메뉴 건너뛰기




Volumn 41, Issue 5, 2011, Pages 334-344

Preclinical assessment of strategies for enhancement of metaiodobenzylguanidine therapy of neuroendocrine tumors

Author keywords

[No Author keywords available]

Indexed keywords

(3 IODOBENZYL)GUANIDINE; (3 IODOBENZYL)GUANIDINE I 123; (3 IODOBENZYL)GUANIDINE I 131; ASTATINE 211; BORTEZOMIB; DISULFIRAM; HISTONE DEACETYLASE INHIBITOR; META ASTATOBENZYLGUANIDINE AT 211; NICOTINAMIDE ADENINE DINUCLEOTIDE ADENOSINE DIPHOSPHATE RIBOSYLTRANSFERASE INHIBITOR; RADIOPHARMACEUTICAL AGENT; TOPOTECAN; UNCLASSIFIED DRUG;

EID: 79960930191     PISSN: 00012998     EISSN: 15584623     Source Type: Journal    
DOI: 10.1053/j.semnuclmed.2011.03.004     Document Type: Review
Times cited : (14)

References (144)
  • 1
    • 0024338618 scopus 로고
    • Active uptake and extravesicular storage of m-iodobenzylguanidine in human neuroblastoma SK-N-SH cells
    • Smets LA, Loesberg C, Janssen M, et al: Active uptake and extravesicular storage of m-iodobenzylguanidine in human neuroblastoma SKNSH cells. Cancer Res 49:2941-2944, 1989 (Pubitemid 19148630)
    • (1989) Cancer Research , vol.49 , Issue.11 , pp. 2941-2944
    • Smets, L.A.1    Loesberg, C.2    Janssen, M.3    Metwally, E.A.4    Huiskamp, R.5
  • 2
    • 0028245761 scopus 로고
    • Radioiodinated metaiodobenzylguanidine: A review of its biodistribution and pharmacokinetics, drug interactions, cytotoxicity and dosimetry
    • Wafelman AR, Hoefnagel CA, Maes RA, et al: Radio-iodinated MIBG: a review of its biodistribution and pharmacokinetics, drug interactions, cytotoxicity and dosimetry. Eur J Nucl Med 21:545-559, 1994 (Pubitemid 24189419)
    • (1994) European Journal of Nuclear Medicine , vol.21 , Issue.6 , pp. 545-559
    • Wafelman, A.R.1    Hoefnagel, C.A.2    Maes, R.A.A.3    Beijnen, J.H.4
  • 3
    • 0025726824 scopus 로고
    • Intracellular localization of mIBG in human neuroblastoma cells by electron spectroscopic imaging
    • Gaze MN, Huxham IM, Mairs RJ, et al: Intracellular localization of mIBG in human neuroblastoma cells by electron spectroscopic imaging. Int J Cancer 47:875-880, 1991
    • (1991) Int J Cancer , vol.47 , pp. 875-880
    • Gaze, M.N.1    Huxham, I.M.2    Mairs, R.J.3
  • 4
    • 0029060571 scopus 로고
    • No-carrier-added 131I-mIBG: Evaluation of a novel preparation of a therapeutic radiopharmaceutical
    • Mairs RJ, Cunningham SH, Russell J, et al: No-carrier-added 131I-mIBG: evaluation of a novel preparation of a therapeutic radiopharmaceutical. J Nucl Med 36:1088-1095, 1995
    • (1995) J Nucl Med , vol.36 , pp. 1088-1095
    • Mairs, R.J.1    Cunningham, S.H.2    Russell, J.3
  • 5
    • 18844423539 scopus 로고    scopus 로고
    • 131I-meta- iodobenzylguanidine with topotecan as a radiosensitizer in children with metastatic neuroblastoma
    • 131I-meta-iodobenzylguanidine with topotecan as a radiosensitizer in children with metastatic neuroblastoma. Cancer Biother Radiopharm 20:195-199, 2005
    • (2005) Cancer Biother Radiopharm , vol.20 , pp. 195-199
    • Gaze, M.N.1    Chang, Y.C.2    Flux, G.D.3
  • 6
    • 33845510523 scopus 로고    scopus 로고
    • Malignant pheochromocytoma in a population-based study
    • Wangberg B, Muth A, Khorram-Manesh A, et al: Malignant pheochromocytoma in a population-based study. Ann N Y Acad Sci 1073:512-516, 2006
    • (2006) Ann N Y Acad Sci , vol.1073 , pp. 512-516
    • Wangberg, B.1    Muth, A.2    Khorram-Manesh, A.3
  • 8
    • 0029061599 scopus 로고
    • Clinical experience with radiation enhancement by hyperbaric oxygen in children with recurrent neuroblastoma stage IV
    • Voute PA, van der Kleij AJ, De Kraker J, et al: Clinical experience with radiation enhancement by hyperbaric oxygen in children with recurrent neuroblastoma stage IV. Eur J Cancer 31A:596-600, 1995
    • (1995) Eur J Cancer , vol.31 A , pp. 596-600
    • Voute, P.A.1    Van Der Kleij, A.J.2    De Kraker, J.3
  • 9
    • 11144273908 scopus 로고    scopus 로고
    • Hyperbaric oxygen to intensify 131-I-MIBG therapy in refractory neuroblastoma stage 4
    • Diehl M, Fußhoeller G, Menzel C, et al: Hyperbaric oxygen to intensify 131-I-MIBG therapy in refractory neuroblastoma stage 4. Nuklearmedizin 6:N77-N80, 2004
    • (2004) Nuklearmedizin , vol.6
    • Diehl, M.1    Fußhoeller, G.2    Menzel, C.3
  • 10
    • 0038345477 scopus 로고    scopus 로고
    • 131I-metaiodobenzylguanidine therapy for 12 patients with malignant pheochromocytoma
    • 131I- metaiodobenzylguanidine therapy for 12 patients with malignant pheochromocytoma. Cancer 98:239-248, 2003
    • (2003) Cancer , vol.98 , pp. 239-248
    • Rose, B.1    Matthay, K.K.2    Price, D.3
  • 11
    • 0028960625 scopus 로고
    • Multi-modality megatherapy with [131I]meta-iodobenzylguanidine, high-dose melphalan and total body irradiation with bone marrow rescue: Feasibility study of a new strategy for advanced neuroblastoma
    • Gaze MN, Wheldon TE, O'Donoghue JA, et al: Multi-modality megatherapy with [131I]meta-iodobenzylguanidine, high-dose melphalan and total body irradiation with bone marrow rescue: feasibility study of a new strategy for advanced neuroblastoma. Eur J Cancer 31A:252-256, 1995
    • (1995) Eur J Cancer , vol.31 , pp. 252-256
    • Gaze, M.N.1    Wheldon, T.E.2    O'Donoghue, J.A.3
  • 12
    • 0000764141 scopus 로고
    • Solid-phase exchange radioiodination of aryl iodides: Facilitation by ammonium sulfate
    • Mangner JT, Wu J-L, Wieland DM: Solid-phase exchange radioiodination of aryl iodides: facilitation by ammonium sulfate. J Org Chem 47:1484-1488, 1982
    • (1982) J Org Chem , vol.47 , pp. 1484-1488
    • Mangner, J.T.1    Wu, J.-L.2    Wieland, D.M.3
  • 15
    • 0029664597 scopus 로고    scopus 로고
    • 131I]MIBG in nude mice bearing SKNSH human neuroblastoma xenografts: Effect of specific activity
    • 131I]MIBG in nude mice bearing SKNSH human neuroblastoma xenografts: effect of specific activity. Br J Cancer 73:1171-1177, 1996
    • (1996) Br J Cancer , vol.73 , pp. 1171-1177
    • Vaidyanathan, G.1    Friedman, H.S.2    Keir, S.T.3
  • 16
    • 0030950342 scopus 로고    scopus 로고
    • Effect of adrenergic receptor ligands on metaiodobenzylguanidine uptake and storage in neuroblastoma cells
    • DOI 10.1007/s002590050087
    • Babich JW, Graham W, Fischman AJ: Effect of adrenergic receptor ligands on metaiodobenzylguanidine uptake and storage in neuroblastoma cells. Eur J Nucl Med 24:538-543, 1997 (Pubitemid 27239213)
    • (1997) European Journal of Nuclear Medicine , vol.24 , Issue.5 , pp. 538-543
    • Babich, J.W.1    Graham, W.2    Fischman, A.J.3
  • 19
    • 0023898658 scopus 로고
    • 125I] iodobenzylguanidine on the human neuroblastoma cell lines SKNSH and SKN-LO
    • 125I]iodobenzylguanidine on the human neuroblastoma cell lines SKNSH and SKN-LO. Cancer Res 48:2993-2997, 1988
    • (1988) Cancer Res , vol.48 , pp. 2993-2997
    • Bruchelt, G.1    Girgert, R.2    Buck, J.3
  • 21
    • 33645869293 scopus 로고    scopus 로고
    • No-carrier-added versus carrier-added 123I-metaiodobenzylguanidine for the assessment of cardiac sympathetic nerve activity
    • Verberne HJ, de Bruin K, Habraken JBA, et al: No-carrier-added versus carrier-added 123I-metaiodobenzylguanidine for the assessment of cardiac sympathetic nerve activity. Eur J Nucl Med 33:483-490, 2006
    • (2006) Eur J Nucl Med , vol.33 , pp. 483-490
    • Verberne, H.J.1    De Bruin, K.2    Habraken, J.B.A.3
  • 22
    • 0343489023 scopus 로고
    • 131I]MIBG in xenografted BALB/c nu/nu mice
    • Evans AE, Biedler JL, Brodeur GM, et al (eds): (ed 4). New York, Wiley-Liss
    • 131I]MIBG in xenografted BALB/c nu/nu mice, in Evans AE, Biedler JL, Brodeur GM, et al (eds):Advances in Neuroblastoma Research (ed 4). New York, Wiley-Liss, 1994, pp 446-453
    • (1994) Advances in Neuroblastoma Research , pp. 446-453
    • Rutgers, M.1    Buitenhuis, C.K.2    Hoefnagel, C.A.3
  • 23
    • 0029006232 scopus 로고
    • Enhanced tumor uptake and in vitro radiotoxicity of no-carrier-added 131I-MIBG: Implications for the radiotherapy of neuroblastoma
    • Mairs RJ, Russell J, Cunningham SH, et al: Enhanced tumor uptake and in vitro radiotoxicity of no-carrier-added 131I-MIBG: implications for the radiotherapy of neuroblastoma. Eur J Cancer 31:576-581, 1995
    • (1995) Eur J Cancer , vol.31 , pp. 576-581
    • Mairs, R.J.1    Russell, J.2    Cunningham, S.H.3
  • 24
    • 27144487807 scopus 로고    scopus 로고
    • 131I]meta- iodobenzyl guanidine, for the treatment of tumours transfected with the noradrenaline transporter gene
    • 131I]meta-iodobenzyl guanidine, for the treatment of tumours transfected with the noradrenaline transporter gene. Lett Drug Des Disc 1:50-57, 2004
    • (2004) Lett Drug des Disc , vol.1 , pp. 50-57
    • Boyd, M.1    Ross, S.2    Owens, J.3
  • 28
    • 0030842506 scopus 로고    scopus 로고
    • Acute liver necrosis induced by iodine-131-MIBG in the treatment of metastatic carcinoid tumors
    • Bongers V, de Klerk JMH, Zonnenberg BA, et al: Acute liver necrosis induced by iodine-131-MIBG in the treatment of metastatic carcinoid tumors. J Nucl Med 38:1024-1026, 1997
    • (1997) J Nucl Med , vol.38 , pp. 1024-1026
    • Bongers, V.1    De Klerk, J.M.H.2    Zonnenberg, B.A.3
  • 30
    • 0029029458 scopus 로고
    • New MIBG preparation to improve targeted radiotherapy and reduce toxic side-effects in neuroblastoma patients undergoing combination treatment
    • Mairs RJ, Zalutsky MR: New MIBG preparation to improve targeted radiotherapy and reduce toxic side-effects in neuroblastoma patients undergoing combination treatment. Br J Cancer 72:250, 1995
    • (1995) Br J Cancer , vol.72 , pp. 250
    • Mairs, R.J.1    Zalutsky, M.R.2
  • 32
    • 17844375396 scopus 로고    scopus 로고
    • 123I] iodobenzylguanidine in clinical studies II
    • 123I]iodobenzylguanidine in clinical studies II. Eur J Nucl Med 28:941, 2001
    • (2001) Eur J Nucl Med , vol.28 , pp. 941
    • Knickmeier, M.1
  • 33
    • 0034184715 scopus 로고    scopus 로고
    • 123I]mIBG: An initial clinical study in patients with phaeochromocytoma
    • 123I]mIBG: an initial clinical study in patients with phaeochromocytoma. Nucl Med Commun21:437-440, 2000
    • (2000) Nucl Med Commun , vol.21 , pp. 437-440
    • Owens, J.1    Bolster, A.A.2    Prosser, J.3
  • 34
    • 49049091151 scopus 로고    scopus 로고
    • Radioiodinated MIBG in paraganglioma and pheochromocytoma: Previous results and early experiences using nocarrier- added MIBG
    • James O, Coleman RE: Radioiodinated MIBG in paraganglioma and pheochromocytoma: previous results and early experiences using nocarrier- added MIBG. Nucl Med Biol 35:S1:63-S1:76, 2009
    • (2009) Nucl Med Biol , vol.35
    • James, O.1    Coleman, R.E.2
  • 35
    • 0022885542 scopus 로고
    • Dosimetric aspects of radiolabeled antibodies for tumor therapy
    • Humm JL: Dosimetric aspects of radiolabeled antibodies for tumor therapy. J Nucl Med 27:1490-1497, 1986
    • (1986) J Nucl Med , vol.27 , pp. 1490-1497
    • Humm, J.L.1
  • 37
    • 0033848363 scopus 로고    scopus 로고
    • Astatine-211-labeled radiotherapeutics: An emerging approach to targeted alpha particle therapy
    • Zalutsky MR, Vaidyanathan G: Astatine-211-labeled radiotherapeutics: an emerging approach to targeted alpha particle therapy. Curr Pharm Des 6:1433-1455, 2000
    • (2000) Curr Pharm des , vol.6 , pp. 1433-1455
    • Zalutsky, M.R.1    Vaidyanathan, G.2
  • 38
    • 37649010802 scopus 로고    scopus 로고
    • Clinical experience with α-particle emitting 211At: Treatment of recurrent brain tumor patients with 211At-labeled chimeric antitenascin monoclonal antibody 81C6
    • Zalutsky MR, Reardon DA, Akabani G, et al: Clinical experience with α-particle emitting 211At: treatment of recurrent brain tumor patients with 211At-labeled chimeric antitenascin monoclonal antibody 81C6. J Nucl Med 49:30-38, 2008
    • (2008) J Nucl Med , vol.49 , pp. 30-38
    • Zalutsky, M.R.1    Reardon, D.A.2    Akabani, G.3
  • 41
    • 0028177363 scopus 로고
    • Meta-[211At]astatobenzylguanidine: Further evaluation of a potential therapeutic agent
    • Vaidyanathan G, Strickland DK, Zalutsky MR: Meta-[211At] astatobenzylguanidine: further evaluation of a potential therapeutic agent. Int J Cancer 57:908-913, 1994
    • (1994) Int J Cancer , vol.57 , pp. 908-913
    • Vaidyanathan, G.1    Strickland, D.K.2    Zalutsky, M.R.3
  • 42
    • 0031747436 scopus 로고    scopus 로고
    • Radiotoxicity to neuroblastoma cells and spheroids of beta-, alpha- and Auger electron-emitting conjugates of benzylguanidine
    • Cunningham SH, Mairs RJ, Wheldon TE, et al: Radiotoxicity to neuroblastoma cells and spheroids of beta-, alpha- and Auger electron-emitting conjugates of benzylguanidine. Br J Cancer 77:2061-2068, 1998
    • (1998) Br J Cancer , vol.77 , pp. 2061-2068
    • Cunningham, S.H.1    Mairs, R.J.2    Wheldon, T.E.3
  • 44
    • 33846052858 scopus 로고    scopus 로고
    • Comparison of radiohaloanalogues of meta-iodobenzylguanidine (MIBG) for a combined gene- And targeted radiotherapy approach to bladder carcinoma
    • Fullerton NE, Boyd M, Ross SC, et al: Comparison of radiohaloanalogues of meta-iodobenzylguanidine (MIBG) for a combined gene- and targeted radiotherapy approach to bladder carcinoma. J Med Chem 1:611-618, 2005
    • (2005) J Med Chem , vol.1 , pp. 611-618
    • Fullerton, N.E.1    Boyd, M.2    Ross, S.C.3
  • 45
    • 33746095486 scopus 로고    scopus 로고
    • Radiation-induced biologic bystander effect elicited in vitro by targeted radiopharmaceuticals labeled with alpha, beta-, and auger electron-emitting radionuclides
    • Boyd M, Ross SC, Dorrens J, et al: Radiation induced biological bystander effect elicited in vitro by targeted radiopharmaceuticals labeled with alpha, beta- and Auger electron emitting radionuclides. J Nucl Med 47:1007-1015, 2006 (Pubitemid 46939954)
    • (2006) Journal of Nuclear Medicine , vol.47 , Issue.6 , pp. 1007-1015
    • Boyd, M.1    Ross, S.C.2    Dorrens, J.3    Fullerton, N.E.4    Ker, W.T.5    Zalutsky, M.R.6    Mairs, R.J.7
  • 47
    • 0036726584 scopus 로고    scopus 로고
    • Transfectant mosaic spheroids: A new model for evaluation of tumour cell killing in targeted radiotherapy and experimental gene therapy
    • DOI 10.1002/jgm.293
    • Boyd M, Mairs SC, Stevenson K, et al: Transfectant mosaic spheroids: a new model for the evaluation of bystander effects in experimental gene therapy. J Gene Med 4:567-576, 2002 (Pubitemid 40268383)
    • (2002) Journal of Gene Medicine , vol.4 , Issue.5 , pp. 567-576
    • Boyd, M.1    Mairs, S.C.2    Stevenson, K.3    Livingstone, A.4    Clark, A.M.5    Ross, S.C.6    Mairs, R.J.7
  • 48
    • 0034980559 scopus 로고    scopus 로고
    • Radiation-induced bystander effects: Past history and future directions
    • Mothersill C, Seymour CB: Radiation induced bystander effects: past history and future directions. Radiat Res 155:759-767, 2001 (Pubitemid 32494601)
    • (2001) Radiation Research , vol.155 , Issue.6 , pp. 759-767
    • Mothersill, C.1    Seymour, C.2
  • 49
    • 1042278763 scopus 로고    scopus 로고
    • Radiation-induced bystander effects - Implications for cancer
    • Mothersill C, Seymour CB: Radiation-induced bystander effects: implications for cancer. Nat Rev Cancer 4:158-164, 2004 (Pubitemid 38198744)
    • (2004) Nature Reviews Cancer , vol.4 , Issue.2 , pp. 158-164
    • Mothersill, C.1    Seymour, C.B.2
  • 50
    • 0036023759 scopus 로고    scopus 로고
    • Early events in the apoptotic cascade initiated in cells treated with medium from the progeny of irradiated cells
    • Lyng FM, Seymour CB, Mothersill C: Early events in the apoptotic cascade initiated in cells treated with medium from the progeny of irradiated cells. Radiat Prot Dosimetry 99:169-172, 2002
    • (2002) Radiat Prot Dosimetry , vol.99 , pp. 169-172
    • Lyng, F.M.1    Seymour, C.B.2    Mothersill, C.3
  • 51
    • 0037250171 scopus 로고    scopus 로고
    • Radiation-induced genomic instability and bystander effects: Related inflammatory-type responses to radiation-induced stress and injury? A review
    • Lorimore SA, Wright EG: Radiation-induced genomic instability and bystander effects: related inflammatory-type responses to radiation-induced stress and injury? a review. Int J Radiat Biol 79:15-25, 2003 (Pubitemid 36140233)
    • (2003) International Journal of Radiation Biology , vol.79 , Issue.1 , pp. 15-25
    • Lorimore, S.A.1    Wright, E.G.2
  • 52
    • 0242658543 scopus 로고    scopus 로고
    • Is there a common mechanism underlying genomic instability, bystander effects and other nontargeted effects of exposure to ionising radiation?
    • Morgan WF: Is there a common mechanism underlying genomic instability, bystander effects and other nontargeted effects of exposure to ionising radiation? Oncogene 22:7094-7099, 2003
    • (2003) Oncogene , vol.22 , pp. 7094-7099
    • Morgan, W.F.1
  • 53
    • 0242658588 scopus 로고    scopus 로고
    • Genomic instability and bystander effects: A historical perspective
    • Little JB: Genomic instability and bystander effects: a historical perspective. Oncogene 22:6978-6987, 2003
    • (2003) Oncogene , vol.22 , pp. 6978-6987
    • Little, J.B.1
  • 55
    • 0035144598 scopus 로고    scopus 로고
    • Free radical-initiated and gap junction-mediated bystander effect due to nonuniform distribution of incorporated radioactivity in a three-dimensional tissue culture model
    • Bishayee A, Hill HZ, Stein D, et al: Free radical-initiated and gap junction-mediated bystander effect due to nonuniform distribution of incorporated radioactivity in a three-dimensional tissue culture model. Radiat Res 155:335-344, 2001 (Pubitemid 32125985)
    • (2001) Radiation Research , vol.155 , Issue.2 , pp. 335-344
    • Bishayee, A.1    Hill, H.Z.2    Stein, D.3    Rao, D.V.4    Howell, R.W.5
  • 57
    • 1642405531 scopus 로고    scopus 로고
    • In vivo validation of the bystander effect
    • Kassis AI: In vivo validation of the bystander effect. Hum Exp Toxicol 23:71-73, 2004
    • (2004) Hum Exp Toxicol , vol.23 , pp. 71-73
    • Kassis, A.I.1
  • 58
    • 0030898609 scopus 로고    scopus 로고
    • Medium from irradiated human epithelial cells but not human fibroblasts reduces the clonogenic survival of unirradiated cells
    • Mothersill C, Seymour CB: Medium from irradiated human epithelial cells but not human fibroblasts reduced clonogenic survival of unirradiated cells. Int J Radiat Biol 71:421-427, 1997 (Pubitemid 27187398)
    • (1997) International Journal of Radiation Biology , vol.71 , Issue.4 , pp. 421-427
    • Mothersill, C.1    Seymour, C.2
  • 59
    • 0034109161 scopus 로고    scopus 로고
    • Relative contribution of bystander and targeted cell killing to the low- dose region of the radiation dose-response curve
    • Seymour CB, Mothersill C: Relative contribution and targeted cell killing to the low-dose region of the radiation dose-response curve. Radiat Res 153:508-511, 2000 (Pubitemid 30252597)
    • (2000) Radiation Research , vol.153 , Issue.5 I , pp. 508-511
    • Seymour, C.B.1    Mothersill, C.2
  • 60
  • 61
    • 0028842671 scopus 로고
    • Relationships between tumour size and curability for uniformly targeted therapy with betaemitting radionuclides
    • O'Donoghue JA, Bardies M, Wheldon TE: Relationships between tumour size and curability for uniformly targeted therapy with betaemitting radionuclides. J Nucl Med 36:1902-1909, 1995
    • (1995) J Nucl Med , vol.36 , pp. 1902-1909
    • O'Donoghue, J.A.1    Bardies, M.2    Wheldon, T.E.3
  • 62
    • 0031050382 scopus 로고    scopus 로고
    • The effect of cisplatin pretreatment on the accumulation of MIBG by neuroblastoma cells in vitro
    • Armour A, Cunningham SH, Gaze MN, et al: The effect of cisplatin pretreatment on the accumulation of MIBG by neuroblastoma cells in vitro. Br J Cancer 75:470-476, 1997 (Pubitemid 27085284)
    • (1997) British Journal of Cancer , vol.75 , Issue.4 , pp. 470-476
    • Armour, A.1    Cunningham, S.H.2    Gaze, M.N.3    Wheldon, T.E.4    Mairs, R.J.5
  • 63
    • 0025903831 scopus 로고
    • Pharmacokinetics and intracellular distribution of the tumor-targeted radiopharmaceutical miodo- benzylguanidine in SKNSH neuroblastoma and PC-12 pheochromocytoma cells
    • Smets LA, Janssen M, Rutgers M, et al: Pharmacokinetics and intracellular distribution of the tumor-targeted radiopharmaceutical miodo- benzylguanidine in SKNSH neuroblastoma and PC-12 pheochromocytoma cells.Int J Cancer 48:609-615, 1991
    • (1991) Int J Cancer , vol.48 , pp. 609-615
    • Smets, L.A.1    Janssen, M.2    Rutgers, M.3
  • 64
    • 0026793893 scopus 로고
    • γInterferon increases metaiodobenzylguanidine incorporation and retention in humanneuroblastoma cells
    • Montaldo PG, Carbone R, Ponzoni M, et al: γInterferon increases metaiodobenzylguanidine incorporation and retention in humanneuroblastoma cells. Cancer Res 52:4960-4964, 1992
    • (1992) Cancer Res , vol.52 , pp. 4960-4964
    • Montaldo, P.G.1    Carbone, R.2    Ponzoni, M.3
  • 65
    • 0030038672 scopus 로고    scopus 로고
    • Increase of metaiodobenzylguanidine uptake and intracellular half-life during differentiation of human neuroblastoma cells
    • DOI 10.1002/(SICI)1097-0215(19960703)67:1<95::AID-IJC16>3.0.CO;2-B
    • Montaldo PG, Raffaghello L, Guarnaccia F, et al: Increase of metaiodobenzylguanidine uptake and intracellular half-life during differentiation of human neuroblastoma cells. Int J Cancer 67:95-100, 1996 (Pubitemid 26236074)
    • (1996) International Journal of Cancer , vol.67 , Issue.1 , pp. 95-100
    • Montaldo, P.G.1    Raffaghello, L.2    Guarnaccia, F.3    Pistoia, V.4    Garaventa, A.5    Ponzoni, M.6
  • 66
    • 3042798291 scopus 로고    scopus 로고
    • A novel approach in the treatment of neuroendocrine gastrointestinal tumors: Additive anti-proliferative effects of interferon-gamma and meta-iodobenzylguanidine
    • Hopfner M, Sutter AP, Huether A, et al: A novel approach in the treatment of neuroendocrine gastrointestinal tumors: additive anti-proliferative effects of interferon-gamma and meta-iodobenzylguanidine. BMC Cancer 4:23-37, 2004
    • (2004) BMC Cancer , vol.4 , pp. 23-37
    • Hopfner, M.1    Sutter, A.P.2    Huether, A.3
  • 67
    • 0008380616 scopus 로고    scopus 로고
    • Experimental tumour therapy with targeted radionuclides in multicellular tumour spheroids
    • Hagen U, Harder D, Jung H,et al (eds): Wurtzburg: Universitatsdruckerei, H. Sturtz AG
    • Mairs RJ, Wheldon TE: Experimental tumour therapy with targeted radionuclides in multicellular tumour spheroids, in Hagen U, Harder D, Jung H,et al (eds): Radiation Research 1895-1995; Congress Proceedings, 1996, Vol 2. Wurtzburg: Universitatsdruckerei, H. Sturtz AG, 1996, pp 819-826
    • (1996) Radiation Research 1895-1995; Congress Proceedings, 1996 , vol.2 , pp. 819-826
    • Mairs, R.J.1    Wheldon, T.E.2
  • 68
    • 0032813193 scopus 로고    scopus 로고
    • 125I-meta- iodobenzylguanidine uptake in human neuroblastoma cell lines
    • DOI 10.1016/S0959-8049(99)00078-7, PII S0959804999000787
    • Meco D, Lasorella A, Riccardi A, et al: Influence of cisplatin and doxorubicin on I-125-meta-iodobenzylguanidine uptake in human neuroblastoma cell lines. Eur J Cancer 35:1227-1234, 1999 (Pubitemid 29359183)
    • (1999) European Journal of Cancer , vol.35 , Issue.8 , pp. 1227-1234
    • Meco, D.1    Lasorella, A.2    Riccardi, A.3    Servidei, T.4    Mastrangelo, R.5    Riccardi, R.6
  • 69
    • 79251514664 scopus 로고    scopus 로고
    • 3H]norepinephrine in mouse pheochromocytoma cells and tumors after treatment with the histone deacetylase inhibitors
    • 3H]norepinephrine in mouse pheochromocytoma cells and tumors after treatment with the histone deacetylase inhibitors. Endocr Relat Cancer 18:143-149, 2011
    • (2011) Endocr Relat Cancer , vol.18 , pp. 143-149
    • Martiniova, L.1    Perera, S.M.2    Brouwers, F.M.3
  • 70
    • 0030826929 scopus 로고    scopus 로고
    • Topoisomerase I inhibitors: Review and update
    • Rothenberg ML: Topoisomerase I inhibitors: review and update.Ann Oncol 8:837-855, 1997
    • (1997) Ann Oncol , vol.8 , pp. 837-855
    • Rothenberg, M.L.1
  • 73
    • 23944497538 scopus 로고    scopus 로고
    • 131I]MIBG and topotecan: A rationale for combination therapy for neuroblastoma
    • 131I]MIBG and topotecan: a rationale for combination therapy for neuroblastoma. Cancer Lett 228:221-227, 2005
    • (2005) Cancer Lett , vol.228 , pp. 221-227
    • McCluskey, A.G.1    Boyd, M.2    Gaze, M.N.3
  • 75
    • 0026328062 scopus 로고
    • Synergistic cell killing by ionizing radiation and topoisomerase I inhibitor topotecan (SK and F 104864)
    • Mattern MR, Hofmann GA, McCabe FL, et al: Synergistic cell killing by ionizing radiation and topoisomerase I inhibitor topotecan (SK and F 104864). Cancer Res 51:5813-5816, 1991
    • (1991) Cancer Res , vol.51 , pp. 5813-5816
    • Mattern, M.R.1    Hofmann, G.A.2    McCabe, F.L.3
  • 76
    • 0027529978 scopus 로고
    • Interaction of topoisomerase I inhibitors with radiation in cis-diamminedichloroplatinum(II)-sensitive and -resistant cells in vitro and in the FSaIIC fibrosarcoma in vivo
    • DOI 10.1002/ijc.2910530122
    • Boscia RE, Korbut T, Holden SA, et al: Interaction of topoisomerase I inhibitors with radiation in cis-diamminedichloroplatinum(II)-sensitive and -resistant cells in vitro and in the FSaIIC fibrosarcoma in vivo. Int J Cancer 53:118-123, 1993 (Pubitemid 23018433)
    • (1993) International Journal of Cancer , vol.53 , Issue.1 , pp. 118-123
    • Boscia, R.E.1    Korbut, T.2    Holden, S.A.3    Ara, G.4    Teicher, B.A.5
  • 77
    • 0028328850 scopus 로고
    • Interaction of ionising radiation with the topoisomerase I poison camptothecin in growing V-79 and HeLa cells
    • Hennequin C, Giocanti N, Balosso J, et al: Interaction of ionising radiation with the topoisomerase I poison camptothecin in growing V-79 and HeLa cells. Cancer Res 54:1720-1728, 1994
    • (1994) Cancer Res , vol.54 , pp. 1720-1728
    • Hennequin, C.1    Giocanti, N.2    Balosso, J.3
  • 79
    • 0030248750 scopus 로고    scopus 로고
    • Concentration and timing dependence of lethality enhancement between topotecan, a topoisomerase I inhibitor, and ionizing radiation
    • DOI 10.1016/S0360-3016(96)00328-8
    • Lamond JP, Wang M, Kinsella TJ, et al: Concentration and timing dependence enhancement between topotecan, a topoisomerase I inhibitor and ionising radiation. Int J Radiat Oncol Biol Phys 36:361-368, 1996 (Pubitemid 26359462)
    • (1996) International Journal of Radiation Oncology Biology Physics , vol.36 , Issue.2 , pp. 361-368
    • Lamond, J.P.1    Wang, M.2    Kinsella, T.J.3    Boothman, D.A.4
  • 83
    • 0042570556 scopus 로고    scopus 로고
    • Oral topotecan for refractory and relapsed neuroblastoma: A retrospective analysis
    • DOI 10.1097/00043426-200308000-00003
    • Kramer K, Kushner BH, Cheung N-KV: Oral topotecan for refractory and relapsed neuroblastoma: a retrospective analysis. J Pediatr Hematol Oncol 25:601-605, 2003 (Pubitemid 36959419)
    • (2003) Journal of Pediatric Hematology/Oncology , vol.25 , Issue.8 , pp. 601-605
    • Kramer, K.1    Kushner, B.H.2    Cheung, N.-K.V.3
  • 85
    • 69549145898 scopus 로고    scopus 로고
    • The potential role and application of PARP inhibitors in cancer treatment
    • Chalmers AC: The potential role and application of PARP inhibitors in cancer treatment. Br Med Bull 89:23-40, 2009
    • (2009) Br Med Bull , vol.89 , pp. 23-40
    • Chalmers, A.C.1
  • 88
    • 54049112348 scopus 로고    scopus 로고
    • Replication-dependent radiosensitisation of human glioma cells by inhibition of poly(ADP-ribose) polymerase: Mechanisms and therapeutic potential
    • Dungey FA, Loeser DA, Chalmers AJ: Replication-dependent radiosensitisation of human glioma cells by inhibition of poly(ADP-ribose) polymerase: mechanisms and therapeutic potential. Int J Radiat Oncol Biol Phys 72:1188-1197, 2008
    • (2008) Int J Radiat Oncol Biol Phys , vol.72 , pp. 1188-1197
    • Dungey, F.A.1    Loeser, D.A.2    Chalmers, A.J.3
  • 89
    • 28544441999 scopus 로고    scopus 로고
    • The novel poly(ADP-ribose) polymerase inhibitor, AG14361 sensitizes cells to topoisomerase I poisons by increasing the persistence of DNA strand breaks
    • DOI 10.1158/1078-0432.CCR-05-1224
    • Smith LM, Willmore E, Austin CA, et al: The novel poly(ADP-Ribose) polymerase inhibitor, AG14361, sensitizes cells to topoisomerase I poisons by increasing the persistence of DNA strand breaks. Clin Cancer Res 11:8449-8457, 2005 (Pubitemid 41746960)
    • (2005) Clinical Cancer Research , vol.11 , Issue.23 , pp. 8449-8457
    • Smith, L.M.1    Willmore, E.2    Austin, C.A.3    Curtin, N.J.4
  • 90
    • 24944544025 scopus 로고    scopus 로고
    • Poly(ADP-ribose)polymerase inhibition: Where now?
    • Woon EC, Threadgill MD: Poly(ADP-ribose)polymerase inhibition: where now? Curr Med Chem 12:2373-2392, 2005
    • (2005) Curr Med Chem , vol.12 , pp. 2373-2392
    • Woon, E.C.1    Threadgill, M.D.2
  • 91
    • 1042303686 scopus 로고    scopus 로고
    • Poly(ADP-ribose) polymerase-1 and ionizing radiation: Sensor, signaller and therapeutic target
    • Chalmers AJ: Poly(ADP-ribose) polymerase-1 and ionizing radiation: sensor, signaller and therapeutic target. Clin Oncol 16:29-39, 2004
    • (2004) Clin Oncol , vol.16 , pp. 29-39
    • Chalmers, A.J.1
  • 92
    • 1242271229 scopus 로고    scopus 로고
    • Radiosensitization of human and rodent cell lines by INO-1001, a novel inhibitor of poly(ADP-ribose) polymerase
    • DOI 10.1016/j.canlet.2003.10.029
    • Brock WA, Milas L, Bergh S, et al: Radiosensitization of human and rodent cell lines by INO-1001, a novel inhibitor of poly(ADP-ribose) polymerase. Cancer Lett 205:155-160, 2004 (Pubitemid 38230122)
    • (2004) Cancer Letters , vol.205 , Issue.2 , pp. 155-160
    • Brock, W.A.1    Milas, L.2    Bergh, S.3    Lo, R.4    Szabo, C.5    Mason, K.A.6
  • 94
    • 54049112348 scopus 로고    scopus 로고
    • Replication-dependent radiosensitization of human glioma cells by inhibition of poly(ADP-Ribose) polymerase: Mechanisms and therapeutic potential
    • Dungey FA, Loser DA, Chalmers AJ: Replication-dependent radiosensitization of human glioma cells by inhibition of poly(ADP-Ribose) polymerase: mechanisms and therapeutic potential. Int J Radiat Oncol Biol Phys 72:1188-1197, 2008
    • (2008) Int J Radiat Oncol Biol Phys , vol.72 , pp. 1188-1197
    • Dungey, F.A.1    Loser, D.A.2    Chalmers, A.J.3
  • 95
    • 4544348052 scopus 로고    scopus 로고
    • Poly(ADP-ribosyl)ation as a DNA damage-induced post-translational modification regulating poly(ADP-ribose) polymerase-1-topoisomerase I interaction
    • DOI 10.1074/jbc.M402729200
    • Yung TM, Sato S, Satoh MS: Poly(ADP-ribosyl)ation as a DNA damage-induced post-translational modification regulating poly(ADP-ribose) polymerase-1-topoisomerase I interaction. J Biol Chem 279:39686-39696, 2004 (Pubitemid 39258237)
    • (2004) Journal of Biological Chemistry , vol.279 , Issue.38 , pp. 39686-39696
    • Yung, T.M.C.1    Sato, S.2    Satoh, M.S.3
  • 96
    • 1242339684 scopus 로고    scopus 로고
    • Poly(ADP-ribose) Reactivates Stalled DNA Topoisomerase I and Induces DNA Strand Break Resealing
    • DOI 10.1074/jbc.C300437200
    • Malanga M, Althaus FR: Poly(ADP-ribose) reactivates stalled DNA topoisomerase I and induces DNA strand break resealing. J Biol Chem 279:5244-5248, 2004 (Pubitemid 38220543)
    • (2004) Journal of Biological Chemistry , vol.279 , Issue.7 , pp. 5244-5248
    • Malanga, M.1    Althaus, F.R.2
  • 97
    • 0030057744 scopus 로고    scopus 로고
    • Camptothecin resistance involving steps subsequent to the formation of protein-linked DNA breaks in human camptothecin-resistant KB cell lines
    • Beidler DR, Chang JY, Zhou BS, et al: Camptothecin resistance involving steps subsequent to the formation of protein-linked DNA breaks in human camptothecin-resistant KB cell lines. Cancer Res 56:345-353, 1996 (Pubitemid 26026364)
    • (1996) Cancer Research , vol.56 , Issue.2 , pp. 345-353
    • Beidler, D.R.1    Chang, J.-Y.2    Zhou, B.-S.3    Cheng, Y.-C.4
  • 100
    • 0035137364 scopus 로고    scopus 로고
    • Differential effects of the poly (ADP-ribose) polymerase (PARP) inhibitor NU1025 on topoisomerase I and II inhibitor cytotoxicity in L1210 cells in vitro
    • DOI 10.1054/bjoc.2000.1555
    • Bowman KJ, Newell DR, Calvert AH, et al: Differential effects of the poly (ADP-ribose) polymerase (PARP) inhibitor NU1025 on topoisomerase I and II inhibitor cytotoxicity in L1210 cells in vitro. Br J Cancer 84:106-112, 2001 (Pubitemid 32102513)
    • (2001) British Journal of Cancer , vol.84 , Issue.1 , pp. 106-112
    • Bowman, K.J.1    Newell, D.R.2    Calvert, A.H.3    Curtin, N.J.4
  • 101
    • 0036447366 scopus 로고    scopus 로고
    • Potential clinical applications of poly(ADP-ribose) polymerase (PARP) inhibitors
    • DOI 10.1006/phrs.2001.0935
    • Tentori L, Portarena I, Graziani G: Potential clinical applications of poly(ADP-ribose) polymerase (PARP) inhibitors. Pharmacol Res45:73-85, 2002 (Pubitemid 35425647)
    • (2002) Pharmacological Research , vol.45 , Issue.2 , pp. 73-85
    • Tentori, L.1    Portarena, I.2    Graziani, G.3
  • 102
    • 4143130089 scopus 로고    scopus 로고
    • Poly(ADP-ribosyl)ation inhibitors: Promising drug candidates for a wide variety of pathophysiologic conditions
    • DOI 10.1002/ijc.20342
    • Beneke S, Diefenbach J, Bürkle A, et al: Poly(ADP-ribosyl)ation inhibitors: promising drug candidates for a wide variety of pathophysiologic conditions. Int J Cancer 111:813-818, 2004 (Pubitemid 39096229)
    • (2004) International Journal of Cancer , vol.111 , Issue.6 , pp. 813-818
    • Beneke, S.1    Diefenbach, J.2    Burkle, A.3
  • 103
    • 19444362951 scopus 로고    scopus 로고
    • Clinical perspectives of PARP inhibitors
    • DOI 10.1016/j.phrs.2005.02.013, PII S1043661805000460
    • Graziani G, Szabo C: Clinical perspectives of PARP inhibitors.Pharmacol Res 52:109-118, 2005 (Pubitemid 40725605)
    • (2005) Pharmacological Research , vol.52 , Issue.1 SPEC. ISS. , pp. 109-118
    • Graziani, G.1    Szabo, C.2
  • 104
    • 18944390248 scopus 로고    scopus 로고
    • Poly(ADP-ribose) polymerase and the therapeutic effects of its inhibitors
    • DOI 10.1038/nrd1718
    • Jagtap P, Szabo C: Poly(ADP-ribose) polymerase and the therapeutic effects of its inhibitors. Nat Rev Drug Discov 4:421-440, 2005 (Pubitemid 40704125)
    • (2005) Nature Reviews Drug Discovery , vol.4 , Issue.5 , pp. 421-440
    • Jagtap, P.1    Szabo, C.2
  • 105
    • 0036181371 scopus 로고    scopus 로고
    • Development of the proteasome inhibitor, PS-341
    • Adams J, Boyd M: Development of the proteasome inhibitor, PS-341. Oncologist 7:9-16, 2002
    • (2002) Oncologist , vol.7 , pp. 9-16
    • Adams, J.1    Boyd, M.2
  • 107
    • 16544389047 scopus 로고    scopus 로고
    • Phase I study of the proteasome inhibitor bortezomib in pediatric patients with refractory solid tumors: A Children's Oncology Group study (ADVL0015)
    • Blaney SM, Bernstein M, Neville K, et al: Phase I study of the proteasome inhibitor bortezomib in pediatric patients with refractory solid tumors: a Children's Oncology Group study (ADVL0015). J Clin Oncol 22:4804-4809, 2004
    • (2004) J Clin Oncol , vol.22 , pp. 4804-4809
    • Blaney, S.M.1    Bernstein, M.2    Neville, K.3
  • 108
    • 0030854371 scopus 로고    scopus 로고
    • Ubiquitin-dependent destruction of topoisomerase I is stimulated by the antitumor drug camptothecin
    • DOI 10.1074/jbc.272.39.24159
    • Desai SD, Liu LF, Vasquez-Abad D, et al: Ubiquitin-dependent destruction of topoisomerase I is stimulated by the antitumour drug camptothecin. J Biol Chem 272:24159-24164, 1997 (Pubitemid 27418613)
    • (1997) Journal of Biological Chemistry , vol.272 , Issue.39 , pp. 24159-24164
    • Desai, S.D.1    Liu, L.F.2    Vazquez-Abad, D.3    D'Arpa, P.4
  • 109
    • 26844545414 scopus 로고    scopus 로고
    • The proteasome inhibitor MG-132 sensitizes PC-3 prostate cancer cells to ionizing radiation by a DNA-PK-independent mechanism
    • Pajonk F, van Ophoven A, Weissenberger C, et al: The proteasome inhibitor MG-132 sensitizes PC-3 prostate cancer cells to ionizing radiation by a DNA-PK-independent mechanism. BMC Cancer 5:76, 2005
    • (2005) BMC Cancer , vol.5 , pp. 76
    • Pajonk, F.1    Van Ophoven, A.2    Weissenberger, C.3
  • 110
    • 77950300115 scopus 로고    scopus 로고
    • Proteasome inhibitor bortezomib increases radiation sensitivity in androgen independent human prostate cancer cells
    • Goktas S, Baran Y, Ural AU, et al: Proteasome inhibitor bortezomib increases radiation sensitivity in androgen independent human prostate cancer cells. Urology 75:793-798, 2010
    • (2010) Urology , vol.75 , pp. 793-798
    • Goktas, S.1    Baran, Y.2    Ural, A.U.3
  • 111
    • 79551503105 scopus 로고    scopus 로고
    • Radiosensitizing properties of bortezomib depend on therapeutic schedule
    • Labussière M, Pinel S, Vandamme M, et al: Radiosensitizing properties of bortezomib depend on therapeutic schedule. Int J Radiat Oncol Biol Phys 79:892-900, 2011
    • (2011) Int J Radiat Oncol Biol Phys , vol.79 , pp. 892-900
    • Labussière, M.1    Pinel, S.2    Vandamme, M.3
  • 113
    • 0028900690 scopus 로고
    • Persistent oxidative stress in cancer
    • Toyokuni S, Okamoto K, Yodoi J, et al: Persistent oxidative stress in cancer. FEBS Lett 358:1-3, 1995
    • (1995) FEBS Lett , vol.358 , pp. 1-3
    • Toyokuni, S.1    Okamoto, K.2    Yodoi, J.3
  • 114
    • 33745489431 scopus 로고    scopus 로고
    • Oxidative and nitrative DNA damage in animals and patients with inflammatory diseases in relation to inflammation-related carcinogenesis
    • Kawanishi S, Hiraku Y, Pinlaor S, et al: Oxidative and nitrative DNA damage in animals and patients with inflammatory diseases in relation to inflammation-related carcinogenesis. Biol Chem 387:365-372, 2006
    • (2006) Biol Chem , vol.387 , pp. 365-372
    • Kawanishi, S.1    Hiraku, Y.2    Pinlaor, S.3
  • 115
    • 0030988524 scopus 로고    scopus 로고
    • Antioxidant enzyme levels in cancer
    • Oberley TD, Oberley LW: Antioxidant enzyme levels in cancer. Histol Histopathol 12:525-535, 1997 (Pubitemid 27240324)
    • (1997) Histology and Histopathology , vol.12 , Issue.2 , pp. 525-535
    • Oberley, T.D.1
  • 117
    • 79951534617 scopus 로고    scopus 로고
    • PKCδ sensitizes neuroblastoma cells to L-buthionine-sulfoximine and etoposide inducing reactive oxygen species overproduction and DNA damage
    • Marengo B, De Ciucis C, Ricciarelli R, et al: PKCδ sensitizes neuroblastoma cells to L-buthionine-sulfoximine and etoposide inducing reactive oxygen species overproduction and DNA damage. PLoS ONE 6:e14661:1-12, 2011
    • (2011) PLoS ONE , vol.6
    • Marengo, B.1    De Ciucis, C.2    Ricciarelli, R.3
  • 118
    • 67650071137 scopus 로고    scopus 로고
    • Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach?
    • Trachootham D, Alexandre J, Huang P, et al: Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Natl Rev 8:579-591, 2009
    • (2009) Natl Rev , vol.8 , pp. 579-591
    • Trachootham, D.1    Alexandre, J.2    Huang, P.3
  • 120
    • 33751285781 scopus 로고    scopus 로고
    • Disulfiram, a clinically used anti-alcoholism drug and copper-binding agent, induces apoptotic cell death in breast cancer cultures and xenografts via inhibition of the proteasome activity
    • DOI 10.1158/0008-5472.CAN-06-2126
    • Chen D, Cui QC, Yang H, et al: Disulfiram, a clinically used anti-alcoholism drug and copper-binding agent, induces apoptotic cell death in breast cancer cultures and xenografts via inhibition of the proteasome activity. Cancer Res 66:10425-10433, 2006 (Pubitemid 44799763)
    • (2006) Cancer Research , vol.66 , Issue.21 , pp. 10425-10433
    • Chen, D.1    Cui, Q.C.2    Yang, H.3    Dou, Q.P.4
  • 121
    • 0026590541 scopus 로고
    • Dithiocarbamates as potent inhibitors of nuclear factor B activation in intact cells
    • Schreck R, Meier B, Mannel DN, et al: Dithiocarbamates as potent inhibitors of nuclear factor B activation in intact cells. J Exp Med 175:1181-1194, 1992
    • (1992) J Exp Med , vol.175 , pp. 1181-1194
    • Schreck, R.1    Meier, B.2    Mannel, D.N.3
  • 122
    • 0036223193 scopus 로고    scopus 로고
    • Retinoids enhance cisplatin-based chemoradiation in cervical cancer cells in vitro
    • Scribner DR, Benbrook DM: Retinoids enhance cisplatin-based chemoradiation in cervical cancer cells in vitro. Gynecol Oncol 85:223-225, 2002
    • (2002) Gynecol Oncol , vol.85 , pp. 223-225
    • Scribner, D.R.1    Benbrook, D.M.2
  • 123
    • 0033233191 scopus 로고    scopus 로고
    • The inhibition of NF-B activation pathways and the induction of apoptosis by dithiocarbamates in T cells are blocked by the glutathione precursor N-acetyl-L-cysteine
    • Fernandez PC, Machado JJ, Heussler VT, et al: The inhibition of NF-B activation pathways and the induction of apoptosis by dithiocarbamates in T cells are blocked by the glutathione precursor N-acetyl-l-cysteine. Biol Chem 380:1383-1394, 1999 (Pubitemid 30021360)
    • (1999) Biological Chemistry , vol.380 , Issue.12 , pp. 1383-1394
    • Fernandez, P.C.1    Machado Jr., J.2    Heussler, V.T.3    Botteron, C.4    Palmer, G.H.5    Dobbelaere, D.A.E.6
  • 124
    • 33745298519 scopus 로고    scopus 로고
    • Nuclear factor-kB in cancer development and progression
    • Karin M: Nuclear factor-kB in cancer development and progression. Nature 441:431-436, 2006
    • (2006) Nature , vol.441 , pp. 431-436
    • Karin, M.1
  • 125
    • 0031855760 scopus 로고    scopus 로고
    • Induction of apoptosis by thiuramdisulfides, the reactive metabolites of dithiocarbamates, through coordinative modulation of NFB, c-fos/c-jun, and p53 proteins
    • DOI 10.1002/(SICI)1098-2744(199808)22:4<235::AID-MC5>3.0.CO;2-I
    • Liu GY, Frank N, Bartsch H, et al: Induction of apoptosis by thiuram-disulfides, the reactive metabolites of dithiocarbamates, through coordinative modulation of NFB, c-fos/c-jun, and p53 proteins. Mol Carcinogen 22:235-246, 1998 (Pubitemid 28394985)
    • (1998) Molecular Carcinogenesis , vol.22 , Issue.4 , pp. 235-246
    • Liu, G.-Y.1    Frank, N.2    Bartsch, H.3    Lin, J.-K.4
  • 126
    • 0036050582 scopus 로고    scopus 로고
    • Disulfiram induces apoptosis in human melanoma cells: A redox-related process
    • Cen D, Gonzalez RI, Buckmeier JA, et al: Disulfiram induces apoptosis in human melanoma cells: a redox-related process. Mol Cancer Ther 1:197-204, 2002
    • (2002) Mol Cancer Ther , vol.1 , pp. 197-204
    • Cen, D.1    Gonzalez, R.I.2    Buckmeier, J.A.3
  • 127
    • 0032905030 scopus 로고    scopus 로고
    • Control of inducible chemoresistance: Enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-B
    • DOI 10.1038/7410
    • Wang C-Y, Cusack JCJ, Liu R, et al: Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-B. Nat Med 5:412-417, 1999 (Pubitemid 29180579)
    • (1999) Nature Medicine , vol.5 , Issue.4 , pp. 412-417
    • Wang, C.-Y.1    Cusack Jr., J.C.2    Liu, R.3    Baldwin Jr., A.S.4
  • 128
    • 0034192390 scopus 로고    scopus 로고
    • Inducible chemoresistance to 7-ethyl-10-[4-(1-piperidino)-1-piperidino]- carbonyloxycamptothecin (CPT-11) in colorectal cancer cells and a xenograft model is overcome by inhibition of nuclear factor-B activation
    • Cusack JCJ, Liu R, Baldwin AS Jr, et al: Inducible chemoresistance to 7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxycamptothe cin (CPT-11) in colorectal cancer cells and a xenograft model is overcome by inhibition of nuclear factor-B activation. Cancer Res 60:2323-2330, 2000 (Pubitemid 30262414)
    • (2000) Cancer Research , vol.60 , Issue.9 , pp. 2323-2330
    • Cusack Jr., J.C.1    Liu, R.2    Baldwin Jr., A.S.3
  • 130
    • 0345149546 scopus 로고    scopus 로고
    • Oxidative stress induces NF-kappaB nuclear translocation without degredation of IkappaBalpha
    • Canty TG, Boyle EM, Farr A, et al: Oxidative stress induces NF-kappaB nuclear translocation without degredation of IkappaBalpha. Circulation 100:11361-11364, 1999
    • (1999) Circulation , vol.100 , pp. 11361-11364
    • Canty, T.G.1    Boyle, E.M.2    Farr, A.3
  • 131
    • 33747892114 scopus 로고    scopus 로고
    • A concise review of DNA damage checkpoints and repair in mammalian cells
    • DOI 10.1016/j.carrev.2006.02.002, PII S1553838906000212
    • Houtgraaf JH, Versmissen J, van der Giessen WJ, et al: A concise review of DNA damage checkpoints and repair in mammalian cells. Cardiovasc Revasc Med 7:165-172, 2006 (Pubitemid 44292065)
    • (2006) Cardiovascular Revascularization Medicine , vol.7 , Issue.3 , pp. 165-172
    • Houtgraaf, J.H.1    Versmissen, J.2    Van Der Giessen, W.J.3
  • 132
    • 38949147038 scopus 로고    scopus 로고
    • G2 checkpoint abrogation and checkpoint kinase-1 targeting in the treatment of cancer
    • DOI 10.1038/sj.bjc.6604208, PII 6604208
    • Bucher N, Britten CD: G2 checkpoint abrogation and checkpoint kinase-1 targeting in the treatment of cancer. Br J Cancer 98:523-528, 2008 (Pubitemid 351214533)
    • (2008) British Journal of Cancer , vol.98 , Issue.3 , pp. 523-528
    • Bucher, N.1    Britten, C.D.2
  • 133
    • 52949139387 scopus 로고    scopus 로고
    • AZD7762, a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies
    • Zabludoff SD, Deng C, Grondine MR, et al: AZD7762, a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies. Mol Cancer Ther 7:2955-2966, 2008
    • (2008) Mol Cancer Ther , vol.7 , pp. 2955-2966
    • Zabludoff, S.D.1    Deng, C.2    Grondine, M.R.3
  • 134
    • 77955653997 scopus 로고    scopus 로고
    • Discovery of a novel class of triazolones as checkpoint kinase inhibitors: Hit to lead exploration
    • Oza V, Ashwell S, Brassil P, et al: Discovery of a novel class of triazolones as checkpoint kinase inhibitors: hit to lead exploration. Bioorg Med Chem Lett 20:5133-5138, 2010
    • (2010) Bioorg Med Chem Lett , vol.20 , pp. 5133-5138
    • Oza, V.1    Ashwell, S.2    Brassil, P.3
  • 135
    • 33847065486 scopus 로고    scopus 로고
    • The Epigenomics of Cancer
    • DOI 10.1016/j.cell.2007.01.029, PII S0092867407001274
    • Jones PA, Baylin SB: The epigenomics of cancer. Cell 128:683-692, 2007 (Pubitemid 46273572)
    • (2007) Cell , vol.128 , Issue.4 , pp. 683-692
    • Jones, P.A.1    Baylin, S.B.2
  • 136
    • 67449158992 scopus 로고    scopus 로고
    • Stepwise DNA methylation changes are linked to escape from defined proliferation barriers and mammary epithelial cell immortalization
    • Novak P, Jensen TJ, Garbe JC, et al: Stepwise DNA methylation changes are linked to escape from defined proliferation barriers and mammary epithelial cell immortalization. Cancer Res 69:5251-5260, 2009
    • (2009) Cancer Res , vol.69 , pp. 5251-5260
    • Novak, P.1    Jensen, T.J.2    Garbe, J.C.3
  • 137
    • 33748451151 scopus 로고    scopus 로고
    • Anticancer activities of histone deacetylase inhibitors
    • DOI 10.1038/nrd2133, PII NRD2133
    • Bolden JE, Peart MJ, Johnstone RW: Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 5:769-784, 2006 (Pubitemid 44348499)
    • (2006) Nature Reviews Drug Discovery , vol.5 , Issue.9 , pp. 769-784
    • Bolden, J.E.1    Peart, M.J.2    Johnstone, R.W.3
  • 139
    • 30344477367 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer
    • DOI 10.1038/nrc1779
    • Minucci S, Pelicci PG: Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6:38-51, 2006 (Pubitemid 43054973)
    • (2006) Nature Reviews Cancer , vol.6 , Issue.1 , pp. 38-51
    • Minucci, S.1    Pelicci, P.G.2
  • 140
    • 33745041170 scopus 로고    scopus 로고
    • Histone Deacetylation as a Target for Radiosensitization
    • DOI 10.1016/S0070-2153(05)73006-4, PII S0070215305730064
    • Cerna D, Camphausen K, Tofilon PJ: Histone deacetylation as a target for radiosensitization. Curr Top Dev Biol 73:173-204, 2006 (Pubitemid 43878133)
    • (2006) Current Topics in Developmental Biology , vol.73 , pp. 173-204
    • Cerna, D.1    Camphausen, K.2    Tofilon, P.J.3
  • 143
    • 0036828101 scopus 로고    scopus 로고
    • MS-27-275, an inhibitor of histone deacetylase, has marked in vitro and in vivo antitumor activity against pediatric solid tumors
    • Jaboin J, Wild J, Hamidi H, et al: MS-27-275, an inhibitor of histone deacetylase, has marked in vitro and in vivo antitumor activity against pediatric solid tumors. Cancer Res 62:6108-6115, 2002 (Pubitemid 35244459)
    • (2002) Cancer Research , vol.62 , Issue.21 , pp. 6108-6115
    • Jaboin, J.1    Wild, J.2    Hamidi, H.3    Khanna, C.4    Kim, C.J.5    Robey, R.6    Bates, S.E.7    Thiele, C.J.8
  • 144
    • 0035328528 scopus 로고    scopus 로고
    • The histone deacetylase inhibitor, CBHA, inhibits growth of human neuroblastoma xenografts in vivo, alone and synergistically with all-trans retinoic acid
    • Coffey DC, Kutko MC, Glick RD, et al: The histone deacetylase inhibitor, CBHA, inhibits growth of human neuroblastoma xenografts in vivo, alone and synergistically with all-trans retinoic acid. Cancer Res 61:3591-3554, 2001
    • (2001) Cancer Res , vol.61 , pp. 3591-13554
    • Coffey, D.C.1    Kutko, M.C.2    Glick, R.D.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.