메뉴 건너뛰기




Volumn 42, Issue 7, 2015, Pages 1093-1105

Identification of imaging biomarkers for the assessment of tumour response to different treatments in a preclinical glioma model

Author keywords

Biomarker; CAIX; HIF 1alpha; Optical imaging; Temozolomide

Indexed keywords

ANTINEOPLASTIC AGENT; CARBONATE DEHYDRATASE; DACARBAZINE; HIF1A PROTEIN, HUMAN; HYPOXIA INDUCIBLE FACTOR 1ALPHA; PHOTOPROTEIN; TEMOZOLOMIDE; TUMOR MARKER;

EID: 84939934765     PISSN: 16197070     EISSN: 16197089     Source Type: Journal    
DOI: 10.1007/s00259-015-3040-7     Document Type: Article
Times cited : (20)

References (54)
  • 1
    • 33747178432 scopus 로고    scopus 로고
    • Hypoxia in the tumorigenesis of gliomas and as a potential target for therapeutic measures
    • PID: 16709030
    • Jensen RL. Hypoxia in the tumorigenesis of gliomas and as a potential target for therapeutic measures. Neurosurg Focus 2006;20(4):E24.
    • (2006) Neurosurg Focus , vol.20 , Issue.4 , pp. 24
    • Jensen, R.L.1
  • 2
    • 51649090078 scopus 로고    scopus 로고
    • Hypoxia-inducible factor-2 regulates vascular tumorigenesis in mice
    • COI: 1:CAS:528:DC%2BD1cXhtV2qtr3K, PID: 18490920
    • Rankin EB, Rha J, Unger TL, Wu CH, Shutt HP, Johnson RS, et al. Hypoxia-inducible factor-2 regulates vascular tumorigenesis in mice. Oncogene 2008;27(40):5354–8.
    • (2008) Oncogene , vol.27 , Issue.40 , pp. 5354-5358
    • Rankin, E.B.1    Rha, J.2    Unger, T.L.3    Wu, C.H.4    Shutt, H.P.5    Johnson, R.S.6
  • 3
    • 4944229705 scopus 로고    scopus 로고
    • Schedule-dependent inhibition of hypoxia-inducible factor-1alpha protein accumulation, angiogenesis, and tumor growth by topotecan in U251-HRE glioblastoma xenografts
    • COI: 1:CAS:528:DC%2BD2cXotFalsr8%3D, PID: 15466170
    • Rapisarda A, Zalek J, Hollingshead M, Braunschweig T, Uranchimeg B, Bonomi CA, et al. Schedule-dependent inhibition of hypoxia-inducible factor-1alpha protein accumulation, angiogenesis, and tumor growth by topotecan in U251-HRE glioblastoma xenografts. Cancer Res 2004;64(19):6845–8.
    • (2004) Cancer Res , vol.64 , Issue.19 , pp. 6845-6848
    • Rapisarda, A.1    Zalek, J.2    Hollingshead, M.3    Braunschweig, T.4    Uranchimeg, B.5    Bonomi, C.A.6
  • 4
    • 0034654174 scopus 로고    scopus 로고
    • Modulation of hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics
    • COI: 1:CAS:528:DC%2BD3cXit1aqt74%3D, PID: 10749120
    • Zhong H, Chiles K, Feldser D, Laughner E, Hanrahan C, Georgescu MM, et al. Modulation of hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res 2000;60(6):1541–5.
    • (2000) Cancer Res , vol.60 , Issue.6 , pp. 1541-1545
    • Zhong, H.1    Chiles, K.2    Feldser, D.3    Laughner, E.4    Hanrahan, C.5    Georgescu, M.M.6
  • 5
    • 61349201437 scopus 로고    scopus 로고
    • In vitro angiogenesis by human umbilical vein endothelial cells (HUVEC) induced by three-dimensional co-culture with glioblastoma cells
    • COI: 1:CAS:528:DC%2BD1MXitFKntLg%3D, PID: 19039523
    • Chen Z, Htay A, Dos Santos W, Gillies GT, Fillmore HL, Sholley MM, et al. In vitro angiogenesis by human umbilical vein endothelial cells (HUVEC) induced by three-dimensional co-culture with glioblastoma cells. J Neurooncol 2009;92(2):121–8.
    • (2009) J Neurooncol , vol.92 , Issue.2 , pp. 121-128
    • Chen, Z.1    Htay, A.2    Dos Santos, W.3    Gillies, G.T.4    Fillmore, H.L.5    Sholley, M.M.6
  • 6
    • 34347206794 scopus 로고    scopus 로고
    • Hypoxia inhibits paclitaxel-induced apoptosis through adenosine-mediated phosphorylation of bad in glioblastoma cells
    • COI: 1:CAS:528:DC%2BD2sXnsVyjsb8%3D, PID: 17400763
    • Merighi S, Benini A, Mirandola P, Gessi S, Varani K, Leung E, et al. Hypoxia inhibits paclitaxel-induced apoptosis through adenosine-mediated phosphorylation of bad in glioblastoma cells. Mol Pharmacol 2007;72(1):162–72.
    • (2007) Mol Pharmacol , vol.72 , Issue.1 , pp. 162-172
    • Merighi, S.1    Benini, A.2    Mirandola, P.3    Gessi, S.4    Varani, K.5    Leung, E.6
  • 7
    • 33748056466 scopus 로고    scopus 로고
    • In vitro drug response and molecular markers associated with drug resistance in malignant gliomas
    • COI: 1:CAS:528:DC%2BD28XnvVGksr8%3D, PID: 16899598
    • Fruehauf JP, Brem H, Brem S, Sloan A, Barger G, Huang W, et al. In vitro drug response and molecular markers associated with drug resistance in malignant gliomas. Clin Cancer Res 2006;12(15):4523–32.
    • (2006) Clin Cancer Res , vol.12 , Issue.15 , pp. 4523-4532
    • Fruehauf, J.P.1    Brem, H.2    Brem, S.3    Sloan, A.4    Barger, G.5    Huang, W.6
  • 8
    • 0036536697 scopus 로고    scopus 로고
    • Biochemical changes associated with a multidrug-resistant phenotype of a human glioma cell line with temozolomide-acquired resistance
    • COI: 1:CAS:528:DC%2BD38XivVajtbw%3D, PID: 11960598
    • Ma J, Murphy M, O’Dwyer PJ, Berman E, Reed K, Gallo JM. Biochemical changes associated with a multidrug-resistant phenotype of a human glioma cell line with temozolomide-acquired resistance. Biochem Pharmacol 2002;63(7):1219–28.
    • (2002) Biochem Pharmacol , vol.63 , Issue.7 , pp. 1219-1228
    • Ma, J.1    Murphy, M.2    O’Dwyer, P.J.3    Berman, E.4    Reed, K.5    Gallo, J.M.6
  • 9
    • 20044372154 scopus 로고    scopus 로고
    • MGMT gene silencing and benefit from temozolomide in glioblastoma
    • COI: 1:CAS:528:DC%2BD2MXit1Wktro%3D, PID: 15758010
    • Hegi ME, Diserens A-C, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 2005;352:997–1003.
    • (2005) N Engl J Med , vol.352 , pp. 997-1003
    • Hegi, M.E.1    Diserens, A.-C.2    Gorlia, T.3    Hamou, M.F.4    de Tribolet, N.5    Weller, M.6
  • 10
    • 1842865745 scopus 로고    scopus 로고
    • Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells
    • COI: 1:CAS:528:DC%2BD2cXit12ntb0%3D, PID: 14713959
    • Kanzawa T, Germano IM, Komata T, Ito H, Kondo Y, Kondo S. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ 2004;11(4):448–57.
    • (2004) Cell Death Differ , vol.11 , Issue.4 , pp. 448-457
    • Kanzawa, T.1    Germano, I.M.2    Komata, T.3    Ito, H.4    Kondo, Y.5    Kondo, S.6
  • 12
    • 84892450078 scopus 로고    scopus 로고
    • Mechanism of temozolomide-induced antitumour effects on glioma cells
    • COI: 1:CAS:528:DC%2BC2cXhs1Omt77P, PID: 24326954
    • Shen W, Hu JA, Zheng JS. Mechanism of temozolomide-induced antitumour effects on glioma cells. J Int Med Res 2014;42(1):164–72.
    • (2014) J Int Med Res , vol.42 , Issue.1 , pp. 164-172
    • Shen, W.1    Hu, J.A.2    Zheng, J.S.3
  • 13
    • 0036840676 scopus 로고    scopus 로고
    • Small animal imaging. Current technology and perspectives for oncological imaging
    • PID: 12387842
    • Lewis JS, Achilefu S, Garbow JR, Laforest R, Welch MJ. Small animal imaging. Current technology and perspectives for oncological imaging. Eur J Cancer 2002;38(16):2173–88.
    • (2002) Eur J Cancer , vol.38 , Issue.16 , pp. 2173-2188
    • Lewis, J.S.1    Achilefu, S.2    Garbow, J.R.3    Laforest, R.4    Welch, M.J.5
  • 14
    • 34247562186 scopus 로고    scopus 로고
    • Integrating noninvasive molecular imaging into molecular medicine: an evolving paradigm
    • COI: 1:CAS:528:DC%2BD2sXlt1aks70%3D, PID: 17403616
    • Massoud TF, Gambhir SS. Integrating noninvasive molecular imaging into molecular medicine: an evolving paradigm. Trends Mol Med 2007;13(5):183–91.
    • (2007) Trends Mol Med , vol.13 , Issue.5 , pp. 183-191
    • Massoud, T.F.1    Gambhir, S.S.2
  • 15
    • 67651166780 scopus 로고    scopus 로고
    • Increased antitumor activity of bevacizumab in combination with hypoxia inducible factor-1 inhibition
    • COI: 1:CAS:528:DC%2BD1MXosVyms7g%3D, PID: 19584228
    • Rapisarda A, Hollingshead M, Uranchimeg B, Bonomi CA, Borgel SD, Carter JP, et al. Increased antitumor activity of bevacizumab in combination with hypoxia inducible factor-1 inhibition. Mol Cancer Ther 2009;8(7):1867–77.
    • (2009) Mol Cancer Ther , vol.8 , Issue.7 , pp. 1867-1877
    • Rapisarda, A.1    Hollingshead, M.2    Uranchimeg, B.3    Bonomi, C.A.4    Borgel, S.D.5    Carter, J.P.6
  • 16
    • 44649152229 scopus 로고    scopus 로고
    • Design and synthesis of novel fluoroquinolone based inhibitors of multiple targets in the PI3K/Akt-mTOR pathway
    • COI: 1:CAS:528:DC%2BD1cXmvFSnt7w%3D, PID: 18501601
    • Lohar MV, Mundada R, Bhonde M, Padgaonkar A, Deore V, Yewalkar N, et al. Design and synthesis of novel fluoroquinolone based inhibitors of multiple targets in the PI3K/Akt-mTOR pathway. Bioorg Med Chem Lett 2008;18(12):3603–6.
    • (2008) Bioorg Med Chem Lett , vol.18 , Issue.12 , pp. 3603-3606
    • Lohar, M.V.1    Mundada, R.2    Bhonde, M.3    Padgaonkar, A.4    Deore, V.5    Yewalkar, N.6
  • 17
    • 33847421328 scopus 로고    scopus 로고
    • Cell type-specific, topoisomerase II-dependent inhibition of hypoxia-inducible factor-1alpha protein accumulation by NSC 644221
    • COI: 1:CAS:528:DC%2BD2sXhsVyku7Y%3D, PID: 17289897
    • Creighton-Gutteridge M, Cardellina 2nd JH, Stephen AG, Rapisarda A, Uranchimeg B, Hite K, et al. Cell type-specific, topoisomerase II-dependent inhibition of hypoxia-inducible factor-1alpha protein accumulation by NSC 644221. Clin Cancer Res 2007;13(3):1010–8.
    • (2007) Clin Cancer Res , vol.13 , Issue.3 , pp. 1010-1018
    • Creighton-Gutteridge, M.1    Cardellina, J.H.2    Stephen, A.G.3    Rapisarda, A.4    Uranchimeg, B.5    Hite, K.6
  • 18
    • 84870481111 scopus 로고    scopus 로고
    • BMP2 sensitizes glioblastoma stem-like cells to temozolomide by affecting HIF-1α stability and MGMT expression
    • COI: 1:STN:280:DC%2BC3s%2FmtVyhsA%3D%3D, PID: 23076220
    • Persano L, Pistollato F, Rampazzo E, Della Puppa A, Abbadi S, Frasson C, et al. BMP2 sensitizes glioblastoma stem-like cells to temozolomide by affecting HIF-1α stability and MGMT expression. Cell Death Dis 2012;3:e412.
    • (2012) Cell Death Dis , vol.3 , pp. 412
    • Persano, L.1    Pistollato, F.2    Rampazzo, E.3    Della Puppa, A.4    Abbadi, S.5    Frasson, C.6
  • 19
    • 84896394612 scopus 로고    scopus 로고
    • Validation of an engineered cell model for in vitro and in vivo HIF-1α evaluation by different imaging modalities
    • COI: 1:STN:280:DC%2BC3sbltVGmtw%3D%3D, PID: 24002614
    • Lo Dico A, Valtorta S, Martelli C, Belloli S, Gianelli U, Tosi D, et al. Validation of an engineered cell model for in vitro and in vivo HIF-1α evaluation by different imaging modalities. Mol Imaging Biol 2014;16(2):210–23.
    • (2014) Mol Imaging Biol , vol.16 , Issue.2 , pp. 210-223
    • Lo Dico, A.1    Valtorta, S.2    Martelli, C.3    Belloli, S.4    Gianelli, U.5    Tosi, D.6
  • 20
    • 29644442764 scopus 로고    scopus 로고
    • Receptor tyrosine kinase signaling in gliomagenesis: pathobiology and therapeutic approaches
    • COI: 1:CAS:528:DC%2BD3sXps1KgsbY%3D, PID: 14508101
    • Kapoor GS, O’Rourke DM. Receptor tyrosine kinase signaling in gliomagenesis: pathobiology and therapeutic approaches. Cancer Biol Ther 2003;2(4):330–42.
    • (2003) Cancer Biol Ther , vol.2 , Issue.4 , pp. 330-342
    • Kapoor, G.S.1    O’Rourke, D.M.2
  • 21
    • 51049109033 scopus 로고    scopus 로고
    • Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity
    • COI: 1:CAS:528:DC%2BD1cXoslCgtb4%3D, PID: 18606717
    • Maira SM, Stauffer F, Brueggen J, Furet P, Schnell C, Fritsch C, et al. Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity. Mol Cancer Ther 2008;7(7):1851–63.
    • (2008) Mol Cancer Ther , vol.7 , Issue.7 , pp. 1851-1863
    • Maira, S.M.1    Stauffer, F.2    Brueggen, J.3    Furet, P.4    Schnell, C.5    Fritsch, C.6
  • 22
    • 79961173138 scopus 로고    scopus 로고
    • Roles of the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensitivity to therapy-implications for cancer and aging
    • COI: 1:CAS:528:DC%2BC3MXkvFKjt70%3D, PID: 21422497
    • Steelman LS, Chappell WH, Abrams SL, Kempf RC, Long J, Laidler P, et al. Roles of the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensitivity to therapy-implications for cancer and aging. Aging 2011;3(3):192–222.
    • (2011) Aging , vol.3 , Issue.3 , pp. 192-222
    • Steelman, L.S.1    Chappell, W.H.2    Abrams, S.L.3    Kempf, R.C.4    Long, J.5    Laidler, P.6
  • 23
    • 84863594749 scopus 로고    scopus 로고
    • Sensitivity of glioblastomas to clinically available MEK inhibitors is defined by neurofibromin 1 deficiency
    • COI: 1:CAS:528:DC%2BC38XpsFKku7c%3D, PID: 22573716
    • See WL, Tan IL, Mukherjee J, Nicolaides T, Pieper RO. Sensitivity of glioblastomas to clinically available MEK inhibitors is defined by neurofibromin 1 deficiency. Cancer Res 2012;72(13):3350–9.
    • (2012) Cancer Res , vol.72 , Issue.13 , pp. 3350-3359
    • See, W.L.1    Tan, I.L.2    Mukherjee, J.3    Nicolaides, T.4    Pieper, R.O.5
  • 24
    • 80054829310 scopus 로고    scopus 로고
    • Pharmacological targeting of the constitutively activated MEK/MAPK-dependent signaling pathway in glioma cells inhibits cell proliferation and migration
    • COI: 1:CAS:528:DC%2BC3MXhs1Slsb%2FO, PID: 21850371
    • Glassmann A, Reichmann K, Scheffler B, Glas M, Veit N, Probstmeier R. Pharmacological targeting of the constitutively activated MEK/MAPK-dependent signaling pathway in glioma cells inhibits cell proliferation and migration. Int J Oncol 2011;39(6):1567–75.
    • (2011) Int J Oncol , vol.39 , Issue.6 , pp. 1567-1575
    • Glassmann, A.1    Reichmann, K.2    Scheffler, B.3    Glas, M.4    Veit, N.5    Probstmeier, R.6
  • 25
    • 84939992209 scopus 로고    scopus 로고
    • A phase I/II study of BEZ235 in patients with advanced solid malignancies enriched by patients with advanced breast cancer. ClinicalTrial.gov Identifier number: NCT00620594
    • A phase I/II study of BEZ235 in patients with advanced solid malignancies enriched by patients with advanced breast cancer. ClinicalTrial.gov Identifier number: NCT00620594.
  • 26
    • 84940005967 scopus 로고    scopus 로고
    • Efficacy and safety of the combination therapy of dabrafenib and trametinib in subjects with BRAF V600E- mutated rare cancers. ClinicalTrial.gov Identifier number: NCT02034110
    • Efficacy and safety of the combination therapy of dabrafenib and trametinib in subjects with BRAF V600E- mutated rare cancers. ClinicalTrial.gov Identifier number: NCT02034110.
  • 27
    • 20044366163 scopus 로고    scopus 로고
    • Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma
    • COI: 1:CAS:528:DC%2BD2MXit1Wksbk%3D, PID: 15758009
    • Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352(10):987–96.
    • (2005) N Engl J Med , vol.352 , Issue.10 , pp. 987-996
    • Stupp, R.1    Mason, W.P.2    van den Bent, M.J.3    Weller, M.4    Fisher, B.5    Taphoorn, M.J.6
  • 28
    • 57449084713 scopus 로고    scopus 로고
    • In vivo bioluminescence imaging in an experimental mouse model for dendritic cell based immunotherapy against malignant glioma
    • COI: 1:STN:280:DC%2BD1cjotVWnsw%3D%3D, PID: 18787761
    • Maes W, Deroose C, Reumers V, Krylyshkina O, Gijsbers R, Baekelandt V, et al. In vivo bioluminescence imaging in an experimental mouse model for dendritic cell based immunotherapy against malignant glioma. J Neurooncol 2009;91:127–39.
    • (2009) J Neurooncol , vol.91 , pp. 127-139
    • Maes, W.1    Deroose, C.2    Reumers, V.3    Krylyshkina, O.4    Gijsbers, R.5    Baekelandt, V.6
  • 29
    • 76749104844 scopus 로고    scopus 로고
    • Patient-tailored, imaging-guided, long-term temozolomide chemotherapy in patients with glioblastoma
    • COI: 1:CAS:528:DC%2BC3cXisFKktL4%3D, PID: 20128997
    • Galldiks N, Kracht LW, Burghaus L, Ullrich RT, Backes H, Brunn A, et al. Patient-tailored, imaging-guided, long-term temozolomide chemotherapy in patients with glioblastoma. Mol Imaging 2010;9(1):40–6.
    • (2010) Mol Imaging , vol.9 , Issue.1 , pp. 40-46
    • Galldiks, N.1    Kracht, L.W.2    Burghaus, L.3    Ullrich, R.T.4    Backes, H.5    Brunn, A.6
  • 30
    • 76049123027 scopus 로고    scopus 로고
    • Specific apoptosis induction by the dual PI3K/mTor inhibitor NVP-BEZ235 in HER2 amplified and PIK3CA mutant breast cancer cells
    • COI: 1:CAS:528:DC%2BC3cXmtlagug%3D%3D, PID: 20007781
    • Brachmann SM, Hofmann I, Schnell C, Fritsch C, Wee S, Lane H, et al. Specific apoptosis induction by the dual PI3K/mTor inhibitor NVP-BEZ235 in HER2 amplified and PIK3CA mutant breast cancer cells. Proc Natl Acad Sci U S A 2009;106:22299–304.
    • (2009) Proc Natl Acad Sci U S A , vol.106 , pp. 22299-22304
    • Brachmann, S.M.1    Hofmann, I.2    Schnell, C.3    Fritsch, C.4    Wee, S.5    Lane, H.6
  • 31
    • 78049241020 scopus 로고    scopus 로고
    • Activity of the novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 against T-cell acute lymphoblastic leukemia
    • COI: 1:CAS:528:DC%2BC3cXht1yqs7fM, PID: 20876803
    • Chiarini F, Grimaldi C, Ricci F, Tazzari PL, Evangelisti C, Ognibene A, et al. Activity of the novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 against T-cell acute lymphoblastic leukemia. Cancer Res 2010;70(20):8097–107.
    • (2010) Cancer Res , vol.70 , Issue.20 , pp. 8097-8107
    • Chiarini, F.1    Grimaldi, C.2    Ricci, F.3    Tazzari, P.L.4    Evangelisti, C.5    Ognibene, A.6
  • 32
    • 68849091778 scopus 로고    scopus 로고
    • NVP-BEZ235, a novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor, elicits multifaceted antitumor activities in human gliomas
    • COI: 1:CAS:528:DC%2BD1MXpvFCnu7o%3D, PID: 19671762
    • Liu TJ, Koul D, LaFortune T, Tiao N, Shen RJ, Maira SM, et al. NVP-BEZ235, a novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor, elicits multifaceted antitumor activities in human gliomas. Mol Cancer Ther 2009;8:2204–10.
    • (2009) Mol Cancer Ther , vol.8 , pp. 2204-2210
    • Liu, T.J.1    Koul, D.2    LaFortune, T.3    Tiao, N.4    Shen, R.J.5    Maira, S.M.6
  • 33
    • 79952261716 scopus 로고    scopus 로고
    • GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition
    • Gilmartin AG, Bleam MR, Groy A, Moss KG, Minthorn EA, et al. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin Cancer Res 2011;17:989–1000.
    • (2011) Clin Cancer Res , vol.17 , pp. 989-1000
    • Gilmartin, A.G.1    Bleam, M.R.2    Groy, A.3    Moss, K.G.4    Minthorn, E.A.5
  • 34
    • 84859412054 scopus 로고    scopus 로고
    • Comprehensive predictive biomarker analysis for MEK inhibitor GSK1120212
    • COI: 1:CAS:528:DC%2BC38XjtlKisLw%3D, PID: 22169769
    • Jing J, Greshock J, Holbrook JD, Gilmartin A, Zhang X, McNeil E, et al. Comprehensive predictive biomarker analysis for MEK inhibitor GSK1120212. Mol Cancer Ther 2012;11(3):720–9.
    • (2012) Mol Cancer Ther , vol.11 , Issue.3 , pp. 720-729
    • Jing, J.1    Greshock, J.2    Holbrook, J.D.3    Gilmartin, A.4    Zhang, X.5    McNeil, E.6
  • 35
    • 79959996485 scopus 로고    scopus 로고
    • Pathway inhibition: emerging molecular targets for treating glioblastoma
    • COI: 1:CAS:528:DC%2BC3MXpsVCqu7g%3D, PID: 21636705
    • Wick W, Weller M, Weiler M, Batchelor T, Yung AW, Platten M. Pathway inhibition: emerging molecular targets for treating glioblastoma. Neuro Oncol 2011;13(6):566–79.
    • (2011) Neuro Oncol , vol.13 , Issue.6 , pp. 566-579
    • Wick, W.1    Weller, M.2    Weiler, M.3    Batchelor, T.4    Yung, A.W.5    Platten, M.6
  • 36
    • 0026075610 scopus 로고
    • Hypoxia-inducible nuclear factors bind to an enhancer element located 3′ to the human erythropoietin gene
    • COI: 1:CAS:528:DyaK3MXlsFWku7w%3D, PID: 2062846
    • Semenza GL, Nejfelt MK, Chi SM, Antonarakis SE. Hypoxia-inducible nuclear factors bind to an enhancer element located 3′ to the human erythropoietin gene. Proc Natl Acad Sci U S A 1991;88(13):5680–4.
    • (1991) Proc Natl Acad Sci U S A , vol.88 , Issue.13 , pp. 5680-5684
    • Semenza, G.L.1    Nejfelt, M.K.2    Chi, S.M.3    Antonarakis, S.E.4
  • 37
    • 33751169387 scopus 로고    scopus 로고
    • Hypoxia-inducible factor-1 (HIF-1)
    • COI: 1:CAS:528:DC%2BD28XhtFygsbjI, PID: 16887934
    • Ke Q, Costa M. Hypoxia-inducible factor-1 (HIF-1). Mol Pharmacol 2006;70(5):1469–80.
    • (2006) Mol Pharmacol , vol.70 , Issue.5 , pp. 1469-1480
    • Ke, Q.1    Costa, M.2
  • 38
    • 33746564193 scopus 로고    scopus 로고
    • The hypoxic inducible stress response as a target for cancer drug discovery
    • COI: 1:CAS:528:DC%2BD28XpsVersbk%3D, PID: 16890803
    • Welsh SJ, Koh MY, Powis G. The hypoxic inducible stress response as a target for cancer drug discovery. Semin Oncol 2006;33(4):486–97.
    • (2006) Semin Oncol , vol.33 , Issue.4 , pp. 486-497
    • Welsh, S.J.1    Koh, M.Y.2    Powis, G.3
  • 39
    • 57149142495 scopus 로고    scopus 로고
    • The impact of O2 availability on human cancer
    • COI: 1:CAS:528:DC%2BD1cXhsVWhu7fO, PID: 18987634
    • Bertout JA, Patel SA, Simon MC. The impact of O2 availability on human cancer. Nat Rev Cancer 2008;8(12):967–75.
    • (2008) Nat Rev Cancer , vol.8 , Issue.12 , pp. 967-975
    • Bertout, J.A.1    Patel, S.A.2    Simon, M.C.3
  • 40
    • 84897457122 scopus 로고    scopus 로고
    • Temozolomide resistance in glioblastoma cells occurs partly through epidermal growth factor receptor-mediated induction of connexin 43
    • COI: 1:CAS:528:DC%2BC2cXltFanurk%3D, PID: 24675463
    • Munoz JL, Rodriguez-Cruz V, Greco SJ, Ramkissoon SH, Ligon KL, Rameshwar P. Temozolomide resistance in glioblastoma cells occurs partly through epidermal growth factor receptor-mediated induction of connexin 43. Cell Death Dis 2014;5:e1145.
    • (2014) Cell Death Dis , vol.5 , pp. 1145
    • Munoz, J.L.1    Rodriguez-Cruz, V.2    Greco, S.J.3    Ramkissoon, S.H.4    Ligon, K.L.5    Rameshwar, P.6
  • 41
    • 57349110570 scopus 로고    scopus 로고
    • Combining bevacizumab with temozolomide increases the antitumor efficacy of temozolomide in a human glioblastoma orthotopic xenograft model
    • COI: 1:CAS:528:DC%2BD1MXhvVWht7g%3D, PID: 19048117
    • Mathieu V, De Nève N, Le Mercier M, Dewelle J, Gaussin JF, Dehoux M, et al. Combining bevacizumab with temozolomide increases the antitumor efficacy of temozolomide in a human glioblastoma orthotopic xenograft model. Neoplasia 2008;10(12):1383–92.
    • (2008) Neoplasia , vol.10 , Issue.12 , pp. 1383-1392
    • Mathieu, V.1    De Nève, N.2    Le Mercier, M.3    Dewelle, J.4    Gaussin, J.F.5    Dehoux, M.6
  • 42
    • 84884813627 scopus 로고    scopus 로고
    • Bortezomib overcomes MGMT-related resistance of glioblastoma cell lines to temozolomide in a schedule-dependent manner
    • COI: 1:CAS:528:DC%2BC3sXhsVOlurfI, PID: 23645448
    • Vlachostergios PJ, Hatzidaki E, Befani CD, Liakos P, Papandreou CN. Bortezomib overcomes MGMT-related resistance of glioblastoma cell lines to temozolomide in a schedule-dependent manner. Invest New Drugs 2013;31(5):1169–81.
    • (2013) Invest New Drugs , vol.31 , Issue.5 , pp. 1169-1181
    • Vlachostergios, P.J.1    Hatzidaki, E.2    Befani, C.D.3    Liakos, P.4    Papandreou, C.N.5
  • 43
    • 34848881062 scopus 로고    scopus 로고
    • FLT: measuring tumor cell proliferation in vivo with positron emission tomography and 3′-deoxy-3′-[18F]fluorothymidine
    • PID: 17920350
    • Salskov A, Tammisetti VS, Grierson J, Vesselle H. FLT: measuring tumor cell proliferation in vivo with positron emission tomography and 3′-deoxy-3′-[18F]fluorothymidine. Semin Nucl Med 2007;37(6):429–39.
    • (2007) Semin Nucl Med , vol.37 , Issue.6 , pp. 429-439
    • Salskov, A.1    Tammisetti, V.S.2    Grierson, J.3    Vesselle, H.4
  • 44
    • 77952298539 scopus 로고    scopus 로고
    • Kinetics of 3′-deoxy-3′-18F-fluorothymidine during treatment monitoring of recurrent high-grade glioma
    • PID: 20395318
    • Schiepers C, Dahlbom M, Chen W, Cloughesy T, Czernin J, Phelps ME, et al. Kinetics of 3′-deoxy-3′-18F-fluorothymidine during treatment monitoring of recurrent high-grade glioma. J Nucl Med 2010;51(5):720–7.
    • (2010) J Nucl Med , vol.51 , Issue.5 , pp. 720-727
    • Schiepers, C.1    Dahlbom, M.2    Chen, W.3    Cloughesy, T.4    Czernin, J.5    Phelps, M.E.6
  • 45
    • 0038037735 scopus 로고    scopus 로고
    • Regulation of angiogenesis by hypoxia: role of the HIF system
    • COI: 1:CAS:528:DC%2BD3sXktFOnur8%3D, PID: 12778166
    • Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med 2003;9:677–84.
    • (2003) Nat Med , vol.9 , pp. 677-684
    • Pugh, C.W.1    Ratcliffe, P.J.2
  • 46
    • 62749124795 scopus 로고    scopus 로고
    • Transcriptional control of the tumor- and hypoxia-marker carbonic anhydrase 9: a one transcription factor (HIF-1) show?
    • COI: 1:CAS:528:DC%2BD1MXjvVWjsro%3D, PID: 19344680
    • Kaluz S, Kaluzová M, Liao SY, Lerman M, Stanbridge EJ. Transcriptional control of the tumor- and hypoxia-marker carbonic anhydrase 9: a one transcription factor (HIF-1) show? Biochim Biophys Acta 2009;1795(2):162–72.
    • (2009) Biochim Biophys Acta , vol.1795 , Issue.2 , pp. 162-172
    • Kaluz, S.1    Kaluzová, M.2    Liao, S.Y.3    Lerman, M.4    Stanbridge, E.J.5
  • 47
    • 77955001665 scopus 로고    scopus 로고
    • PET of hypoxia with 89Zr-labeled cG250-F(ab′)2 in head and neck tumors
    • COI: 1:CAS:528:DC%2BC3cXhtVWmu7rM, PID: 20554724
    • Hoeben BA, Kaanders JH, Franssen GM, Troost EG, Rijken PF, Oosterwijk E, et al. PET of hypoxia with 89Zr-labeled cG250-F(ab′)2 in head and neck tumors. J Nucl Med 2010;51(7):1076–83.
    • (2010) J Nucl Med , vol.51 , Issue.7 , pp. 1076-1083
    • Hoeben, B.A.1    Kaanders, J.H.2    Franssen, G.M.3    Troost, E.G.4    Rijken, P.F.5    Oosterwijk, E.6
  • 48
    • 84973235519 scopus 로고    scopus 로고
    • Synthesis and evaluation of monomeric, dimeric and trimeric benzenesulfonamide derivatives for imaging carbonic anhydrase IX with PET
    • Lau J, Zhang Z, Hundal-Jabal N, Liu Z, Benard F, Lin KS. Synthesis and evaluation of monomeric, dimeric and trimeric benzenesulfonamide derivatives for imaging carbonic anhydrase IX with PET. J Nucl Med 2014;55(Suppl 1):444.
    • (2014) J Nucl Med , vol.55 , pp. 444
    • Lau, J.1    Zhang, Z.2    Hundal-Jabal, N.3    Liu, Z.4    Benard, F.5    Lin, K.S.6
  • 49
    • 0030017852 scopus 로고    scopus 로고
    • Anti-renal-cell carcinoma chimeric antibody G250 facilitates antibody-dependent cellular cytotoxicity with in vitro and in vivo interleukin-2-activated effectors
    • COI: 1:CAS:528:DyaK28Xkslagtbg%3D, PID: 8811493
    • Surfus JE, Hank JA, Oosterwijk E, Welt S, Lindstrom MJ, Albertini MR, et al. Anti-renal-cell carcinoma chimeric antibody G250 facilitates antibody-dependent cellular cytotoxicity with in vitro and in vivo interleukin-2-activated effectors. J Immunother Emphasis Tumor Immunol 1996;19(3):184–91.
    • (1996) J Immunother Emphasis Tumor Immunol , vol.19 , Issue.3 , pp. 184-191
    • Surfus, J.E.1    Hank, J.A.2    Oosterwijk, E.3    Welt, S.4    Lindstrom, M.J.5    Albertini, M.R.6
  • 50
    • 11144354471 scopus 로고    scopus 로고
    • A phase II trial of chimeric monoclonal antibody G250 for advanced renal cell carcinoma patients
    • COI: 1:CAS:528:DC%2BD2cXhslWrs7c%3D, PID: 14997194
    • Bleumer I, Knuth A, Oosterwijk E, Hofmann R, Varga Z, Lamers C, et al. A phase II trial of chimeric monoclonal antibody G250 for advanced renal cell carcinoma patients. Br J Cancer 2004;90(5):985–90.
    • (2004) Br J Cancer , vol.90 , Issue.5 , pp. 985-990
    • Bleumer, I.1    Knuth, A.2    Oosterwijk, E.3    Hofmann, R.4    Varga, Z.5    Lamers, C.6
  • 51
    • 84876266454 scopus 로고    scopus 로고
    • Chaperone-mediated autophagy targets hypoxia-inducible factor-1α (HIF-1α) for lysosomal degradation
    • COI: 1:CAS:528:DC%2BC3sXlvV2rtLk%3D, PID: 23457305
    • Hubbi ME, Hu H, Kshitiz, Ahmed I, Levchenko A, Semenza GL. Chaperone-mediated autophagy targets hypoxia-inducible factor-1α (HIF-1α) for lysosomal degradation. J Biol Chem 2013;288(15):10703–14.
    • (2013) J Biol Chem , vol.288 , Issue.15 , pp. 10703-10714
    • Hubbi, M.E.1    Hu, H.2    Kshitiz, A.I.3    Levchenko, A.4    Semenza, G.L.5
  • 52
    • 84884315657 scopus 로고    scopus 로고
    • STUB1/CHIP is required for HIF1A degradation by chaperone-mediated autophagy
    • COI: 1:CAS:528:DC%2BC2cXktF2rtb4%3D, PID: 23880665
    • Ferreira JV, Fôfo H, Bejarano E, Bento CF, Ramalho JS, Girão H, et al. STUB1/CHIP is required for HIF1A degradation by chaperone-mediated autophagy. Autophagy 2013;9(9):1349–66.
    • (2013) Autophagy , vol.9 , Issue.9 , pp. 1349-1366
    • Ferreira, J.V.1    Fôfo, H.2    Bejarano, E.3    Bento, C.F.4    Ramalho, J.S.5    Girão, H.6
  • 53
    • 33645812852 scopus 로고    scopus 로고
    • Effects of small molecules on chaperone-mediated autophagy
    • COI: 1:CAS:528:DC%2BD2MXht12lt7vF, PID: 16874031
    • Finn PF, Mesires NT, Vine M, Dice JF. Effects of small molecules on chaperone-mediated autophagy. Autophagy 2005;1(3):141–5.
    • (2005) Autophagy , vol.1 , Issue.3 , pp. 141-145
    • Finn, P.F.1    Mesires, N.T.2    Vine, M.3    Dice, J.F.4
  • 54
    • 0028834626 scopus 로고
    • Inhibition of the association with nuclear matrix of pRB, p70 and p40 proteins along with the specific suppression of c-MYC expression by geldanamycin, an inhibitor of Src tyrosine kinase
    • COI: 1:CAS:528:DyaK2MXotlKmtb4%3D, PID: 7592047
    • Yamaki H, Nakajima M, Seimiya H, Saya H, Sugita M, Tsuruo TJ. Inhibition of the association with nuclear matrix of pRB, p70 and p40 proteins along with the specific suppression of c-MYC expression by geldanamycin, an inhibitor of Src tyrosine kinase. J Antibiot 1995;48(9):1021–6.
    • (1995) J Antibiot , vol.48 , Issue.9 , pp. 1021-1026
    • Yamaki, H.1    Nakajima, M.2    Seimiya, H.3    Saya, H.4    Sugita, M.5    Tsuruo, T.J.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.