 10) in an LKB1-dependent manner
    • Song P., Wu Y., Xu J., Xie Z., Dong Y., Zhang M., Zou M.H. Reactive nitrogen species induced by hyperglycemia suppresses Akt signaling and triggers apoptosis by upregulating phosphatase PTEN (phosphatase and tensin homologue deleted on chromosome 10) in an LKB1-dependent manner. Circulation 2007, 116:1585-1595.
    • (2007) Circulation , vol.116 , pp. 1585-1595
    • Song, P.1    Wu, Y.2    Xu, J.3    Xie, Z.4    Dong, Y.5    Zhang, M.6    Zou, M.H.7
  • 138
    • 80052314345 scopus 로고    scopus 로고
    • A novel de novo mutation in LKB1 gene in a Chinese Peutz Jeghers syndrome patient significantly diminished p53 activity
    • Liu L., Du X., Nie J. A novel de novo mutation in LKB1 gene in a Chinese Peutz Jeghers syndrome patient significantly diminished p53 activity. Clin. Res. Hepatol. Gastroenterol. 2011, 35:221-226.
    • (2011) Clin. Res. Hepatol. Gastroenterol. , vol.35 , pp. 221-226
    • Liu, L.1    Du, X.2    Nie, J.3
  • 139
    • 79251614530 scopus 로고    scopus 로고
    • Lkb1 and Pten synergise to suppress mTOR-mediated tumorigenesis and epithelial-mesenchymal transition in the mouse bladder
    • Shorning B.Y., Griffiths D., Clarke A.R. Lkb1 and Pten synergise to suppress mTOR-mediated tumorigenesis and epithelial-mesenchymal transition in the mouse bladder. PLoS One 2011, 6:e16209.
    • (2011) PLoS One , vol.6
    • Shorning, B.Y.1    Griffiths, D.2    Clarke, A.R.3
  • 141
    • 0028963260 scopus 로고
    • 13: Relative efficacy of randomly allocated diet, sulphonylurea, insulin, or metformin in patients with newly diagnosed non-insulin dependent diabetes followed for three years
    • United Kingdom Prospective Diabetes Study (UKPDS)
    • United Kingdom Prospective Diabetes Study (UKPDS) 13: Relative efficacy of randomly allocated diet, sulphonylurea, insulin, or metformin in patients with newly diagnosed non-insulin dependent diabetes followed for three years. BMJ 1995, 310:83-88.
    • (1995) BMJ , vol.310 , pp. 83-88
  • 150
    • 80052397117 scopus 로고    scopus 로고
    • Potent anti-proliferative effects of metformin on trastuzumab-resistant breast cancer cells via inhibition of erbB2/IGF-1 receptor interactions
    • Liu B., Fan Z., Edgerton S.M., Yang X., Lind S.E., Thor A.D. Potent anti-proliferative effects of metformin on trastuzumab-resistant breast cancer cells via inhibition of erbB2/IGF-1 receptor interactions. Cell Cycle 2011, 10:2959-2966.
    • (2011) Cell Cycle , vol.10 , pp. 2959-2966
    • Liu, B.1    Fan, Z.2    Edgerton, S.M.3    Yang, X.4    Lind, S.E.5    Thor, A.D.6
  • 151
    • 77951708711 scopus 로고    scopus 로고
    • Crosstalk between insulin/insulin-like growth factor-1 receptors and G protein-coupled receptor signaling systems: a novel target for the antidiabetic drug metformin in pancreatic cancer
    • Rozengurt E., Sinnett-Smith J., Kisfalvi K. Crosstalk between insulin/insulin-like growth factor-1 receptors and G protein-coupled receptor signaling systems: a novel target for the antidiabetic drug metformin in pancreatic cancer. Clin. Cancer Res. 2010, 16:2505-2511.
    • (2010) Clin. Cancer Res. , vol.16 , pp. 2505-2511
    • Rozengurt, E.1    Sinnett-Smith, J.2    Kisfalvi, K.3
  • 152
    • 69249162223 scopus 로고    scopus 로고
    • Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth
    • Kisfalvi K., Eibl G., Sinnett-Smith J., Rozengurt E. Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res. 2009, 69:6539-6545.
    • (2009) Cancer Res. , vol.69 , pp. 6539-6545
    • Kisfalvi, K.1    Eibl, G.2    Sinnett-Smith, J.3    Rozengurt, E.4
  • 153
    • 66849093704 scopus 로고    scopus 로고
    • Genome-wide inhibitory impact of the AMPK activator metformin on [kinesins, tubulins, histones, auroras and polo-like kinases] M-phase cell cycle genes in human breast cancer cells
    • Oliveras-Ferraros C., Vazquez-Martin A., Menendez J.A. Genome-wide inhibitory impact of the AMPK activator metformin on [kinesins, tubulins, histones, auroras and polo-like kinases] M-phase cell cycle genes in human breast cancer cells. Cell Cycle 2009, 8:1633-1636.
    • (2009) Cell Cycle , vol.8 , pp. 1633-1636
    • Oliveras-Ferraros, C.1    Vazquez-Martin, A.2    Menendez, J.A.3
  • 154
    • 65349175605 scopus 로고    scopus 로고
    • Metformin inhibits breast cancer cell growth, colony formation and induces cell cycle arrest in vitro
    • Alimova I.N., Liu B., Fan Z., Edgerton S.M., Dillon T., Lind S.E., Thor A.D. Metformin inhibits breast cancer cell growth, colony formation and induces cell cycle arrest in vitro. Cell Cycle 2009, 8:909-915.
    • (2009) Cell Cycle , vol.8 , pp. 909-915
    • Alimova, I.N.1    Liu, B.2    Fan, Z.3    Edgerton, S.M.4    Dillon, T.5    Lind, S.E.6    Thor, A.D.7
  • 155
    • 58149354390 scopus 로고    scopus 로고
    • Cell cycle arrest in metformin treated breast cancer cells involves activation of AMPK, downregulation of cyclin D1, and requires p27Kip1 or p21Cip1
    • Zhuang Y., Miskimins W.K. Cell cycle arrest in metformin treated breast cancer cells involves activation of AMPK, downregulation of cyclin D1, and requires p27Kip1 or p21Cip1. J. Mol. Signal. 2008, 3:18.
    • (2008) J. Mol. Signal. , vol.3 , pp. 18
    • Zhuang, Y.1    Miskimins, W.K.2
  • 156
    • 84857467120 scopus 로고    scopus 로고
    • Metformin induces apoptosis of lung cancer cells through activating JNK/p38 MAPK pathway and GADD153
    • Wu N., Gu C., Gu H., Hu H., Han Y., Li Q. Metformin induces apoptosis of lung cancer cells through activating JNK/p38 MAPK pathway and GADD153. Neoplasma 2011, 58:482-490.
    • (2011) Neoplasma , vol.58 , pp. 482-490
    • Wu, N.1    Gu, C.2    Gu, H.3    Hu, H.4    Han, Y.5    Li, Q.6
  • 157
    • 80051607846 scopus 로고    scopus 로고
    • Antidiabetic drug metformin induces apoptosis in human MCF breast cancer via targeting ERK signaling
    • Malki A., Youssef A. Antidiabetic drug metformin induces apoptosis in human MCF breast cancer via targeting ERK signaling. Oncol. Res. 2011, 19:275-285.
    • (2011) Oncol. Res. , vol.19 , pp. 275-285
    • Malki, A.1    Youssef, A.2
  • 159
    • 79957438835 scopus 로고    scopus 로고
    • Induction of apoptosis by metformin in epithelial ovarian cancer: involvement of the Bcl-2 family proteins
    • Yasmeen A., Beauchamp M.C., Piura E., Segal E., Pollak M., Gotlieb W.H. Induction of apoptosis by metformin in epithelial ovarian cancer: involvement of the Bcl-2 family proteins. Gynecol. Oncol. 2011, 121:492-498.
    • (2011) Gynecol. Oncol. , vol.121 , pp. 492-498
    • Yasmeen, A.1    Beauchamp, M.C.2    Piura, E.3    Segal, E.4    Pollak, M.5    Gotlieb, W.H.6
  • 160
  • 161
    • 79955834631 scopus 로고    scopus 로고
    • Metformin suppresses ovarian cancer growth and metastasis with enhancement of cisplatin cytotoxicity in vivo
    • Rattan R., Graham R.P., Maguire J.L., Giri S., Shridhar V. Metformin suppresses ovarian cancer growth and metastasis with enhancement of cisplatin cytotoxicity in vivo. Neoplasia 2011, 13:483-491.
    • (2011) Neoplasia , vol.13 , pp. 483-491
    • Rattan, R.1    Graham, R.P.2    Maguire, J.L.3    Giri, S.4    Shridhar, V.5
  • 162
    • 58149156184 scopus 로고    scopus 로고
    • Therapeutic metformin/AMPK activation promotes the angiogenic phenotype in the ERalpha negative MDA-MB-435 breast cancer model
    • Phoenix K.N., Vumbaca F., Claffey K.P. Therapeutic metformin/AMPK activation promotes the angiogenic phenotype in the ERalpha negative MDA-MB-435 breast cancer model. Breast Cancer Res. Treat. 2009, 113:101-111.
    • (2009) Breast Cancer Res. Treat. , vol.113 , pp. 101-111
    • Phoenix, K.N.1    Vumbaca, F.2    Claffey, K.P.3
  • 163
    • 77956185559 scopus 로고    scopus 로고
    • Dietary energy availability affects primary and metastatic breast cancer and metformin efficacy
    • Phoenix K.N., Vumbaca F., Fox M.M., Evans R., Claffey K.P. Dietary energy availability affects primary and metastatic breast cancer and metformin efficacy. Breast Cancer Res. Treat. 2010, 123:333-344.
    • (2010) Breast Cancer Res. Treat. , vol.123 , pp. 333-344
    • Phoenix, K.N.1    Vumbaca, F.2    Fox, M.M.3    Evans, R.4    Claffey, K.P.5
  • 164
    • 84859526277 scopus 로고    scopus 로고
    • Metformin inhibits growth of thyroid carcinoma cells, suppresses self-renewal of derived cancer stem cells, and potentiates the effect of chemotherapeutic agents
    • Chen G., Xu S., Renko K., Derwahl M. Metformin inhibits growth of thyroid carcinoma cells, suppresses self-renewal of derived cancer stem cells, and potentiates the effect of chemotherapeutic agents. J. Clin. Endocrinol. Metab. 2012, 97:E510-520.
    • (2012) J. Clin. Endocrinol. Metab. , vol.97
    • Chen, G.1    Xu, S.2    Renko, K.3    Derwahl, M.4
  • 165
    • 70350236538 scopus 로고    scopus 로고
    • Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission
    • Hirsch H.A., Iliopoulos D., Tsichlis P.N., Struhl K. Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res. 2009, 69:7507-7511.
    • (2009) Cancer Res. , vol.69 , pp. 7507-7511
    • Hirsch, H.A.1    Iliopoulos, D.2    Tsichlis, P.N.3    Struhl, K.4
  • 166
    • 84858745832 scopus 로고    scopus 로고
    • Metformin potentiates the effects of paclitaxel in endometrial cancer cells through inhibition of cell proliferation and modulation of the mTOR pathway
    • Hanna R.K., Zhou C., Malloy K.M., Gehrig P.A., Bae-Jump V.L. Metformin potentiates the effects of paclitaxel in endometrial cancer cells through inhibition of cell proliferation and modulation of the mTOR pathway. Gynecol. Oncol. 2012, 125:458-469.
    • (2012) Gynecol. Oncol. , vol.125 , pp. 458-469
    • Hanna, R.K.1    Zhou, C.2    Malloy, K.M.3    Gehrig, P.A.4    Bae-Jump, V.L.5
  • 167
    • 84856287736 scopus 로고    scopus 로고
    • A humanized anti-IGF-1R monoclonal antibody (R1507) and/or metformin
    • Kawanami T., Takiguchi S., Ikeda N., Funakoshi A. A humanized anti-IGF-1R monoclonal antibody (R1507) and/or metformin. Oncol. Rep. 2012, 27:867-872.
    • (2012) Oncol. Rep. , vol.27 , pp. 867-872
    • Kawanami, T.1    Takiguchi, S.2    Ikeda, N.3    Funakoshi, A.4
  • 169
    • 79955490053 scopus 로고    scopus 로고
    • Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types
    • Iliopoulos D., Hirsch H.A., Struhl K. Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types. Cancer Res. 2011, 71:3196-3201.
    • (2011) Cancer Res. , vol.71 , pp. 3196-3201
    • Iliopoulos, D.1    Hirsch, H.A.2    Struhl, K.3
  • 171
    • 61449113987 scopus 로고    scopus 로고
    • The effects of adiponectin and metformin on prostate and colon neoplasia involve activation of AMP-activated protein kinase
    • Zakikhani M., Dowling R.J., Sonenberg N., Pollak M.N. The effects of adiponectin and metformin on prostate and colon neoplasia involve activation of AMP-activated protein kinase. Cancer Prev. Res. (Phila) 2008, 1:369-375.
    • (2008) Cancer Prev. Res. (Phila) , vol.1 , pp. 369-375
    • Zakikhani, M.1    Dowling, R.J.2    Sonenberg, N.3    Pollak, M.N.4
  • 172
    • 75149186480 scopus 로고    scopus 로고
    • Metformin decreases IGF1-induced cell proliferation and protein synthesis through AMP-activated protein kinase in cultured bovine granulosa cells
    • Tosca L., Rame C., Chabrolle C., Tesseraud S., Dupont J. Metformin decreases IGF1-induced cell proliferation and protein synthesis through AMP-activated protein kinase in cultured bovine granulosa cells. Reproduction 2010, 139:409-418.
    • (2010) Reproduction , vol.139 , pp. 409-418
    • Tosca, L.1    Rame, C.2    Chabrolle, C.3    Tesseraud, S.4    Dupont, J.5
  • 173
    • 77956150294 scopus 로고    scopus 로고
    • Metformin inhibits aromatase expression in human breast adipose stromal cells via stimulation of AMP-activated protein kinase
    • Brown K.A., Hunger N.I., Docanto M., Simpson E.R. Metformin inhibits aromatase expression in human breast adipose stromal cells via stimulation of AMP-activated protein kinase. Breast Cancer Res. Treat. 2010, 123:591-596.
    • (2010) Breast Cancer Res. Treat. , vol.123 , pp. 591-596
    • Brown, K.A.1    Hunger, N.I.2    Docanto, M.3    Simpson, E.R.4
  • 175
    • 78751692291 scopus 로고    scopus 로고
    • Metformin attenuates ovarian cancer cell growth in an AMP-kinase dispensable manner
    • Rattan R., Giri S., Hartmann L.C., Shridhar V. Metformin attenuates ovarian cancer cell growth in an AMP-kinase dispensable manner. J. Cell. Mol. Med. 2011, 15:166-178.
    • (2011) J. Cell. Mol. Med. , vol.15 , pp. 166-178
    • Rattan, R.1    Giri, S.2    Hartmann, L.C.3    Shridhar, V.4
  • 177
    • 0034614420 scopus 로고    scopus 로고
    • Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I
    • El-Mir M.Y., Nogueira V., Fontaine E., Averet N., Rigoulet M., Leverve X. Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J. Biol. Chem. 2000, 275:223-228.
    • (2000) J. Biol. Chem. , vol.275 , pp. 223-228
    • El-Mir, M.Y.1    Nogueira, V.2    Fontaine, E.3    Averet, N.4    Rigoulet, M.5    Leverve, X.6
  • 178
    • 0034659785 scopus 로고    scopus 로고
    • Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain
    • Owen M.R., Doran E., Halestrap A.P. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem. J. 2000, 348(Pt 3):607-614.
    • (2000) Biochem. J. , vol.348 , Issue.PART 3 , pp. 607-614
    • Owen, M.R.1    Doran, E.2    Halestrap, A.P.3
  • 179
    • 39449096289 scopus 로고    scopus 로고
    • Phosphorylation of LKB1 at serine 428 by protein kinase C-zeta is required for metformin-enhanced activation of the AMP-activated protein kinase in endothelial cells
    • Xie Z., Dong Y., Scholz R., Neumann D., Zou M.H. Phosphorylation of LKB1 at serine 428 by protein kinase C-zeta is required for metformin-enhanced activation of the AMP-activated protein kinase in endothelial cells. Circulation 2008, 117:952-962.
    • (2008) Circulation , vol.117 , pp. 952-962
    • Xie, Z.1    Dong, Y.2    Scholz, R.3    Neumann, D.4    Zou, M.H.5
  • 181
    • 36348950449 scopus 로고    scopus 로고
    • Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells
    • Dowling R.J., Zakikhani M., Fantus I.G., Pollak M., Sonenberg N. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res. 2007, 67:10804-10812.
    • (2007) Cancer Res. , vol.67 , pp. 10804-10812
    • Dowling, R.J.1    Zakikhani, M.2    Fantus, I.G.3    Pollak, M.4    Sonenberg, N.5
  • 182
    • 78650931836 scopus 로고    scopus 로고
    • Metformin activates AMP kinase through inhibition of AMP deaminase
    • Ouyang J., Parakhia R.A., Ochs R.S. Metformin activates AMP kinase through inhibition of AMP deaminase. J. Biol. Chem. 2011, 286:1-11.
    • (2011) J. Biol. Chem. , vol.286 , pp. 1-11
    • Ouyang, J.1    Parakhia, R.A.2    Ochs, R.S.3
  • 187
    • 33845707278 scopus 로고    scopus 로고
    • Statins activate AMP-activated protein kinase in vitro and in vivo
    • Sun W., Lee T.S., Zhu M., Gu C., Wang Y., Zhu Y., Shyy J.Y. Statins activate AMP-activated protein kinase in vitro and in vivo. Circulation 2006, 114:2655-2662.
    • (2006) Circulation , vol.114 , pp. 2655-2662
    • Sun, W.1    Lee, T.S.2    Zhu, M.3    Gu, C.4    Wang, Y.5    Zhu, Y.6    Shyy, J.Y.7
  • 189
    • 33749349202 scopus 로고    scopus 로고
    • Polyphenols stimulate AMP-activated protein kinase, lower lipids, and inhibit accelerated atherosclerosis in diabetic LDL receptor-deficient mice
    • Zang M., Xu S., Maitland-Toolan K.A., Zuccollo A., Hou X., Jiang B., Wierzbicki M., Verbeuren T.J., Cohen R.A. Polyphenols stimulate AMP-activated protein kinase, lower lipids, and inhibit accelerated atherosclerosis in diabetic LDL receptor-deficient mice. Diabetes 2006, 55:2180-2191.
    • (2006) Diabetes , vol.55 , pp. 2180-2191
    • Zang, M.1    Xu, S.2    Maitland-Toolan, K.A.3    Zuccollo, A.4    Hou, X.5    Jiang, B.6    Wierzbicki, M.7    Verbeuren, T.J.8    Cohen, R.A.9
  • 190
    • 33744804815 scopus 로고    scopus 로고
    • Inhibition of lipid synthesis through activation of AMP kinase: an additional mechanism for the hypolipidemic effects of berberine
    • Brusq J.M., Ancellin N., Grondin P., Guillard R., Martin S., Saintillan Y., Issandou M. Inhibition of lipid synthesis through activation of AMP kinase: an additional mechanism for the hypolipidemic effects of berberine. J. Lipid Res. 2006, 47:1281-1288.
    • (2006) J. Lipid Res. , vol.47 , pp. 1281-1288
    • Brusq, J.M.1    Ancellin, N.2    Grondin, P.3    Guillard, R.4    Martin, S.5    Saintillan, Y.6    Issandou, M.7
  • 191
    • 0028073143 scopus 로고
    • Inhibition of lipolysis and lipogenesis in isolated rat adipocytes with AICAR, a cell-permeable activator of AMP-activated protein kinase
    • Sullivan J.E., Brocklehurst K.J., Marley A.E., Carey F., Carling D., Beri R.K. Inhibition of lipolysis and lipogenesis in isolated rat adipocytes with AICAR, a cell-permeable activator of AMP-activated protein kinase. FEBS Lett. 1994, 353:33-36.
    • (1994) FEBS Lett. , vol.353 , pp. 33-36
    • Sullivan, J.E.1    Brocklehurst, K.J.2    Marley, A.E.3    Carey, F.4    Carling, D.5    Beri, R.K.6
  • 192
    • 85047689953 scopus 로고
    • 5-aminoimidazole-4-carboxamide ribonucleoside. A specific method for activating AMP-activated protein kinase in intact cells?
    • Corton J.M., Gillespie J.G., Hawley S.A., Hardie D.G. 5-aminoimidazole-4-carboxamide ribonucleoside. A specific method for activating AMP-activated protein kinase in intact cells?. Eur. J. Biochem. 1995, 229:558-565.
    • (1995) Eur. J. Biochem. , vol.229 , pp. 558-565
    • Corton, J.M.1    Gillespie, J.G.2    Hawley, S.A.3    Hardie, D.G.4
  • 196
    • 3543025724 scopus 로고    scopus 로고
    • AMP-activated protein kinase activators can inhibit the growth of prostate cancer cells by multiple mechanisms
    • Xiang X., Saha A.K., Wen R., Ruderman N.B., Luo Z. AMP-activated protein kinase activators can inhibit the growth of prostate cancer cells by multiple mechanisms. Biochem. Biophys. Res. Commun. 2004, 321:161-167.
    • (2004) Biochem. Biophys. Res. Commun. , vol.321 , pp. 161-167
    • Xiang, X.1    Saha, A.K.2    Wen, R.3    Ruderman, N.B.4    Luo, Z.5
  • 197
    • 33644978143 scopus 로고    scopus 로고
    • Rosiglitazone suppresses human lung carcinoma cell growth through PPARgamma-dependent and PPARgamma-independent signal pathways
    • Han S., Roman J. Rosiglitazone suppresses human lung carcinoma cell growth through PPARgamma-dependent and PPARgamma-independent signal pathways. Mol. Cancer Ther. 2006, 5:430-437.
    • (2006) Mol. Cancer Ther. , vol.5 , pp. 430-437
    • Han, S.1    Roman, J.2
  • 198
    • 77957367974 scopus 로고    scopus 로고
    • Inhibition of autophagy enhances anticancer effects of atorvastatin in digestive malignancies
    • Yang P.M., Liu Y.L., Lin Y.C., Shun C.T., Wu M.S., Chen C.C. Inhibition of autophagy enhances anticancer effects of atorvastatin in digestive malignancies. Cancer Res. 2010, 70:7699-7709.
    • (2010) Cancer Res. , vol.70 , pp. 7699-7709
    • Yang, P.M.1    Liu, Y.L.2    Lin, Y.C.3    Shun, C.T.4    Wu, M.S.5    Chen, C.C.6
  • 200
    • 84455208086 scopus 로고    scopus 로고
    • Berberine-induced AMPK activation inhibits the metastatic potential of melanoma cells via reduction of ERK activity and COX-2 protein expression
    • Kim H.S., Kim M.J., Kim E.J., Yang Y., Lee M.S., Lim J.S. Berberine-induced AMPK activation inhibits the metastatic potential of melanoma cells via reduction of ERK activity and COX-2 protein expression. Biochem. Pharmacol. 2012, 83:385-394.
    • (2012) Biochem. Pharmacol. , vol.83 , pp. 385-394
    • Kim, H.S.1    Kim, M.J.2    Kim, E.J.3    Yang, Y.4    Lee, M.S.5    Lim, J.S.6
  • 201
    • 34247844379 scopus 로고    scopus 로고
    • Resveratrol induces apoptosis in chemoresistant cancer cells via modulation of AMPK signaling pathway
    • Hwang J.T., Kwak D.W., Lin S.K., Kim H.M., Kim Y.M., Park O.J. Resveratrol induces apoptosis in chemoresistant cancer cells via modulation of AMPK signaling pathway. Ann. N. Y. Acad. Sci. 2007, 1095:441-448.
    • (2007) Ann. N. Y. Acad. Sci. , vol.1095 , pp. 441-448
    • Hwang, J.T.1    Kwak, D.W.2    Lin, S.K.3    Kim, H.M.4    Kim, Y.M.5    Park, O.J.6
  • 202
    • 34548653197 scopus 로고    scopus 로고
    • Activation of adenosine monophosphate activated protein kinase inhibits growth of multiple myeloma cells
    • Baumann P., Mandl-Weber S., Emmerich B., Straka C., Schmidmaier R. Activation of adenosine monophosphate activated protein kinase inhibits growth of multiple myeloma cells. Exp. Cell Res. 2007, 313:3592-3603.
    • (2007) Exp. Cell Res. , vol.313 , pp. 3592-3603
    • Baumann, P.1    Mandl-Weber, S.2    Emmerich, B.3    Straka, C.4    Schmidmaier, R.5
  • 203
    • 67650514082 scopus 로고    scopus 로고
    • Metformin induces unique biological and molecular responses in triple negative breast cancer cells
    • Liu B., Fan Z., Edgerton S.M., Deng X.S., Alimova I.N., Lind S.E., Thor A.D. Metformin induces unique biological and molecular responses in triple negative breast cancer cells. Cell Cycle 2009, 8:2031-2040.
    • (2009) Cell Cycle , vol.8 , pp. 2031-2040
    • Liu, B.1    Fan, Z.2    Edgerton, S.M.3    Deng, X.S.4    Alimova, I.N.5    Lind, S.E.6    Thor, A.D.7
  • 204
    • 33751284806 scopus 로고    scopus 로고
    • Metformin is an AMP kinase-dependent growth inhibitor for breast cancer cells
    • Zakikhani M., Dowling R., Fantus I.G., Sonenberg N., Pollak M. Metformin is an AMP kinase-dependent growth inhibitor for breast cancer cells. Cancer Res. 2006, 66:10269-10273.
    • (2006) Cancer Res. , vol.66 , pp. 10269-10273
    • Zakikhani, M.1    Dowling, R.2    Fantus, I.G.3    Sonenberg, N.4    Pollak, M.5
  • 205
    • 79955980904 scopus 로고    scopus 로고
    • Metformin induces both caspase-dependent and poly(ADP-ribose) polymerase-dependent cell death in breast cancer cells
    • Zhuang Y., Miskimins W.K. Metformin induces both caspase-dependent and poly(ADP-ribose) polymerase-dependent cell death in breast cancer cells. Mol. Cancer Res. 2011, 9:603-615.
    • (2011) Mol. Cancer Res. , vol.9 , pp. 603-615
    • Zhuang, Y.1    Miskimins, W.K.2
  • 208
  • 211
    • 79952290187 scopus 로고    scopus 로고
    • Metformin treatment exerts antiinvasive and antimetastatic effects in human endometrial carcinoma cells
    • Tan B.K., Adya R., Chen J., Lehnert H., Sant Cassia L.J., Randeva H.S. Metformin treatment exerts antiinvasive and antimetastatic effects in human endometrial carcinoma cells. J. Clin. Endocrinol. Metab. 2011, 96:808-816.
    • (2011) J. Clin. Endocrinol. Metab. , vol.96 , pp. 808-816
    • Tan, B.K.1    Adya, R.2    Chen, J.3    Lehnert, H.4    Sant Cassia, L.J.5    Randeva, H.S.6
  • 212
    • 83455225509 scopus 로고    scopus 로고
    • Antileukemic effects of AMPK activators on BCR-ABL-expressing cells
    • Vakana E., Altman J.K., Glaser H., Donato N.J., Platanias L.C. Antileukemic effects of AMPK activators on BCR-ABL-expressing cells. Blood 2011, 118:6399-6402.
    • (2011) Blood , vol.118 , pp. 6399-6402
    • Vakana, E.1    Altman, J.K.2    Glaser, H.3    Donato, N.J.4    Platanias, L.C.5
  • 215
    • 77953589054 scopus 로고    scopus 로고
    • Metformin blocks the stimulative effect of a high-energy diet on colon carcinoma growth in vivo and is associated with reduced expression of fatty acid synthase
    • Algire C., Amrein L., Zakikhani M., Panasci L., Pollak M. Metformin blocks the stimulative effect of a high-energy diet on colon carcinoma growth in vivo and is associated with reduced expression of fatty acid synthase. Endocr. Relat. Cancer 2010, 17:351-360.
    • (2010) Endocr. Relat. Cancer , vol.17 , pp. 351-360
    • Algire, C.1    Amrein, L.2    Zakikhani, M.3    Panasci, L.4    Pollak, M.5
  • 221
    • 0035897909 scopus 로고    scopus 로고
    • Advances in transplantation tolerance
    • Yu X., Carpenter P., Anasetti C. Advances in transplantation tolerance. Lancet 2001, 357:1959-1963.
    • (2001) Lancet , vol.357 , pp. 1959-1963
    • Yu, X.1    Carpenter, P.2    Anasetti, C.3
  • 222
    • 0026659046 scopus 로고
    • Rapamycin-FKBP specifically blocks growth-dependent activation of and signaling by the 70 kd S6 protein kinases
    • Chung J., Kuo C.J., Crabtree G.R., Blenis J. Rapamycin-FKBP specifically blocks growth-dependent activation of and signaling by the 70 kd S6 protein kinases. Cell 1992, 69:1227-1236.
    • (1992) Cell , vol.69 , pp. 1227-1236
    • Chung, J.1    Kuo, C.J.2    Crabtree, G.R.3    Blenis, J.4
  • 223
  • 224
    • 0034790016 scopus 로고    scopus 로고
    • MTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer
    • Yu K., Toral-Barza L., Discafani C., Zhang W.G., Skotnicki J., Frost P., Gibbons J.J. mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocr. Relat. Cancer 2001, 8:249-258.
    • (2001) Endocr. Relat. Cancer , vol.8 , pp. 249-258
    • Yu, K.1    Toral-Barza, L.2    Discafani, C.3    Zhang, W.G.4    Skotnicki, J.5    Frost, P.6    Gibbons, J.J.7
  • 226
    • 16844371494 scopus 로고    scopus 로고
    • Effects of the mammalian target of rapamycin inhibitor CCI-779 used alone or with chemotherapy on human prostate cancer cells and xenografts
    • Wu L., Birle D.C., Tannock I.F. Effects of the mammalian target of rapamycin inhibitor CCI-779 used alone or with chemotherapy on human prostate cancer cells and xenografts. Cancer Res. 2005, 65:2825-2831.
    • (2005) Cancer Res. , vol.65 , pp. 2825-2831
    • Wu, L.1    Birle, D.C.2    Tannock, I.F.3
  • 228
    • 33645690146 scopus 로고    scopus 로고
    • In vivo antitumor effect of the mTOR inhibitor CCI-779 and gemcitabine in xenograft models of human pancreatic cancer
    • Ito D., Fujimoto K., Mori T., Kami K., Koizumi M., Toyoda E., Kawaguchi Y., Doi R. In vivo antitumor effect of the mTOR inhibitor CCI-779 and gemcitabine in xenograft models of human pancreatic cancer. Int. J. Cancer 2006, 118:2337-2343.
    • (2006) Int. J. Cancer , vol.118 , pp. 2337-2343
    • Ito, D.1    Fujimoto, K.2    Mori, T.3    Kami, K.4    Koizumi, M.5    Toyoda, E.6    Kawaguchi, Y.7    Doi, R.8
  • 229
    • 37749051218 scopus 로고    scopus 로고
    • Rapamycin enhances the anti-tumor effect of gemcitabine in pancreatic cancer cells
    • Okada T., Sawada T., Kubota K. Rapamycin enhances the anti-tumor effect of gemcitabine in pancreatic cancer cells. Hepatogastroenterology 2007, 54:2129-2133.
    • (2007) Hepatogastroenterology , vol.54 , pp. 2129-2133
    • Okada, T.1    Sawada, T.2    Kubota, K.3
  • 232
    • 79956088595 scopus 로고    scopus 로고
    • The long and winding road to rational treatment of cancer associated with LKB1/AMPK/TSC/mTORC1 signaling
    • van Veelen W., Korsse S.E., van de Laar L., Peppelenbosch M.P. The long and winding road to rational treatment of cancer associated with LKB1/AMPK/TSC/mTORC1 signaling. Oncogene 2011, 30:2289-2303.
    • (2011) Oncogene , vol.30 , pp. 2289-2303
    • van Veelen, W.1    Korsse, S.E.2    van de Laar, L.3    Peppelenbosch, M.P.4
  • 243
    • 83255167036 scopus 로고    scopus 로고
    • Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study
    • Pavel M.E., Hainsworth J.D., Baudin E., Peeters M., Horsch D., Winkler R.E., Klimovsky J., Lebwohl D., Jehl V., Wolin E.M., Oberg K., Van Cutsem E., Yao J.C. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 2011, 378:2005-2012.
    • (2011) Lancet , vol.378 , pp. 2005-2012
    • Pavel, M.E.1    Hainsworth, J.D.2    Baudin, E.3    Peeters, M.4    Horsch, D.5    Winkler, R.E.6    Klimovsky, J.7    Lebwohl, D.8    Jehl, V.9    Wolin, E.M.10    Oberg, K.11    Van Cutsem, E.12    Yao, J.C.13
  • 248
    • 9244219598 scopus 로고    scopus 로고
    • Breast cancer-loss of PTEN predicts resistance to treatment
    • Pandolfi P.P. Breast cancer-loss of PTEN predicts resistance to treatment. N. Engl. J. Med. 2004, 351:2337-2338.
    • (2004) N. Engl. J. Med. , vol.351 , pp. 2337-2338
    • Pandolfi, P.P.1
  • 251
    • 34247127783 scopus 로고    scopus 로고
    • Phosphorylation of ribosomal p70 S6 kinase and rapamycin sensitivity in human colorectal cancer
    • Nozawa H., Watanabe T., Nagawa H. Phosphorylation of ribosomal p70 S6 kinase and rapamycin sensitivity in human colorectal cancer. Cancer Lett. 2007, 251:105-113.
    • (2007) Cancer Lett. , vol.251 , pp. 105-113
    • Nozawa, H.1    Watanabe, T.2    Nagawa, H.3
  • 255
    • 79958806173 scopus 로고    scopus 로고
    • MTOR kinase inhibitor AZD8055 enhances the immunotherapeutic activity of an agonist CD40 antibody in cancer treatment
    • Jiang Q., Weiss J.M., Back T., Chan T., Ortaldo J.R., Guichard S., Wiltrout R.H. mTOR kinase inhibitor AZD8055 enhances the immunotherapeutic activity of an agonist CD40 antibody in cancer treatment. Cancer Res. 2011, 71:4074-4084.
    • (2011) Cancer Res. , vol.71 , pp. 4074-4084
    • Jiang, Q.1    Weiss, J.M.2    Back, T.3    Chan, T.4    Ortaldo, J.R.5    Guichard, S.6    Wiltrout, R.H.7
  • 260
    • 84862779787 scopus 로고    scopus 로고
    • Inhibition of tumor cell growth, proliferation and migration by X-387, a novel active-site inhibitor of mTOR
    • Chen S.M., Liu J.L., Wang X., Liang C., Ding J., Meng L.H. Inhibition of tumor cell growth, proliferation and migration by X-387, a novel active-site inhibitor of mTOR. Biochem. Pharmacol. 2012, 83:1183-1194.
    • (2012) Biochem. Pharmacol. , vol.83 , pp. 1183-1194
    • Chen, S.M.1    Liu, J.L.2    Wang, X.3    Liang, C.4    Ding, J.5    Meng, L.H.6
  • 262
    • 67749110462 scopus 로고    scopus 로고
    • Oral rapamycin reduces tumour burden and vascularization in Lkb1(+/-) mice
    • Robinson J., Lai C., Martin A., Nye E., Tomlinson I., Silver A. Oral rapamycin reduces tumour burden and vascularization in Lkb1(+/-) mice. J. Pathol. 2009, 219:35-40.
    • (2009) J. Pathol. , vol.219 , pp. 35-40
    • Robinson, J.1    Lai, C.2    Martin, A.3    Nye, E.4    Tomlinson, I.5    Silver, A.6
  • 264
    • 62749135926 scopus 로고    scopus 로고
    • Chemopreventive efficacy of rapamycin on Peutz-Jeghers syndrome in a mouse model
    • Wei C., Amos C.I., Zhang N., Zhu J., Wang X., Frazier M.L. Chemopreventive efficacy of rapamycin on Peutz-Jeghers syndrome in a mouse model. Cancer Lett. 2009, 277:149-154.
    • (2009) Cancer Lett. , vol.277 , pp. 149-154
    • Wei, C.1    Amos, C.I.2    Zhang, N.3    Zhu, J.4    Wang, X.5    Frazier, M.L.6
  • 265
    • 51649126752 scopus 로고    scopus 로고
    • Inhibition of the mTORC1 pathway suppresses intestinal polyp formation and reduces mortality in ApcDelta716 mice
    • Fujishita T., Aoki K., Lane H.A., Aoki M., Taketo M.M. Inhibition of the mTORC1 pathway suppresses intestinal polyp formation and reduces mortality in ApcDelta716 mice. Proc. Natl. Acad. Sci. U. S. A. 2008, 105:13544-13549.
    • (2008) Proc. Natl. Acad. Sci. U. S. A. , vol.105 , pp. 13544-13549
    • Fujishita, T.1    Aoki, K.2    Lane, H.A.3    Aoki, M.4    Taketo, M.M.5
  • 270
    • 65449116467 scopus 로고    scopus 로고
    • Rapamycin weekly maintenance dosing and the potential efficacy of combination sorafenib plus rapamycin but not atorvastatin or doxycycline in tuberous sclerosis preclinical models
    • Lee N., Woodrum C.L., Nobil A.M., Rauktys A.E., Messina M.P., Dabora S.L. Rapamycin weekly maintenance dosing and the potential efficacy of combination sorafenib plus rapamycin but not atorvastatin or doxycycline in tuberous sclerosis preclinical models. BMC Pharmacol. 2009, 9:8.
    • (2009) BMC Pharmacol. , vol.9 , pp. 8
    • Lee, N.1    Woodrum, C.L.2    Nobil, A.M.3    Rauktys, A.E.4    Messina, M.P.5    Dabora, S.L.6
  • 271
    • 67650077581 scopus 로고    scopus 로고
    • Equivalent benefit of mTORC1 blockade and combined PI3K-mTOR blockade in a mouse model of tuberous sclerosis
    • Pollizzi K., Malinowska-Kolodziej I., Stumm M., Lane H., Kwiatkowski D. Equivalent benefit of mTORC1 blockade and combined PI3K-mTOR blockade in a mouse model of tuberous sclerosis. Mol. Cancer 2009, 8:38.
    • (2009) Mol. Cancer , vol.8 , pp. 38
    • Pollizzi, K.1    Malinowska-Kolodziej, I.2    Stumm, M.3    Lane, H.4    Kwiatkowski, D.5
  • 275
    • 82555205486 scopus 로고    scopus 로고
    • Preclinical testing of PI3K/AKT/mTOR signaling inhibitors in a mouse model of ovarian endometrioid adenocarcinoma
    • Wu R., Hu T.C., Rehemtulla A., Fearon E.R., Cho K.R. Preclinical testing of PI3K/AKT/mTOR signaling inhibitors in a mouse model of ovarian endometrioid adenocarcinoma. Clin. Cancer Res. 2011, 17:7359-7372.
    • (2011) Clin. Cancer Res. , vol.17 , pp. 7359-7372
    • Wu, R.1    Hu, T.C.2    Rehemtulla, A.3    Fearon, E.R.4    Cho, K.R.5
  • 276
    • 77954370712 scopus 로고    scopus 로고
    • Inhibition of mTORC1 signaling reduces tumor growth but does not prevent cancer progression in a mouse model of thyroid cancer
    • Guigon C.J., Fozzatti L., Lu C., Willingham M.C., Cheng S.Y. Inhibition of mTORC1 signaling reduces tumor growth but does not prevent cancer progression in a mouse model of thyroid cancer. Carcinogenesis 2010, 31:1284-1291.
    • (2010) Carcinogenesis , vol.31 , pp. 1284-1291
    • Guigon, C.J.1    Fozzatti, L.2    Lu, C.3    Willingham, M.C.4    Cheng, S.Y.5
  • 281
    • 34548089170 scopus 로고    scopus 로고
    • Rapamycin inhibits multiple stages of c-Neu/ErbB2 induced tumor progression in a transgenic mouse model of HER2-positive breast cancer
    • Mosley J.D., Poirier J.T., Seachrist D.D., Landis M.D., Keri R.A. Rapamycin inhibits multiple stages of c-Neu/ErbB2 induced tumor progression in a transgenic mouse model of HER2-positive breast cancer. Mol. Cancer Ther. 2007, 6:2188-2197.
    • (2007) Mol. Cancer Ther. , vol.6 , pp. 2188-2197
    • Mosley, J.D.1    Poirier, J.T.2    Seachrist, D.D.3    Landis, M.D.4    Keri, R.A.5
  • 282
    • 80054791210 scopus 로고    scopus 로고
    • Multimodal biomarker investigation on efficacy and mechanism of action for the mammalian target of rapamycin inhibitor, temsirolimus, in a preclinical mammary carcinoma OncoMouse model: a translational medicine study in support for early clinical development
    • Wang X., Zhan Y., Zhao L., Alvarez J., Chaudhary I., Zhou B.B., Abraham R.T., Feuerstein G.Z. Multimodal biomarker investigation on efficacy and mechanism of action for the mammalian target of rapamycin inhibitor, temsirolimus, in a preclinical mammary carcinoma OncoMouse model: a translational medicine study in support for early clinical development. J. Pharmacol. Exp. Ther. 2011, 339:421-429.
    • (2011) J. Pharmacol. Exp. Ther. , vol.339 , pp. 421-429
    • Wang, X.1    Zhan, Y.2    Zhao, L.3    Alvarez, J.4    Chaudhary, I.5    Zhou, B.B.6    Abraham, R.T.7    Feuerstein, G.Z.8
  • 284
    • 18744407590 scopus 로고    scopus 로고
    • A genetically defined mouse ovarian carcinoma model for the molecular characterization of pathway-targeted therapy and tumor resistance
    • Xing D., Orsulic S. A genetically defined mouse ovarian carcinoma model for the molecular characterization of pathway-targeted therapy and tumor resistance. Proc. Natl. Acad. Sci. U. S. A. 2005, 102:6936-6941.
    • (2005) Proc. Natl. Acad. Sci. U. S. A. , vol.102 , pp. 6936-6941
    • Xing, D.1    Orsulic, S.2
  • 286
    • 33845227643 scopus 로고    scopus 로고
    • Rapamycin synergizes with the epidermal growth factor receptor inhibitor erlotinib in non-small-cell lung, pancreatic, colon, and breast tumors
    • Buck E., Eyzaguirre A., Brown E., Petti F., McCormack S., Haley J.D., Iwata K.K., Gibson N.W., Griffin G. Rapamycin synergizes with the epidermal growth factor receptor inhibitor erlotinib in non-small-cell lung, pancreatic, colon, and breast tumors. Mol. Cancer Ther. 2006, 5:2676-2684.
    • (2006) Mol. Cancer Ther. , vol.5 , pp. 2676-2684
    • Buck, E.1    Eyzaguirre, A.2    Brown, E.3    Petti, F.4    McCormack, S.5    Haley, J.D.6    Iwata, K.K.7    Gibson, N.W.8    Griffin, G.9
  • 287
    • 38949141134 scopus 로고    scopus 로고
    • Synergic antiproliferative and antiangiogenic effects of EGFR and mTor inhibitors on pancreatic cancer cells
    • Azzariti A., Porcelli L., Gatti G., Nicolin A., Paradiso A. Synergic antiproliferative and antiangiogenic effects of EGFR and mTor inhibitors on pancreatic cancer cells. Biochem. Pharmacol. 2008, 75:1035-1044.
    • (2008) Biochem. Pharmacol. , vol.75 , pp. 1035-1044
    • Azzariti, A.1    Porcelli, L.2    Gatti, G.3    Nicolin, A.4    Paradiso, A.5
  • 289
    • 73549121061 scopus 로고    scopus 로고
    • Effects of combined inhibition of MEK and mTOR on downstream signaling and tumor growth in pancreatic cancer xenograft models
    • Chang Q., Chen E., Hedley D.W. Effects of combined inhibition of MEK and mTOR on downstream signaling and tumor growth in pancreatic cancer xenograft models. Cancer Biol. Ther. 2009, 8:1893-1901.
    • (2009) Cancer Biol. Ther. , vol.8 , pp. 1893-1901
    • Chang, Q.1    Chen, E.2    Hedley, D.W.3
  • 290
    • 40349108627 scopus 로고    scopus 로고
    • Inhibition of mTOR pathway by everolimus cooperates with EGFR inhibitors in human tumours sensitive and resistant to anti-EGFR drugs
    • Bianco R., Garofalo S., Rosa R., Damiano V., Gelardi T., Daniele G., Marciano R., Ciardiello F., Tortora G. Inhibition of mTOR pathway by everolimus cooperates with EGFR inhibitors in human tumours sensitive and resistant to anti-EGFR drugs. Br. J. Cancer 2008, 98:923-930.
    • (2008) Br. J. Cancer , vol.98 , pp. 923-930
    • Bianco, R.1    Garofalo, S.2    Rosa, R.3    Damiano, V.4    Gelardi, T.5    Daniele, G.6    Marciano, R.7    Ciardiello, F.8    Tortora, G.9
  • 291
    • 34248576166 scopus 로고    scopus 로고
    • Rapamycin together with herceptin significantly increased anti-tumor efficacy compared to either alone in ErbB2 over expressing breast cancer cells
    • Wang L.H., Chan J.L., Li W. Rapamycin together with herceptin significantly increased anti-tumor efficacy compared to either alone in ErbB2 over expressing breast cancer cells. Int. J. Cancer 2007, 121:157-164.
    • (2007) Int. J. Cancer , vol.121 , pp. 157-164
    • Wang, L.H.1    Chan, J.L.2    Li, W.3
  • 292
    • 67149131861 scopus 로고    scopus 로고
    • Efficacy of combined inhibition of mTOR and ERK/MAPK pathways in treating a tuberous sclerosis complex cell model
    • Mi R., Ma J., Zhang D., Li L., Zhang H. Efficacy of combined inhibition of mTOR and ERK/MAPK pathways in treating a tuberous sclerosis complex cell model. J. Genet. Genomics 2009, 36:355-361.
    • (2009) J. Genet. Genomics , vol.36 , pp. 355-361
    • Mi, R.1    Ma, J.2    Zhang, D.3    Li, L.4    Zhang, H.5
  • 294
    • 79960946383 scopus 로고    scopus 로고
    • Combinatorial treatments that overcome PDGFRbeta-driven resistance of melanoma cells to V600EB-RAF inhibition
    • Shi H., Kong X., Ribas A., Lo R.S. Combinatorial treatments that overcome PDGFRbeta-driven resistance of melanoma cells to V600EB-RAF inhibition. Cancer Res. 2011, 71:5067-5074.
    • (2011) Cancer Res. , vol.71 , pp. 5067-5074
    • Shi, H.1    Kong, X.2    Ribas, A.3    Lo, R.S.4
  • 296
    • 77953609118 scopus 로고    scopus 로고
    • Compensatory activation of Akt in response to mTOR and Raf inhibitors - a rationale for dual-targeted therapy approaches in neuroendocrine tumor disease
    • Zitzmann K., Ruden J., Brand S., Goke B., Lichtl J., Spottl G., Auernhammer C.J. Compensatory activation of Akt in response to mTOR and Raf inhibitors - a rationale for dual-targeted therapy approaches in neuroendocrine tumor disease. Cancer Lett. 2010, 295:100-109.
    • (2010) Cancer Lett. , vol.295 , pp. 100-109
    • Zitzmann, K.1    Ruden, J.2    Brand, S.3    Goke, B.4    Lichtl, J.5    Spottl, G.6    Auernhammer, C.J.7
  • 297
    • 79951839137 scopus 로고    scopus 로고
    • Interpreting mammalian target of rapamycin and cell growth inhibition in a genetically engineered mouse model of Nf1-deficient astrocytes
    • Banerjee S., Gianino S.M., Gao F., Christians U., Gutmann D.H. Interpreting mammalian target of rapamycin and cell growth inhibition in a genetically engineered mouse model of Nf1-deficient astrocytes. Mol. Cancer Ther. 2011, 10:279-291.
    • (2011) Mol. Cancer Ther. , vol.10 , pp. 279-291
    • Banerjee, S.1    Gianino, S.M.2    Gao, F.3    Christians, U.4    Gutmann, D.H.5
  • 299
    • 80051537004 scopus 로고    scopus 로고
    • Characterization and targeting of phosphatidylinositol-3 kinase (PI3K) and mammalian target of rapamycin (mTOR) in renal cell cancer
    • Elfiky A.A., Aziz S.A., Conrad P.J., Siddiqui S., Hackl W., Maira M., Robert C.L., Kluger H.M. Characterization and targeting of phosphatidylinositol-3 kinase (PI3K) and mammalian target of rapamycin (mTOR) in renal cell cancer. J. Transl. Med 2011, 9:133.
    • (2011) J. Transl. Med , vol.9 , pp. 133
    • Elfiky, A.A.1    Aziz, S.A.2    Conrad, P.J.3    Siddiqui, S.4    Hackl, W.5    Maira, M.6    Robert, C.L.7    Kluger, H.M.8
  • 302
    • 64949155000 scopus 로고    scopus 로고
    • Activity of a novel, dual PI3-kinase/mTor inhibitor NVP-BEZ235 against primary human pancreatic cancers grown as orthotopic xenografts
    • Cao P., Maira S.M., Garcia-Echeverria C., Hedley D.W. Activity of a novel, dual PI3-kinase/mTor inhibitor NVP-BEZ235 against primary human pancreatic cancers grown as orthotopic xenografts. Br. J. Cancer 2009, 100:1267-1276.
    • (2009) Br. J. Cancer , vol.100 , pp. 1267-1276
    • Cao, P.1    Maira, S.M.2    Garcia-Echeverria, C.3    Hedley, D.W.4
  • 304
    • 77953932867 scopus 로고    scopus 로고
    • Dual inhibition of PI3K and mTOR inhibits autocrine and paracrine proliferative loops in PI3K/Akt/mTOR-addicted lymphomas
    • Bhatt A.P., Bhende P.M., Sin S.H., Roy D., Dittmer D.P., Damania B. Dual inhibition of PI3K and mTOR inhibits autocrine and paracrine proliferative loops in PI3K/Akt/mTOR-addicted lymphomas. Blood 2010, 115:4455-4463.
    • (2010) Blood , vol.115 , pp. 4455-4463
    • Bhatt, A.P.1    Bhende, P.M.2    Sin, S.H.3    Roy, D.4    Dittmer, D.P.5    Damania, B.6
  • 308
    • 79954603804 scopus 로고    scopus 로고
    • Dual targeting of phosphoinositide 3-kinase and mammalian target of rapamycin using NVP-BEZ235 as a novel therapeutic approach in human ovarian carcinoma
    • Santiskulvong C., Konecny G.E., Fekete M., Chen K.Y., Karam A., Mulholland D., Eng C., Wu H., Song M., Dorigo O. Dual targeting of phosphoinositide 3-kinase and mammalian target of rapamycin using NVP-BEZ235 as a novel therapeutic approach in human ovarian carcinoma. Clin. Cancer Res. 2011, 17:2373-2384.
    • (2011) Clin. Cancer Res. , vol.17 , pp. 2373-2384
    • Santiskulvong, C.1    Konecny, G.E.2    Fekete, M.3    Chen, K.Y.4    Karam, A.5    Mulholland, D.6    Eng, C.7    Wu, H.8    Song, M.9    Dorigo, O.10
  • 313
    • 70350228529 scopus 로고    scopus 로고
    • Dual phosphoinositide 3-kinase/mammalian target of rapamycin blockade is an effective radiosensitizing strategy for the treatment of non-small cell lung cancer harboring K-RAS mutations
    • Konstantinidou G., Bey E.A., Rabellino A., Schuster K., Maira M.S., Gazdar A.F., Amici A., Boothman D.A., Scaglioni P.P. Dual phosphoinositide 3-kinase/mammalian target of rapamycin blockade is an effective radiosensitizing strategy for the treatment of non-small cell lung cancer harboring K-RAS mutations. Cancer Res. 2009, 69:7644-7652.
    • (2009) Cancer Res. , vol.69 , pp. 7644-7652
    • Konstantinidou, G.1    Bey, E.A.2    Rabellino, A.3    Schuster, K.4    Maira, M.S.5    Gazdar, A.F.6    Amici, A.7    Boothman, D.A.8    Scaglioni, P.P.9
  • 315
    • 84856819520 scopus 로고    scopus 로고
    • The dual kinase inhibitor NVP-BEZ235 in combination with cytotoxic drugs exerts anti-proliferative activity towards acute lymphoblastic leukemia cells
    • Schult C., Dahlhaus M., Glass A., Fischer K., Lange S., Freund M., Junghanss C. The dual kinase inhibitor NVP-BEZ235 in combination with cytotoxic drugs exerts anti-proliferative activity towards acute lymphoblastic leukemia cells. Anticancer. Res. 2012, 32:463-474.
    • (2012) Anticancer. Res. , vol.32 , pp. 463-474
    • Schult, C.1    Dahlhaus, M.2    Glass, A.3    Fischer, K.4    Lange, S.5    Freund, M.6    Junghanss, C.7
  • 317
    • 84857579424 scopus 로고    scopus 로고
    • The efficacy of a novel, dual PI3K/mTOR inhibitor NVP-BEZ235 to enhance chemotherapy and antiangiogenic response in pancreatic cancer
    • Awasthi N., Yen P.L., Schwarz M.A., Schwarz R.E. The efficacy of a novel, dual PI3K/mTOR inhibitor NVP-BEZ235 to enhance chemotherapy and antiangiogenic response in pancreatic cancer. J. Cell. Biochem. 2012, 113:784-791.
    • (2012) J. Cell. Biochem. , vol.113 , pp. 784-791
    • Awasthi, N.1    Yen, P.L.2    Schwarz, M.A.3    Schwarz, R.E.4
  • 321
    • 80054118460 scopus 로고    scopus 로고
    • Synergistic action of a RAF inhibitor and a dual PI3K/mTOR inhibitor in thyroid cancer
    • Jin N., Jiang T., Rosen D.M., Nelkin B.D., Ball D.W. Synergistic action of a RAF inhibitor and a dual PI3K/mTOR inhibitor in thyroid cancer. Clin. Cancer Res. 2011, 17:6482-6489.
    • (2011) Clin. Cancer Res. , vol.17 , pp. 6482-6489
    • Jin, N.1    Jiang, T.2    Rosen, D.M.3    Nelkin, B.D.4    Ball, D.W.5
  • 322
    • 79960683561 scopus 로고    scopus 로고
    • Targeting renal cell carcinoma with NVP-BEZ235, a dual PI3K/mTOR inhibitor, in combination with sorafenib
    • Roulin D., Waselle L., Dormond-Meuwly A., Dufour M., Demartines N., Dormond O. Targeting renal cell carcinoma with NVP-BEZ235, a dual PI3K/mTOR inhibitor, in combination with sorafenib. Mol. Cancer 2011, 10:90.
    • (2011) Mol. Cancer , vol.10 , pp. 90
    • Roulin, D.1    Waselle, L.2    Dormond-Meuwly, A.3    Dufour, M.4    Demartines, N.5    Dormond, O.6
  • 323
    • 51349108718 scopus 로고    scopus 로고
    • Ablation of PI3K blocks BCR-ABL leukemogenesis in mice, and a dual PI3K/mTOR inhibitor prevents expansion of human BCR-ABL+leukemia cells
    • Kharas M.G., Janes M.R., Scarfone V.M., Lilly M.B., Knight Z.A., Shokat K.M., Fruman D.A. Ablation of PI3K blocks BCR-ABL leukemogenesis in mice, and a dual PI3K/mTOR inhibitor prevents expansion of human BCR-ABL+leukemia cells. J. Clin. Invest. 2008, 118:3038-3050.
    • (2008) J. Clin. Invest. , vol.118 , pp. 3038-3050
    • Kharas, M.G.1    Janes, M.R.2    Scarfone, V.M.3    Lilly, M.B.4    Knight, Z.A.5    Shokat, K.M.6    Fruman, D.A.7
  • 325
    • 78651388423 scopus 로고    scopus 로고
    • Vertical inhibition of the mTORC1/mTORC2/PI3K pathway shows synergistic effects against melanoma in vitro and in vivo
    • Werzowa J., Koehrer S., Strommer S., Cejka D., Fuereder T., Zebedin E., Wacheck V. Vertical inhibition of the mTORC1/mTORC2/PI3K pathway shows synergistic effects against melanoma in vitro and in vivo. J. Investig. Dermatol. 2011, 131:495-503.
    • (2011) J. Investig. Dermatol. , vol.131 , pp. 495-503
    • Werzowa, J.1    Koehrer, S.2    Strommer, S.3    Cejka, D.4    Fuereder, T.5    Zebedin, E.6    Wacheck, V.7
  • 330
    • 82555166037 scopus 로고    scopus 로고
    • Simultaneous targeting of PI3K and mTOR with NVP-BGT226 is highly effective in multiple myeloma
    • Baumann P., Schneider L., Mandl-Weber S., Oduncu F., Schmidmaier R. Simultaneous targeting of PI3K and mTOR with NVP-BGT226 is highly effective in multiple myeloma. Anticancer Drugs 2012, 23:131-138.
    • (2012) Anticancer Drugs , vol.23 , pp. 131-138
    • Baumann, P.1    Schneider, L.2    Mandl-Weber, S.3    Oduncu, F.4    Schmidmaier, R.5
  • 331
    • 81255129000 scopus 로고    scopus 로고
    • Novel phosphoinositide 3-kinase/mTOR dual inhibitor, NVP-BGT226, displays potent growth-inhibitory activity against human head and neck cancer cells in vitro and in vivo
    • Chang K.Y., Tsai S.Y., Wu C.M., Yen C.J., Chuang B.F., Chang J.Y. Novel phosphoinositide 3-kinase/mTOR dual inhibitor, NVP-BGT226, displays potent growth-inhibitory activity against human head and neck cancer cells in vitro and in vivo. Clin. Cancer Res. 2011, 17:7116-7126.
    • (2011) Clin. Cancer Res. , vol.17 , pp. 7116-7126
    • Chang, K.Y.1    Tsai, S.Y.2    Wu, C.M.3    Yen, C.J.4    Chuang, B.F.5    Chang, J.Y.6
  • 332
    • 84863576741 scopus 로고    scopus 로고
    • The dual PI3K/mTOR inhibitor NVP-BGT226 induces cell cycle arrest and regulates Survivin gene expression in human pancreatic cancer cell lines
    • Glienke W., Maute L., Wicht J., Bergmann L. The dual PI3K/mTOR inhibitor NVP-BGT226 induces cell cycle arrest and regulates Survivin gene expression in human pancreatic cancer cell lines. Tumour Biol. 2011, 33:757-765.
    • (2011) Tumour Biol. , vol.33 , pp. 757-765
    • Glienke, W.1    Maute, L.2    Wicht, J.3    Bergmann, L.4
  • 333
    • 79957818433 scopus 로고    scopus 로고
    • Comparison of the effects of the PI3K/mTOR inhibitors NVP-BEZ235 and GSK2126458 on tamoxifen-resistant breast cancer cells
    • Leung E., Kim J.E., Rewcastle G.W., Finlay G.J., Baguley B.C. Comparison of the effects of the PI3K/mTOR inhibitors NVP-BEZ235 and GSK2126458 on tamoxifen-resistant breast cancer cells. Cancer Biol. Ther. 2011, 11:938-946.
    • (2011) Cancer Biol. Ther. , vol.11 , pp. 938-946
    • Leung, E.1    Kim, J.E.2    Rewcastle, G.W.3    Finlay, G.J.4    Baguley, B.C.5
  • 337
    • 0344862109 scopus 로고    scopus 로고
    • Correlation of staining for LKB1 and COX-2 in hamartomatous polyps and carcinomas from patients with Peutz-Jeghers syndrome
    • Wei C., Amos C.I., Rashid A., Sabripour M., Nations L., McGarrity T.J., Frazier M.L. Correlation of staining for LKB1 and COX-2 in hamartomatous polyps and carcinomas from patients with Peutz-Jeghers syndrome. J. Histochem. Cytochem. 2003, 51:1665-1672.
    • (2003) J. Histochem. Cytochem. , vol.51 , pp. 1665-1672
    • Wei, C.1    Amos, C.I.2    Rashid, A.3    Sabripour, M.4    Nations, L.5    McGarrity, T.J.6    Frazier, M.L.7


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.