|
1
|
Jacobs SR, Herman CE, Maciver NJ, et al:
Glucose uptake is limiting in T cell activation and requires
CD28-mediated Akt-dependent and independent pathways. J Immunol.
180:4476–4486. 2008. View Article : Google Scholar : PubMed/NCBI
|
|
2
|
Kubicek GJ, Champ C, Fogh S, et al:
FDG-PET staging and importance of lymph node SUV in head and neck
cancer. Head Neck Oncol. 2:192010. View Article : Google Scholar : PubMed/NCBI
|
|
3
|
Peng NJ, Liou WS, Liu RS, Hu C, Tsay DG
and Liu CB: Early detection of recurrent ovarian cancer in patients
with low-level increases in serum CA-125 levels by
2-[F-18]fluoro-2-deoxy-D-glucose-positron emission
tomography/computed tomography. Cancer Biother Radiopharm.
26:175–181. 2011. View Article : Google Scholar : PubMed/NCBI
|
|
4
|
Masui T, Doi R, Ito T, et al: Diagnostic
value of (18)F -fluorodeoxyglucose positron emission tomography for
pancreatic neuroendocrine tumors with reference to the World Health
Organization classification. Oncol Lett. 1:155–159. 2010.PubMed/NCBI
|
|
5
|
Li LF, Zhou SH, Zhao K, et al: Clinical
significance of FDG single-photon emission computed tomography:
Computed tomography in the diagnosis of head and neck cancers and
study of its mechanism. Cancer Biother Radiopharm. 23:701–714.
2008. View Article : Google Scholar
|
|
6
|
Esen Akkas B, Gökaslan D, Güner L and
Ilgin Karabacak N: FDG uptake in brown adipose tissue-a brief
report on brown fat with FDG uptake mechanisms and quantitative
analysis using dual-time-point FDG PET/CT. Rev Esp Med Nucl.
30:14–18. 2011. View Article : Google Scholar
|
|
7
|
Ko BH, Paik JY, Jung KH and Lee KH:
17beta-estradiol augments 18F-FDG uptake and glycolysis of T47D
breast cancer cells via membrane-initiated rapid PI3K-Akt
activation. J Nucl Med. 51:1740–1747. 2010. View Article : Google Scholar : PubMed/NCBI
|
|
8
|
Prante O, Maschauer S, Fremont V, et al:
Regulation of uptake of 18F-FDG by a follicular human thyroid
cancer cell line with mutation-activated K-ras. J Nucl Med.
50:1364–1370. 2009. View Article : Google Scholar : PubMed/NCBI
|
|
9
|
Wood IS and Trayhurn P: Glucose
transporters (Glut and SGLT): expanded families of sugar transport
proteins. Br J Nutr. 89:3–9. 2003. View Article : Google Scholar : PubMed/NCBI
|
|
10
|
Sun L, Zeng X, Yan C, et al: Crystal
structure of a bacterial homologue of glucose transporters GLUT1–4.
Nature. 490:361–366. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
11
|
Lastraioli E, Bencini L, Bianchini E, et
al: hERG1 channels and Glut-1 as independent prognostic indicators
of worse outcome in stage I and II colorectal cancer: A pilot
study. Transl Oncol. 5:105–112. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
12
|
Tong SY, Lee JM, Ki KD, et al: Correlation
between FDG uptake by PET/CT and the expressions of glucose
transporter type 1 and hexokinase II in cervical cancer. Int J
Gynecol Cancer. 22:654–658. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
13
|
Sasaki H, Shitara M, Yokota K, et al:
Overexpression of GLUT1 correlates with Kras mutations in lung
carcinomas. Mol Med Rep. 5:599–602. 2012.
|
|
14
|
Liu TQ, Fan J, Zhou L and Zheng SS:
Effects of suppressing glucose transporter-1 by an antisense
oligodeoxynucleotide on the growth of human hepatocellular
carcinoma cells. Hepatobiliary Pancreat Dis Int. 10:72–77. 2011.
View Article : Google Scholar : PubMed/NCBI
|
|
15
|
Melstrom LG, Salabat MR, Ding XZ, et al:
Apigenin down-regulates the hypoxia response genes: HIF-1α, GLUT-1,
and VEGF in human pancreatic cancer cells. J Surg Res. 167:173–181.
2011. View Article : Google Scholar : PubMed/NCBI
|
|
16
|
Fan J, Zhou JQ, Yu GR and Lu DD: Glucose
transporter protein 1-targeted RNA interference inhibits growth and
invasion of the osteosarcoma cell line MG63 in vitro. Cancer
Biother Radiopharm. 25:521–527. 2010. View Article : Google Scholar : PubMed/NCBI
|
|
17
|
Rademakers SE, Lok J, van der Kogel AJ,
Bussink J and Kaanders JH: Metabolic markers in relation to
hypoxia; staining patterns and colocalization of pimonidazole,
HIF-1α, CAIX, LDH-5, GLUT-1, MCT1 and MCT4. BMC Cancer. 11:1672011.
View Article : Google Scholar
|
|
18
|
Eckert AW, Lautner MH, Taubert H, Schubert
J and Bilkenroth U: Expression of Glut-1 is a prognostic marker for
oral squamous cell carcinoma patients. Oncol Rep. 20:1381–1385.
2008.PubMed/NCBI
|
|
19
|
Kunkel M, Reichert TE, Benz P, et al:
Overexpression of Glut-1 and increased glucose metabolism in tumors
are associated with a poor prognosis in patients with oral squamous
cell carcinoma. Cancer. 97:1015–1024. 2003. View Article : Google Scholar : PubMed/NCBI
|
|
20
|
Zhou S, Wang S, Wu Q, Fan J and Wang Q:
Expression of glucose transporter-1 and -3 in the head and neck
carcinoma - the correlation of the expression with the biological
behaviors. ORL J Otorhinolaryngol Relat Spec. 70:189–194. 2008.
View Article : Google Scholar
|
|
21
|
Choi JW, Yoon DJ, Lee HW, Han DP and Ahn
YH: Antisense GLUT1 RNA suppresses the transforming phenotypes of
NIH 3T3 cells transformed by N-Ras. Yonsei Med J. 36:480–486. 1995.
View Article : Google Scholar : PubMed/NCBI
|
|
22
|
Chan JY, Kong SK, Choy YM, Lee CY and Fung
KP: Inhibition of glucose transporter gene expression by antisense
nucleic acids in HL-60 leukemia cells. Life Sci. 65:63–70. 1999.
View Article : Google Scholar : PubMed/NCBI
|
|
23
|
Ito S, Nemoto T, Satoh S, Sekihara H,
Seyama Y and Kubota S: Human rhabdomyosarcoma cells retain
insulin-regulated glucose transport activity through glucose
transporter 1. Arch Biochem Biophys. 373:72–82. 2000. View Article : Google Scholar : PubMed/NCBI
|
|
24
|
Noguchi Y, Saito A, Miyagi Y, et al:
Suppression of facilitative glucose transporter 1 mRNA can suppress
tumor growth. Cancer Lett. 154:175–182. 2000. View Article : Google Scholar : PubMed/NCBI
|
|
25
|
Bussink J, van der Kogel AJ and Kaanders
JH: Activation of the PI3-K/AKT pathway and implications for
radioresistance mechanisms in head and neck cancer. Lancet Oncol.
9:288–296. 2008. View Article : Google Scholar : PubMed/NCBI
|
|
26
|
Stuschke M and Thames HD: Fractionation
sensitivities and dose-control relations of head and neck
carcinomas: analysis of the randomized hyperfractionation trials.
Radiother Oncol. 51:113–121. 1999. View Article : Google Scholar : PubMed/NCBI
|
|
27
|
Denekamp J, Daşu A, Waites A and Littbrand
B: Hyperfractionation as an effective way of overcoming
radioresistance. Int J Radiat Oncol Biol Phys. 42:705–709. 1998.
View Article : Google Scholar : PubMed/NCBI
|
|
28
|
Hingorani M, Colley WP, Dixit S and Beavis
AM: Hypofractionated radiotherapy for glioblastoma: strategy for
poor-risk patients or hope for the future? Br J Radiol.
85:e770–e781. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
29
|
Bollschweiler E, Hölscher AH and Metzger
R: Histologic tumor type and the rate of complete response after
neoadjuvant therapy for esophageal cancer. Future Oncol. 6:25–35.
2010. View Article : Google Scholar
|
|
30
|
Sho M, Akahori T, Tanaka T, et al:
Pathological and clinical impact of neoadjuvant chemoradiotherapy
using full-dose gemcitabine and concurrent radiation for resectable
pancreatic cancer. J Hepatobiliary Pancreat Sci. 20:197–205. 2013.
View Article : Google Scholar
|
|
31
|
Yamazaki H, Nakamura S, Nishimura T, et
al: Hypofractionated stereotactic radiotherapy with the hypoxic
sensitizer AK-2123 (sanazole) for reirradiation of brain
metastases: a preliminary feasibility report. Anticancer Res.
33:1773–1776. 2013.PubMed/NCBI
|
|
32
|
Chen FH, Chiang CS, Wang CC, et al:
Vasculatures in tumors growing from preirradiated tissues: formed
by vasculogenesis and resistant to radiation and antiangiogenic
therapy. Int J Radiat Oncol Biol Phys. 80:1512–1521. 2011.
View Article : Google Scholar : PubMed/NCBI
|
|
33
|
Saigusa S, Toiyama Y, Tanaka K, et al:
Prognostic significance of glucose transporter-1 (GLUT1) gene
expression in rectal cancer after preoperative chemoradiotherapy.
Surg Today. 42:460–469. 2012. View Article : Google Scholar
|
|
34
|
Kunkel M, Moergel M, Stockinger M, et al:
Overexpression of GLUT-1 is associated with resistance to
radiotherapy and adverse prognosis in squamous cell carcinoma of
the oral cavity. Oral Oncol. 43:796–803. 2007. View Article : Google Scholar : PubMed/NCBI
|
|
35
|
Doki Y, Takachi K, Ishikawa O, et al:
Reduced tumor vessel density and high expression of glucose
transporter 1 suggest tumor hypoxia of squamous cell carcinoma of
the esophagus surviving after radiotherapy. Surgery. 137:536–544.
2005. View Article : Google Scholar : PubMed/NCBI
|
|
36
|
Pedersen MW, Holm S, Lund EL, Højgaard L
and Kristjansen PE: Coregulation of glucose uptake and vascular
endothelial growth factor (VEGF) in two small-cell lung cancer
(SCLC) sublines in vivo and in vitro. Neoplasia. 3:80–87. 2001.
View Article : Google Scholar : PubMed/NCBI
|
|
37
|
Korkeila E, Jaakkola PM, Syrjänen K,
Pyrhönen S and Sundström J: Pronounced tumour regression after
radiotherapy is associated with negative/weak glucose transporter-1
expression in rectal cancer. Anticancer Res. 31:311–315.
2011.PubMed/NCBI
|
|
38
|
Korkeila EA, Sundström J, Pyrhönen S and
Syrjänen K: Carbonic anhydrase IX, hypoxia-inducible factor-1α,
ezrin and glucose transporter-1 as predictors of disease outcome in
rectal cancer: multivariate Cox survival models following data
reduction by principal component analysis of the
clinicopathological predctors. Anticancer Res. 31:4529–4535.
2011.PubMed/NCBI
|
|
39
|
Luo XM, Zhou SH and Fan J: Glucose
transporter-1 as a new therapeutic target in laryngeal carcinoma. J
Int Med Res. 38:1885–1892. 2010. View Article : Google Scholar
|
|
40
|
Bai J, Guo XG and Bai XP: Epidermal growth
factor receptor-related DNA repair and radiation-resistance
regulatory mechanisms: a mini-review. Asian Pac J Cancer Prev.
13:4879–4881. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
41
|
Theys J, Yahyanejad S, Habets R, et al:
High NOTCH activity induces radiation resistance in non small cell
lung cancer. Radiother Oncol. 108:440–445. 2013. View Article : Google Scholar : PubMed/NCBI
|
|
42
|
Yun J, Rago C, Cheong I, et al: Glucose
deprivation contributes to the development of KRAS pathway
mutations in tumor cells. Science. 325:1555–1559. 2009. View Article : Google Scholar : PubMed/NCBI
|
|
43
|
Yasuda M, Miyazawa M, Fujita M, et al:
Expression of hypoxia inducible factor-1alpha (HIF-1alpha) and
glucose transporter-1 (GLUT-1) in ovarian adenocarcinomas:
difference in hypoxic status depending on histological character.
Oncol Rep. 19:111–116. 2008.
|
|
44
|
Mayer A, Höckel M, Wree A and Vaupel P:
Microregional expression of glucose transporter-1 and oxygenation
status: lack of correlation in locally advanced cervical cancers.
Clin Cancer Res. 11:2768–2773. 2005. View Article : Google Scholar : PubMed/NCBI
|
|
45
|
Ding XZ, Fehsenfeld DM, Murphy LO, Permert
J and Adrian TE: Physiological concentrations of insulin augment
pancreatic cancer cell proliferation and glucose utilization by
activating MAP kinase, PI3 kinase and enhancing GLUT-1 expression.
Pancreas. 21:310–320. 2000. View Article : Google Scholar : PubMed/NCBI
|
|
46
|
Sommermann TG, O’Neill K, Plas DR and
Cahir-McFarland E: IKKβ and NF-κB transcription govern lymphoma
cell survival through AKT-induced plasma membrane trafficking of
GLUT1. Cancer Res. 71:7291–7300. 2011. View Article : Google Scholar : PubMed/NCBI
|
|
47
|
Wofford JA, Wieman HL, Jacobs SR, Zhao Y
and Rathmell JC: IL-7 promotes Glut1 trafficking and glucose uptake
via STAT5-mediated activation of Akt to support T-cell survival.
Blood. 111:2101–2111. 2008. View Article : Google Scholar
|
|
48
|
Wieman HL, Wofford JA and Rathmell JC:
Cytokine stimulation promotes glucose uptake via
phosphatidylinositol-3 kinase/Akt regulation of Glut1 activity and
trafficking. Mol Biol Cell. 18:1437–1446. 2007. View Article : Google Scholar : PubMed/NCBI
|
|
49
|
Melstrom LG, Salabat MR, Ding XZ, et al:
Apigenin inhibits the GLUT-1 glucose transporter and the
phosphoinositide 3-Kinase/Akt pathway in human pancreatic cancer
cells. Pancreas. 37:426–431. 2008. View Article : Google Scholar : PubMed/NCBI
|
|
50
|
Schuurbiers OC, Kaanders JH, van der
Heijden HF, Dekhuijzen RP, Oyen WJ and Bussink J: The
PI3-K/AKT-pathway and radiation resistance mechanisms in non-small
cell lung cancer. J Thorac Oncol. 4:761–767. 2009. View Article : Google Scholar : PubMed/NCBI
|
|
51
|
Söderlund K, Pérez-Tenorio G and Stål O:
Activation of the phosphatidylinositol 3-kinase/Akt pathway
prevents radiation-induced apoptosis in breast cancer cells. Int J
Oncol. 26:25–32. 2005.
|
|
52
|
Florczak U, Toulany M, Kehlbach R and
Peter Rodemann H: 2-Methoxyestradiol-induced radiosensitization is
independent of SOD but depends on inhibition of Akt and DNA-PKcs
activities. Radiother Oncol. 92:334–338. 2009. View Article : Google Scholar : PubMed/NCBI
|
|
53
|
Brophy S, Sheehan KM, McNamara DA, Deasy
J, Bouchier-Hayes DJ and Kay EW: GLUT-1 expression and response to
chemoradiotherapy in rectal cancer. Int J Cancer. 125:2778–2782.
2009. View Article : Google Scholar : PubMed/NCBI
|
|
54
|
Zhou SH, Fan J, Chen XM, Cheng KJ and Wang
SQ: Inhibition of cell proliferation and glucose uptake in human
laryngeal carcinoma cells by antisense oligonucleotides against
glucose transporter-1. Head Neck. 31:1624–1633. 2009. View Article : Google Scholar : PubMed/NCBI
|
|
55
|
Yan SX, Luo XM, Zhou SH, et al: Effect of
antisense oligodeoxynucleotides glucose transporter-1 on
enhancement of radiosensitivity of laryngeal carcinoma. Int J Med
Sci. 10:1375–1386. 2013. View Article : Google Scholar : PubMed/NCBI
|
|
56
|
Upadhyay M, Samal J, Kandpal M, Singh OV
and Vivekanandan P: The Warburg effect: insights from the past
decade. Pharmacol Ther. 137:318–330. 2013. View Article : Google Scholar
|
|
57
|
Bensinger SJ and Christofk HR: New aspects
of the Warburg effect in cancer cell biology. Semin Cell Dev Biol.
23:352–361. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
58
|
Hirschhaeuser F, Sattler UG and
Mueller-Klieser W: Lactate: a metabolic key player in cancer.
Cancer Res. 71:6921–6925. 2011. View Article : Google Scholar : PubMed/NCBI
|
|
59
|
Krzeslak A, Wojcik-Krowiranda K, Forma E,
et al: Expression of GLUT1 and GLUT3 glucose transporters in
endometrial and breast cancers. Pathol Oncol Res. 18:721–728. 2012.
View Article : Google Scholar : PubMed/NCBI
|
|
60
|
Sheehan JP, Shaffrey ME, Gupta B, Larner
J, Rich JN and Park DM: Improving the radiosensitivity of
radioresistant and hypoxic glioblastoma. Future Oncol. 6:1591–1601.
2010. View Article : Google Scholar : PubMed/NCBI
|
|
61
|
Overgaard J: Hypoxic modification of
radiotherapy in squamous cell carcinoma of the head and neck - a
systematic review and meta-analysis. Radiother Oncol. 100:22–32.
2011. View Article : Google Scholar : PubMed/NCBI
|
|
62
|
Shimanishi M, Ogi K, Sogabe Y, et al:
Silencing of GLUT-1 inhibits sensitization of oral cancer cells to
cisplatin during hypoxia. J Oral Pathol Med. 42:382–388. 2013.
View Article : Google Scholar
|
|
63
|
Pez F, Dayan F, Durivault J, et al: The
HIF-1-inducible lysyl oxidase activates HIF-1 via the Akt pathway
in a positive regulation loop and synergizes with HIF-1 in
promoting tumor cell growth. Cancer Res. 71:1647–1657. 2011.
View Article : Google Scholar : PubMed/NCBI
|
|
64
|
Yasuda M, Miyazawa M, Fujita M, et al:
Expression of hypoxia inducible factor-1alpha (HIF-1alpha) and
glucose transporter-1 (GLUT-1) in ovarian adenocarcinomas:
difference in hypoxic status depending on histological character.
Oncol Rep. 19:111–116. 2008.
|
|
65
|
Wu XH, Chen SP, Mao JY, Ji XX, Yao HT and
Zhou SH: Expression and significance of hypoxia-inducible factor-1α
and glucose transporter-1 in laryngeal carcinoma. Oncol Lett.
5:261–266. 2013.
|
|
66
|
Evans A, Bates V, Troy H, et al: Glut-1 as
a therapeutic target: increased chemoresistance and
HIF-1-independent link with cell turnover is revealed through
COMPARE analysis and metabolomic studies. Cancer Chemother
Pharmacol. 61:377–393. 2008. View Article : Google Scholar
|
|
67
|
Ke CC, Liu RS, Yang AH, et al:
CD133-expressing thyroid cancer cells are undifferentiated,
radioresistant and survive radioiodide therapy. Eur J Nucl Med Mol
Imaging. 40:61–71. 2013. View Article : Google Scholar
|
|
68
|
Piao LS, Hur W, Kim TK, et al:
CD133+ liver cancer stem cells modulate radioresistance
in human hepatocellular carcinoma. Cancer Lett. 315:129–137. 2012.
View Article : Google Scholar
|
|
69
|
Mai HM, Zheng JW, Wang YA, et al: CD133
selected stem cells from proliferating infantile hemangioma and
establishment of an in vivo mice model of hemangioma. Chin Med J
(Engl). 126:88–94. 2013.
|
|
70
|
Chen XH, Bao YY, Zhou SH, Wang QY, Wei Y
and Fan J: Glucose transporter-1 expression in CD133+
laryngeal carcinoma Hep-2 cells. Mol Med Rep. 8:1695–1700.
2013.PubMed/NCBI
|
|
71
|
Stein I, Neeman M, Shweiki D, Itin A and
Keshet E: Stabilization of vascular endothelial growth factor mRNA
by hypoxia and hypoglycemia and coregulation with other
ischemia-induced genes. Mol Cell Biol. 15:5363–5368.
1995.PubMed/NCBI
|
|
72
|
Gogineni VR, Nalla AK, Gupta R, Dinh DH,
Klopfenstein JD and Rao JS: Chk2-mediated G2/M cell cycle arrest
maintains radiation resistance in malignant meningioma cells.
Cancer Lett. 313:64–75. 2011. View Article : Google Scholar : PubMed/NCBI
|
|
73
|
Hematulin A, Meethang S, Ingkaninan K and
Sagan D: Derris scandens Benth extract potentiates radioresistance
of Hep-2 laryngeal cancer cells. Asian Pac J Cancer Prev.
13:1289–1295. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
74
|
Young EF, Smilenov LB, Lieberman HB and
Hall EJ: Combined haploinsufficiency and genetic control of the
G2/M checkpoint in irradiated cells. Radiat Res. 177:743–750. 2012.
View Article : Google Scholar : PubMed/NCBI
|
|
75
|
Zhou Q, Lui VW and Yeo W: Targeting the
PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Future Oncol.
7:1149–1167. 2011. View Article : Google Scholar : PubMed/NCBI
|
|
76
|
Keck S, Glencer AC and Rugo HS: Everolimus
and its role in hormone-resistant and trastuzumab-resistant
metastatic breast cancer. Future Oncol. 8:1383–1396. 2012.
View Article : Google Scholar : PubMed/NCBI
|
|
77
|
Webster L, Hodgkiss RJ and Wilson GD: Cell
cycle distribution of hypoxia and progression of hypoxic tumour
cells in vivo. Br J Cancer. 77:227–234. 1998. View Article : Google Scholar : PubMed/NCBI
|
|
78
|
Koritzinsky M, Wouters BG, Amellem O and
Pettersen EO: Cell cycle progression and radiation survival
following prolonged hypoxia and re-oxygenation. Int J Radiat Biol.
77:319–328. 2001. View Article : Google Scholar : PubMed/NCBI
|
|
79
|
Kumareswaran R, Ludkovski O, Meng A, Sykes
J, Pintilie M and Bristow RG: Chronic hypoxia compromises repair of
DNA double-strand breaks to drive genetic instability. J Cell Sci.
125:189–199. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
80
|
Toustrup K, Sørensen BS, Nordsmark M, et
al: Development of a hypoxia gene expression classifier with
predictive impact for hypoxic modification of radiotherapy in head
and neck cancer. Cancer Res. 71:5923–5931. 2011. View Article : Google Scholar : PubMed/NCBI
|
|
81
|
Koumenis C: ER stress, hypoxia tolerance
and tumor progression. Curr Mol Med. 6:55–69. 2006. View Article : Google Scholar : PubMed/NCBI
|
|
82
|
Kitagawa N, Kondo S, Wakisaka N, et al:
Expression of seven-in-absentia homologue 1 and hypoxia-inducible
factor 1 alpha: novel prognostic factors of nasopharyngeal
carcinoma. Cancer Lett. 331:52–57. 2013. View Article : Google Scholar
|
|
83
|
Pentheroudakis G, Nicolaou I, Kotoula V,
et al: Prognostic utility of angiogenesis and hypoxia effectors in
patients with operable squamous cell cancer of the larynx. Oral
Oncol. 48:709–716. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
84
|
Moeller BJ, Cao Y, Li CY and Dewhirst MW:
Radiation activates HIF-1 to regulate vascular radiosensitivity in
tumors: role of reoxygenation, free radicals, and stress granules.
Cancer Cell. 5:429–441. 2004. View Article : Google Scholar : PubMed/NCBI
|
|
85
|
Kim WY, Oh SH, Woo JK, Hong WK and Lee HY:
Targeting heat shock protein 90 overrides the resistance of lung
cancer cells by blocking radiation-induced stabilization of
hypoxia-inducible factor-1alpha. Cancer Res. 69:1624–1632. 2009.
View Article : Google Scholar : PubMed/NCBI
|
|
86
|
Lee SM, Lee CT, Kim YW, Han SK, Shim YS
and Yoo CG: Hypoxia confers protection against apoptosis via
PI3K/Akt and ERK pathways in lung cancer cells. Cancer Lett.
242:231–238. 2006. View Article : Google Scholar : PubMed/NCBI
|
|
87
|
Edwards E, Geng L, Tan J, Onishko H,
Donnelly E and Hallahan DE: Phosphatidylinositol 3-kinase/Akt
signaling in the response of vascular endothelium to ionizing
radiation. Cancer Res. 62:4671–4677. 2002.PubMed/NCBI
|
|
88
|
Kumar P, Miller AI and Polverini PJ: p38
MAPK mediates gamma-irradiation-induced endothelial cell apoptosis
and vascular endothelial growth factor protects endothelial cells
through phosphoinositide 3-kinase-Akt-Bcl-2 pathway. J Biol Chem.
279:43352–43360. 2004. View Article : Google Scholar : PubMed/NCBI
|
|
89
|
Kumar P, Benedict R, Urzua F, Fischbach C,
Mooney D and Polverini P: Combination treatment significantly
enhances the efficacy of antitumor therapy by preferentially
targeting angiogenesis. Lab Invest. 85:756–767. 2005. View Article : Google Scholar : PubMed/NCBI
|
|
90
|
Wu J, Chen C and Zhao KN:
Phosphatidylinositol 3-kinase signaling as a therapeutic target for
cervical cancer. Curr Cancer Drug Targets. 13:143–156. 2013.
View Article : Google Scholar : PubMed/NCBI
|
|
91
|
Shinohara ET and Maity A: Increasing
sensitivity to radiotherapy and chemotherapy by using novel
biological agents that alter the tumor microenvironment. Curr Mol
Med. 9:1034–1045. 2009. View Article : Google Scholar : PubMed/NCBI
|
|
92
|
Fokas E, McKenna WG and Muschel RJ: The
impact of tumor microenvironment on cancer treatment and its
modulation by direct and indirect antivascular strategies. Cancer
Metastasis Rev. 31:823–842. 2012. View Article : Google Scholar : PubMed/NCBI
|
|
93
|
Zhan M and Han ZC: Phosphatidylinositide
3-kinase/AKT in radiation responses. Histol Histopathol.
19:915–923. 2004.PubMed/NCBI
|
|
94
|
Park JK, Jung HY, Park SH, et al:
Combination of PTEN and gamma-ionizing radiation enhances cell
death and G(2)/M arrest through regulation of AKT activity and p21
induction in non-small-cell lung cancer cells. Int J Radiat Oncol
Biol Phys. 70:1552–1560. 2008. View Article : Google Scholar : PubMed/NCBI
|
|
95
|
Kandel ES, Skeen J, Majewski N, et al:
Activation of Akt/protein kinase B overcomes a G(2)/m cell cycle
checkpoint induced by DNA damage. Mol Cell Biol. 22:7831–7841.
2002. View Article : Google Scholar : PubMed/NCBI
|
|
96
|
Mukherjee B, Tomimatsu N, Amancherla K,
Camacho CV, Pichamoorthy N and Burma S: The dual PI3K/mTOR
inhibitor NVP-BEZ235 is a potent inhibitor of ATM- and
DNA-PKCs-mediated DNA damage responses. Neoplasia. 14:34–43.
2012.PubMed/NCBI
|
|
97
|
Yang J, Xu X, Hao Y, et al: Expression of
DNA-PKcs and BRCA1 as prognostic indicators in nasopharyngeal
carcinoma following intensity-modulated radiation therapy. Oncol
Lett. 5:1199–1204. 2013.PubMed/NCBI
|
|
98
|
Millet P, Granotier C, Etienne O and
Boussin FD: Radiation-induced upregulation of telomerase activity
escapes PI3-kinase inhibition in two malignant glioma cell lines.
Int J Oncol. 43:375–382. 2013.PubMed/NCBI
|
|
99
|
Qu YY, Hu SL, Xu XY, et al: Nimotuzumab
enhances the radiosensitivity of cancer cells in vitro by
inhibiting radiation-induced DNA damage repair. PLoS One.
8:e707272013. View Article : Google Scholar : PubMed/NCBI
|
|
100
|
Zhang T, Cui GB, Zhang J, et al:
Inhibition of PI3 kinases enhances the sensitivity of non-small
cell lung cancer cells to ionizing radiation. Oncol Rep.
24:1683–1689. 2010.PubMed/NCBI
|
|
101
|
Azad A, Jackson S, Cullinane C, et al:
Inhibition of DNA-dependent protein kinase induces accelerated
senescence in irradiated human cancer cells. Mol Cancer Res.
9:1696–1707. 2011. View Article : Google Scholar : PubMed/NCBI
|
|
102
|
Li HF, Kim JS and Waldman T:
Radiation-induced Akt activation modulates radioresistance in human
glioblastoma cells. Radiat Oncol. 4:432009. View Article : Google Scholar : PubMed/NCBI
|
|
103
|
Minjgee M, Toulany M, Kehlbach R, Giehl K
and Rodemann HP: K-RAS(V12) induces autocrine production of EGFR
ligands and mediates radioresistance through EGFR-dependent Akt
signaling and activation of DNA-PKcs. Int J Radiat Oncol Biol Phys.
81:1506–1514. 2011. View Article : Google Scholar : PubMed/NCBI
|
|
104
|
Petrás M, Lajtos T, Friedländer E, et al:
Molecular interactions of ErbB1 (EGFR) and integrin-β1 in
astrocytoma frozen sections predict clinical outcome and correlate
with Akt-mediated in vitro radioresistance. Neuro Oncol.
15:1027–1040. 2013. View Article : Google Scholar
|
|
105
|
Gautier EL, Westerterp M, Bhagwat N, et
al: HDL and Glut1 inhibition reverse a hypermetabolic state in
mouse models of myeloproliferative disorders. J Exp Med.
210:339–353. 2013. View Article : Google Scholar : PubMed/NCBI
|
|
106
|
Swainson L, Kinet S, Mongellaz C,
Sourisseau M, Henriques T and Taylor N: IL-7-induced proliferation
of recent thymic emigrants requires activation of the PI3K pathway.
Blood. 109:1034–1042. 2007. View Article : Google Scholar
|
|
107
|
Rathmell JC, Fox CJ, Plas DR, Hammerman
PS, Cinalli RM and Thompson CB: Akt-directed glucose metabolism can
prevent Bax conformation change and promote growth
factor-independent survival. Mol Cell Biol. 23:7315–7328. 2003.
View Article : Google Scholar : PubMed/NCBI
|
|
108
|
Zhao Y, Altman BJ, Coloff JL, et al:
Glycogen synthase kinase 3alpha and 3beta mediate a
glucose-sensitive antiapoptotic signaling pathway to stabilize
Mcl-1. Mol Cell Biol. 27:4328–4339. 2007. View Article : Google Scholar : PubMed/NCBI
|
|
109
|
Roberts MS, Woods AJ, Dale TC, Van Der
Sluijs P and Norman JC: Protein kinase B/Akt acts via glycogen
synthase kinase 3 to regulate recycling of alpha v beta 3 and alpha
5 beta 1 integrins. Mol Cell Biol. 24:1505–1515. 2004. View Article : Google Scholar : PubMed/NCBI
|
|
110
|
Doughty CA, Bleiman BF, Wagner DJ, et al:
Antigen receptor-mediated changes in glucose metabolism in B
lymphocytes: role of phosphatidylinositol 3-kinase signaling in the
glycolytic control of growth. Blood. 107:4458–4465. 2006.
View Article : Google Scholar : PubMed/NCBI
|
|
111
|
Marko AJ, Miller RA, Kelman A and
Frauwirth KA: Induction of glucose metabolism in stimulated T
lymphocytes is regulated by mitogen-activated protein kinase
signaling. PLoS One. 5:e154252010. View Article : Google Scholar : PubMed/NCBI
|
|
112
|
Radhakrishnan P, Baraneedharan U,
Veluchamy S, et al: Inhibition of rapamycin-induced AKT activation
elicits differential antitumor response in head and neck cancers.
Cancer Res. 73:1118–1127. 2013. View Article : Google Scholar : PubMed/NCBI
|
|
113
|
Samih N, Hovsepian S, Aouani A, Lombardo D
and Fayet G: Glut-1 translocation in FRTL-5 thyroid cells: role of
phosphatidylinositol 3-kinase and N-glycosylation. Endrocrinology.
141:4146–4155. 2000. View Article : Google Scholar
|
|
114
|
Clarke JF, Young PW, Yonezawa K, Kasuga M
and Holman GD: Inhibition of the translocation of GLUT1 and GLUT4
in 3T3-L1 cells by the phosphatidylinositol 3-kinase inhibitor,
wortmannin. Biochem J. 300:631–635. 1994.PubMed/NCBI
|
|
115
|
Golkar L, Salabat MR, Ding XZ, et al:
Apigenin inhibits pancreatic cancer cell proliferation via
down-regulation of the GLUT-1 glucose transporter through the
phosphoinositide 3-kinase (PI3K)/Akt signaling pathway (Abstract).
Pancreas. 33:4642006. View Article : Google Scholar
|
|
116
|
Pore N, Jiang Z, Shu HK, Bernhard E, Kao
GD and Maity A: Akt1 activation can augment hypoxia-inducible
factor-1alpha expression by increasing protein translation through
a mammalian target of rapamycin-independent pathway. Mol Cancer
Res. 4:471–479. 2006. View Article : Google Scholar : PubMed/NCBI
|
|
117
|
Plas DR, Talapatra S, Edinger AL, Rathmell
JC and Thompson CB: Akt and Bcl-xL promote growth
factor-independent survival through distinct effects on
mitochondrial physiology. J Biol Chem. 276:12041–12048. 2001.
View Article : Google Scholar : PubMed/NCBI
|
|
118
|
Fang J, Bao YY, Zhou SH, et al: Recurrent
prognostic factors and expression of GLUT-1, PI3K and p-Akt in
adenoid cystic carcinomas of the head and neck: Clinicopathological
features and biomarkers of adenoid cystic carcinoma. Oncol Lett.
4:1234–1240. 2012.PubMed/NCBI
|
|
119
|
Shen WQ, Cheng KJ, Bao YY, Zhou SH and Yao
HT: Expression of Glut-1, HIF-1α, PI3K and p-Akt in a case of
ceruminous adenoma. Head Neck Oncol. 4:182012. View Article : Google Scholar
|
|
120
|
Silva A, Gírio A, Cebola I, Santos CI,
Antunes F and Barata JT: Intracellular reactive oxygen species are
essential for PI3K/Akt/mTOR-dependent IL-7-mediated viability of
T-cell acute lymphoblastic leukemia cells. Leukemia. 25:960–967.
2011. View Article : Google Scholar : PubMed/NCBI
|
|
121
|
Barata JT, Silva A, Brandao JG, Nadler LM,
Cardoso AA and Boussiotis VA: Activation of PI3K is indispensable
for interleukin 7-mediated viability, proliferation, glucose use,
and growth of T cell acute lymphoblastic leukemia cells. J Exp Med.
200:659–669. 2004. View Article : Google Scholar : PubMed/NCBI
|
|
122
|
Wahl H, Daudi S, Kshirsagar M, et al:
Expression of metabolically targeted biomarkers in endometrial
carcinoma. Gynecol Oncol. 116:21–27. 2010. View Article : Google Scholar
|
|
123
|
Fumarola C, Caffarra C, La Monica S, et
al: Effects of sorafenib on energy metabolism in breast cancer
cells: role of AMPK-mTORC1 signaling. Breast Cancer Res Treat.
141:67–78. 2013. View Article : Google Scholar : PubMed/NCBI
|
|
124
|
Ou J, Luan W, Deng J, Sa R and Liang H: αV
integrin induces multicellular radioresistance in human
nasopharyngeal carcinoma via activating SAPK/JNK pathway. Plos One.
7:e387372012. View Article : Google Scholar
|
|
125
|
Xiao H, Zhang Q, Shen J, Bindokas V and
Xing HR: Pharmacologic inactivation of kinase suppressor of Ras1
sensitizes epidermal growth factor receptor and oncogenic
Ras-dependent tumors to ionizing radiation treatment. Mol Cancer
Ther. 9:2724–2736. 2010. View Article : Google Scholar : PubMed/NCBI
|
|
126
|
Li P, Zhang Q, Torossian A, et al:
Simultaneous inhibition of EGFR and PI3K enhances radiosensitivity
in human breast cancer. Int J Radiat Oncol Biol Phys. 83:e391–e397.
2012. View Article : Google Scholar : PubMed/NCBI
|
|
127
|
Tan C, de Noronha RG, Roecker AJ, et al:
Identification of a novel small-molecule inhibitor of the
hypoxia-inducible factor 1 pathway. Cancer Res. 65:605–612.
2005.PubMed/NCBI
|
|
128
|
Romeo Y and Roux PP: Paving the way for
targeting RSK in cancer. Expert Opin Ther Targets. 15:5–9. 2011.
View Article : Google Scholar
|
|
129
|
Cataldi A, di Giacomo V, Rapino M,
Genovesi D and Rana RA: Cyclic nucleotide Response Element Binding
protein (CREB) activation promotes survival signal in human K562
erythroleukemia cells exposed to ionising radiation/etoposide
combined treatment. J Radiat Res. 47:113–120. 2006. View Article : Google Scholar : PubMed/NCBI
|
|
130
|
Moretti L, Yang ES, Kim KW and Lu B:
Autophagy signaling in cancer and its potential as novel target to
improve anticancer therapy. Drug Resist Updat. 10:135–143. 2007.
View Article : Google Scholar : PubMed/NCBI
|
|
131
|
Shinohara ET, Cao C, Niermann K, et al:
Enhanced radiation damage of tumor vasculature by mTOR inhibitors.
Oncogene. 24:5414–5422. 2005. View Article : Google Scholar : PubMed/NCBI
|
|
132
|
Hudes G, Carducci M, Tomczak P, et al:
Temsirolimus, interferon alpha, or both for advanced renal-cell
carcinoma. N Engl J Med. 356:2271–2281. 2007. View Article : Google Scholar : PubMed/NCBI
|