메뉴 건너뛰기




Volumn 26, Issue 4, 2016, Pages 484-498

Drugging the undruggables: Exploring the ubiquitin system for drug development

Author keywords

degrader; drug target; hydrophobic tag; PROTAC; SERD; ubiquitin proteasome system

Indexed keywords

PROTEASOME; PROTEASOME INHIBITOR; UBIQUITIN;

EID: 84963540125     PISSN: 10010602     EISSN: 17487838     Source Type: Journal    
DOI: 10.1038/cr.2016.31     Document Type: Review
Times cited : (355)

References (133)
  • 3
    • 49549117842 scopus 로고    scopus 로고
    • Ubiquitin chain editing revealed by polyubiquitin linkage-specific antibodies
    • Newton K, Matsumoto ML, Wertz IE, et al. Ubiquitin chain editing revealed by polyubiquitin linkage-specific antibodies. Cell 2008; 134:668-678.
    • (2008) Cell , vol.134 , pp. 668-678
    • Newton, K.1    Matsumoto, M.L.2    Wertz, I.E.3
  • 4
    • 43049162227 scopus 로고    scopus 로고
    • Mechanism of ubiquitin-chain formation by the human anaphase-promoting complex
    • Jin L, Williamson A, Banerjee S, Philipp I, Rape M. Mechanism of ubiquitin-chain formation by the human anaphase-promoting complex. 2008; Cell 133:653-665.
    • (2008) Cell , vol.133 , pp. 653-665
    • Jin, L.1    Williamson, A.2    Banerjee, S.3    Philipp, I.4    Rape, M.5
  • 5
    • 84858124845 scopus 로고    scopus 로고
    • Generation and physiological roles of linear ubiquitin chains
    • Walczak H, Iwai K, Dikic I. Generation and physiological roles of linear ubiquitin chains. BMC Biol 2012; 10:23-23.
    • (2012) BMC Biol , vol.10 , pp. 23
    • Walczak, H.1    Iwai, K.2    Dikic, I.3
  • 6
    • 28344456279 scopus 로고    scopus 로고
    • A genomic and functional inventory of deubiquitinating enzymes
    • Nijman SM, Luna-Vargas MP, Velds A, et al. A genomic and functional inventory of deubiquitinating enzymes. Cell 2005; 123:773-786.
    • (2005) Cell , vol.123 , pp. 773-786
    • Nijman, S.M.1    Luna-Vargas, M.P.2    Velds, A.3
  • 7
    • 68049084674 scopus 로고    scopus 로고
    • Breaking the chains: Structure and function of the deubiquitinases
    • Komander D, Clague MJ, Urbe S. Breaking the chains: structure and function of the deubiquitinases. Nat Rev Mol Cell Biol 2009; 10:550-563.
    • (2009) Nat Rev Mol Cell Biol , vol.10 , pp. 550-563
    • Komander, D.1    Clague, M.J.2    Urbe, S.3
  • 8
    • 84922393807 scopus 로고    scopus 로고
    • Ubiquitination in disease pathogenesis and treatment
    • Popovic D, Vucic D, Dikic I. Ubiquitination in disease pathogenesis and treatment. Nat Med 2014; 20:1242-1253.
    • (2014) Nat Med , vol.20 , pp. 1242-1253
    • Popovic, D.1    Vucic, D.2    Dikic, I.3
  • 9
    • 0032499264 scopus 로고    scopus 로고
    • Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism
    • Kitada T, Asakawa S, Hattori N, et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998; 392:605-608.
    • (1998) Nature , vol.392 , pp. 605-608
    • Kitada, T.1    Asakawa, S.2    Hattori, N.3
  • 10
    • 1542615085 scopus 로고    scopus 로고
    • USP6 (Tre2) fusion oncogenes in aneurysmal bone cyst
    • Oliveira AM, Hsi BL, Weremowicz S, et al. USP6 (Tre2) fusion oncogenes in aneurysmal bone cyst. Cancer Res 2004; 64:1920-1923;
    • (2004) Cancer Res , vol.64 , pp. 1920-1923
    • Oliveira, A.M.1    Hsi, B.L.2    Weremowicz, S.3
  • 11
    • 57749188299 scopus 로고    scopus 로고
    • Targeting cancer with small molecule kinase inhibitors
    • Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer 2009; 9:28-39.
    • (2009) Nat Rev Cancer , vol.9 , pp. 28-39
    • Zhang, J.1    Yang, P.L.2    Gray, N.S.3
  • 12
    • 75149130051 scopus 로고    scopus 로고
    • Targeting the cancer kinome through polypharmacology
    • Knight ZA, Lin H, Shokat KM. Targeting the cancer kinome through polypharmacology. Nat Rev Cancer 2010; 10:130-137.
    • (2010) Nat Rev Cancer , vol.10 , pp. 130-137
    • Knight, Z.A.1    Lin, H.2    Shokat, K.M.3
  • 13
    • 67349256160 scopus 로고    scopus 로고
    • Ubiquitin-like protein activation by E1 enzymes: The apex for downstream signalling pathways
    • Schulman BA, Wade Harper J. Ubiquitin-like protein activation by E1 enzymes: the apex for downstream signalling pathways. Nat Rev Mol Cell Biol 2009; 10:319-331.
    • (2009) Nat Rev Mol Cell Biol , vol.10 , pp. 319-331
    • Schulman, B.A.1    Wade Harper, J.2
  • 14
    • 34347329214 scopus 로고    scopus 로고
    • Dual E1 activation systems for ubiquitin differentially regulate E2 enzyme charging
    • Jin J, Li X, Gygi SP, Harper JW. Dual E1 activation systems for ubiquitin differentially regulate E2 enzyme charging. Nature 2007; 447:1135-1138.
    • (2007) Nature , vol.447 , pp. 1135-1138
    • Jin, J.1    Li, X.2    Gygi, S.P.3    Harper, J.W.4
  • 15
    • 35148886143 scopus 로고    scopus 로고
    • Inhibitors of ubiquitin-activating enzyme (E1), a new class of potential cancer therapeutics
    • Yang, Y, Kitagaki J, Dai RM, et al. Inhibitors of ubiquitin-activating enzyme (E1), a new class of potential cancer therapeutics. Cancer Res 2007; 67:9472-9481.
    • (2007) Cancer Res , vol.67 , pp. 9472-9481
    • Yang, Y.1    Kitagaki, J.2    Dai, R.M.3
  • 16
    • 77950421253 scopus 로고    scopus 로고
    • The ubiquitin-activating enzyme E1 as a therapeutic target for the treatment of leukemia and multiple myeloma
    • Xu GW, Ali M, Wood TE, et al. The ubiquitin-activating enzyme E1 as a therapeutic target for the treatment of leukemia and multiple myeloma. Blood 2010; 115:2251-2259.
    • (2010) Blood , vol.115 , pp. 2251-2259
    • Xu, G.W.1    Ali, M.2    Wood, T.E.3
  • 17
    • 84901988917 scopus 로고    scopus 로고
    • Structural and functional insights to ubiquitin-like protein conjugation
    • Streich FC, Lima CD. Structural and functional insights to ubiquitin-like protein conjugation. Annu Rev Biophys 2014; 43:357-379.
    • (2014) Annu Rev Biophys , vol.43 , pp. 357-379
    • Streich, F.C.1    Lima, C.D.2
  • 18
    • 64749098830 scopus 로고    scopus 로고
    • An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer
    • Soucy TA, Smith PG, Milhollen MA, et al. An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature 2009; 458:732-736.
    • (2009) Nature , vol.458 , pp. 732-736
    • Soucy, T.A.1    Smith, P.G.2    Milhollen, M.A.3
  • 19
    • 73649110303 scopus 로고    scopus 로고
    • Substrate-assisted inhibition of ubiquitin-like protein-activating enzymes: The NEDD8 E1 inhibitor MLN4924 forms a NEDD8-AMP mimetic in situ
    • Brownell JE, Sintchak MD, Gavin JM, et al. Substrate-assisted inhibition of ubiquitin-like protein-activating enzymes: the NEDD8 E1 inhibitor MLN4924 forms a NEDD8-AMP mimetic in situ. Mol Cell 2010; 37:102-111.
    • (2010) Mol Cell , vol.37 , pp. 102-111
    • Brownell, J.E.1    Sintchak, M.D.2    Gavin, J.M.3
  • 20
    • 50449108516 scopus 로고    scopus 로고
    • Structural insights into NEDD8 activation of cullin-RING ligases: Conformational control of conjugation
    • Duda DM, Borg LA, Scott DC, Hunt HW, Hammel M, Schulman BA. Structural insights into NEDD8 activation of cullin-RING ligases: conformational control of conjugation. Cell 2008; 134:995-1006.
    • (2008) Cell , vol.134 , pp. 995-1006
    • Duda, D.M.1    Borg, L.A.2    Scott, D.C.3    Hunt, H.W.4    Hammel, M.5    Schulman, B.A.6
  • 21
    • 78649974984 scopus 로고    scopus 로고
    • Dynamics of cullin-RING ubiquitin ligase network revealed by systematic quantitative proteomics
    • Bennett EJ, Rush J, Gygi SP, Harper JW. Dynamics of cullin-RING ubiquitin ligase network revealed by systematic quantitative proteomics. Cell 2010; 143:951-965.
    • (2010) Cell , vol.143 , pp. 951-965
    • Bennett, E.J.1    Rush, J.2    Gygi, S.P.3    Harper, J.W.4
  • 22
    • 82455179484 scopus 로고    scopus 로고
    • Systematic and quantitative assessment of the ubiquitin-modified proteome
    • Kim W, Bennett EJ, Huttlin EL, et al. Systematic and quantitative assessment of the ubiquitin-modified proteome. Mol Cell 2011; 44:325-340.
    • (2011) Mol Cell , vol.44 , pp. 325-340
    • Kim, W.1    Bennett, E.J.2    Huttlin, E.L.3
  • 23
    • 80054694510 scopus 로고    scopus 로고
    • Global identification of modular cullin-RING ligase substrates
    • Emanuele MJ, Elia AE, Xu Q, et al. Global identification of modular cullin-RING ligase substrates. Cell 2011; 147, 459-474.
    • (2011) Cell , vol.147 , pp. 459-474
    • Emanuele, M.J.1    Elia, A.E.2    Xu, Q.3
  • 24
    • 70350461507 scopus 로고    scopus 로고
    • Building ubiquitin chains: E2 enzymes at work
    • Ye Y, Rape M. Building ubiquitin chains: E2 enzymes at work. Nat Rev Mol Cell Biol 2009; 10:755-764.
    • (2009) Nat Rev Mol Cell Biol , vol.10 , pp. 755-764
    • Ye, Y.1    Rape, M.2
  • 25
    • 79959656754 scopus 로고    scopus 로고
    • An allosteric inhibitor of the human Cdc34 ubiquitin-conjugating enzyme
    • Ceccarelli DF, Tang X, Pelletier B, et al. An allosteric inhibitor of the human Cdc34 ubiquitin-conjugating enzyme. Cell 2011; 145:1075-1087.
    • (2011) Cell , vol.145 , pp. 1075-1087
    • Ceccarelli, D.F.1    Tang, X.2    Pelletier, B.3
  • 26
    • 0034644474 scopus 로고    scopus 로고
    • Activation of the IκB kinase complex by TRAF6 requires a dimeric ubiquitin-conjugating enzyme complex and a unique polyubiquitin chain
    • Deng L, Wang C, Spencer E, et al. Activation of the IκB kinase complex by TRAF6 requires a dimeric ubiquitin-conjugating enzyme complex and a unique polyubiquitin chain. Cell 2000; 103:351-361.
    • (2000) Cell , vol.103 , pp. 351-361
    • Deng, L.1    Wang, C.2    Spencer, E.3
  • 27
    • 84865415118 scopus 로고    scopus 로고
    • Inhibition of proliferation and survival of diffuse large B-cell lymphoma cells by a small-molecule inhibitor of the ubiquitin-conjugating enzyme Ubc13-Uev1A
    • Pulvino M, Liang Y, Oleksyn D, et al. Inhibition of proliferation and survival of diffuse large B-cell lymphoma cells by a small-molecule inhibitor of the ubiquitin-conjugating enzyme Ubc13-Uev1A. Blood 2012; 120:1668-1677.
    • (2012) Blood , vol.120 , pp. 1668-1677
    • Pulvino, M.1    Liang, Y.2    Oleksyn, D.3
  • 28
    • 0030772376 scopus 로고    scopus 로고
    • Novel inhibitors of cytokine-induced IκBα phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo
    • Pierce JW, Schoenleber R, Jesmok G, et al. Novel inhibitors of cytokine-induced IκBα phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo. J Biol Chem 1997; 272:21096-21103.
    • (1997) J Biol Chem , vol.272 , pp. 21096-21103
    • Pierce, J.W.1    Schoenleber, R.2    Jesmok, G.3
  • 29
    • 84876240899 scopus 로고    scopus 로고
    • The anti-inflammatory drug BAY 11-7082 suppresses the MyD88-dependent signalling network by targeting the ubiquitin system
    • Strickson S, Campbell DG, Emmerich CH, et al. The anti-inflammatory drug BAY 11-7082 suppresses the MyD88-dependent signalling network by targeting the ubiquitin system. Biochem J 2013; 451:427-437.
    • (2013) Biochem J , vol.451 , pp. 427-437
    • Strickson, S.1    Campbell, D.G.2    Emmerich, C.H.3
  • 30
    • 44949231368 scopus 로고    scopus 로고
    • Genome-wide and functional annotation of human E3 ubiquitin ligases identifies MULAN, a mitochondrial E3 that regulates the organelle's dynamics and signaling
    • Li W, Bengtson MH, Ulbrich A, et al. Genome-wide and functional annotation of human E3 ubiquitin ligases identifies MULAN, a mitochondrial E3 that regulates the organelle's dynamics and signaling. PLoS One 2008; 3:e1487.
    • (2008) PLoS One , vol.3
    • Li, W.1    Bengtson, M.H.2    Ulbrich, A.3
  • 31
    • 84898754901 scopus 로고    scopus 로고
    • New insights into ubiquitin E3 ligase mechanism
    • Berndsen CE, Wolberger C. New insights into ubiquitin E3 ligase mechanism. Nat Struct Mol Biol 2014; 21:301-307.
    • (2014) Nat Struct Mol Biol , vol.21 , pp. 301-307
    • Berndsen, C.E.1    Wolberger, C.2
  • 33
    • 84896870884 scopus 로고    scopus 로고
    • RBR E3 ubiquitin ligases: New structures, new insights, new questions
    • Spratt Donald E, Walden H, Shaw GS. RBR E3 ubiquitin ligases: new structures, new insights, new questions. Biochem J 2014; 458:421-437.
    • (2014) Biochem J , vol.458 , pp. 421-437
    • Spratt Donald, E.1    Walden, H.2    Shaw, G.S.3
  • 35
    • 0033176887 scopus 로고    scopus 로고
    • SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27
    • Carrano AC, Eytan E, Hershko A, Pagano M. SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27. Nat Cell Biol 1999; 1:193-199.
    • (1999) Nat Cell Biol , vol.1 , pp. 193-199
    • Carrano, A.C.1    Eytan, E.2    Hershko, A.3    Pagano, M.4
  • 36
    • 0033174070 scopus 로고    scopus 로고
    • P45SKP2 promotes p27Kip1 degradation and induces S phase in quiescent cells
    • Sutterluty H, Chatelain E, Marti A, et al. p45SKP2 promotes p27Kip1 degradation and induces S phase in quiescent cells. Nat Cell Biol 1999; 1:207-214.
    • (1999) Nat Cell Biol , vol.1 , pp. 207-214
    • Sutterluty, H.1    Chatelain, E.2    Marti, A.3
  • 37
    • 0032530151 scopus 로고    scopus 로고
    • Human CUL-1 associates with the SKP1/SKP2 complex and regulates p21(CIP1/WAF1) and cyclin D proteins
    • Yu ZK, Gervais JL, Zhang H. Human CUL-1 associates with the SKP1/SKP2 complex and regulates p21(CIP1/WAF1) and cyclin D proteins. Proc Natl Acad Sci USA 1998; 95:11324-11329.
    • (1998) Proc Natl Acad Sci USA , vol.95 , pp. 11324-11329
    • Yu, Z.K.1    Gervais, J.L.2    Zhang, H.3
  • 38
    • 84861552793 scopus 로고    scopus 로고
    • The Skp2-SCF E3 ligase regulates Akt ubiquitination, glycolysis, herceptin sensitivity, and tumorigenesis
    • Chan CH, Li CF, Yang WL, et al. The Skp2-SCF E3 ligase regulates Akt ubiquitination, glycolysis, herceptin sensitivity, and tumorigenesis. Cell 2012; 149:1098-1111.
    • (2012) Cell , vol.149 , pp. 1098-1111
    • Chan, C.H.1    Li, C.F.2    Yang, W.L.3
  • 39
    • 19444367346 scopus 로고    scopus 로고
    • Differential expression of the F-box proteins Skp2 and Skp2B in breast cancer
    • Radke S, Pirkmaier A, Germain D. Differential expression of the F-box proteins Skp2 and Skp2B in breast cancer. Oncogene 2005; 24:3448-3458.
    • (2005) Oncogene , vol.24 , pp. 3448-3458
    • Radke, S.1    Pirkmaier, A.2    Germain, D.3
  • 40
    • 21644448195 scopus 로고    scopus 로고
    • Correlation of Skp2 with carcinogenesis, invasion, metastasis, and prognosis in colorectal tumors
    • Li J-Q, Wu F, Masaki T, et al. Correlation of Skp2 with carcinogenesis, invasion, metastasis, and prognosis in colorectal tumors. Int J Oncol 2004; 25:87-95.
    • (2004) Int J Oncol , vol.25 , pp. 87-95
    • Li, J.-Q.1    Wu, F.2    Masaki, T.3
  • 41
    • 77950916977 scopus 로고    scopus 로고
    • Prognostic significance of S-phase kinase-associated protein 2 and p27kip1 in patients with diffuse large B-cell lymphoma: Effects of rituximab
    • Seki R, Ohshima K, Fujisaki T, et al. Prognostic significance of S-phase kinase-associated protein 2 and p27kip1 in patients with diffuse large B-cell lymphoma: effects of rituximab. Ann Oncol 2010; 21:833-841.
    • (2010) Ann Oncol , vol.21 , pp. 833-841
    • Seki, R.1    Ohshima, K.2    Fujisaki, T.3
  • 42
    • 0035942224 scopus 로고    scopus 로고
    • Skp2 is oncogenic and overexpressed in human cancers
    • Gstaiger M, Jordan R, Lim M, et al. Skp2 is oncogenic and overexpressed in human cancers. Proc Natl Acad Sci USA 2001; 98:5043-5048.
    • (2001) Proc Natl Acad Sci USA , vol.98 , pp. 5043-5048
    • Gstaiger, M.1    Jordan, R.2    Lim, M.3
  • 43
    • 75049085839 scopus 로고    scopus 로고
    • The expression and prognosis of FOXO3a and Skp2 in human hepatocellular carcinoma
    • Lu M, Ma J, Xue W, et al. The expression and prognosis of FOXO3a and Skp2 in human hepatocellular carcinoma. Pathol Oncol Res 2009; 15:679-687.
    • (2009) Pathol Oncol Res , vol.15 , pp. 679-687
    • Lu, M.1    Ma, J.2    Xue, W.3
  • 44
    • 84873540049 scopus 로고    scopus 로고
    • ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets
    • Souers AJ, Leverson JD, Boghaert ER, et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med 2013; 19:202-208.
    • (2013) Nat Med , vol.19 , pp. 202-208
    • Souers, A.J.1    Leverson, J.D.2    Boghaert, E.R.3
  • 45
    • 84871569969 scopus 로고    scopus 로고
    • Specific small molecule inhibitors of Skp2-mediated p27 degradation
    • Wu L, Grigoryan AV, Li Y, Hao B, Pagano M, Cardozo TJ. Specific small molecule inhibitors of Skp2-mediated p27 degradation. Chem Biol 2012; 19:1515-1524.
    • (2012) Chem Biol , vol.19 , pp. 1515-1524
    • Wu, L.1    Grigoryan, A.V.2    Li, Y.3    Hao, B.4    Pagano, M.5    Cardozo, T.J.6
  • 46
    • 84881192827 scopus 로고    scopus 로고
    • Pharmacological inactivation of Skp2 SCF ubiquitin ligase restricts cancer stem cell traits and cancer progression
    • Chan CH, Morrow JK, Li CF, et al. Pharmacological inactivation of Skp2 SCF ubiquitin ligase restricts cancer stem cell traits and cancer progression. Cell 2013; 154:556-568.
    • (2013) Cell , vol.154 , pp. 556-568
    • Chan, C.H.1    Morrow, J.K.2    Li, C.F.3
  • 47
    • 18344391432 scopus 로고    scopus 로고
    • Structure of the Cul1-Rbx1-Skp1-F boxSkp2 SCF ubiquitin ligase complex
    • Zheng N, Schulman BA, Song L, et al. Structure of the Cul1-Rbx1-Skp1-F boxSkp2 SCF ubiquitin ligase complex. Nature 2002; 416:703-709.
    • (2002) Nature , vol.416 , pp. 703-709
    • Zheng, N.1    Schulman, B.A.2    Song, L.3
  • 48
    • 0030941458 scopus 로고    scopus 로고
    • P53, the cellular gatekeeper for growth and division
    • Levine AJ. p53, the cellular gatekeeper for growth and division. Cell 1997; 88:323-331.
    • (1997) Cell , vol.88 , pp. 323-331
    • Levine, A.J.1
  • 49
    • 0026649648 scopus 로고
    • The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation
    • Momand J, Zambetti GP, Olson DC, George D, Levine AJ. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 1992; 69:1237-1245.
    • (1992) Cell , vol.69 , pp. 1237-1245
    • Momand, J.1    Zambetti, G.P.2    Olson, D.C.3    George, D.4    Levine, A.J.5
  • 50
    • 0030575937 scopus 로고    scopus 로고
    • Structure of the MDM2 oncoprotein bound to the p53 tumor suppressor transactivation domain
    • Kussie PH, Gorina S, Marechal V, et al. Structure of the MDM2 oncoprotein bound to the p53 tumor suppressor transactivation domain. Science 1996; 274:948-953.
    • (1996) Science , vol.274 , pp. 948-953
    • Kussie, P.H.1    Gorina, S.2    Marechal, V.3
  • 51
    • 0030905284 scopus 로고    scopus 로고
    • Mdm2 promotes the rapid degradation of p53
    • Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature 1997; 387:296-299.
    • (1997) Nature , vol.387 , pp. 296-299
    • Haupt, Y.1    Maya, R.2    Kazaz, A.3    Oren, M.4
  • 53
    • 0028172753 scopus 로고
    • Over-expression of the MDM2 gene is found in some cases of haematological malignancies
    • Quesnel B, Preudhomme C, Oscier D, et al. Over-expression of the MDM2 gene is found in some cases of haematological malignancies. Br J Haematol 1994; 88:415-418.
    • (1994) Br J Haematol , vol.88 , pp. 415-418
    • Quesnel, B.1    Preudhomme, C.2    Oscier, D.3
  • 54
    • 0029020147 scopus 로고
    • Amplification of the MDM2 gene in human breast cancer and its association with MDM2 and p53 protein status
    • McCann AH, Kirley A, Carney DN, et al. Amplification of the MDM2 gene in human breast cancer and its association with MDM2 and p53 protein status. Br J Cancer 1995; 71:981-985.
    • (1995) Br J Cancer , vol.71 , pp. 981-985
    • McCann, A.H.1    Kirley, A.2    Carney, D.N.3
  • 55
    • 10744221485 scopus 로고    scopus 로고
    • In vivo activation of the p53 pathway by small-molecule antagonists of MDM2
    • Vassilev LT, Vu BT, Graves B, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 2004; 303:844-848.
    • (2004) Science , vol.303 , pp. 844-848
    • Vassilev, L.T.1    Vu, B.T.2    Graves, B.3
  • 56
    • 41649102468 scopus 로고    scopus 로고
    • Temporal activation of p53 by a specific MDM2 inhibitor is selectively toxic to tumors and leads to complete tumor growth inhibition
    • Shangary S, Qin D, McEachern D, et al. Temporal activation of p53 by a specific MDM2 inhibitor is selectively toxic to tumors and leads to complete tumor growth inhibition. Proc Natl Acad Sci USA 2008; 105:3933-3938.
    • (2008) Proc Natl Acad Sci USA , vol.105 , pp. 3933-3938
    • Shangary, S.1    Qin, D.2    McEachern, D.3
  • 57
    • 11144315535 scopus 로고    scopus 로고
    • Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors
    • Issaeva N, Bozko P, Enge M, et al. Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors. Nat Med 2004; 10:1321-1328.
    • (2004) Nat Med , vol.10 , pp. 1321-1328
    • Issaeva, N.1    Bozko, P.2    Enge, M.3
  • 58
    • 36849035345 scopus 로고    scopus 로고
    • Ubiquitination and degradation of mutant p53
    • Lukashchuk N, Vousden KH. Ubiquitination and degradation of mutant p53. Mol Cell Biol 2007; 27:8284-8295.
    • (2007) Mol Cell Biol , vol.27 , pp. 8284-8295
    • Lukashchuk, N.1    Vousden, K.H.2
  • 59
    • 65449144050 scopus 로고    scopus 로고
    • PRIMA-1 reactivates mutant p53 by covalent binding to the core domain
    • Lambert JM, Gorzov P, Veprintsev DB, et al. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell 2009; 15:376-388.
    • (2009) Cancer Cell , vol.15 , pp. 376-388
    • Lambert, J.M.1    Gorzov, P.2    Veprintsev, D.B.3
  • 60
    • 0033082995 scopus 로고    scopus 로고
    • IAP family proteins-suppressors of apoptosis
    • Deveraux QL, Reed JC. IAP family proteins-suppressors of apoptosis. Genes Dev 1999; 13:239-252.
    • (1999) Genes Dev , vol.13 , pp. 239-252
    • Deveraux, Q.L.1    Reed, J.C.2
  • 61
  • 62
    • 84905014915 scopus 로고    scopus 로고
    • IAP family of cell death and signaling regulators
    • Ashkenazi A, Wells JA, Yuan JY, eds. Academic Press
    • Silke J, Vucic D. IAP family of cell death and signaling regulators. In: Ashkenazi A, Wells JA, Yuan JY, eds. Methods in Enzymology Vol 545. Academic Press, 2014: 35-65.
    • (2014) Methods in Enzymology , vol.545 , pp. 35-65
    • Silke, J.1    Vucic, D.2
  • 63
    • 84856495152 scopus 로고    scopus 로고
    • Targeting IAP proteins for therapeutic intervention in cancer
    • Fulda S, Vucic D. Targeting IAP proteins for therapeutic intervention in cancer. Nat Rev Drug Discov 2012; 11:109-124.
    • (2012) Nat Rev Drug Discov , vol.11 , pp. 109-124
    • Fulda, S.1    Vucic, D.2
  • 64
    • 0034616945 scopus 로고    scopus 로고
    • Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition
    • Du C, Fang M, Li Y, Li L, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000; 102:33-42.
    • (2000) Cell , vol.102 , pp. 33-42
    • Du, C.1    Fang, M.2    Li, Y.3    Li, L.4    Wang, X.5
  • 65
    • 0034616942 scopus 로고    scopus 로고
    • Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins
    • Verhagen AM, Ekert PG, Pakusch M, et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000; 102:43-53.
    • (2000) Cell , vol.102 , pp. 43-53
    • Verhagen, A.M.1    Ekert, P.G.2    Pakusch, M.3
  • 66
    • 36048999753 scopus 로고    scopus 로고
    • IAP antagonists induce autoubiquitination of c-IAPs, NF-kappaB activation, and TNFalpha-dependent apoptosis
    • Varfolomeev E, Blankenship JW, Wayson SM, et al. IAP antagonists induce autoubiquitination of c-IAPs, NF-kappaB activation, and TNFalpha-dependent apoptosis. Cell 2007; 131:669-681.
    • (2007) Cell , vol.131 , pp. 669-681
    • Varfolomeev, E.1    Blankenship, J.W.2    Wayson, S.M.3
  • 67
    • 80054837173 scopus 로고    scopus 로고
    • Antagonists induce a conformational change in cIAP1 that promotes autoubiquitination
    • Dueber EC, Schoeffler AJ, Lingel A, et al. Antagonists induce a conformational change in cIAP1 that promotes autoubiquitination. Science 2011; 334:376-380.
    • (2011) Science , vol.334 , pp. 376-380
    • Dueber, E.C.1    Schoeffler, A.J.2    Lingel, A.3
  • 68
    • 0036295676 scopus 로고    scopus 로고
    • Molecular regulation of muscle cachexia: It may be more than the proteasome
    • Hasselgren PO, Wray C, Mammen J. Molecular regulation of muscle cachexia: it may be more than the proteasome. Biochem Biophys Res Commun 2002; 290:1-10.
    • (2002) Biochem Biophys Res Commun , vol.290 , pp. 1-10
    • Hasselgren, P.O.1    Wray, C.2    Mammen, J.3
  • 69
    • 0036830917 scopus 로고    scopus 로고
    • Cachexia in cancer patients
    • Tisdale MJ. Cachexia in cancer patients. Nat Rev Cancer 2002; 2:862-871.
    • (2002) Nat Rev Cancer , vol.2 , pp. 862-871
    • Tisdale, M.J.1
  • 70
    • 2342667387 scopus 로고    scopus 로고
    • The development of proteasome inhibitors as anticancer drugs
    • Adams J. The development of proteasome inhibitors as anticancer drugs. Cancer Cell 2004; 5:417-421.
    • (2004) Cancer Cell , vol.5 , pp. 417-421
    • Adams, J.1
  • 71
    • 0035300479 scopus 로고    scopus 로고
    • The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells
    • Hideshima T, Richardson P, Chauhan D, et al. The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res 2001; 61:3071-3076.
    • (2001) Cancer Res , vol.61 , pp. 3071-3076
    • Hideshima, T.1    Richardson, P.2    Chauhan, D.3
  • 73
    • 0037441760 scopus 로고    scopus 로고
    • Molecular mechanisms mediating antimyeloma activity of proteasome inhibitor PS-341
    • Hideshima T, Richardson P, Chauhan D, et al. Molecular mechanisms mediating antimyeloma activity of proteasome inhibitor PS-341. Blood 2002; 101:1530-1534.
    • (2002) Blood , vol.101 , pp. 1530-1534
    • Hideshima, T.1    Richardson, P.2    Chauhan, D.3
  • 74
    • 41949136647 scopus 로고    scopus 로고
    • Bortezomib blocks Bax degradation in malignant B cells during treatment with TRAIL
    • Liu FT, Agrawal SG, Gribben JG, et al. Bortezomib blocks Bax degradation in malignant B cells during treatment with TRAIL. Blood 2007; 111:2797-2805.
    • (2007) Blood , vol.111 , pp. 2797-2805
    • Liu, F.T.1    Agrawal, S.G.2    Gribben, J.G.3
  • 75
    • 2542523228 scopus 로고    scopus 로고
    • Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors
    • Pei XY, Dai Y, Grant S. Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clin Cancer Res 2004; 10:3839-3852.
    • (2004) Clin Cancer Res , vol.10 , pp. 3839-3852
    • Pei, X.Y.1    Dai, Y.2    Grant, S.3
  • 77
    • 0033621047 scopus 로고    scopus 로고
    • Epoxomicin, a potent and selective proteasome inhibitor, exhibits in vivo antiinflammatory activity
    • Meng L, Mohan R, Kwok BH, Elofsson M, Sin N, Crews CM. Epoxomicin, a potent and selective proteasome inhibitor, exhibits in vivo antiinflammatory activity. Proc Natl Acad Sci USA 1999; 96:10403-10408.
    • (1999) Proc Natl Acad Sci USA , vol.96 , pp. 10403-10408
    • Meng, L.1    Mohan, R.2    Kwok, B.H.3    Elofsson, M.4    Sin, N.5    Crews, C.M.6
  • 78
    • 34447116376 scopus 로고    scopus 로고
    • Antitumor activity of PR-171, a novel irreversible inhibitor of the proteasome
    • Demo SD, Kirk CJ, Aujay MA, et al. Antitumor activity of PR-171, a novel irreversible inhibitor of the proteasome. Cancer Res 2007; 67:6383-6391.
    • (2007) Cancer Res , vol.67 , pp. 6383-6391
    • Demo, S.D.1    Kirk, C.J.2    Aujay, M.A.3
  • 79
    • 0343262654 scopus 로고    scopus 로고
    • Crystal structure of epoxomicin:20S proteasome reveals a molecular basis for selectivity of α',β'-epoxyketone proteasome inhibitors
    • Groll M, Kim KB, Kairies N, Huber R, Crews CM. Crystal structure of epoxomicin:20S proteasome reveals a molecular basis for selectivity of α',β'-epoxyketone proteasome inhibitors. J Am Chem Soc 2000; 122:1237-1238.
    • (2000) J Am Chem Soc , vol.122 , pp. 1237-1238
    • Groll, M.1    Kim, K.B.2    Kairies, N.3    Huber, R.4    Crews, C.M.5
  • 81
    • 80051691845 scopus 로고    scopus 로고
    • In vitro and in vivo selective antitumor activity of a novel orally ioavailable proteasome inhibitor MLN9708 against multiple myeloma cells
    • Chauhan D, Tian Z, Zhou B, et al. In vitro and in vivo selective antitumor activity of a novel orally ioavailable proteasome inhibitor MLN9708 against multiple myeloma cells. Clin Cancer Res 2011; 17:5311-5321.
    • (2011) Clin Cancer Res , vol.17 , pp. 5311-5321
    • Chauhan, D.1    Tian, Z.2    Zhou, B.3
  • 82
    • 84856103470 scopus 로고    scopus 로고
    • Novel, orally active, proteasome inhibitor, delanzomib (CEP-18770, ameliorates disease symptoms and glomerulonephritis in two preclinical mouse models of SLE
    • Seavey MM, Lu LD, Stump KL, Wallace NH, Ruggeri BA. Novel, orally active, proteasome inhibitor, delanzomib (CEP-18770, ameliorates disease symptoms and glomerulonephritis in two preclinical mouse models of SLE. Int Immunopharmacol 2012; 12:257-270.
    • (2012) Int Immunopharmacol , vol.12 , pp. 257-270
    • Seavey, M.M.1    Lu, L.D.2    Stump, K.L.3    Wallace, N.H.4    Ruggeri, B.A.5
  • 83
    • 59649114341 scopus 로고    scopus 로고
    • Inactivation of murine Usp1 results in genomic instability and a fanconi anemia phenotype
    • Kim JM, Parmar K, Huang M, et al. Inactivation of murine Usp1 results in genomic instability and a fanconi anemia phenotype. Dev Cell 2009; 16:314-320.
    • (2009) Dev Cell , vol.16 , pp. 314-320
    • Kim, J.M.1    Parmar, K.2    Huang, M.3
  • 84
    • 73849083434 scopus 로고    scopus 로고
    • Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival
    • Schwickart M, Huang X, Lill JR, et al. Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival. Nature 2010; 463:103-107.
    • (2010) Nature , vol.463 , pp. 103-107
    • Schwickart, M.1    Huang, X.2    Lill, J.R.3
  • 85
    • 79955977893 scopus 로고    scopus 로고
    • Deubiquitinase USP37 is activated by CDK2 to antagonize APC(CDH1) and promote S phase entry
    • Huang X, Summers MK, Pham V, et al. Deubiquitinase USP37 is activated by CDK2 to antagonize APC(CDH1) and promote S phase entry. Mol Cell 2011; 42:511-523.
    • (2011) Mol Cell , vol.42 , pp. 511-523
    • Huang, X.1    Summers, M.K.2    Pham, V.3
  • 86
    • 84937966446 scopus 로고    scopus 로고
    • USP37 directly deubiquitinates and stabilizes c-Myc in lung cancer
    • Pan J, Deng Q, Jiang C, et al. USP37 directly deubiquitinates and stabilizes c-Myc in lung cancer. Oncogene 2015; 34:3957-3967.
    • (2015) Oncogene , vol.34 , pp. 3957-3967
    • Pan, J.1    Deng, Q.2    Jiang, C.3
  • 87
    • 84898802930 scopus 로고    scopus 로고
    • Toward understanding ubiquitin-modifying enzymes: From pharmacological targeting to proteomics
    • Lill JR, Wertz IE. Toward understanding ubiquitin-modifying enzymes: from pharmacological targeting to proteomics. Trends Pharmacol Sci 2014; 35:187-207.
    • (2014) Trends Pharmacol Sci , vol.35 , pp. 187-207
    • Lill, J.R.1    Wertz, I.E.2
  • 88
    • 84860112066 scopus 로고    scopus 로고
    • Discovery of specific inhibitors of human USP7/HAUSP deubiquitinating enzyme
    • Reverdy C, Conrath S, Lopez R, et al. Discovery of specific inhibitors of human USP7/HAUSP deubiquitinating enzyme. Chem Biol 2012; 19:467-477.
    • (2012) Chem Biol , vol.19 , pp. 467-477
    • Reverdy, C.1    Conrath, S.2    Lopez, R.3
  • 89
    • 84866021069 scopus 로고    scopus 로고
    • A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance
    • Chauhan D, Tian Z, Nicholson B, et al. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell 2012; 22:345-358.
    • (2012) Cancer Cell , vol.22 , pp. 345-358
    • Chauhan, D.1    Tian, Z.2    Nicholson, B.3
  • 90
    • 84894555108 scopus 로고    scopus 로고
    • Regulated protein turnover: Snapshots of the proteasome in action
    • Bhattacharyya S, Yu H, Mim C, Matouschek A. Regulated protein turnover: snapshots of the proteasome in action. Nat Rev Mol Cell Biol 2014; 15:122-133.
    • (2014) Nat Rev Mol Cell Biol , vol.15 , pp. 122-133
    • Bhattacharyya, S.1    Yu, H.2    Mim, C.3    Matouschek, A.4
  • 91
    • 84856085129 scopus 로고    scopus 로고
    • Inhibition of proteasome deubiquitinating activity as a new cancer therapy
    • D'Arcy P, Brnjic S, Olofsson MH, et al. Inhibition of proteasome deubiquitinating activity as a new cancer therapy. Nat Med 2011; 17:1636-1640.
    • (2011) Nat Med , vol.17 , pp. 1636-1640
    • D'Arcy, P.1    Brnjic, S.2    Olofsson, M.H.3
  • 92
    • 77956527159 scopus 로고    scopus 로고
    • Enhancement of proteasome activity by a small-molecule inhibitor of USP14
    • Lee BH, Lee MJ, Park S, et al. Enhancement of proteasome activity by a small-molecule inhibitor of USP14. Nature 2010; 467:179-184.
    • (2010) Nature , vol.467 , pp. 179-184
    • Lee, B.H.1    Lee, M.J.2    Park, S.3
  • 94
    • 0035890987 scopus 로고    scopus 로고
    • Auxin regulates SCFTIR1-dependent degradation of AUX/IAA proteins
    • Gray WM, Kepinski S, Rouse D, Leyser O, Estelle M. Auxin regulates SCFTIR1-dependent degradation of AUX/IAA proteins. Nature 2001; 414:271-276.
    • (2001) Nature , vol.414 , pp. 271-276
    • Gray, W.M.1    Kepinski, S.2    Rouse, D.3    Leyser, O.4    Estelle, M.5
  • 95
    • 0031975589 scopus 로고    scopus 로고
    • The TIR1 protein of Arabidopsis functions in auxin response and is related to human SKP2 and yeast Grr1p
    • Ruegger M, Dewey E, Gray WM, Hobbie L, Turner J, Estelle M et al. The TIR1 protein of Arabidopsis functions in auxin response and is related to human SKP2 and yeast Grr1p. Genes Dev 1998; 12:198-207.
    • (1998) Genes Dev , vol.12 , pp. 198-207
    • Ruegger, M.1    Dewey, E.2    Gray, W.M.3    Hobbie, L.4    Turner, J.5    Estelle, M.6
  • 96
    • 78549274705 scopus 로고    scopus 로고
    • Jasmonate perception by inositol-phosphate-potentiated COI1-JAZ co-receptor
    • Sheard LB, Tan X, Mao H, et al. Jasmonate perception by inositol-phosphate-potentiated COI1-JAZ co-receptor. Nature 2010; 468:400-405.
    • (2010) Nature , vol.468 , pp. 400-405
    • Sheard, L.B.1    Tan, X.2    Mao, H.3
  • 97
    • 79955617241 scopus 로고    scopus 로고
    • RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling
    • Zhang Y, Liu S, Mickanin C, et al. RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling. Nat Cell Biol 2011; 13:623-629.
    • (2011) Nat Cell Biol , vol.13 , pp. 623-629
    • Zhang, Y.1    Liu, S.2    Mickanin, C.3
  • 98
    • 79960727271 scopus 로고    scopus 로고
    • Ubiquitin ligase RNF146 regulates tankyrase and axin to promote Wnt signaling
    • Callow MG, Tran H, Phu L, et al. Ubiquitin ligase RNF146 regulates tankyrase and axin to promote Wnt signaling. PLoS One 2011; 6:e22595.
    • (2011) PLoS One , vol.6
    • Callow, M.G.1    Tran, H.2    Phu, L.3
  • 99
    • 84920024622 scopus 로고    scopus 로고
    • Allosteric activation of the RNF146 ubiquitin ligase by a poly(ADP-ribosyl)ation signal
    • DaRosa PA, Wang Z2, Jiang X, et al. Allosteric activation of the RNF146 ubiquitin ligase by a poly(ADP-ribosyl)ation signal. Nature 2015; 517:223-226.
    • (2015) Nature , vol.517 , pp. 223-226
    • DaRosa, P.A.1    Wang, Z.2    Jiang, X.3
  • 100
    • 84877787623 scopus 로고    scopus 로고
    • Lenalidomide treatment for multiple myeloma: Systematic review and meta-analysis of randomized controlled trials
    • Yang B, Yu RL, Chi XH, Lu XC. Lenalidomide treatment for multiple myeloma: systematic review and meta-analysis of randomized controlled trials. PLoS One 2013; 8:e64354.
    • (2013) PLoS One , vol.8
    • Yang, B.1    Yu, R.L.2    Chi, X.H.3    Lu, X.C.4
  • 101
    • 77949350034 scopus 로고    scopus 로고
    • Identification of a primary target of thalidomide teratogenicity
    • Ito T, Ando H, Suzuki T, et al. Identification of a primary target of thalidomide teratogenicity. Science 2010; 327:1345-1350.
    • (2010) Science , vol.327 , pp. 1345-1350
    • Ito, T.1    Ando, H.2    Suzuki, T.3
  • 102
    • 84892593087 scopus 로고    scopus 로고
    • The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins
    • Lu G, Middleton RE, Sun H, et al. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science 2014; 343:305-309.
    • (2014) Science , vol.343 , pp. 305-309
    • Lu, G.1    Middleton, R.E.2    Sun, H.3
  • 103
    • 84892576029 scopus 로고    scopus 로고
    • Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells
    • Kronke J, Udeshi ND, Narla A, et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 2014; 343:301-305.
    • (2014) Science , vol.343 , pp. 301-305
    • Kronke, J.1    Udeshi, N.D.2    Narla, A.3
  • 104
    • 84893289821 scopus 로고    scopus 로고
    • A phase 2 trial of lenalidomide, bortezomib, and dexamethasone in patients with relapsed and relapsed/refractory myeloma
    • Richardson PG, Xie W, Jagannath S, et al. A phase 2 trial of lenalidomide, bortezomib, and dexamethasone in patients with relapsed and relapsed/refractory myeloma. Blood 2014; 123:1461-1469.
    • (2014) Blood , vol.123 , pp. 1461-1469
    • Richardson, P.G.1    Xie, W.2    Jagannath, S.3
  • 105
    • 73349115580 scopus 로고    scopus 로고
    • Multicenter, phase I, dose-escalation trial of lenalidomide plus bortezomib for relapsed and relapsed/refractory multiple myeloma
    • Richardson PG, Weller E, Jagannath S, et al. Multicenter, phase I, dose-escalation trial of lenalidomide plus bortezomib for relapsed and relapsed/refractory multiple myeloma. J Clin Oncol 2009; 27:5713-5719.
    • (2009) J Clin Oncol , vol.27 , pp. 5713-5719
    • Richardson, P.G.1    Weller, E.2    Jagannath, S.3
  • 106
    • 84905568369 scopus 로고    scopus 로고
    • Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide
    • Fischer ES, Böhm K, Lydeard JR, et al. Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide. Nature 2014; 512:49-53.
    • (2014) Nature , vol.512 , pp. 49-53
    • Fischer, E.S.1    Böhm, K.2    Lydeard, J.R.3
  • 107
    • 0035902475 scopus 로고    scopus 로고
    • PROTACs: Chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation
    • Sakamoto KM, Kim KB, Kumagai A, Mercurio F, Crews CM, Deshaies RJ. PROTACs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc Natl Acad Sci USA 2001; 98:8554-8559.
    • (2001) Proc Natl Acad Sci USA , vol.98 , pp. 8554-8559
    • Sakamoto, K.M.1    Kim, K.B.2    Kumagai, A.3    Mercurio, F.4    Crews, C.M.5    Deshaies, R.J.6
  • 108
    • 84916880505 scopus 로고    scopus 로고
    • Structure of the human Cereblon-DDB1-lenalidomide complex reveals basis for responsiveness to thalidomide analogs
    • Chamberlain PP, Lopez-Girona A, Miller K, et al. Structure of the human Cereblon-DDB1-lenalidomide complex reveals basis for responsiveness to thalidomide analogs. Nat Struct Mol Biol 2014; 21:803-809.
    • (2014) Nat Struct Mol Biol , vol.21 , pp. 803-809
    • Chamberlain, P.P.1    Lopez-Girona, A.2    Miller, K.3
  • 109
    • 78650847770 scopus 로고    scopus 로고
    • Selective inhibition of BET bromodomains
    • Filippakopoulos P, Qi J, Picaud S, et al. Selective inhibition of BET bromodomains. Nature 2010; 468:1067-1073.
    • (2010) Nature , vol.468 , pp. 1067-1073
    • Filippakopoulos, P.1    Qi, J.2    Picaud, S.3
  • 110
    • 80052955256 scopus 로고    scopus 로고
    • BET bromodomain inhibition as a therapeutic strategy to target c-Myc
    • Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 2011; 146:904-917.
    • (2011) Cell , vol.146 , pp. 904-917
    • Delmore, J.E.1    Issa, G.C.2    Lemieux, M.E.3
  • 111
    • 84932634729 scopus 로고    scopus 로고
    • Phthalimide conjugation as a strategy for in vivo target protein degradation
    • Winter GE, Buckley DL, Paulk J, et al. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 2015; 348:1376-1381.
    • (2015) Science , vol.348 , pp. 1376-1381
    • Winter, G.E.1    Buckley, D.L.2    Paulk, J.3
  • 112
    • 84931560527 scopus 로고    scopus 로고
    • Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4
    • Lu J, Qian Y, Altieri M, et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem Biol 2015; 22:755-763.
    • (2015) Chem Biol , vol.22 , pp. 755-763
    • Lu, J.1    Qian, Y.2    Altieri, M.3
  • 113
    • 84939794726 scopus 로고    scopus 로고
    • HaloPROTACS: Use of small molecule PROTACs to induce degradation of HaloTag fusion proteins
    • Buckley DL, Raina K, Darricarrere N, et al. HaloPROTACS: use of small molecule PROTACs to induce degradation of HaloTag fusion proteins. ACS Chem Biol 2015; 10:1831-1837.
    • (2015) ACS Chem Biol , vol.10 , pp. 1831-1837
    • Buckley, D.L.1    Raina, K.2    Darricarrere, N.3
  • 114
    • 84939788143 scopus 로고    scopus 로고
    • Selective small molecule induced degradation of the BET bromodomain protein BRD4
    • Zengerle M, Chan KH, Ciulli A. Selective small molecule induced degradation of the BET bromodomain protein BRD4. ACS Chem Biol 2015; 10:1770-1777.
    • (2015) ACS Chem Biol , vol.10 , pp. 1770-1777
    • Zengerle, M.1    Chan, K.H.2    Ciulli, A.3
  • 115
    • 84937514576 scopus 로고    scopus 로고
    • Catalytic in vivo protein knockdown by small-molecule PROTACs
    • Bondeson DP, Mares A, Smith IE, et al. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat Chem Biol 2015; 11:611-617.
    • (2015) Nat Chem Biol , vol.11 , pp. 611-617
    • Bondeson, D.P.1    Mares, A.2    Smith, I.E.3
  • 116
    • 0015859467 scopus 로고
    • Principles that govern the folding of protein chains
    • Anfinsen CB. Principles that govern the folding of protein chains. Science 1973; 181:223-230.
    • (1973) Science , vol.181 , pp. 223-230
    • Anfinsen, C.B.1
  • 117
    • 0030059689 scopus 로고    scopus 로고
    • Forces contributing to the conformational stability of proteins
    • Pace CN, Shirley BA, McNutt M, Gajiwala K. Forces contributing to the conformational stability of proteins. FASEB J 1996; 10:75-83.
    • (1996) FASEB J , vol.10 , pp. 75-83
    • Pace, C.N.1    Shirley, B.A.2    McNutt, M.3    Gajiwala, K.4
  • 118
    • 0029002720 scopus 로고
    • The hydrophobic effect in protein folding
    • Lins L, Brasseur R. The hydrophobic effect in protein folding. FASEB J 1995; 9:535-540.
    • (1995) FASEB J , vol.9 , pp. 535-540
    • Lins, L.1    Brasseur, R.2
  • 119
    • 79960562309 scopus 로고    scopus 로고
    • Small-molecule hydrophobic tagging-induced degradation of HaloTag fusion proteins
    • Neklesa TK, Tae HS, Schneekloth AR, et al. Small-molecule hydrophobic tagging-induced degradation of HaloTag fusion proteins. Nat Chem Biol 2011; 7:538-543.
    • (2011) Nat Chem Biol , vol.7 , pp. 538-543
    • Neklesa, T.K.1    Tae, H.S.2    Schneekloth, A.R.3
  • 120
    • 23144443884 scopus 로고    scopus 로고
    • Protein quality control: Chaperones culling corrupt conformations
    • McClellan AJ, Tam S, Kaganovich D, Frydman J. Protein quality control: chaperones culling corrupt conformations. Nat Cell Biol 2005; 7:736-741.
    • (2005) Nat Cell Biol , vol.7 , pp. 736-741
    • McClellan, A.J.1    Tam, S.2    Kaganovich, D.3    Frydman, J.4
  • 121
    • 84934299225 scopus 로고    scopus 로고
    • Targeted protein destabilization reveals an estrogen-mediated ER stress response
    • Raina K, Noblin DJ, Serebrenik YV, et al. Targeted protein destabilization reveals an estrogen-mediated ER stress response. Nat Chem Biol 2014; 10:957-962.
    • (2014) Nat Chem Biol , vol.10 , pp. 957-962
    • Raina, K.1    Noblin, D.J.2    Serebrenik, Y.V.3
  • 122
    • 84920590896 scopus 로고    scopus 로고
    • Pharmacological targeting of the pseudokinase Her3
    • Xie T, Lim SM, Westover KD, et al. Pharmacological targeting of the pseudokinase Her3. Nat Chem Biol 2014; 10:1006-1012.
    • (2014) Nat Chem Biol , vol.10 , pp. 1006-1012
    • Xie, T.1    Lim, S.M.2    Westover, K.D.3
  • 123
  • 124
    • 0037501319 scopus 로고    scopus 로고
    • The estrogen receptor: A model for molecular medicine
    • Jensen EV, Jordan VC. The estrogen receptor: a model for molecular medicine. Clin Cancer Res 2003; 9:1980-1989.
    • (2003) Clin Cancer Res , vol.9 , pp. 1980-1989
    • Jensen, E.V.1    Jordan, V.C.2
  • 125
    • 84888391516 scopus 로고    scopus 로고
    • ESR1 ligand-binding domain mutations in hormone-resistant breast cancer
    • Toy W, Shen Y, Won H, et al. ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat Genet 2013; 45:1439-1445.
    • (2013) Nat Genet , vol.45 , pp. 1439-1445
    • Toy, W.1    Shen, Y.2    Won, H.3
  • 126
    • 84888381937 scopus 로고    scopus 로고
    • Activating ESR1 mutations in hormone-resistant metastatic breast cancer
    • Robinson DR, Wu YM, Vats P, et al. Activating ESR1 mutations in hormone-resistant metastatic breast cancer. Nat Genet 2013; 45:1446-1451.
    • (2013) Nat Genet , vol.45 , pp. 1446-1451
    • Robinson, D.R.1    Wu, Y.M.2    Vats, P.3
  • 128
    • 0026395885 scopus 로고
    • A potent specific pure antiestrogen with clinical potential
    • Wakeling AE, Dukes M, Bowler J. A potent specific pure antiestrogen with clinical potential. Cancer Res 1991; 51:3867-3873.
    • (1991) Cancer Res , vol.51 , pp. 3867-3873
    • Wakeling, A.E.1    Dukes, M.2    Bowler, J.3
  • 129
    • 84933059419 scopus 로고    scopus 로고
    • Identification of GDC-0810 (ARN-810), an orally bioavailable selective estrogen receptor degrader (SERD) that demonstrates robust activity in tamoxifen-resistant breast cancer xenografts
    • Lai A, Kahraman M, Govek S, et al. Identification of GDC-0810 (ARN-810), an orally bioavailable selective estrogen receptor degrader (SERD) that demonstrates robust activity in tamoxifen-resistant breast cancer xenografts. J Med Chem 2015; 58:4888-4904.
    • (2015) J Med Chem , vol.58 , pp. 4888-4904
    • Lai, A.1    Kahraman, M.2    Govek, S.3
  • 130
    • 0032446607 scopus 로고    scopus 로고
    • The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen
    • Shiau AK, Barstad D, Loria PM, et al. The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell 1998; 95:927-937.
    • (1998) Cell , vol.95 , pp. 927-937
    • Shiau, A.K.1    Barstad, D.2    Loria, P.M.3
  • 131
    • 0036234964 scopus 로고    scopus 로고
    • Structural characterization of a subtype-selective ligand reveals a novel mode of estrogen receptor antagonism
    • Shiau AK, Barstad D, Radek JT, et al. Structural characterization of a subtype-selective ligand reveals a novel mode of estrogen receptor antagonism. Nat Struct Mol Biol 2002; 9:359-364.
    • (2002) Nat Struct Mol Biol , vol.9 , pp. 359-364
    • Shiau, A.K.1    Barstad, D.2    Radek, J.T.3
  • 132
    • 18944381947 scopus 로고    scopus 로고
    • Structural basis for an unexpected mode of SERM-mediated ER antagonism
    • Wu YL, Yang X, Ren Z, et al. Structural basis for an unexpected mode of SERM-mediated ER antagonism. Mol Cell 2005; 18:413-424.
    • (2005) Mol Cell , vol.18 , pp. 413-424
    • Wu, Y.L.1    Yang, X.2    Ren, Z.3
  • 133
    • 84873085753 scopus 로고    scopus 로고
    • A strategy for modulation of enzymes in the ubiquitin system
    • Ernst A, Avvakumov G, Tong J, et al. A strategy for modulation of enzymes in the ubiquitin system. Science 2013; 339:590-595.
    • (2013) Science , vol.339 , pp. 590-595
    • Ernst, A.1    Avvakumov, G.2    Tong, J.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.