메뉴 건너뛰기




Volumn 37, Issue 1, 2018, Pages

Correction to: Emerging roles of Myc in stem cell biology and novel tumor therapies (Journal of Experimental and Clinical Cancer Research (2018) 37 (173) DOI: 10.1186/s13046-018-0835-y);Emerging roles of Myc in stem cell biology and novel tumor therapies

Author keywords

Cancer stem cells; CD44 variant; Drug repositioning; Epigenetics; FBW7; Induced pluripotent stem cells; Metabolic reprogramming; Myc family; Neuroendocrine tumors; Ubiquitination

Indexed keywords

ANTINEOPLASTIC AGENT; BORTEZOMIB; F BOX/WD REPEAT CONTAINING PROTEIN 7; GLUTAMINE; HERMES ANTIGEN; MAX PROTEIN; MYC PROTEIN; N MYC PROTO ONCOGENE PROTEIN; REDUCED NICOTINAMIDE ADENINE DINUCLEOTIDE PHOSPHATE; RETINOIC ACID; SALAZOSULFAPYRIDINE; TRANSCRIPTION FACTOR EZH2; VALPROIC ACID; MYC PROTEIN, HUMAN;

EID: 85050749737     PISSN: None     EISSN: 17569966     Source Type: Journal    
DOI: 10.1186/s13046-018-0964-3     Document Type: Erratum
Times cited : (184)

References (236)
  • 1
    • 0033551387 scopus 로고    scopus 로고
    • Mechanisms of apoptosis by c-Myc
    • 10378693
    • Prendergast GC. Mechanisms of apoptosis by c-Myc. Oncogene. 1999;18(19):2967-87.
    • (1999) Oncogene , vol.18 , Issue.19 , pp. 2967-2987
    • Prendergast, G.C.1
  • 2
    • 56749184298 scopus 로고    scopus 로고
    • Reflecting on 25 years with MYC
    • 19029958
    • Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nat Rev Cancer. 2008;8(12):976-90.
    • (2008) Nat Rev Cancer , vol.8 , Issue.12 , pp. 976-990
    • Meyer, N.1    Penn, L.Z.2
  • 5
    • 0029864934 scopus 로고    scopus 로고
    • Discrimination between different E-box-binding proteins at an endogenous target gene of c-myc
    • 8600028
    • Desbarats L, Gaubatz S, Eilers M. Discrimination between different E-box-binding proteins at an endogenous target gene of c-myc. Genes Dev. 1996;10(4):447-60.
    • (1996) Genes Dev , vol.10 , Issue.4 , pp. 447-460
    • Desbarats, L.1    Gaubatz, S.2    Eilers, M.3
  • 6
    • 0030762087 scopus 로고    scopus 로고
    • C-Myc/Max heterodimers bind cooperatively to the E-box sequences located in the first intron of the rat ornithine decarboxylase (ODC) gene
    • 9162900 146624
    • Walhout AJ, Gubbels JM, Bernards R, van der Vliet PC, Timmers HT. c-Myc/Max heterodimers bind cooperatively to the E-box sequences located in the first intron of the rat ornithine decarboxylase (ODC) gene. Nucleic Acids Res. 1997;25(8):1493-501.
    • (1997) Nucleic Acids Res , vol.25 , Issue.8 , pp. 1493-1501
    • Walhout, A.J.1    Gubbels, J.M.2    Bernards, R.3    Van Der Vliet, P.C.4    Timmers, H.T.5
  • 7
    • 0000538165 scopus 로고
    • Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells
    • 6961453 347441
    • Dalla-Favera R, Bregni M, Erikson J, Patterson D, Gallo RC, Croce CM. Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells. Proc Natl Acad Sci U S A. 1982;79(24):7824-7.
    • (1982) Proc Natl Acad Sci U S A , vol.79 , Issue.24 , pp. 7824-7827
    • Dalla-Favera, R.1    Bregni, M.2    Erikson, J.3    Patterson, D.4    Gallo, R.C.5    Croce, C.M.6
  • 8
    • 0001292985 scopus 로고
    • Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells
    • 6818551 347444
    • Taub R, Kirsch I, Morton C, Lenoir G, Swan D, Tronick S, Aaronson S, Leder P. Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells. Proc Natl Acad Sci U S A. 1982;79(24):7837-41.
    • (1982) Proc Natl Acad Sci U S A , vol.79 , Issue.24 , pp. 7837-7841
    • Taub, R.1    Kirsch, I.2    Morton, C.3    Lenoir, G.4    Swan, D.5    Tronick, S.6    Aaronson, S.7    Leder, P.8
  • 11
    • 84859171807 scopus 로고    scopus 로고
    • MYC on the path to cancer
    • 22464321 3345192
    • Dang CV. MYC on the path to cancer. Cell. 2012;149(1):22-35.
    • (2012) Cell , vol.149 , Issue.1 , pp. 22-35
    • Dang, C.V.1
  • 12
    • 85050763688 scopus 로고    scopus 로고
    • Targeting oncogenic Myc as a strategy for cancer treatment
    • 29527331 5837124
    • Chen H, Liu H, Qing G. Targeting oncogenic Myc as a strategy for cancer treatment. Signal Transduct Target Ther. 2018;3:5.
    • (2018) Signal Transduct Target Ther , vol.3 , pp. 5
    • Chen, H.1    Liu, H.2    Qing, G.3
  • 14
    • 85019688331 scopus 로고    scopus 로고
    • Genetic profiling of MYC and BCL2 in diffuse large B-cell lymphoma determines cell-of-origin-specific clinical impact
    • 28351934
    • Ennishi D, Mottok A, Ben-Neriah S, Shulha HP, Farinha P, Chan FC, Meissner B, Boyle M, Hother C, Kridel R, et al. Genetic profiling of MYC and BCL2 in diffuse large B-cell lymphoma determines cell-of-origin-specific clinical impact. Blood. 2017;129(20):2760-2770.
    • (2017) Blood , vol.129 , Issue.20 , pp. 2760-2770
    • Ennishi, D.1    Mottok, A.2    Ben-Neriah, S.3    Shulha, H.P.4    Farinha, P.5    Chan, F.C.6    Meissner, B.7    Boyle, M.8    Hother, C.9    Kridel, R.10
  • 15
    • 79952841250 scopus 로고    scopus 로고
    • MYC and Breast Cancer
    • 21779462 3092228
    • Xu J, Chen Y, Olopade OI. MYC and Breast Cancer. Genes Cancer. 2010;1(6):629-40.
    • (2010) Genes Cancer , vol.1 , Issue.6 , pp. 629-640
    • Xu, J.1    Chen, Y.2    Olopade, O.I.3
  • 17
    • 0034524132 scopus 로고    scopus 로고
    • The Myc/Max/Mad network and the transcriptional control of cell behavior
    • 11031250
    • Grandori C, Cowley SM, James LP, Eisenman RN. The Myc/Max/Mad network and the transcriptional control of cell behavior. Annu Rev Cell Dev Biol. 2000;16:653-99.
    • (2000) Annu Rev Cell Dev Biol , vol.16 , pp. 653-699
    • Grandori, C.1    Cowley, S.M.2    James, L.P.3    Eisenman, R.N.4
  • 18
    • 0035904463 scopus 로고    scopus 로고
    • Function and regulation of the transcription factors of the Myc/Max/Mad network
    • 11602341
    • Luscher B. Function and regulation of the transcription factors of the Myc/Max/Mad network. Gene. 2001;277(1-2):1-14.
    • (2001) Gene , vol.277 , Issue.1-2 , pp. 1-14
    • Luscher, B.1
  • 19
    • 0027315650 scopus 로고
    • A null c-myc mutation causes lethality before 10.5 days of gestation in homozygotes and reduced fertility in heterozygous female mice
    • 8458579
    • Davis AC, Wims M, Spotts GD, Hann SR, Bradley A. A null c-myc mutation causes lethality before 10.5 days of gestation in homozygotes and reduced fertility in heterozygous female mice. Genes Dev. 1993;7(4):671-82.
    • (1993) Genes Dev , vol.7 , Issue.4 , pp. 671-682
    • Davis, A.C.1    Wims, M.2    Spotts, G.D.3    Hann, S.R.4    Bradley, A.5
  • 21
    • 0026628060 scopus 로고
    • A targeted mutation reveals a role for N-myc in branching morphogenesis in the embryonic mouse lung
    • 1577267
    • Moens CB, Auerbach AB, Conlon RA, Joyner AL, Rossant J. A targeted mutation reveals a role for N-myc in branching morphogenesis in the embryonic mouse lung. Genes Dev. 1992;6(5):691-704.
    • (1992) Genes Dev , vol.6 , Issue.5 , pp. 691-704
    • Moens, C.B.1    Auerbach, A.B.2    Conlon, R.A.3    Joyner, A.L.4    Rossant, J.5
  • 22
    • 0027066738 scopus 로고
    • Loss of N-myc function results in embryonic lethality and failure of the epithelial component of the embryo to develop
    • 1459449
    • Stanton BR, Perkins AS, Tessarollo L, Sassoon DA, Parada LF. Loss of N-myc function results in embryonic lethality and failure of the epithelial component of the embryo to develop. Genes Dev. 1992;6(12A):2235-47.
    • (1992) Genes Dev , vol.6 , Issue.12 A , pp. 2235-2247
    • Stanton, B.R.1    Perkins, A.S.2    Tessarollo, L.3    Sassoon, D.A.4    Parada, L.F.5
  • 23
    • 0037108268 scopus 로고    scopus 로고
    • N-myc is essential during neurogenesis for the rapid expansion of progenitor cell populations and the inhibition of neuronal differentiation
    • 12381668 187459
    • Knoepfler PS, Cheng PF, Eisenman RN. N-myc is essential during neurogenesis for the rapid expansion of progenitor cell populations and the inhibition of neuronal differentiation. Genes Dev. 2002;16(20):2699-712.
    • (2002) Genes Dev , vol.16 , Issue.20 , pp. 2699-2712
    • Knoepfler, P.S.1    Cheng, P.F.2    Eisenman, R.N.3
  • 25
    • 84883759641 scopus 로고    scopus 로고
    • Control of vertebrate development by MYC
    • 24003246 3753724
    • Hurlin PJ. Control of vertebrate development by MYC. Cold Spring Harb Perspect Med. 2013;3(9):a014332.
    • (2013) Cold Spring Harb Perspect Med , vol.3 , Issue.9 , pp. a014332
    • Hurlin, P.J.1
  • 29
    • 16844364151 scopus 로고    scopus 로고
    • LIF/STAT3 controls ES cell self-renewal and pluripotency by a Myc-dependent mechanism
    • 15673569
    • Cartwright P, McLean C, Sheppard A, Rivett D, Jones K, Dalton S. LIF/STAT3 controls ES cell self-renewal and pluripotency by a Myc-dependent mechanism. Development. 2005;132(5):885-96.
    • (2005) Development , vol.132 , Issue.5 , pp. 885-896
    • Cartwright, P.1    McLean, C.2    Sheppard, A.3    Rivett, D.4    Jones, K.5    Dalton, S.6
  • 31
    • 33744917003 scopus 로고    scopus 로고
    • Phosphorylation regulates Myc expression via prolonged activation of the mitogen-activated protein kinase pathway
    • 16596619
    • Wang Z, Ge L, Wang M, Carr BI. Phosphorylation regulates Myc expression via prolonged activation of the mitogen-activated protein kinase pathway. J Cell Physiol. 2006;208(1):133-40.
    • (2006) J Cell Physiol , vol.208 , Issue.1 , pp. 133-140
    • Wang, Z.1    Ge, L.2    Wang, M.3    Carr, B.I.4
  • 32
    • 77956214711 scopus 로고    scopus 로고
    • Myc represses primitive endoderm differentiation in pluripotent stem cells
    • 20804970 2954754
    • Smith KN, Singh AM, Dalton S. Myc represses primitive endoderm differentiation in pluripotent stem cells. Cell Stem Cell. 2010;7(3):343-54.
    • (2010) Cell Stem Cell , vol.7 , Issue.3 , pp. 343-354
    • Smith, K.N.1    Singh, A.M.2    Dalton, S.3
  • 34
    • 0033569429 scopus 로고    scopus 로고
    • Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF
    • 10541554 317101
    • Jacobs JJ, Scheijen B, Voncken JW, Kieboom K, Berns A, van Lohuizen M. Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev. 1999;13(20):2678-90.
    • (1999) Genes Dev , vol.13 , Issue.20 , pp. 2678-2690
    • Jacobs, J.J.1    Scheijen, B.2    Voncken, J.W.3    Kieboom, K.4    Berns, A.5    Van Lohuizen, M.6
  • 35
    • 84873320012 scopus 로고    scopus 로고
    • A positive feedback loop regulates the expression of polycomb group protein BMI1 via WNT signaling pathway
    • 23239878
    • Cho JH, Dimri M, Dimri GP. A positive feedback loop regulates the expression of polycomb group protein BMI1 via WNT signaling pathway. J Biol Chem. 2013;288(5):3406-18.
    • (2013) J Biol Chem , vol.288 , Issue.5 , pp. 3406-3418
    • Cho, J.H.1    Dimri, M.2    Dimri, G.P.3
  • 36
    • 84978096840 scopus 로고    scopus 로고
    • Bmi-1: At the crossroads of physiological and pathological biology
    • 26448339 4593320
    • Bhattacharya R, Mustafi SB, Street M, Dey A, Dwivedi SK. Bmi-1: At the crossroads of physiological and pathological biology. Genes Dis. 2015;2(3):225-39.
    • (2015) Genes Dis , vol.2 , Issue.3 , pp. 225-239
    • Bhattacharya, R.1    Mustafi, S.B.2    Street, M.3    Dey, A.4    Dwivedi, S.K.5
  • 37
    • 33747195353 scopus 로고    scopus 로고
    • Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors
    • 16904174
    • Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663-76.
    • (2006) Cell , vol.126 , Issue.4 , pp. 663-676
    • Takahashi, K.1    Yamanaka, S.2
  • 38
    • 34249880066 scopus 로고    scopus 로고
    • Generation of germline-competent induced pluripotent stem cells
    • 17554338
    • Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature. 2007;448(7151):313-7.
    • (2007) Nature , vol.448 , Issue.7151 , pp. 313-317
    • Okita, K.1    Ichisaka, T.2    Yamanaka, S.3
  • 43
    • 55849117368 scopus 로고    scopus 로고
    • Generation of mouse induced pluripotent stem cells without viral vectors
    • 18845712
    • Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse induced pluripotent stem cells without viral vectors. Science. 2008;322(5903):949-53.
    • (2008) Science , vol.322 , Issue.5903 , pp. 949-953
    • Okita, K.1    Nakagawa, M.2    Hyenjong, H.3    Ichisaka, T.4    Yamanaka, S.5
  • 44
    • 55849115999 scopus 로고    scopus 로고
    • Induced pluripotent stem cells generated without viral integration
    • 18818365 3987909
    • Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K. Induced pluripotent stem cells generated without viral integration. Science. 2008;322(5903):945-9.
    • (2008) Science , vol.322 , Issue.5903 , pp. 945-949
    • Stadtfeld, M.1    Nagaya, M.2    Utikal, J.3    Weir, G.4    Hochedlinger, K.5
  • 45
    • 65649116572 scopus 로고    scopus 로고
    • Human induced pluripotent stem cells free of vector and transgene sequences
    • 19325077 2758053
    • Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin II, Thomson JA. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009;324(5928):797-801.
    • (2009) Science , vol.324 , Issue.5928 , pp. 797-801
    • Yu, J.1    Hu, K.2    Smuga-Otto, K.3    Tian, S.4    Stewart, R.5    Slukvin, I.I.6    Thomson, J.A.7
  • 46
    • 65449154892 scopus 로고    scopus 로고
    • Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon
    • 19337237 2677165
    • Yusa K, Rad R, Takeda J, Bradley A. Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon. Nat Methods. 2009;6(5):363-9.
    • (2009) Nat Methods , vol.6 , Issue.5 , pp. 363-369
    • Yusa, K.1    Rad, R.2    Takeda, J.3    Bradley, A.4
  • 48
    • 0037417130 scopus 로고    scopus 로고
    • Functional analysis of the N-terminal domain of the Myc oncoprotein
    • 12673205
    • Oster SK, Mao DY, Kennedy J, Penn LZ. Functional analysis of the N-terminal domain of the Myc oncoprotein. Oncogene. 2003;22(13):1998-2010.
    • (2003) Oncogene , vol.22 , Issue.13 , pp. 1998-2010
    • Oster, S.K.1    Mao, D.Y.2    Kennedy, J.3    Penn, L.Z.4
  • 49
    • 52449101436 scopus 로고    scopus 로고
    • Transcription-independent functions of MYC: Regulation of translation and DNA replication
    • 18698328
    • Cole MD, Cowling VH. Transcription-independent functions of MYC: regulation of translation and DNA replication. Nat Rev Mol Cell Biol. 2008;9(10):810-5.
    • (2008) Nat Rev Mol Cell Biol , vol.9 , Issue.10 , pp. 810-815
    • Cole, M.D.1    Cowling, V.H.2
  • 50
    • 0018182764 scopus 로고
    • Identification of nucleotide sequences which may encode the oncogenic capacity of avian retrovirus MC29
    • 214581 354308
    • Sheiness D, Fanshier L, Bishop JM. Identification of nucleotide sequences which may encode the oncogenic capacity of avian retrovirus MC29. J Virol. 1978;28(2):600-10.
    • (1978) J Virol , vol.28 , Issue.2 , pp. 600-610
    • Sheiness, D.1    Fanshier, L.2    Bishop, J.M.3
  • 52
    • 85045907266 scopus 로고    scopus 로고
    • Enhancer reprogramming in tumor progression: A new route towards cancer cell plasticity
    • 29691590
    • Fagnocchi L, Poli V, Zippo A. Enhancer reprogramming in tumor progression: a new route towards cancer cell plasticity. Cell Mol Life Sci. 2018;75(14):2537-2555.
    • (2018) Cell Mol Life Sci , vol.75 , Issue.14 , pp. 2537-2555
    • Fagnocchi, L.1    Poli, V.2    Zippo, A.3
  • 53
    • 0842309106 scopus 로고    scopus 로고
    • MYCN in neuronal tumours
    • 15013217
    • Schwab M. MYCN in neuronal tumours. Cancer Lett. 2004;204(2):179-87.
    • (2004) Cancer Lett , vol.204 , Issue.2 , pp. 179-187
    • Schwab, M.1
  • 55
    • 85041428603 scopus 로고    scopus 로고
    • The Expanding World of N-MYC-Driven Tumors
    • 29358508
    • Rickman DS, Schulte JH, Eilers M. The Expanding World of N-MYC-Driven Tumors. Cancer Discov. 2018;8(2):150-63.
    • (2018) Cancer Discov , vol.8 , Issue.2 , pp. 150-163
    • Rickman, D.S.1    Schulte, J.H.2    Eilers, M.3
  • 57
    • 85020278745 scopus 로고    scopus 로고
    • Biology and evolution of poorly differentiated neuroendocrine tumors
    • 28586335
    • Rickman DS, Beltran H, Demichelis F, Rubin MA. Biology and evolution of poorly differentiated neuroendocrine tumors. Nat Med. 2017;23(6):1-10.
    • (2017) Nat Med , vol.23 , Issue.6 , pp. 1-10
    • Rickman, D.S.1    Beltran, H.2    Demichelis, F.3    Rubin, M.A.4
  • 59
    • 84905244381 scopus 로고    scopus 로고
    • Regulation and Role of EZH2 in Cancer
    • 25038756 4132442
    • Yamaguchi H, Hung MC. Regulation and Role of EZH2 in Cancer. Cancer Res Treat. 2014;46(3):209-22.
    • (2014) Cancer Res Treat , vol.46 , Issue.3 , pp. 209-222
    • Yamaguchi, H.1    Hung, M.C.2
  • 62
    • 0023263664 scopus 로고
    • Increased expression of N-myc in human small cell lung cancer biopsies predicts lack of response to chemotherapy and poor prognosis
    • 3039835
    • Funa K, Steinholtz L, Nou E, Bergh J. Increased expression of N-myc in human small cell lung cancer biopsies predicts lack of response to chemotherapy and poor prognosis. Am J Clin Pathol. 1987;88(2):216-20.
    • (1987) Am J Clin Pathol , vol.88 , Issue.2 , pp. 216-220
    • Funa, K.1    Steinholtz, L.2    Nou, E.3    Bergh, J.4
  • 63
    • 0031049728 scopus 로고    scopus 로고
    • N-Myc gene amplification is a major prognostic factor in localized neuroblastoma: Results of the French NBL 90 study. Neuroblastoma Study Group of the Societe Francaise d'Oncologie Pediatrique
    • Rubie H, Hartmann O, Michon J, Frappaz D, Coze C, Chastagner P, Baranzelli MC, Plantaz D, Avet-Loiseau H, Benard J, et al. N-Myc gene amplification is a major prognostic factor in localized neuroblastoma: results of the French NBL 90 study. Neuroblastoma Study Group of the Societe Francaise d'Oncologie Pediatrique. J Clin Oncol. 1997;15(3):1171-1182.
    • (1997) J Clin Oncol , vol.15 , Issue.3 , pp. 1171-1182
    • Rubie, H.1    Hartmann, O.2    Michon, J.3    Frappaz, D.4    Coze, C.5    Chastagner, P.6    Baranzelli, M.C.7    Plantaz, D.8    Avet-Loiseau, H.9    Benard, J.10
  • 66
    • 63749118521 scopus 로고    scopus 로고
    • Outcome prediction in pediatric medulloblastoma based on DNA copy-number aberrations of chromosomes 6q and 17q and the MYC and MYCN loci
    • 19255330
    • Pfister S, Remke M, Benner A, Mendrzyk F, Toedt G, Felsberg J, Wittmann A, Devens F, Gerber NU, Joos S, et al. Outcome prediction in pediatric medulloblastoma based on DNA copy-number aberrations of chromosomes 6q and 17q and the MYC and MYCN loci. J Clin Oncol. 2009;27(10):1627-36.
    • (2009) J Clin Oncol , vol.27 , Issue.10 , pp. 1627-1636
    • Pfister, S.1    Remke, M.2    Benner, A.3    Mendrzyk, F.4    Toedt, G.5    Felsberg, J.6    Wittmann, A.7    Devens, F.8    Gerber, N.U.9    Joos, S.10
  • 69
    • 85020839679 scopus 로고    scopus 로고
    • Treatment advances in small cell lung cancer (SCLC)
    • 28579387
    • Waqar SN, Morgensztern D. Treatment advances in small cell lung cancer (SCLC). Pharmacol Ther. 2017;180:16-23.
    • (2017) Pharmacol Ther , vol.180 , pp. 16-23
    • Waqar, S.N.1    Morgensztern, D.2
  • 70
    • 85045296183 scopus 로고    scopus 로고
    • Current and future therapeutic approaches for the treatment of small cell lung cancer
    • 29544351
    • Rossi A, Tay R, Chiramel J, Prelaj A, Califano R. Current and future therapeutic approaches for the treatment of small cell lung cancer. Expert Rev Anticancer Ther. 2018;18(5):473-86.
    • (2018) Expert Rev Anticancer Ther , vol.18 , Issue.5 , pp. 473-486
    • Rossi, A.1    Tay, R.2    Chiramel, J.3    Prelaj, A.4    Califano, R.5
  • 71
    • 80052265739 scopus 로고    scopus 로고
    • Anti-tumour/metastasis effects of the potassium-sparing diuretic amiloride: An orally active anti-cancer drug waiting for its call-of-duty?
    • 21544803
    • Matthews H, Ranson M, Kelso MJ. Anti-tumour/metastasis effects of the potassium-sparing diuretic amiloride: an orally active anti-cancer drug waiting for its call-of-duty? Int J Cancer. 2011;129(9):2051-61.
    • (2011) Int J Cancer , vol.129 , Issue.9 , pp. 2051-2061
    • Matthews, H.1    Ranson, M.2    Kelso, M.J.3
  • 72
    • 84976620845 scopus 로고    scopus 로고
    • Potential Roles of Amiloride-Sensitive Sodium Channels in Cancer Development
    • 27403419 4926023
    • Xu S, Liu C, Ma Y, Ji HL, Li X. Potential Roles of Amiloride-Sensitive Sodium Channels in Cancer Development. Biomed Res Int. 2016;2016:2190216.
    • (2016) Biomed Res Int , vol.2016 , pp. 2190216
    • Xu, S.1    Liu, C.2    Ma, Y.3    Ji, H.L.4    Li, X.5
  • 74
    • 85015149861 scopus 로고    scopus 로고
    • Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: From pathophysiology to treatment
    • 28279189 5345270
    • Yoshida GJ. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol. 2017;10(1):67.
    • (2017) J Hematol Oncol , vol.10 , Issue.1 , pp. 67
    • Yoshida, G.J.1
  • 75
    • 67651153048 scopus 로고    scopus 로고
    • New clinical developments in histone deacetylase inhibitors for epigenetic therapy of cancer
    • 19486511 2695818
    • Cang S, Ma Y, Liu D. New clinical developments in histone deacetylase inhibitors for epigenetic therapy of cancer. J Hematol Oncol. 2009;2:22.
    • (2009) J Hematol Oncol , vol.2 , pp. 22
    • Cang, S.1    Ma, Y.2    Liu, D.3
  • 76
    • 84880184328 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor potentiated the ability of MTOR inhibitor to induce autophagic cell death in Burkitt leukemia/lymphoma
    • 23866964 3722002
    • Dong LH, Cheng S, Zheng Z, Wang L, Shen Y, Shen ZX, Chen SJ, Zhao WL. Histone deacetylase inhibitor potentiated the ability of MTOR inhibitor to induce autophagic cell death in Burkitt leukemia/lymphoma. J Hematol Oncol. 2013;6:53.
    • (2013) J Hematol Oncol , vol.6 , pp. 53
    • Dong, L.H.1    Cheng, S.2    Zheng, Z.3    Wang, L.4    Shen, Y.5    Shen, Z.X.6    Chen, S.J.7    Zhao, W.L.8
  • 77
    • 0035889955 scopus 로고    scopus 로고
    • Histopathology (International Neuroblastoma Pathology Classification) and MYCN status in patients with peripheral neuroblastic tumors: A report from the Children's Cancer Group
    • 11745206
    • Goto S, Umehara S, Gerbing RB, Stram DO, Brodeur GM, Seeger RC, Lukens JN, Matthay KK, Shimada H. Histopathology (International Neuroblastoma Pathology Classification) and MYCN status in patients with peripheral neuroblastic tumors: a report from the Children's Cancer Group. Cancer. 2001;92(10):2699-708.
    • (2001) Cancer , vol.92 , Issue.10 , pp. 2699-2708
    • Goto, S.1    Umehara, S.2    Gerbing, R.B.3    Stram, D.O.4    Brodeur, G.M.5    Seeger, R.C.6    Lukens, J.N.7    Matthay, K.K.8    Shimada, H.9
  • 78
    • 78651189519 scopus 로고
    • Tumors of Neural Crest Origin; Biochemical and Pathological Correlation
    • 14318483
    • Greer M, Anton AH, Williams CM, Echevarria RA. Tumors of Neural Crest Origin; Biochemical and Pathological Correlation. Arch Neurol. 1965;13:139-48.
    • (1965) Arch Neurol , vol.13 , pp. 139-148
    • Greer, M.1    Anton, A.H.2    Williams, C.M.3    Echevarria, R.A.4
  • 79
    • 0037366067 scopus 로고    scopus 로고
    • Neuroblastoma: Biological insights into a clinical enigma
    • 12612655
    • Brodeur GM. Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer. 2003;3(3):203-16.
    • (2003) Nat Rev Cancer , vol.3 , Issue.3 , pp. 203-216
    • Brodeur, G.M.1
  • 80
    • 84975299347 scopus 로고    scopus 로고
    • Reactivation of p53 via MDM2 inhibition
    • 26492369 4632320
    • Kim ES, Shohet JM. Reactivation of p53 via MDM2 inhibition. Cell Death Dis. 2015;6:e1936.
    • (2015) Cell Death Dis , vol.6 , pp. e1936
    • Kim, E.S.1    Shohet, J.M.2
  • 86
  • 88
    • 84883488965 scopus 로고    scopus 로고
    • Kruppel-like factor 4 (KLF4) suppresses neuroblastoma cell growth and determines non-tumorigenic lineage differentiation
    • 23045286
    • Shum CK, Lau ST, Tsoi LL, Chan LK, Yam JW, Ohira M, Nakagawara A, Tam PK, Ngan ES. Kruppel-like factor 4 (KLF4) suppresses neuroblastoma cell growth and determines non-tumorigenic lineage differentiation. Oncogene. 2013;32(35):4086-99.
    • (2013) Oncogene , vol.32 , Issue.35 , pp. 4086-4099
    • Shum, C.K.1    Lau, S.T.2    Tsoi, L.L.3    Chan, L.K.4    Yam, J.W.5    Ohira, M.6    Nakagawara, A.7    Tam, P.K.8    Ngan, E.S.9
  • 89
    • 0025288707 scopus 로고
    • N-myc mRNA forms an RNA-RNA duplex with endogenous antisense transcripts
    • 1695323 360949
    • Krystal GW, Armstrong BC, Battey JF. N-myc mRNA forms an RNA-RNA duplex with endogenous antisense transcripts. Mol Cell Biol. 1990;10(8):4180-91.
    • (1990) Mol Cell Biol , vol.10 , Issue.8 , pp. 4180-4191
    • Krystal, G.W.1    Armstrong, B.C.2    Battey, J.F.3
  • 90
    • 0026881782 scopus 로고
    • Isolation and characterization of complementary DNA for N-cym, a gene encoded by the DNA strand opposite to N-myc
    • 1419902
    • Armstrong BC, Krystal GW. Isolation and characterization of complementary DNA for N-cym, a gene encoded by the DNA strand opposite to N-myc. Cell Growth Differ. 1992;3(6):385-90.
    • (1992) Cell Growth Differ , vol.3 , Issue.6 , pp. 385-390
    • Armstrong, B.C.1    Krystal, G.W.2
  • 91
    • 84896730858 scopus 로고    scopus 로고
    • NCYM, a Cis-antisense gene of MYCN, encodes a de novo evolved protein that inhibits GSK3beta resulting in the stabilization of MYCN in human neuroblastomas
    • Suenaga Y, Islam SM, Alagu J, Kaneko Y, Kato M, Tanaka Y, Kawana H, Hossain S, Matsumoto D, Yamamoto M, et al. NCYM, a Cis-antisense gene of MYCN, encodes a de novo evolved protein that inhibits GSK3beta resulting in the stabilization of MYCN in human neuroblastomas. PLoS Genet. 2014;10(1):e1003996.
    • (2014) PLoS Genet , vol.10 , Issue.1 , pp. e1003996
    • Suenaga, Y.1    Islam, S.M.2    Alagu, J.3    Kaneko, Y.4    Kato, M.5    Tanaka, Y.6    Kawana, H.7    Hossain, S.8    Matsumoto, D.9    Yamamoto, M.10
  • 92
    • 0033554684 scopus 로고    scopus 로고
    • Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children's Cancer Group
    • Matthay KK, Villablanca JG, Seeger RC, Stram DO, Harris RE, Ramsay NK, Swift P, Shimada H, Black CT, Brodeur GM, et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children's Cancer Group. N Engl J Med. 1999;341(16):1165-1173.
    • (1999) N Engl J Med. , vol.341 , Issue.16 , pp. 1165-1173
    • Matthay, K.K.1    Villablanca, J.G.2    Seeger, R.C.3    Stram, D.O.4    Harris, R.E.5    Ramsay, N.K.6    Swift, P.7    Shimada, H.8    Black, C.T.9    Brodeur, G.M.10
  • 94
    • 0035969112 scopus 로고    scopus 로고
    • All trans retinoic acid in acute promyelocytic leukemia
    • 11704842
    • Degos L, Wang ZY. All trans retinoic acid in acute promyelocytic leukemia. Oncogene. 2001;20(49):7140-5.
    • (2001) Oncogene , vol.20 , Issue.49 , pp. 7140-7145
    • Degos, L.1    Wang, Z.Y.2
  • 95
    • 0037974596 scopus 로고    scopus 로고
    • Treatment of acute promyelocytic leukemia with ATRA and As2O3: A model of molecular target-based cancer therapy
    • 12642682
    • Fang J, Chen SJ, Tong JH, Wang ZG, Chen GQ, Chen Z. Treatment of acute promyelocytic leukemia with ATRA and As2O3: a model of molecular target-based cancer therapy. Cancer Biol Ther. 2002;1(6):614-20.
    • (2002) Cancer Biol Ther , vol.1 , Issue.6 , pp. 614-620
    • Fang, J.1    Chen, S.J.2    Tong, J.H.3    Wang, Z.G.4    Chen, G.Q.5    Chen, Z.6
  • 96
    • 34247614521 scopus 로고    scopus 로고
    • HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity
    • 17482131
    • Zhang H, Gao P, Fukuda R, Kumar G, Krishnamachary B, Zeller KI, Dang CV, Semenza GL. HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity. Cancer Cell. 2007;11(5):407-20.
    • (2007) Cancer Cell , vol.11 , Issue.5 , pp. 407-420
    • Zhang, H.1    Gao, P.2    Fukuda, R.3    Kumar, G.4    Krishnamachary, B.5    Zeller, K.I.6    Dang, C.V.7    Semenza, G.L.8
  • 97
    • 70350728803 scopus 로고    scopus 로고
    • MYC-induced cancer cell energy metabolism and therapeutic opportunities
    • 19861459 2783410
    • Dang CV, Le A, Gao P. MYC-induced cancer cell energy metabolism and therapeutic opportunities. Clin Cancer Res. 2009;15(21):6479-83.
    • (2009) Clin Cancer Res , vol.15 , Issue.21 , pp. 6479-6483
    • Dang, C.V.1    Le, A.2    Gao, P.3
  • 98
    • 84861891911 scopus 로고    scopus 로고
    • Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC
    • 22615405 3384197
    • Liu W, Le A, Hancock C, Lane AN, Dang CV, Fan TW, Phang JM. Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC. Proc Natl Acad Sci U S A. 2012;109(23):8983-8.
    • (2012) Proc Natl Acad Sci U S A , vol.109 , Issue.23 , pp. 8983-8988
    • Liu, W.1    Le, A.2    Hancock, C.3    Lane, A.N.4    Dang, C.V.5    Fan, T.W.6    Phang, J.M.7
  • 99
    • 84887430714 scopus 로고    scopus 로고
    • MTOR complex 2 controls glycolytic metabolism in glioblastoma through FoxO acetylation and upregulation of c-Myc
    • 24140020
    • Masui K, Tanaka K, Akhavan D, Babic I, Gini B, Matsutani T, Iwanami A, Liu F, Villa GR, Gu Y, et al. mTOR complex 2 controls glycolytic metabolism in glioblastoma through FoxO acetylation and upregulation of c-Myc. Cell Metab. 2013;18(5):726-39.
    • (2013) Cell Metab , vol.18 , Issue.5 , pp. 726-739
    • Masui, K.1    Tanaka, K.2    Akhavan, D.3    Babic, I.4    Gini, B.5    Matsutani, T.6    Iwanami, A.7    Liu, F.8    Villa, G.R.9    Gu, Y.10
  • 101
    • 84943246936 scopus 로고    scopus 로고
    • Metabolic reprogramming: The emerging concept and associated therapeutic strategies
    • 26445347 4595070
    • Yoshida GJ. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. J Exp Clin Cancer Res. 2015;34:111.
    • (2015) J Exp Clin Cancer Res , vol.34 , pp. 111
    • Yoshida, G.J.1
  • 104
    • 84930221860 scopus 로고    scopus 로고
    • The c-Myc-LDHA axis positively regulates aerobic glycolysis and promotes tumor progression in pancreatic cancer
    • 26021472 4452209
    • He TL, Zhang YJ, Jiang H, Li XH, Zhu H, Zheng KL. The c-Myc-LDHA axis positively regulates aerobic glycolysis and promotes tumor progression in pancreatic cancer. Med Oncol. 2015;32(7):187.
    • (2015) Med Oncol , vol.32 , Issue.7 , pp. 187
    • He, T.L.1    Zhang, Y.J.2    Jiang, H.3    Li, X.H.4    Zhu, H.5    Zheng, K.L.6
  • 107
    • 43549098260 scopus 로고    scopus 로고
    • The oncogene c-Myc coordinates regulation of metabolic networks to enable rapid cell cycle entry
    • 18414044 2518168
    • Morrish F, Neretti N, Sedivy JM, Hockenbery DM. The oncogene c-Myc coordinates regulation of metabolic networks to enable rapid cell cycle entry. Cell Cycle. 2008;7(8):1054-66.
    • (2008) Cell Cycle , vol.7 , Issue.8 , pp. 1054-1066
    • Morrish, F.1    Neretti, N.2    Sedivy, J.M.3    Hockenbery, D.M.4
  • 112
    • 77954945639 scopus 로고    scopus 로고
    • MiR-17-92 cluster: Ups and downs in cancer and aging
    • 20437201 2899009
    • Grillari J, Hackl M, Grillari-Voglauer R. miR-17-92 cluster: ups and downs in cancer and aging. Biogerontology. 2010;11(4):501-6.
    • (2010) Biogerontology , vol.11 , Issue.4 , pp. 501-506
    • Grillari, J.1    Hackl, M.2    Grillari-Voglauer, R.3
  • 113
    • 84899755840 scopus 로고    scopus 로고
    • MiR-17-92 cluster targets phosphatase and tensin homology and Ikaros Family Zinc Finger 4 to promote TH17-mediated inflammation
    • 24644282 4007439
    • Liu SQ, Jiang S, Li C, Zhang B, Li QJ. miR-17-92 cluster targets phosphatase and tensin homology and Ikaros Family Zinc Finger 4 to promote TH17-mediated inflammation. J Biol Chem. 2014;289(18):12446-56.
    • (2014) J Biol Chem , vol.289 , Issue.18 , pp. 12446-12456
    • Liu, S.Q.1    Jiang, S.2    Li, C.3    Zhang, B.4    Li, Q.J.5
  • 114
    • 84992520625 scopus 로고    scopus 로고
    • A c-Myc/miR17-92/Pten Axis Controls PI3K-Mediated Positive and Negative Selection in B Cell Development and Reconstitutes CD19 Deficiency
    • 27346348
    • Benhamou D, Labi V, Novak R, Dai I, Shafir-Alon S, Weiss A, Gaujoux R, Arnold R, Shen-Orr SS, Rajewsky K, et al. A c-Myc/miR17-92/Pten Axis Controls PI3K-Mediated Positive and Negative Selection in B Cell Development and Reconstitutes CD19 Deficiency. Cell Rep. 2016;16(2):419-31.
    • (2016) Cell Rep , vol.16 , Issue.2 , pp. 419-431
    • Benhamou, D.1    Labi, V.2    Novak, R.3    Dai, I.4    Shafir-Alon, S.5    Weiss, A.6    Gaujoux, R.7    Arnold, R.8    Shen-Orr, S.S.9    Rajewsky, K.10
  • 115
    • 34347402459 scopus 로고    scopus 로고
    • Deficiency in glutamine but not glucose induces MYC-dependent apoptosis in human cells
    • 17606868 2064426
    • Yuneva M, Zamboni N, Oefner P, Sachidanandam R, Lazebnik Y. Deficiency in glutamine but not glucose induces MYC-dependent apoptosis in human cells. J Cell Biol. 2007;178(1):93-105.
    • (2007) J Cell Biol , vol.178 , Issue.1 , pp. 93-105
    • Yuneva, M.1    Zamboni, N.2    Oefner, P.3    Sachidanandam, R.4    Lazebnik, Y.5
  • 117
    • 79952528125 scopus 로고    scopus 로고
    • CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth
    • 21397861
    • Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, Oshima M, Ikeda T, Asaba R, Yagi H, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth. Cancer Cell. 2011;19(3):387-400.
    • (2011) Cancer Cell , vol.19 , Issue.3 , pp. 387-400
    • Ishimoto, T.1    Nagano, O.2    Yae, T.3    Tamada, M.4    Motohara, T.5    Oshima, H.6    Oshima, M.7    Ikeda, T.8    Asaba, R.9    Yagi, H.10
  • 118
    • 84892432945 scopus 로고    scopus 로고
    • Inversed relationship between CD44 variant and c-Myc due to oxidative stress-induced canonical Wnt activation
    • 24333422
    • Yoshida GJ, Saya H. Inversed relationship between CD44 variant and c-Myc due to oxidative stress-induced canonical Wnt activation. Biochem Biophys Res Commun. 2014;443(2):622-7.
    • (2014) Biochem Biophys Res Commun , vol.443 , Issue.2 , pp. 622-627
    • Yoshida, G.J.1    Saya, H.2
  • 119
    • 84956579802 scopus 로고    scopus 로고
    • Therapeutic strategies targeting cancer stem cells
    • 26362755
    • Yoshida GJ, Saya H. Therapeutic strategies targeting cancer stem cells. Cancer Sci. 2016;107(1):5-11.
    • (2016) Cancer Sci , vol.107 , Issue.1 , pp. 5-11
    • Yoshida, G.J.1    Saya, H.2
  • 120
    • 85012951878 scopus 로고    scopus 로고
    • The heterogeneity of cancer stem-like cells at the invasive front
    • 28289330 5307924
    • Yoshida GJ. The heterogeneity of cancer stem-like cells at the invasive front. Cancer Cell Int. 2017;17:23.
    • (2017) Cancer Cell Int , vol.17 , pp. 23
    • Yoshida, G.J.1
  • 121
    • 34249803325 scopus 로고    scopus 로고
    • The transport of glutamine into mammalian cells
    • 17127344
    • McGivan JD, Bungard CI. The transport of glutamine into mammalian cells. Front Biosci. 2007;12:874-82.
    • (2007) Front Biosci , vol.12 , pp. 874-882
    • McGivan, J.D.1    Bungard, C.I.2
  • 122
    • 85038036739 scopus 로고    scopus 로고
    • Glutamine Transport and Mitochondrial Metabolism in Cancer Cell Growth
    • 29376023 5770653
    • Scalise M, Pochini L, Galluccio M, Console L, Indiveri C. Glutamine Transport and Mitochondrial Metabolism in Cancer Cell Growth. Front Oncol. 2017;7:306.
    • (2017) Front Oncol , vol.7 , pp. 306
    • Scalise, M.1    Pochini, L.2    Galluccio, M.3    Console, L.4    Indiveri, C.5
  • 123
    • 84954410074 scopus 로고    scopus 로고
    • Metabolic reprogramming in cancer cells: Glycolysis, glutaminolysis, and Bcl-2 proteins as novel therapeutic targets for cancer
    • 26791262 4721116
    • Li C, Zhang G, Zhao L, Ma Z, Chen H. Metabolic reprogramming in cancer cells: glycolysis, glutaminolysis, and Bcl-2 proteins as novel therapeutic targets for cancer. World J Surg Oncol. 2016;14(1):15.
    • (2016) World J Surg Oncol , vol.14 , Issue.1 , pp. 15
    • Li, C.1    Zhang, G.2    Zhao, L.3    Ma, Z.4    Chen, H.5
  • 124
    • 0034647570 scopus 로고    scopus 로고
    • G6PD deficient cells and the bioreduction of disulfides: Effects of DHEA, GSH depletion and phenylarsine oxide
    • 10891335
    • Biaglow JE, Ayene IS, Koch CJ, Donahue J, Stamato TD, Tuttle SW. G6PD deficient cells and the bioreduction of disulfides: effects of DHEA, GSH depletion and phenylarsine oxide. Biochem Biophys Res Commun. 2000;273(3):846-52.
    • (2000) Biochem Biophys Res Commun , vol.273 , Issue.3 , pp. 846-852
    • Biaglow, J.E.1    Ayene, I.S.2    Koch, C.J.3    Donahue, J.4    Stamato, T.D.5    Tuttle, S.W.6
  • 125
    • 84862278857 scopus 로고    scopus 로고
    • The Role of Mitochondrial NADPH-Dependent Isocitrate Dehydrogenase in Cancer Cells
    • 273947 22675360 3363418
    • Smolkova K, Jezek P. The Role of Mitochondrial NADPH-Dependent Isocitrate Dehydrogenase in Cancer Cells. Int J Cell Biol. 2012;2012:273947.
    • (2012) Int J Cell Biol , vol.2012
    • Smolkova, K.1    Jezek, P.2
  • 126
    • 12444279265 scopus 로고
    • On the origin of cancer cells
    • 13298683
    • Warburg O. On the origin of cancer cells. Science. 1956;123(3191):309-14.
    • (1956) Science , vol.123 , Issue.3191 , pp. 309-314
    • Warburg, O.1
  • 127
    • 85006768050 scopus 로고
    • The Metabolism of Tumors in the Body
    • 19872213 2140820
    • Warburg O, Wind F, Negelein E. The Metabolism of Tumors in the Body. J Gen Physiol. 1927;8(6):519-30.
    • (1927) J Gen Physiol , vol.8 , Issue.6 , pp. 519-530
    • Warburg, O.1    Wind, F.2    Negelein, E.3
  • 128
    • 26444469502 scopus 로고    scopus 로고
    • Detection of resistance to imatinib by metabolic profiling: Clinical and drug development implications
    • 16196499
    • Serkova N, Boros LG. Detection of resistance to imatinib by metabolic profiling: clinical and drug development implications. Am J Pharmacogenomics. 2005;5(5):293-302.
    • (2005) Am J Pharmacogenomics , vol.5 , Issue.5 , pp. 293-302
    • Serkova, N.1    Boros, L.G.2
  • 129
    • 79952227361 scopus 로고    scopus 로고
    • CD44 splice isoform switching in human and mouse epithelium is essential for epithelial-mesenchymal transition and breast cancer progression
    • 21393860 3049398
    • Brown RL, Reinke LM, Damerow MS, Perez D, Chodosh LA, Yang J, Cheng C. CD44 splice isoform switching in human and mouse epithelium is essential for epithelial-mesenchymal transition and breast cancer progression. J Clin Invest. 2011;121(3):1064-74.
    • (2011) J Clin Invest , vol.121 , Issue.3 , pp. 1064-1074
    • Brown, R.L.1    Reinke, L.M.2    Damerow, M.S.3    Perez, D.4    Chodosh, L.A.5    Yang, J.6    Cheng, C.7
  • 131
    • 0242404063 scopus 로고    scopus 로고
    • Identification and characterisation of human xCT that co-expresses, with 4F2 heavy chain, the amino acid transport activity system xc
    • 11417227
    • Bassi MT, Gasol E, Manzoni M, Pineda M, Riboni M, Martin R, Zorzano A, Borsani G, Palacin M. Identification and characterisation of human xCT that co-expresses, with 4F2 heavy chain, the amino acid transport activity system xc. Pflugers Arch. 2001;442(2):286-96.
    • (2001) Pflugers Arch , vol.442 , Issue.2 , pp. 286-296
    • Bassi, M.T.1    Gasol, E.2    Manzoni, M.3    Pineda, M.4    Riboni, M.5    Martin, R.6    Zorzano, A.7    Borsani, G.8    Palacin, M.9
  • 132
    • 77949593175 scopus 로고    scopus 로고
    • CD44+ slow-cycling tumor cell expansion is triggered by cooperative actions of Wnt and prostaglandin E2 in gastric tumorigenesis
    • 20028388
    • Ishimoto T, Oshima H, Oshima M, Kai K, Torii R, Masuko T, Baba H, Saya H, Nagano O. CD44+ slow-cycling tumor cell expansion is triggered by cooperative actions of Wnt and prostaglandin E2 in gastric tumorigenesis. Cancer Sci. 2010;101(3):673-8.
    • (2010) Cancer Sci , vol.101 , Issue.3 , pp. 673-678
    • Ishimoto, T.1    Oshima, H.2    Oshima, M.3    Kai, K.4    Torii, R.5    Masuko, T.6    Baba, H.7    Saya, H.8    Nagano, O.9
  • 133
    • 80052566377 scopus 로고    scopus 로고
    • Tumor entrained neutrophils inhibit seeding in the premetastatic lung
    • 21907922 3172582
    • Granot Z, Henke E, Comen EA, King TA, Norton L, Benezra R. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell. 2011;20(3):300-14.
    • (2011) Cancer Cell , vol.20 , Issue.3 , pp. 300-314
    • Granot, Z.1    Henke, E.2    Comen, E.A.3    King, T.A.4    Norton, L.5    Benezra, R.6
  • 136
    • 84962637563 scopus 로고    scopus 로고
    • Mechanisms of ferroptosis
    • 27048822 4887533
    • Cao JY, Dixon SJ. Mechanisms of ferroptosis. Cell Mol Life Sci. 2016;73(11-12):2195-209.
    • (2016) Cell Mol Life Sci , vol.73 , Issue.11-12 , pp. 2195-2209
    • Cao, J.Y.1    Dixon, S.J.2
  • 138
    • 85018380560 scopus 로고    scopus 로고
    • ATF4 promotes angiogenesis and neuronal cell death and confers ferroptosis in a xCT-dependent manner
    • 28553953 5633655
    • Chen D, Fan Z, Rauh M, Buchfelder M, Eyupoglu IY, Savaskan N. ATF4 promotes angiogenesis and neuronal cell death and confers ferroptosis in a xCT-dependent manner. Oncogene. 2017;36(40):5593-608.
    • (2017) Oncogene , vol.36 , Issue.40 , pp. 5593-5608
    • Chen, D.1    Fan, Z.2    Rauh, M.3    Buchfelder, M.4    Eyupoglu, I.Y.5    Savaskan, N.6
  • 140
    • 0020958437 scopus 로고
    • Structural and kinetic differences between the M2 type pyruvate kinases from lung and various tumors
    • 6326772
    • Eigenbrodt E, Leib S, Kramer W, Friis RR, Schoner W. Structural and kinetic differences between the M2 type pyruvate kinases from lung and various tumors. Biomed Biochim Acta. 1983;42(11-12):S278-82.
    • (1983) Biomed Biochim Acta , vol.42 , Issue.11-12 , pp. S278-S282
    • Eigenbrodt, E.1    Leib, S.2    Kramer, W.3    Friis, R.R.4    Schoner, W.5
  • 143
    • 85009874531 scopus 로고    scopus 로고
    • The Warburg effect: 80 years on
    • 27911732 5095922
    • Potter M, Newport E, Morten KJ. The Warburg effect: 80 years on. Biochem Soc Trans. 2016;44(5):1499-505.
    • (2016) Biochem Soc Trans , vol.44 , Issue.5 , pp. 1499-1505
    • Potter, M.1    Newport, E.2    Morten, K.J.3
  • 145
    • 23044500915 scopus 로고    scopus 로고
    • Pyruvate kinase type M2 and its role in tumor growth and spreading
    • 15908230
    • Mazurek S, Boschek CB, Hugo F, Eigenbrodt E. Pyruvate kinase type M2 and its role in tumor growth and spreading. Semin Cancer Biol. 2005;15(4):300-8.
    • (2005) Semin Cancer Biol , vol.15 , Issue.4 , pp. 300-308
    • Mazurek, S.1    Boschek, C.B.2    Hugo, F.3    Eigenbrodt, E.4
  • 147
    • 84867559423 scopus 로고    scopus 로고
    • Pyruvate kinase M2: Multiple faces for conferring benefits on cancer cells
    • 23071357
    • Tamada M, Suematsu M, Saya H. Pyruvate kinase M2: multiple faces for conferring benefits on cancer cells. Clin Cancer Res. 2012;18(20):5554-61.
    • (2012) Clin Cancer Res , vol.18 , Issue.20 , pp. 5554-5561
    • Tamada, M.1    Suematsu, M.2    Saya, H.3
  • 149
    • 84958073577 scopus 로고    scopus 로고
    • PKM2 and cancer: The function of PKM2 beyond glycolysis
    • 26998110 4774429
    • Dong G, Mao Q, Xia W, Xu Y, Wang J, Xu L, Jiang F. PKM2 and cancer: The function of PKM2 beyond glycolysis. Oncol Lett. 2016;11(3):1980-6.
    • (2016) Oncol Lett , vol.11 , Issue.3 , pp. 1980-1986
    • Dong, G.1    Mao, Q.2    Xia, W.3    Xu, Y.4    Wang, J.5    Xu, L.6    Jiang, F.7
  • 151
    • 35649014840 scopus 로고    scopus 로고
    • Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1
    • 17785433 2169056
    • Kim JW, Gao P, Liu YC, Semenza GL, Dang CV. Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1. Mol Cell Biol. 2007;27(21):7381-93.
    • (2007) Mol Cell Biol , vol.27 , Issue.21 , pp. 7381-7393
    • Kim, J.W.1    Gao, P.2    Liu, Y.C.3    Semenza, G.L.4    Dang, C.V.5
  • 155
    • 85046740404 scopus 로고    scopus 로고
    • Metabolic Hallmarks of Metastasis Formation
    • [Epub ahead of print]
    • Elia I, Doglioni G, Fendt SM. Metabolic Hallmarks of Metastasis Formation. Trends Cell Biol. 2018. https://doi.org/10.1016/j.tcb.2018.04.002. [Epub ahead of print].
    • (2018) Trends Cell Biol.
    • Elia, I.1    Doglioni, G.2    Fendt, S.M.3
  • 160
    • 85049652092 scopus 로고    scopus 로고
    • How to eliminate MYCN-positive hepatic cancer stem cells to prevent the recurrence?
    • 29941608
    • Yoshida GJ. How to eliminate MYCN-positive hepatic cancer stem cells to prevent the recurrence? Proc Natl Acad Sci U S A. 2018;115(28):E6388-E6389.
    • (2018) Proc Natl Acad Sci U S A , vol.115 , Issue.28 , pp. E6388-E6389
    • Yoshida, G.J.1
  • 164
    • 48849108368 scopus 로고    scopus 로고
    • Control of nucleotide biosynthesis by the MYC oncoprotein
    • 18682683 3118556
    • McMahon SB. Control of nucleotide biosynthesis by the MYC oncoprotein. Cell Cycle. 2008;7(15):2275-6.
    • (2008) Cell Cycle , vol.7 , Issue.15 , pp. 2275-2276
    • McMahon, S.B.1
  • 166
    • 84978984470 scopus 로고    scopus 로고
    • Clinical development of cancer therapeutics that target metabolism
    • 26428335
    • Clem BF, O'Neal J, Klarer AC, Telang S, Chesney J. Clinical development of cancer therapeutics that target metabolism. QJM. 2016;109(6):367-72.
    • (2016) QJM , vol.109 , Issue.6 , pp. 367-372
    • Clem, B.F.1    O'Neal, J.2    Klarer, A.C.3    Telang, S.4    Chesney, J.5
  • 167
    • 84978900742 scopus 로고    scopus 로고
    • Glutaminolysis as a target for cancer therapy
    • 26592449
    • Jin L, Alesi GN, Kang S. Glutaminolysis as a target for cancer therapy. Oncogene. 2016;35(28):3619-25.
    • (2016) Oncogene , vol.35 , Issue.28 , pp. 3619-3625
    • Jin, L.1    Alesi, G.N.2    Kang, S.3
  • 168
    • 85012889903 scopus 로고    scopus 로고
    • Glutaminolysis gets the spotlight in cancer
    • 28053289
    • Herranz D. Glutaminolysis gets the spotlight in cancer. Oncotarget. 2017;8(7):10761-2.
    • (2017) Oncotarget , vol.8 , Issue.7 , pp. 10761-10762
    • Herranz, D.1
  • 172
    • 85043699777 scopus 로고    scopus 로고
    • Characterization of the interactions of potent allosteric inhibitors with glutaminase C, a key enzyme in cancer cell glutamine metabolism
    • 29317493
    • Huang Q, Stalnecker C, Zhang C, McDermott LA, Iyer P, O'Neill J, Reimer S, Cerione RA, Katt WP. Characterization of the interactions of potent allosteric inhibitors with glutaminase C, a key enzyme in cancer cell glutamine metabolism. J Biol Chem. 2018;293(10):3535-45.
    • (2018) J Biol Chem , vol.293 , Issue.10 , pp. 3535-3545
    • Huang, Q.1    Stalnecker, C.2    Zhang, C.3    McDermott, L.A.4    Iyer, P.5    O'Neill, J.6    Reimer, S.7    Cerione, R.A.8    Katt, W.P.9
  • 173
    • 78650824534 scopus 로고    scopus 로고
    • Ubiquitin-like protein conjugation and the ubiquitin-proteasome system as drug targets
    • 21151032
    • Bedford L, Lowe J, Dick LR, Mayer RJ, Brownell JE. Ubiquitin-like protein conjugation and the ubiquitin-proteasome system as drug targets. Nat Rev Drug Discov. 2011;10(1):29-46.
    • (2011) Nat Rev Drug Discov , vol.10 , Issue.1 , pp. 29-46
    • Bedford, L.1    Lowe, J.2    Dick, L.R.3    Mayer, R.J.4    Brownell, J.E.5
  • 176
    • 85016923851 scopus 로고    scopus 로고
    • Proteasome expression and activity in cancer and cancer stem cells
    • 28345458
    • Voutsadakis IA. Proteasome expression and activity in cancer and cancer stem cells. Tumour Biol. 2017;39(3):1010428317692248.
    • (2017) Tumour Biol , vol.39 , Issue.3 , pp. 248
    • Voutsadakis, I.A.1
  • 177
    • 84940479138 scopus 로고    scopus 로고
    • Regulating the Regulators: Recent Revelations in the Control of E3 Ubiquitin Ligases
    • 26187467 4571856
    • Vittal V, Stewart MD, Brzovic PS, Klevit RE. Regulating the Regulators: Recent Revelations in the Control of E3 Ubiquitin Ligases. J Biol Chem. 2015;290(35):21244-51.
    • (2015) J Biol Chem , vol.290 , Issue.35 , pp. 21244-21251
    • Vittal, V.1    Stewart, M.D.2    Brzovic, P.S.3    Klevit, R.E.4
  • 178
    • 80051978811 scopus 로고    scopus 로고
    • The predator becomes the prey: Regulating the ubiquitin system by ubiquitylation and degradation
    • 21860393 3545438
    • Weissman AM, Shabek N, Ciechanover A. The predator becomes the prey: regulating the ubiquitin system by ubiquitylation and degradation. Nat Rev Mol Cell Biol. 2011;12(9):605-20.
    • (2011) Nat Rev Mol Cell Biol , vol.12 , Issue.9 , pp. 605-620
    • Weissman, A.M.1    Shabek, N.2    Ciechanover, A.3
  • 179
    • 85028121020 scopus 로고    scopus 로고
    • Role of the ubiquitin ligase Fbw7 in cancer progression
    • 22124735
    • Cheng Y, Li G. Role of the ubiquitin ligase Fbw7 in cancer progression. Cancer Metastasis Rev. 2012;31(1-2):75-87.
    • (2012) Cancer Metastasis Rev , vol.31 , Issue.1-2 , pp. 75-87
    • Cheng, Y.1    Li, G.2
  • 180
    • 84875498138 scopus 로고    scopus 로고
    • Role of SKP1-CUL1-F-box-protein (SCF) E3 ubiquitin ligases in skin cancer
    • 23522382
    • Xie CM, Wei W, Sun Y. Role of SKP1-CUL1-F-box-protein (SCF) E3 ubiquitin ligases in skin cancer. J Genet Genomics. 2013;40(3):97-106.
    • (2013) J Genet Genomics , vol.40 , Issue.3 , pp. 97-106
    • Xie, C.M.1    Wei, W.2    Sun, Y.3
  • 181
    • 84877107994 scopus 로고    scopus 로고
    • Genetically engineered mouse models for functional studies of SKP1-CUL1-F-box-protein (SCF) E3 ubiquitin ligases
    • 23528706 3641602
    • Zhou W, Wei W, Sun Y. Genetically engineered mouse models for functional studies of SKP1-CUL1-F-box-protein (SCF) E3 ubiquitin ligases. Cell Res. 2013;23(5):599-619.
    • (2013) Cell Res , vol.23 , Issue.5 , pp. 599-619
    • Zhou, W.1    Wei, W.2    Sun, Y.3
  • 182
    • 85018342631 scopus 로고    scopus 로고
    • Enzyme-substrate relationships in the ubiquitin system: Approaches for identifying substrates of ubiquitin ligases
    • 28455558
    • O'Connor HF, Huibregtse JM. Enzyme-substrate relationships in the ubiquitin system: approaches for identifying substrates of ubiquitin ligases. Cell Mol Life Sci. 2017;74(18):3363-75.
    • (2017) Cell Mol Life Sci , vol.74 , Issue.18 , pp. 3363-3375
    • O'Connor, H.F.1    Huibregtse, J.M.2
  • 183
    • 38549086019 scopus 로고    scopus 로고
    • FBW7 ubiquitin ligase: A tumour suppressor at the crossroads of cell division, growth and differentiation
    • 18094723
    • Welcker M, Clurman BE. FBW7 ubiquitin ligase: a tumour suppressor at the crossroads of cell division, growth and differentiation. Nat Rev Cancer. 2008;8(2):83-93.
    • (2008) Nat Rev Cancer , vol.8 , Issue.2 , pp. 83-93
    • Welcker, M.1    Clurman, B.E.2
  • 184
    • 17044370346 scopus 로고    scopus 로고
    • Regulation of the cell cycle by SCF-type ubiquitin ligases
    • 15840441
    • Nakayama KI, Nakayama K. Regulation of the cell cycle by SCF-type ubiquitin ligases. Semin Cell Dev Biol. 2005;16(3):323-33.
    • (2005) Semin Cell Dev Biol , vol.16 , Issue.3 , pp. 323-333
    • Nakayama, K.I.1    Nakayama, K.2
  • 185
    • 84978898661 scopus 로고    scopus 로고
    • To wake up cancer stem cells, or to let them sleep, that is the question
    • 27116333 4946711
    • Takeishi S, Nakayama KI. To wake up cancer stem cells, or to let them sleep, that is the question. Cancer Sci. 2016;107(7):875-81.
    • (2016) Cancer Sci , vol.107 , Issue.7 , pp. 875-881
    • Takeishi, S.1    Nakayama, K.I.2
  • 186
    • 0017401702 scopus 로고
    • Effects of the mitotic cell-cycle mutation cdc4 on yeast meiosis
    • 328339 1213672
    • Simchen G, Hirschberg J. Effects of the mitotic cell-cycle mutation cdc4 on yeast meiosis. Genetics. 1977;86(1):57-72.
    • (1977) Genetics , vol.86 , Issue.1 , pp. 57-72
    • Simchen, G.1    Hirschberg, J.2
  • 187
    • 6944227551 scopus 로고    scopus 로고
    • A nucleolar isoform of the Fbw7 ubiquitin ligase regulates c-Myc and cell size
    • 15498494
    • Welcker M, Orian A, Grim JE, Eisenman RN, Clurman BE. A nucleolar isoform of the Fbw7 ubiquitin ligase regulates c-Myc and cell size. Curr Biol. 2004;14(20):1852-7.
    • (2004) Curr Biol , vol.14 , Issue.20 , pp. 1852-1857
    • Welcker, M.1    Orian, A.2    Grim, J.E.3    Eisenman, R.N.4    Clurman, B.E.5
  • 189
    • 33846266628 scopus 로고    scopus 로고
    • Fbw7 isoform interaction contributes to cyclin e proteolysis
    • 17189384
    • Zhang W, Koepp DM. Fbw7 isoform interaction contributes to cyclin E proteolysis. Mol Cancer Res. 2006;4(12):935-43.
    • (2006) Mol Cancer Res , vol.4 , Issue.12 , pp. 935-943
    • Zhang, W.1    Koepp, D.M.2
  • 190
    • 78149456945 scopus 로고    scopus 로고
    • WD40 repeat propellers define a ubiquitin-binding domain that regulates turnover of F box proteins
    • 21070969 3266742
    • Pashkova N, Gakhar L, Winistorfer SC, Yu L, Ramaswamy S, Piper RC. WD40 repeat propellers define a ubiquitin-binding domain that regulates turnover of F box proteins. Mol Cell. 2010;40(3):433-43.
    • (2010) Mol Cell , vol.40 , Issue.3 , pp. 433-443
    • Pashkova, N.1    Gakhar, L.2    Winistorfer, S.C.3    Yu, L.4    Ramaswamy, S.5    Piper, R.C.6
  • 193
    • 36549071481 scopus 로고    scopus 로고
    • Conditional inactivation of Fbxw7 impairs cell-cycle exit during T cell differentiation and results in lymphomatogenesis
    • 17984302 2118521
    • Onoyama I, Tsunematsu R, Matsumoto A, Kimura T, de Alboran IM, Nakayama K, Nakayama KI. Conditional inactivation of Fbxw7 impairs cell-cycle exit during T cell differentiation and results in lymphomatogenesis. J Exp Med. 2007;204(12):2875-88.
    • (2007) J Exp Med , vol.204 , Issue.12 , pp. 2875-2888
    • Onoyama, I.1    Tsunematsu, R.2    Matsumoto, A.3    Kimura, T.4    De Alboran, I.M.5    Nakayama, K.6    Nakayama, K.I.7
  • 194
    • 42149132696 scopus 로고    scopus 로고
    • Fbxw7 acts as a critical fail-safe against premature loss of hematopoietic stem cells and development of T-ALL
    • 18367647 2335330
    • Matsuoka S, Oike Y, Onoyama I, Iwama A, Arai F, Takubo K, Mashimo Y, Oguro H, Nitta E, Ito K, et al. Fbxw7 acts as a critical fail-safe against premature loss of hematopoietic stem cells and development of T-ALL. Genes Dev. 2008;22(8):986-991.
    • (2008) Genes Dev , vol.22 , Issue.8 , pp. 986-991
    • Matsuoka, S.1    Oike, Y.2    Onoyama, I.3    Iwama, A.4    Arai, F.5    Takubo, K.6    Mashimo, Y.7    Oguro, H.8    Nitta, E.9    Ito, K.10
  • 195
    • 85014911930 scopus 로고    scopus 로고
    • The NOTCH1-MYC highway toward T-cell acute lymphoblastic leukemia
    • 28115368
    • Sanchez-Martin M, Ferrando A. The NOTCH1-MYC highway toward T-cell acute lymphoblastic leukemia. Blood. 2017;129(9):1124-33.
    • (2017) Blood , vol.129 , Issue.9 , pp. 1124-1133
    • Sanchez-Martin, M.1    Ferrando, A.2
  • 197
    • 80052276418 scopus 로고    scopus 로고
    • Breaking the cell cycle of HSCs by p57 and friends
    • 21885016
    • Tesio M, Trumpp A. Breaking the cell cycle of HSCs by p57 and friends. Cell Stem Cell. 2011;9(3):187-92.
    • (2011) Cell Stem Cell , vol.9 , Issue.3 , pp. 187-192
    • Tesio, M.1    Trumpp, A.2
  • 198
    • 84878240136 scopus 로고    scopus 로고
    • Molecular regulation of stem cell quiescence
    • 23698583
    • Cheung TH, Rando TA. Molecular regulation of stem cell quiescence. Nat Rev Mol Cell Biol. 2013;14(6):329-40.
    • (2013) Nat Rev Mol Cell Biol , vol.14 , Issue.6 , pp. 329-340
    • Cheung, T.H.1    Rando, T.A.2
  • 199
    • 54049087334 scopus 로고    scopus 로고
    • Notch-dependent cell cycle arrest and apoptosis in mouse embryonic fibroblasts lacking Fbxw7
    • 18641686
    • Ishikawa Y, Onoyama I, Nakayama KI, Nakayama K. Notch-dependent cell cycle arrest and apoptosis in mouse embryonic fibroblasts lacking Fbxw7. Oncogene. 2008;27(47):6164-74.
    • (2008) Oncogene , vol.27 , Issue.47 , pp. 6164-6174
    • Ishikawa, Y.1    Onoyama, I.2    Nakayama, K.I.3    Nakayama, K.4
  • 200
    • 84859496041 scopus 로고    scopus 로고
    • Notch inhibition as a promising new approach to cancer therapy
    • 22399357 3361718
    • Purow B. Notch inhibition as a promising new approach to cancer therapy. Adv Exp Med Biol. 2012;727:305-19.
    • (2012) Adv Exp Med Biol , vol.727 , pp. 305-319
    • Purow, B.1
  • 201
    • 55949136859 scopus 로고    scopus 로고
    • Anoikis resistance and tumor metastasis
    • 18579285
    • Simpson CD, Anyiwe K, Schimmer AD. Anoikis resistance and tumor metastasis. Cancer Lett. 2008;272(2):177-85.
    • (2008) Cancer Lett , vol.272 , Issue.2 , pp. 177-185
    • Simpson, C.D.1    Anyiwe, K.2    Schimmer, A.D.3
  • 202
    • 84897085768 scopus 로고    scopus 로고
    • Roles of F-box proteins in cancer
    • 24658274 4306233
    • Wang Z, Liu P, Inuzuka H, Wei W. Roles of F-box proteins in cancer. Nat Rev Cancer. 2014;14(4):233-47.
    • (2014) Nat Rev Cancer , vol.14 , Issue.4 , pp. 233-247
    • Wang, Z.1    Liu, P.2    Inuzuka, H.3    Wei, W.4
  • 204
    • 84861189396 scopus 로고    scopus 로고
    • Tumor suppressor functions of FBW7 in cancer development and progression
    • 22673505 3372850
    • Wang Z, Inuzuka H, Zhong J, Wan L, Fukushima H, Sarkar FH, Wei W. Tumor suppressor functions of FBW7 in cancer development and progression. FEBS Lett. 2012;586(10):1409-18.
    • (2012) FEBS Lett , vol.586 , Issue.10 , pp. 1409-1418
    • Wang, Z.1    Inuzuka, H.2    Zhong, J.3    Wan, L.4    Fukushima, H.5    Sarkar, F.H.6    Wei, W.7
  • 205
    • 85013304766 scopus 로고    scopus 로고
    • E3 ubiquitin ligase FBW7alpha inhibits cholangiocarcinoma cell proliferation by downregulating c-Myc and cyclin e
    • 28184929
    • Li M, Ouyang L, Zheng Z, Xiang D, Ti A, Li L, Dan Y, Yu C, Li W. E3 ubiquitin ligase FBW7alpha inhibits cholangiocarcinoma cell proliferation by downregulating c-Myc and cyclin E. Oncol Rep. 2017;37(3):1627-36.
    • (2017) Oncol Rep , vol.37 , Issue.3 , pp. 1627-1636
    • Li, M.1    Ouyang, L.2    Zheng, Z.3    Xiang, D.4    Ti, A.5    Li, L.6    Dan, Y.7    Yu, C.8    Li, W.9
  • 208
    • 84958159750 scopus 로고    scopus 로고
    • Regulating Fbw7 on the road to cancer
    • 26459133
    • Xu W, Taranets L, Popov N. Regulating Fbw7 on the road to cancer. Semin Cancer Biol. 2016;36:62-70.
    • (2016) Semin Cancer Biol , vol.36 , pp. 62-70
    • Xu, W.1    Taranets, L.2    Popov, N.3
  • 209
    • 4644318882 scopus 로고    scopus 로고
    • Tyrosine kinase inhibitors in renal cell carcinoma
    • 15448033
    • Potti A, George DJ. Tyrosine kinase inhibitors in renal cell carcinoma. Clin Cancer Res. 2004;10(18 Pt 2):6371S-6S.
    • (2004) Clin Cancer Res , vol.10 , Issue.18 , pp. 6371S-6376S
    • Potti, A.1    George, D.J.2
  • 210
    • 33745459794 scopus 로고    scopus 로고
    • Renal cell carcinoma and the use of sorafenib
    • 18360584 1661649
    • Larkin JM, Eisen T. Renal cell carcinoma and the use of sorafenib. Ther Clin Risk Manag. 2006;2(1):87-98.
    • (2006) Ther Clin Risk Manag , vol.2 , Issue.1 , pp. 87-98
    • Larkin, J.M.1    Eisen, T.2
  • 211
    • 33646345376 scopus 로고    scopus 로고
    • Ubiquitin ligases: Cell-cycle control and cancer
    • 16633365
    • Nakayama KI, Nakayama K. Ubiquitin ligases: cell-cycle control and cancer. Nat Rev Cancer. 2006;6(5):369-81.
    • (2006) Nat Rev Cancer , vol.6 , Issue.5 , pp. 369-381
    • Nakayama, K.I.1    Nakayama, K.2
  • 212
    • 67349136004 scopus 로고    scopus 로고
    • Mcl-1 is a potential therapeutic target in multiple types of cancer
    • 19099185
    • Akgul C. Mcl-1 is a potential therapeutic target in multiple types of cancer. Cell Mol Life Sci. 2009;66(8):1326-36.
    • (2009) Cell Mol Life Sci , vol.66 , Issue.8 , pp. 1326-1336
    • Akgul, C.1
  • 214
    • 84908381929 scopus 로고    scopus 로고
    • Role of Fbxw7 in the maintenance of normal stem cells and cancer-initiating cells
    • 24853181 4453837
    • Takeishi S, Nakayama KI. Role of Fbxw7 in the maintenance of normal stem cells and cancer-initiating cells. Br J Cancer. 2014;111(6):1054-9.
    • (2014) Br J Cancer , vol.111 , Issue.6 , pp. 1054-1059
    • Takeishi, S.1    Nakayama, K.I.2
  • 215
    • 84958114871 scopus 로고    scopus 로고
    • Fbw7 and its counteracting forces in stem cells and cancer: Oncoproteins in the balance
    • 26410034
    • Cremona CA, Sancho R, Diefenbacher ME, Behrens A. Fbw7 and its counteracting forces in stem cells and cancer: Oncoproteins in the balance. Semin Cancer Biol. 2016;36:52-61.
    • (2016) Semin Cancer Biol , vol.36 , pp. 52-61
    • Cremona, C.A.1    Sancho, R.2    Diefenbacher, M.E.3    Behrens, A.4
  • 216
    • 52549087785 scopus 로고    scopus 로고
    • Cancer stem cells in solid tumours: Accumulating evidence and unresolved questions
    • 18784658
    • Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008;8(10):755-68.
    • (2008) Nat Rev Cancer , vol.8 , Issue.10 , pp. 755-768
    • Visvader, J.E.1    Lindeman, G.J.2
  • 217
    • 85015610004 scopus 로고    scopus 로고
    • The secret life of quiescent cancer stem cells
    • e968067 27308385
    • Zeuner A. The secret life of quiescent cancer stem cells. Mol Cell Oncol. 2015;2(1):e968067.
    • (2015) Mol Cell Oncol , vol.2 , Issue.1
    • Zeuner, A.1
  • 218
    • 84978765993 scopus 로고    scopus 로고
    • Cancer Stem Cell Quiescence and Plasticity as Major Challenges in Cancer Therapy
    • 27418931 4932171
    • Chen W, Dong J, Haiech J, Kilhoffer MC, Zeniou M. Cancer Stem Cell Quiescence and Plasticity as Major Challenges in Cancer Therapy. Stem Cells Int. 2016;2016:1740936.
    • (2016) Stem Cells Int , vol.2016 , pp. 1740936
    • Chen, W.1    Dong, J.2    Haiech, J.3    Kilhoffer, M.C.4    Zeniou, M.5
  • 219
    • 0035110959 scopus 로고    scopus 로고
    • Dual antitumor effects of 5-fluorouracil on the cell cycle in colorectal carcinoma cells: A novel target mechanism concept for pharmacokinetic modulating chemotherapy
    • 11221829
    • Yoshikawa R, Kusunoki M, Yanagi H, Noda M, Furuyama JI, Yamamura T, Hashimoto-Tamaoki T. Dual antitumor effects of 5-fluorouracil on the cell cycle in colorectal carcinoma cells: a novel target mechanism concept for pharmacokinetic modulating chemotherapy. Cancer Res. 2001;61(3):1029-37.
    • (2001) Cancer Res , vol.61 , Issue.3 , pp. 1029-1037
    • Yoshikawa, R.1    Kusunoki, M.2    Yanagi, H.3    Noda, M.4    Furuyama, J.I.5    Yamamura, T.6    Hashimoto-Tamaoki, T.7
  • 220
    • 0043095544 scopus 로고    scopus 로고
    • Cell cycle phase-specific chemotherapy: Computational methods for guiding treatment
    • 12548007
    • Gardner SN. Cell cycle phase-specific chemotherapy: computational methods for guiding treatment. Cell Cycle. 2002;1(6):369-74.
    • (2002) Cell Cycle , vol.1 , Issue.6 , pp. 369-374
    • Gardner, S.N.1
  • 221
    • 0842308823 scopus 로고    scopus 로고
    • Progress in treatment of small-cell lung cancer: Role of CPT-11
    • 14676791 2395289
    • Saijo N. Progress in treatment of small-cell lung cancer: role of CPT-11. Br J Cancer. 2003;89(12):2178-83.
    • (2003) Br J Cancer , vol.89 , Issue.12 , pp. 2178-2183
    • Saijo, N.1
  • 222
    • 77954187741 scopus 로고    scopus 로고
    • DNA topoisomerases and their poisoning by anticancer and antibacterial drugs
    • 20534341
    • Pommier Y, Leo E, Zhang H, Marchand C. DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem Biol. 2010;17(5):421-33.
    • (2010) Chem Biol , vol.17 , Issue.5 , pp. 421-433
    • Pommier, Y.1    Leo, E.2    Zhang, H.3    Marchand, C.4
  • 223
    • 84893664923 scopus 로고    scopus 로고
    • Cancer stem cell characteristics, ALDH1 expression in the invasive front of nasopharyngeal carcinoma
    • 24248285
    • Luo WR, Yao KT. Cancer stem cell characteristics, ALDH1 expression in the invasive front of nasopharyngeal carcinoma. Virchows Arch. 2014;464(1):35-43.
    • (2014) Virchows Arch , vol.464 , Issue.1 , pp. 35-43
    • Luo, W.R.1    Yao, K.T.2
  • 225
    • 85026824000 scopus 로고    scopus 로고
    • Strategies to Inhibit Myc and Their Clinical Applicability
    • 28280720 5322154
    • Whitfield JR, Beaulieu ME, Soucek L. Strategies to Inhibit Myc and Their Clinical Applicability. Front Cell Dev Biol. 2017;5:10.
    • (2017) Front Cell Dev Biol , vol.5 , pp. 10
    • Whitfield, J.R.1    Beaulieu, M.E.2    Soucek, L.3
  • 226
    • 0037096903 scopus 로고    scopus 로고
    • Omomyc, a potential Myc dominant negative, enhances Myc-induced apoptosis
    • 12067996
    • Soucek L, Jucker R, Panacchia L, Ricordy R, Tato F, Nasi S. Omomyc, a potential Myc dominant negative, enhances Myc-induced apoptosis. Cancer Res. 2002;62(12):3507-10.
    • (2002) Cancer Res , vol.62 , Issue.12 , pp. 3507-3510
    • Soucek, L.1    Jucker, R.2    Panacchia, L.3    Ricordy, R.4    Tato, F.5    Nasi, S.6
  • 229
    • 84962683838 scopus 로고    scopus 로고
    • BET Bromodomain Inhibition Releases the Mediator Complex from Select cis-Regulatory Elements
    • 27068464 4838499
    • Bhagwat AS, Roe JS, Mok BYL, Hohmann AF, Shi J, Vakoc CR. BET Bromodomain Inhibition Releases the Mediator Complex from Select cis-Regulatory Elements. Cell Rep. 2016;15(3):519-30.
    • (2016) Cell Rep , vol.15 , Issue.3 , pp. 519-530
    • Bhagwat, A.S.1    Roe, J.S.2    Mok, B.Y.L.3    Hohmann, A.F.4    Shi, J.5    Vakoc, C.R.6
  • 231
    • 84943747585 scopus 로고    scopus 로고
    • Disruption of BRD4 at H3K27Ac-enriched enhancer region correlates with decreased c-Myc expression in Merkel cell carcinoma
    • 25941994 4622756
    • Sengupta D, Kannan A, Kern M, Moreno MA, Vural E, Stack B Jr, Suen JY, Tackett AJ, Gao L. Disruption of BRD4 at H3K27Ac-enriched enhancer region correlates with decreased c-Myc expression in Merkel cell carcinoma. Epigenetics. 2015;10(6):460-6.
    • (2015) Epigenetics , vol.10 , Issue.6 , pp. 460-466
    • Sengupta, D.1    Kannan, A.2    Kern, M.3    Moreno, M.A.4    Vural, E.5    Stack, B.6    Suen, J.Y.7    Tackett, A.J.8    Gao, L.9
  • 232
    • 0031455754 scopus 로고    scopus 로고
    • Interferon-gamma and retinoic acid down-regulate N-myc in neuroblastoma through complementary mechanisms of action
    • 9570357
    • Wada RK, Pai DS, Huang J, Yamashiro JM, Sidell N. Interferon-gamma and retinoic acid down-regulate N-myc in neuroblastoma through complementary mechanisms of action. Cancer Lett. 1997;121(2):181-8.
    • (1997) Cancer Lett , vol.121 , Issue.2 , pp. 181-188
    • Wada, R.K.1    Pai, D.S.2    Huang, J.3    Yamashiro, J.M.4    Sidell, N.5
  • 233
    • 35148882962 scopus 로고    scopus 로고
    • A phase 2 trial of all-trans-retinoic acid in combination with interferon-alpha2a in children with recurrent neuroblastoma or Wilms tumor: A Pediatric Oncology Branch, NCI and Children's Oncology Group Study
    • 16900483
    • Adamson PC, Matthay KK, O'Brien M, Reaman GH, Sato JK, Balis FM. A phase 2 trial of all-trans-retinoic acid in combination with interferon-alpha2a in children with recurrent neuroblastoma or Wilms tumor: A Pediatric Oncology Branch, NCI and Children's Oncology Group Study. Pediatr Blood Cancer. 2007;49(5):661-5.
    • (2007) Pediatr Blood Cancer , vol.49 , Issue.5 , pp. 661-665
    • Adamson, P.C.1    Matthay, K.K.2    O'Brien, M.3    Reaman, G.H.4    Sato, J.K.5    Balis, F.M.6
  • 235
    • 84944446748 scopus 로고    scopus 로고
    • Bortezomib inhibits Burkitt's lymphoma cell proliferation by downregulating sumoylated hnRNP K and c-Myc expression
    • 26317903 4694880
    • Suk FM, Lin SY, Lin RJ, Hsine YH, Liao YJ, Fang SU, Liang YC. Bortezomib inhibits Burkitt's lymphoma cell proliferation by downregulating sumoylated hnRNP K and c-Myc expression. Oncotarget. 2015;6(28):25988-6001.
    • (2015) Oncotarget , vol.6 , Issue.28 , pp. 25988-26001
    • Suk, F.M.1    Lin, S.Y.2    Lin, R.J.3    Hsine, Y.H.4    Liao, Y.J.5    Fang, S.U.6    Liang, Y.C.7


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.