메뉴 건너뛰기




Volumn 55, Issue , 2015, Pages 35-54

Predicting toxicities of reactive metabolite-positive drug candidates

Author keywords

ADRs; Adverse drug reactions; Bioactivation; Covalent binding; Cytochrome P450; Electrophile; Glutathione; Hepatotoxicity; Idiosyncratic; Liver microsomes; Precision medicine; Prediction in pharmacology; Structural alert

Indexed keywords

DRUG METABOLITE; DRUG;

EID: 84920848626     PISSN: 03621642     EISSN: 15454304     Source Type: Book Series    
DOI: 10.1146/annurev-pharmtox-010814-124720     Document Type: Review
Times cited : (91)

References (77)
  • 1
    • 84862278754 scopus 로고    scopus 로고
    • Mechanism-based inactivation (MBI) of cytochrome P450 enzymes: Structure-activity relationships and discovery strategies to mitigate drug-drug interaction risks
    • Orr ST, Ripp SL, Ballard TE, Henderson JL, Scott DO, et al. 2012. Mechanism-based inactivation (MBI) of cytochrome P450 enzymes: structure-activity relationships and discovery strategies to mitigate drug-drug interaction risks. J. Med. Chem. 55:4896-933
    • (2012) J. Med. Chem. , vol.55 , pp. 4896-4933
    • Orr, S.T.1    Ripp, S.L.2    Ballard, T.E.3    Henderson, J.L.4    Scott, D.O.5
  • 3
    • 12244284627 scopus 로고    scopus 로고
    • Minimizing the potential for metabolic activation as an integral part of drug design
    • Evans DC, Baillie TA. 2005. Minimizing the potential for metabolic activation as an integral part of drug design. Curr. Opin. Drug Discov. Dev. 8:44-50
    • (2005) Curr. Opin. Drug Discov. Dev. , vol.8 , pp. 44-50
    • Evans, D.C.1    Baillie, T.A.2
  • 4
    • 34047266790 scopus 로고    scopus 로고
    • Applying mechanisms of chemical toxicity to predict drug safety
    • Guengerich FP, MacDonald JS. 2007. Applying mechanisms of chemical toxicity to predict drug safety. Chem. Res. Toxicol. 20:344-69
    • (2007) Chem. Res. Toxicol. , vol.20 , pp. 344-369
    • Guengerich, F.P.1    Macdonald, J.S.2
  • 5
    • 79953314978 scopus 로고    scopus 로고
    • Managing the challenge of chemically reactive metabolites in drug development
    • Park BK, Boobis A, Clarke S, Goldring CE, Jones D, et al. 2011. Managing the challenge of chemically reactive metabolites in drug development. Nat. Rev. Drug Discov. 10:292-306
    • (2011) Nat. Rev. Drug Discov. , vol.10 , pp. 292-306
    • Park, B.K.1    Boobis, A.2    Clarke, S.3    Goldring, C.E.4    Jones, D.5
  • 7
    • 34250002084 scopus 로고    scopus 로고
    • HLA-B5701 screening for susceptibility to abacavir hypersensitivity
    • Lucas A, Nolan D, Mallal S. 2007. HLA-B5701 screening for susceptibility to abacavir hypersensitivity. J. Antimicrob. Chemother. 59:591-93
    • (2007) J. Antimicrob. Chemother. , vol.59 , pp. 591-593
    • Lucas, A.1    Nolan, D.2    Mallal, S.3
  • 8
    • 77955082302 scopus 로고    scopus 로고
    • A genome-wide study identifies HLA alleles associated with lumiracoxib-related liver injury
    • Singer JB, Lewitzky S, Leroy E, Yang F, Zhao X, et al. 2010. A genome-wide study identifies HLA alleles associated with lumiracoxib-related liver injury. Nat. Genet. 42:711-14
    • (2010) Nat. Genet. , vol.42 , pp. 711-714
    • Singer, J.B.1    Lewitzky, S.2    Leroy, E.3    Yang, F.4    Zhao, X.5
  • 9
    • 84866406306 scopus 로고    scopus 로고
    • Clinical perspectives on human genetic screening to prevent nevirapine toxicity
    • Nevirapine Toxicogenomics Study Team
    • Haas DW, Mootsikapun P, Ruxrungtham K, Podzamczer D, Nevirapine Toxicogenomics Study Team. 2012. Clinical perspectives on human genetic screening to prevent nevirapine toxicity. Pers. Med. 9:773-82
    • (2012) Pers. Med. , vol.9 , pp. 773-782
    • Haas, D.W.1    Mootsikapun, P.2    Ruxrungtham, K.3    Podzamczer, D.4
  • 10
    • 84880763009 scopus 로고    scopus 로고
    • Idiosyncratic adverse drug reactions: Current concepts
    • Uetrecht J, Naisbitt DJ. 2013. Idiosyncratic adverse drug reactions: current concepts. Pharmacol. Rev. 65:779-808
    • (2013) Pharmacol. Rev. , vol.65 , pp. 779-808
    • Uetrecht, J.1    Naisbitt, D.J.2
  • 13
    • 1642281756 scopus 로고    scopus 로고
    • Drug-protein adducts: An industry perspective on minimizing the potential for drug bioactivation in drug discovery and development
    • Evans DC, Watt AP, Nicoll-Griffith DA, Baillie TA. 2004. Drug-protein adducts: an industry perspective on minimizing the potential for drug bioactivation in drug discovery and development. Chem. Res. Toxicol. 17:3-16
    • (2004) Chem. Res. Toxicol. , vol.17 , pp. 3-16
    • Evans, D.C.1    Watt, A.P.2    Nicoll-Griffith, D.A.3    Baillie, T.A.4
  • 14
    • 79551679354 scopus 로고    scopus 로고
    • Strategies and chemical design approaches to reduce the potential for formation of reactive metabolic species
    • Argikar UA, Mangold JB, Harriman SP. 2011. Strategies and chemical design approaches to reduce the potential for formation of reactive metabolic species. Curr. Top. Med. Chem. 11:419-49
    • (2011) Curr. Top. Med. Chem. , vol.11 , pp. 419-449
    • Argikar, U.A.1    Mangold, J.B.2    Harriman, S.P.3
  • 16
    • 80053004364 scopus 로고    scopus 로고
    • Structural alert/reactive metabolite concept as applied in medicinal chemistry to mitigate the risk of idiosyncratic drug toxicity: A perspective based on the critical examination of trends in the top 200 drugs marketed in the United States
    • Stepan AF, Walker DP, Bauman J, Price DA, Baillie TA, et al. 2011. Structural alert/reactive metabolite concept as applied in medicinal chemistry to mitigate the risk of idiosyncratic drug toxicity: A perspective based on the critical examination of trends in the top 200 drugs marketed in the United States. Chem. Res. Toxicol. 24:1345-410
    • (2011) Chem. Res. Toxicol. , vol.24 , pp. 1345-1410
    • Stepan, A.F.1    Walker, D.P.2    Bauman, J.3    Price, D.A.4    Baillie, T.A.5
  • 17
    • 84867534923 scopus 로고    scopus 로고
    • Theoretical studies of chemical reactivity ofmetabolically activated forms of aromatic amines towardDNA
    • Shamovsky I, Ripa L, Blomberg N, Eriksson LA, Hansen P, et al. 2012. Theoretical studies of chemical reactivity ofmetabolically activated forms of aromatic amines towardDNA. Chem. Res. Toxicol. 25:2236-52
    • (2012) Chem. Res. Toxicol. , vol.25 , pp. 2236-2252
    • Shamovsky, I.1    Ripa, L.2    Blomberg, N.3    Eriksson, L.A.4    Hansen, P.5
  • 18
    • 0029555342 scopus 로고
    • Clozapine is oxidized by activated human neutrophils to a reactive nitrenium ion that irreversibly binds to the cells
    • Liu ZC, Uetrecht JP. 1995. Clozapine is oxidized by activated human neutrophils to a reactive nitrenium ion that irreversibly binds to the cells. J. Pharmacol. Exp. Ther. 275:1476-83
    • (1995) J. Pharmacol. Exp. Ther. , vol.275 , pp. 1476-1483
    • Liu, Z.C.1    Uetrecht, J.P.2
  • 19
    • 0031814439 scopus 로고    scopus 로고
    • A comparison of the covalent binding of clozapine and olanzapine to human neutrophils in vitro and in vivo
    • Gardner I, Leeder JS, Chin T, Zahid N, Uetrecht JP. 1998. A comparison of the covalent binding of clozapine and olanzapine to human neutrophils in vitro and in vivo. Mol. Pharmacol. 53:999-1008
    • (1998) Mol. Pharmacol. , vol.53 , pp. 999-1008
    • Gardner, I.1    Leeder, J.S.2    Chin, T.3    Zahid, N.4    Uetrecht, J.P.5
  • 20
    • 0030913557 scopus 로고    scopus 로고
    • Structural features associated with reactive metabolite formation in clozapine analogues
    • Uetrecht J, Zahid N, Tehim A, Fu JM, Rakhit S. 1997. Structural features associated with reactive metabolite formation in clozapine analogues. Chem. Biol. Interact. 104:117-29
    • (1997) Chem. Biol. Interact. , vol.104 , pp. 117-129
    • Uetrecht, J.1    Zahid, N.2    Tehim, A.3    Fu, J.M.4    Rakhit, S.5
  • 21
    • 20844454730 scopus 로고    scopus 로고
    • Bioactivation of the nontricyclic antidepressant nefazodone to a reactive quinone-imine species in human liver microsomes and recombinant cytochrome P450 3A4
    • Kalgutkar AS, Vaz AD, Lame ME, Henne KR, Soglia J, et al. 2005. Bioactivation of the nontricyclic antidepressant nefazodone to a reactive quinone-imine species in human liver microsomes and recombinant cytochrome P450 3A4. Drug Metab. Dispos. 33:243-53
    • (2005) Drug Metab. Dispos. , vol.33 , pp. 243-253
    • Kalgutkar, A.S.1    Vaz, A.D.2    Lame, M.E.3    Henne, K.R.4    Soglia, J.5
  • 23
    • 53549107196 scopus 로고    scopus 로고
    • In vitro metabolism and covalent binding of enol-carboxamide derivatives and anti-inflammatory agents sudoxicam and meloxicam: Insights into the hepatotoxicity of sudoxicam
    • Obach RS, Kalgutkar AS, Ryder TF, Walker GS. 2008. In vitro metabolism and covalent binding of enol-carboxamide derivatives and anti-inflammatory agents sudoxicam and meloxicam: insights into the hepatotoxicity of sudoxicam. Chem. Res. Toxicol. 21:1890-99
    • (2008) Chem. Res. Toxicol. , vol.21 , pp. 1890-1899
    • Obach, R.S.1    Kalgutkar, A.S.2    Ryder, T.F.3    Walker, G.S.4
  • 24
    • 0032994274 scopus 로고    scopus 로고
    • Clinical pharmacokinetics of meloxicam: A cyclo-oxygenase-2 preferential nonsteroidal anti-inflammatory drug
    • Davies NM, Skjodt NM. 1999. Clinical pharmacokinetics of meloxicam: A cyclo-oxygenase-2 preferential nonsteroidal anti-inflammatory drug. Clin. Pharmacokinet. 36:115-26
    • (1999) Clin. Pharmacokinet. , vol.36 , pp. 115-126
    • Davies, N.M.1    Skjodt, N.M.2
  • 26
    • 77955246397 scopus 로고    scopus 로고
    • Uptake of pramipexole by human organic cation transporters
    • Diao L, Shu Y, Polli JE. 2010. Uptake of pramipexole by human organic cation transporters. Mol. Pharm. 7:1342-47
    • (2010) Mol. Pharm. , vol.7 , pp. 1342-1347
    • Diao, L.1    Shu, Y.2    Polli, J.E.3
  • 28
    • 0033012622 scopus 로고    scopus 로고
    • Quantification in patient urine samples of felbamate and three metabolites: Acid carbamate and two mercapturic acids
    • Thompson CD, Barthen MT, Hopper DW, Miller TA, Quigg M, et al. 1999. Quantification in patient urine samples of felbamate and three metabolites: acid carbamate and two mercapturic acids. Epilepsia 40:769-76
    • (1999) Epilepsia , vol.40 , pp. 769-776
    • Thompson, C.D.1    Barthen, M.T.2    Hopper, D.W.3    Miller, T.A.4    Quigg, M.5
  • 29
    • 34548473336 scopus 로고    scopus 로고
    • The direct thrombin inhibitor ximelagatran/melagatran: A systematic review on clinical applications and an evidence based assessment of risk benefit profile
    • Testa L, Bhindi R, Agostoni P, Abbate A, Zoccai GG, van Gaal WJ. 2007. The direct thrombin inhibitor ximelagatran/melagatran: A systematic review on clinical applications and an evidence based assessment of risk benefit profile. Exp. Opin. Drug Saf. 6:397-406
    • (2007) Exp. Opin. Drug Saf. , vol.6 , pp. 397-406
    • Testa, L.1    Bhindi, R.2    Agostoni, P.3    Abbate, A.4    Zoccai, G.G.5    Van Gaal, W.J.6
  • 30
    • 0028057319 scopus 로고
    • A comparison of the efficacy and toxic effects of sustained- versus immediate-release niacin in hypercholesterolemic patients
    • McKenney JM, Proctor JD, Harris S, Chinchili VM. 1994. A comparison of the efficacy and toxic effects of sustained- versus immediate-release niacin in hypercholesterolemic patients. JAMA 271:672-77
    • (1994) JAMA , vol.271 , pp. 672-677
    • McKenney, J.M.1    Proctor, J.D.2    Harris, S.3    Chinchili, V.M.4
  • 31
    • 0642344305 scopus 로고    scopus 로고
    • Overview of niacin formulations: Differences in pharmacokinetics, efficacy, and safety
    • Pieper JA. 2003. Overview of niacin formulations: differences in pharmacokinetics, efficacy, and safety. Am. J. Health Syst. Pharm. 60:S9-14
    • (2003) Am. J. Health Syst. Pharm. , vol.60 , pp. S9-14
    • Pieper, J.A.1
  • 33
    • 72249097682 scopus 로고    scopus 로고
    • A strategy to minimize reactive metabolite formation: Discovery of (S)-4-(1-cyclopropyl-2-methoxyethyl)-6-[6-(difluoromethoxy)- 2, 5-dimethylpyridin-3-ylamino]-5-oxo-4, 5-dihydropyrazine-2-carbonitrile as a potent, orally bioavailable corticotropin-releasing factor-1 receptor antagonist
    • Hartz RA, Ahuja VT, Zhuo X, Mattson RJ, Denhart DJ, et al. 2009. A strategy to minimize reactive metabolite formation: discovery of (S)-4-(1-cyclopropyl-2-methoxyethyl)-6-[6-(difluoromethoxy)- 2, 5-dimethylpyridin-3-ylamino]-5-oxo-4, 5-dihydropyrazine-2-carbonitrile as a potent, orally bioavailable corticotropin-releasing factor-1 receptor antagonist. J. Med. Chem. 52:7653-68
    • (2009) J. Med. Chem. , vol.52 , pp. 7653-7668
    • Hartz, R.A.1    Ahuja, V.T.2    Zhuo, X.3    Mattson, R.J.4    Denhart, D.J.5
  • 34
    • 77949407166 scopus 로고    scopus 로고
    • Cytochrome P450-mediated epoxidation of 2-aminothiazole-based AKT inhibitors: Identification of novel GSH adducts and reduction of metabolic activation through structural changes guided by in silico and in vitro screening
    • Subramanian R, Lee MR, Allen JG, Bourbeau MP, Fotsch C, et al. 2010. Cytochrome P450-mediated epoxidation of 2-aminothiazole-based AKT inhibitors: identification of novel GSH adducts and reduction of metabolic activation through structural changes guided by in silico and in vitro screening. Chem. Res. Toxicol. 23:653-63
    • (2010) Chem. Res. Toxicol. , vol.23 , pp. 653-663
    • Subramanian, R.1    Lee, M.R.2    Allen, J.G.3    Bourbeau, M.P.4    Fotsch, C.5
  • 35
    • 79951831048 scopus 로고    scopus 로고
    • Intrinsic electrophilicity of a 4-substituted-5-cyano-6-(2-methylpyridin-3-yloxy)pyrimidine derivative: Structural characterization of glutathione conjugates in vitro
    • Kalgutkar AS, Mascitti V, Sharma R, Walker GW, Ryder T, et al. 2011. Intrinsic electrophilicity of a 4-substituted-5-cyano-6-(2-methylpyridin-3-yloxy)pyrimidine derivative: structural characterization of glutathione conjugates in vitro. Chem. Res. Toxicol. 24:269-78
    • (2011) Chem. Res. Toxicol. , vol.24 , pp. 269-278
    • Kalgutkar, A.S.1    Mascitti, V.2    Sharma, R.3    Walker, G.W.4    Ryder, T.5
  • 36
    • 84879103355 scopus 로고    scopus 로고
    • Reactive metabolite trapping studies on imidazo- and 2-methylimidazo[2, 1-b]thiazole-based inverse agonists of the ghrelin receptor
    • Kalgutkar AR, Ryder TF, Walker GS, Orr ST, Cabral S, et al. 2013. Reactive metabolite trapping studies on imidazo- and 2-methylimidazo[2, 1-b]thiazole-based inverse agonists of the ghrelin receptor. Drug Metab. Dispos. 41:1375-88
    • (2013) Drug Metab. Dispos. , vol.41 , pp. 1375-1388
    • Kalgutkar, A.R.1    Ryder, T.F.2    Walker, G.S.3    Orr, S.T.4    Cabral, S.5
  • 37
    • 52449108446 scopus 로고    scopus 로고
    • The discovery of taranabant, a selective cannabinoid-1 receptor inverse agonist for the treatment of obesity
    • Hagmann WK. 2008. The discovery of taranabant, a selective cannabinoid-1 receptor inverse agonist for the treatment of obesity. Arch. Pharm. 341:405-11
    • (2008) Arch. Pharm. , vol.341 , pp. 405-411
    • Hagmann, W.K.1
  • 38
    • 84866927400 scopus 로고    scopus 로고
    • Discovery of piragliatin-first glucokinase activator studied in type 2 diabetic patients
    • Sarabu R, Bizzarro FT, Corbett WL, Dvorozniak MT, Geng W, et al. 2012. Discovery of piragliatin-first glucokinase activator studied in type 2 diabetic patients. J. Med. Chem. 55:7021-36
    • (2012) J. Med. Chem. , vol.55 , pp. 7021-7036
    • Sarabu, R.1    Bizzarro, F.T.2    Corbett, W.L.3    Dvorozniak, M.T.4    Geng, W.5
  • 39
    • 0242383181 scopus 로고    scopus 로고
    • Clinical pharmacokinetics of atorvastatin
    • Lennernas H. 2003. Clinical pharmacokinetics of atorvastatin. Clin. Pharmacokinet. 42:1141-60
    • (2003) Clin. Pharmacokinet. , vol.42 , pp. 1141-1160
    • Lennernas, H.1
  • 40
    • 70349103856 scopus 로고    scopus 로고
    • A zone classification system for risk assessment of idiosyncratic drug toxicity using daily dose and covalent binding
    • Nakayama S, Atsumi R, Takakusa H, Kobayashi Y, Kurihara A, et al. 2009. A zone classification system for risk assessment of idiosyncratic drug toxicity using daily dose and covalent binding. Drug Metab. Dispos. 37:1970-77
    • (2009) Drug Metab. Dispos. , vol.37 , pp. 1970-1977
    • Nakayama, S.1    Atsumi, R.2    Takakusa, H.3    Kobayashi, Y.4    Kurihara, A.5
  • 41
    • 62249112083 scopus 로고    scopus 로고
    • Metabolic oxidative cleavage of thioesters: Evidence for the formation of sulfenic acid intermediates in the bioactivation of the antithrombotic prodrugs ticlopidine and clopidogrel
    • Dansette PM, Libraire J, Bertho G, Mansuy D. 2009. Metabolic oxidative cleavage of thioesters: evidence for the formation of sulfenic acid intermediates in the bioactivation of the antithrombotic prodrugs ticlopidine and clopidogrel. Chem. Res. Toxicol. 22:369-73
    • (2009) Chem. Res. Toxicol. , vol.22 , pp. 369-373
    • Dansette, P.M.1    Libraire, J.2    Bertho, G.3    Mansuy, D.4
  • 42
    • 33746659807 scopus 로고    scopus 로고
    • The active metabolite of clopidogrel disrupts P2Y12 receptor oligomers and partitions them out of lipid rafts
    • Savi P, Zachayus JL, Delesque-Touchard N, Labouret C, Herve C, et al. 2006. The active metabolite of clopidogrel disrupts P2Y12 receptor oligomers and partitions them out of lipid rafts. Proc. Natl. Acad. Sci. USA 103:11069-74
    • (2006) Proc. Natl. Acad. Sci. USA , vol.103 , pp. 11069-11074
    • Savi, P.1    Zachayus, J.L.2    Delesque-Touchard, N.3    Labouret, C.4    Herve, C.5
  • 43
    • 77955676570 scopus 로고    scopus 로고
    • Characterization of glutathione conjugates of duloxetine by mass spectrometry and evaluation of in silico approaches to rationalize the site of conjugation for thiophene containing drugs
    • Wu G, Vashishtha SC, Erve JC. 2010. Characterization of glutathione conjugates of duloxetine by mass spectrometry and evaluation of in silico approaches to rationalize the site of conjugation for thiophene containing drugs. Chem. Res. Toxicol. 23:1393-404
    • (2010) Chem. Res. Toxicol. , vol.23 , pp. 1393-1404
    • Wu, G.1    Vashishtha, S.C.2    Erve, J.C.3
  • 44
    • 0024435503 scopus 로고
    • Metabolic pathways of paroxetine in animals andman and the comparative pharmacological properties of the metabolites
    • Haddock RE, Johnson AM, Langley PF, Nelson DR, Pope JA, et al. 1989. Metabolic pathways of paroxetine in animals andman and the comparative pharmacological properties of the metabolites. Acta Psychiatr. Scand. Suppl. 350:24-26
    • (1989) Acta Psychiatr. Scand. Suppl. , vol.350 , pp. 24-26
    • Haddock, R.E.1    Johnson, A.M.2    Langley, P.F.3    Nelson, D.R.4    Pope, J.A.5
  • 45
    • 18844426008 scopus 로고    scopus 로고
    • In vitro-in vivo extrapolation of CYP2D6 inactivation by paroxetine: Prediction of nonstationary pharmacokinetics and drug interaction magnitude
    • Venkatakrishnan K, Obach RS. 2005. In vitro-in vivo extrapolation of CYP2D6 inactivation by paroxetine: prediction of nonstationary pharmacokinetics and drug interaction magnitude. Drug Metab. Dispos. 33:845-52
    • (2005) Drug Metab. Dispos. , vol.33 , pp. 845-852
    • Venkatakrishnan, K.1    Obach, R.S.2
  • 46
    • 37249018714 scopus 로고    scopus 로고
    • NADPH-dependent covalent binding of [3H]paroxetine to human liver microsomes and S-9 fractions: Identification of an electrophilic quinone metabolite of paroxetine
    • Zhao SX, Dalvie DK, Kelly JM, Soglia JR, Frederick KS, et al. 2007. NADPH-dependent covalent binding of [3H]paroxetine to human liver microsomes and S-9 fractions: identification of an electrophilic quinone metabolite of paroxetine. Chem. Res. Toxicol. 20:1649-57
    • (2007) Chem. Res. Toxicol. , vol.20 , pp. 1649-1657
    • Zhao, S.X.1    Dalvie, D.K.2    Kelly, J.M.3    Soglia, J.R.4    Frederick, K.S.5
  • 47
    • 0035996330 scopus 로고    scopus 로고
    • Cytochrome P450 3A4-mediated bioactivation of raloxifene: Irreversible enzyme inhibition and thiol adduct formation
    • Chen Q, Ngui JS, Doss GA, Wang RW, Cai X, et al. 2002. Cytochrome P450 3A4-mediated bioactivation of raloxifene: irreversible enzyme inhibition and thiol adduct formation. Chem. Res. Toxicol. 15:907-14
    • (2002) Chem. Res. Toxicol. , vol.15 , pp. 907-914
    • Chen, Q.1    Ngui, J.S.2    Doss, G.A.3    Wang, R.W.4    Cai, X.5
  • 49
    • 84880320337 scopus 로고    scopus 로고
    • Pharmacokinetics and pharmacodynamics of canagliflozin, a sodium glucose co-transporter 2 inhibitor, in subjects with type 2 diabetes mellitus
    • Devineni D, Curtin CR, Polidori D, Gutierrez MJ, Murphy J, et al. 2013. Pharmacokinetics and pharmacodynamics of canagliflozin, a sodium glucose co-transporter 2 inhibitor, in subjects with type 2 diabetes mellitus. J. Clin. Pharmacol. 53:601-10
    • (2013) J. Clin. Pharmacol. , vol.53 , pp. 601-610
    • Devineni, D.1    Curtin, C.R.2    Polidori, D.3    Gutierrez, M.J.4    Murphy, J.5
  • 50
    • 67650809062 scopus 로고    scopus 로고
    • Comparative metabolism of 14C-labeled apixaban in mice, rats, rabbits, dogs and humans
    • Zhang D, He K, Raghavan N, Wang L, Mitroka J, et al. 2009. Comparative metabolism of 14C-labeled apixaban in mice, rats, rabbits, dogs and humans. Drug Metab. Dispos. 37:1738-48
    • (2009) Drug Metab. Dispos. , vol.37 , pp. 1738-1748
    • Zhang, D.1    He, K.2    Raghavan, N.3    Wang, L.4    Mitroka, J.5
  • 51
    • 66449099711 scopus 로고    scopus 로고
    • Metabolism and excretion of rivaroxaban, an oral, direct factor Xa inhibitor, in rats, dogs, and humans
    • Weinz C, Schwarz T, Kubiza D, Mueck W, Lang D. 2009. Metabolism and excretion of rivaroxaban, an oral, direct factor Xa inhibitor, in rats, dogs, and humans. Drug Metab. Dispos. 37:1056-64
    • (2009) Drug Metab. Dispos. , vol.37 , pp. 1056-1064
    • Weinz, C.1    Schwarz, T.2    Kubiza, D.3    Mueck, W.4    Lang, D.5
  • 52
    • 41949116970 scopus 로고    scopus 로고
    • Clinical pharmacokinetics and pharmacodynamics of the oral direct thrombin inhibitor dabigatran etexilate
    • Stangier J. 2008. Clinical pharmacokinetics and pharmacodynamics of the oral direct thrombin inhibitor dabigatran etexilate. Clin. Pharmacokinet. 47:285-95
    • (2008) Clin. Pharmacokinet. , vol.47 , pp. 285-295
    • Stangier, J.1
  • 53
    • 84879225503 scopus 로고    scopus 로고
    • Northbrook IL, Astellas Pharma Inc.
    • Xtandi® [package insert]. Northbrook, IL: Astellas Pharma Inc. ; 2012
    • (2012) Xtandi® [Package Insert]
  • 54
    • 84874110472 scopus 로고    scopus 로고
    • Absorption, metabolism and excretion of [14C]pomalidomide in humans following oral administration
    • Hoffmann M, Kasserra C, Reyes J, Schafer P, Kosek J, et al. 2013. Absorption, metabolism and excretion of [14C]pomalidomide in humans following oral administration. Cancer Chemother. Pharmacol. 71:489-501
    • (2013) Cancer Chemother. Pharmacol. , vol.71 , pp. 489-501
    • Hoffmann, M.1    Kasserra, C.2    Reyes, J.3    Schafer, P.4    Kosek, J.5
  • 55
    • 84892899479 scopus 로고    scopus 로고
    • Human cytochrome P450 oxidation of 5- hydroxythalidomide and pomalidomide, an amino analogue of thalidomide
    • Chowdhury G, Shibata N, Yamazaki H, Guengerich FP. 2014. Human cytochrome P450 oxidation of 5- hydroxythalidomide and pomalidomide, an amino analogue of thalidomide. Chem. Res. Toxicol. 27:147-56
    • (2014) Chem. Res. Toxicol. , vol.27 , pp. 147-156
    • Chowdhury, G.1    Shibata, N.2    Yamazaki, H.3    Guengerich, F.P.4
  • 56
    • 84883229213 scopus 로고    scopus 로고
    • South San Francisco, CA: Exelixis Inc.
    • Cometriq ® [package insert]. South San Francisco, CA: Exelixis Inc. ; 2012
    • (2012) Cometriq ® [Package Insert]
  • 57
    • 84905261740 scopus 로고    scopus 로고
    • Research Triangle Park, NC: GlaxoSmithKline
    • Votrient® [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2013
    • (2013) Votrient® [Package Insert]
  • 58
    • 84883281056 scopus 로고    scopus 로고
    • Cambridge, MA: Ariad Pharmaceuticals Inc.
    • Iclusig® [package insert]. Cambridge, MA: Ariad Pharmaceuticals Inc. ; 2012
    • (2012) Iclusig® [Package Insert]
  • 59
    • 84862807013 scopus 로고    scopus 로고
    • Drug-drug interaction potential of marketed oncology drugs: In vitro assessment of time-dependent cytochrome P450 inhibition, reactive metabolite formation and drug-drug interaction
    • Kenny JR, Mukadam S, Zhang C, Tay S, Collins C, et al. 2012. Drug-drug interaction potential of marketed oncology drugs: in vitro assessment of time-dependent cytochrome P450 inhibition, reactive metabolite formation and drug-drug interaction. Pharm. Res. 29:1960-76
    • (2012) Pharm. Res. , vol.29 , pp. 1960-1976
    • Kenny, J.R.1    Mukadam, S.2    Zhang, C.3    Tay, S.4    Collins, C.5
  • 61
    • 84863393782 scopus 로고    scopus 로고
    • Pharmacokinetics of vandetanib: Three phase i studies in healthy subjects
    • Martin P, Oliver S, Kennedy SJ, Partridge E, Hutchison M, et al. 2012. Pharmacokinetics of vandetanib: three phase I studies in healthy subjects. Clin. Ther. 34:221-37
    • (2012) Clin. Ther. , vol.34 , pp. 221-237
    • Martin, P.1    Oliver, S.2    Kennedy, S.J.3    Partridge, E.4    Hutchison, M.5
  • 62
    • 65649139708 scopus 로고    scopus 로고
    • Design and synthesis of an orally bioavailable and selective peptide epoxyketone proteasome inhibitor (PR-047)
    • Zhou HJ, Aujay MA, Bennett MK, Dajee M, Demo SD, et al. 2009. Design and synthesis of an orally bioavailable and selective peptide epoxyketone proteasome inhibitor (PR-047). J. Med. Chem. 52:3028-38
    • (2009) J. Med. Chem. , vol.52 , pp. 3028-3038
    • Zhou, H.J.1    Aujay, M.A.2    Bennett, M.K.3    Dajee, M.4    Demo, S.D.5
  • 63
    • 84871545425 scopus 로고    scopus 로고
    • Clinical pharmacokinetics, metabolism, and drug-drug interaction of carfilzomib
    • Wang Z, Yang J, Kirk C, Fang Y, Alsina M, et al. 2013. Clinical pharmacokinetics, metabolism, and drug-drug interaction of carfilzomib. Drug Metab. Dispos. 41:230-37
    • (2013) Drug Metab. Dispos. , vol.41 , pp. 230-237
    • Wang, Z.1    Yang, J.2    Kirk, C.3    Fang, Y.4    Alsina, M.5
  • 64
    • 79959404661 scopus 로고    scopus 로고
    • Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765
    • Herman SE, Gordon AL, Hertlein E, Ramanunni A, Zhang X, et al. 2011. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood 117:6287-96
    • (2011) Blood , vol.117 , pp. 6287-6296
    • Herman, S.E.1    Gordon, A.L.2    Hertlein, E.3    Ramanunni, A.4    Zhang, X.5
  • 65
    • 58149103921 scopus 로고    scopus 로고
    • Effect of intestinal glucuronidation in limiting hepatic exposure and bioactivation of raloxifene in humans and rats
    • Dalvie D, Kang P, Zientek M, Xiang C, Zhou S, Obach RS. 2008. Effect of intestinal glucuronidation in limiting hepatic exposure and bioactivation of raloxifene in humans and rats. Chem. Res. Toxicol. 21:2260-71
    • (2008) Chem. Res. Toxicol. , vol.21 , pp. 2260-2271
    • Dalvie, D.1    Kang, P.2    Zientek, M.3    Xiang, C.4    Zhou, S.5    Obach, R.S.6
  • 66
    • 53549100467 scopus 로고    scopus 로고
    • Can in vitro metabolism-dependent covalent binding data in liver microsomes distinguish hepatotoxic from nonhepatotoxic drugs? An analysis of 18 drugs with consideration of intrinsic clearance and daily dose
    • Obach RS, Kalgutkar AS, Soglia JR, Zhao SX. 2008. Can in vitro metabolism-dependent covalent binding data in liver microsomes distinguish hepatotoxic from nonhepatotoxic drugs? An analysis of 18 drugs with consideration of intrinsic clearance and daily dose. Chem. Res. Toxicol. 21:1814-22
    • (2008) Chem. Res. Toxicol. , vol.21 , pp. 1814-1822
    • Obach, R.S.1    Kalgutkar, A.S.2    Soglia, J.R.3    Zhao, S.X.4
  • 67
    • 62249093071 scopus 로고    scopus 로고
    • Assessment of three human in vitro systems in the generation of major human excretory and circulating metabolites
    • Dalvie D, Obach RS, Kang P, Prakash C, Loi CM, et al. 2009. Assessment of three human in vitro systems in the generation of major human excretory and circulating metabolites. Chem. Res. Toxicol. 22:357-68
    • (2009) Chem. Res. Toxicol. , vol.22 , pp. 357-368
    • Dalvie, D.1    Obach, R.S.2    Kang, P.3    Prakash, C.4    Loi, C.M.5
  • 68
    • 0023199001 scopus 로고
    • Metabolic activation of the tricyclic antidepressant amineptine: II. Protective role of glutathione against in vitro and in vivo covalent binding
    • Geneve J, Degott C, Letteron P, Tinel M, Descatoire V, et al. 1987. Metabolic activation of the tricyclic antidepressant amineptine: II. Protective role of glutathione against in vitro and in vivo covalent binding. Biochem. Pharmacol. 36:331-37
    • (1987) Biochem. Pharmacol. , vol.36 , pp. 331-337
    • Geneve, J.1    Degott, C.2    Letteron, P.3    Tinel, M.4    Descatoire, V.5
  • 69
    • 0029127584 scopus 로고
    • Inhibition of mitochondrial beta-oxidation as a mechanism of hepatotoxicity
    • Fromenty B, Pessayre D. 1995. Inhibition of mitochondrial beta-oxidation as a mechanism of hepatotoxicity. Pharmacol. Ther. 67:101-54
    • (1995) Pharmacol. Ther. , vol.67 , pp. 101-154
    • Fromenty, B.1    Pessayre, D.2
  • 70
    • 75749102996 scopus 로고    scopus 로고
    • Metabolism and disposition of the thienopyridine antiplatelet drugs ticlopidine, clopidogrel, and prasugrel in humans
    • Farid NA, Kurihara A, Wrighton SA. 2010. Metabolism and disposition of the thienopyridine antiplatelet drugs ticlopidine, clopidogrel, and prasugrel in humans. J. Clin. Pharmacol. 50:126-42
    • (2010) J. Clin. Pharmacol. , vol.50 , pp. 126-142
    • Farid, N.A.1    Kurihara, A.2    Wrighton, S.A.3
  • 71
    • 84920784358 scopus 로고    scopus 로고
    • Bridgewater NJ: Sanofi-Aventis
    • Plavix® [package insert]. Bridgewater, NJ: Sanofi-Aventis; 2013
    • (2013) Plavix® [Package Insert]
  • 72
    • 84920760187 scopus 로고    scopus 로고
    • Research Triangle Park NC: GlaxoSmithKline;
    • MekinistTM [package insert]. Research Triangle Park, NC: GlaxoSmithKline; 2014
    • (2014) MekinistTM [Package Insert]
  • 73
    • 62249155473 scopus 로고    scopus 로고
    • Can in vitro metabolism-dependent covalent binding data distinguish hepatotoxic from nonhepatotoxic drugs? An analysis using human hepatocytes and liver S-9 fraction
    • Bauman JS, Kelly JM, Tripathy S, Zhao SX, Lam WM, et al. 2009. Can in vitro metabolism-dependent covalent binding data distinguish hepatotoxic from nonhepatotoxic drugs? An analysis using human hepatocytes and liver S-9 fraction. Chem. Res. Toxicol. 22:332-40
    • (2009) Chem. Res. Toxicol. , vol.22 , pp. 332-340
    • Bauman, J.S.1    Kelly, J.M.2    Tripathy, S.3    Zhao, S.X.4    Lam, W.M.5
  • 74
    • 84865208356 scopus 로고    scopus 로고
    • In vitro approach to assess the potential for risk of idiosyncratic adverse reactions caused by candidate drugs
    • Thompson RA, Isin EM, Li Y, Weidolf L, Page K, et al. 2012. In vitro approach to assess the potential for risk of idiosyncratic adverse reactions caused by candidate drugs. Chem. Res. Toxicol. 25:1616-32
    • (2012) Chem. Res. Toxicol. , vol.25 , pp. 1616-1632
    • Thompson, R.A.1    Isin, E.M.2    Li, Y.3    Weidolf, L.4    Page, K.5
  • 75
  • 76
    • 67651115761 scopus 로고    scopus 로고
    • Short-acting 5- (trifluoromethyl)pyrido[4, 3-d]pyrimidin-4(3H)-one derivatives as orally-active calcium-sensing receptor antagonists
    • Didiuk MT, Griffith DA, Benbow JW, Liu KK, Walker DP, et al. 2009. Short-acting 5- (trifluoromethyl)pyrido[4, 3-d]pyrimidin-4(3H)-one derivatives as orally-active calcium-sensing receptor antagonists. Bioorg. Med. Chem. Lett. 19:4555-59
    • (2009) Bioorg. Med. Chem. Lett. , vol.19 , pp. 4555-4559
    • Didiuk, M.T.1    Griffith, D.A.2    Benbow, J.W.3    Liu, K.K.4    Walker, D.P.5
  • 77
    • 77953804348 scopus 로고    scopus 로고
    • Discovery tactics to mitigate toxicity risks due to reactive metabolite formation with 2-(2-hydroxyaryl)-5-(trifluoromethyl)pyrido[4, 3-d]pyrimidin- 4(3H)-one derivatives, potent calcium-sensing receptor antagonists and clinical candidate(s) for the treatment of osteoporosis
    • Kalgutkar AS, Griffith DA, Ryder T, Sun H, Miao Z, et al. 2010. Discovery tactics to mitigate toxicity risks due to reactive metabolite formation with 2-(2-hydroxyaryl)-5-(trifluoromethyl)pyrido[4, 3-d]pyrimidin- 4(3H)-one derivatives, potent calcium-sensing receptor antagonists and clinical candidate(s) for the treatment of osteoporosis. Chem. Res. Toxicol. 23:1115-26
    • (2010) Chem. Res. Toxicol. , vol.23 , pp. 1115-1126
    • Kalgutkar, A.S.1    Griffith, D.A.2    Ryder, T.3    Sun, H.4    Miao, Z.5


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.