Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cancer Res Clin Oncol ; 149(10): 7837-7848, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37036506

RESUMO

PURPOSE: Toremifene (TOR) is widely used as an antineoplastic drug and has an inhibitory effect on angiogenesis in mesenteric desmoid tumors and vascular intracranial solitary fibrous tumors. However, no study has investigated the direct effect of TOR on vascular cells. This study aimed at exploring the effect of TOR on the behaviors of vascular smooth muscle cells (VSMCs). METHODS: Human aortic umbilical vascular smooth muscle cells (HAVSMCs) were treated by TOR. Cell morphology, migration, adhesion, and proliferation assay were investigated. The cell cycle, apoptosis, mitochondrial membrane potential, and reactive oxygen species were assessed using flow cytometry. Caspase-3 and 9 activities were assayed using Caspase-3 and Caspase-9 Activity Assay kits, respectively. Immunofluorescence and Western blot assays were carried out to characterize protein expressions of PCNA, p53, and Rho/ROCK signaling pathway. RESULTS: TOR damaged cytoskeleton, inhibited VSMC proliferation, migration, and adhesion, and induced abnormal cell morphology and apoptosis. The antiproliferative activity of TOR was associated with the induction of G0/G1 phase arrest, blocking the cell cycle. TOR disrupted intracellular reactive oxygen species and mitochondrial membrane potential, and enhanced p53 expression and the activities of caspase-3 and caspase-9. Thus, TOR-induced apoptosis by the mitochondrial signaling pathway. Additionally, TOR induced decreased Rho, ROCK, MLC, and pMLC proteins. Collectively, TOR may affect multiple behaviors of VSMCs by damaging cytoskeleton through the Rho/ROCK pathway. CONCLUSION: The adverse effect of TOR on VSMCs could be considered as an important aspect of tumor growth inhibition.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Proliferação de Células , Músculo Liso Vascular/metabolismo , Toremifeno/metabolismo , Toremifeno/farmacologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Caspase 9/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Movimento Celular , Antineoplásicos/efeitos adversos , Neoplasias/metabolismo , Células Cultivadas
2.
J Proteome Res ; 19(11): 4670-4677, 2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-32907334

RESUMO

The global pandemic of Coronavirus Disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to the death of more than 675,000 worldwide and over 150,000 in the United States alone. However, there are currently no approved effective pharmacotherapies for COVID-19. Here, we combine homology modeling, molecular docking, molecular dynamics simulation, and binding affinity calculations to determine potential targets for toremifene, a selective estrogen receptor modulator which we have previously identified as a SARS-CoV-2 inhibitor. Our results indicate the possibility of inhibition of the spike glycoprotein by toremifene, responsible for aiding in fusion of the viral membrane with the cell membrane, via a perturbation to the fusion core. An interaction between the dimethylamine end of toremifene and residues Q954 and N955 in heptad repeat 1 (HR1) perturbs the structure, causing a shift from what is normally a long, helical region to short helices connected by unstructured regions. Additionally, we found a strong interaction between toremifene and the methyltransferase nonstructural protein (NSP) 14, which could be inhibitory to viral replication via its active site. These results suggest potential structural mechanisms for toremifene by blocking the spike protein and NSP14 of SARS-CoV-2, offering a drug candidate for COVID-19.


Assuntos
Betacoronavirus/química , Infecções por Coronavirus/virologia , Exorribonucleases , Pneumonia Viral/virologia , Glicoproteína da Espícula de Coronavírus , Toremifeno , Proteínas não Estruturais Virais , Antivirais/química , Antivirais/metabolismo , COVID-19 , Reposicionamento de Medicamentos , Exorribonucleases/química , Exorribonucleases/metabolismo , Humanos , Simulação de Acoplamento Molecular , Pandemias , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo , Toremifeno/química , Toremifeno/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo
3.
J Med Chem ; 63(13): 7211-7225, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32490678

RESUMO

The recent Ebola epidemics in West Africa underscore the great need for effective and practical therapies for future Ebola virus outbreaks. We have discovered a new series of remarkably potent small molecule inhibitors of Ebola virus entry. These 4-(aminomethyl)benzamide-based inhibitors are also effective against Marburg virus. Synthetic routes to these compounds allowed for the preparation of a wide variety of structures, including a conformationally restrained subset of indolines (compounds 41-50). Compounds 20, 23, 32, 33, and 35 are superior inhibitors of Ebola (Mayinga) and Marburg (Angola) infectious viruses. Representative compounds (20, 32, and 35) have shown good metabolic stability in plasma and liver microsomes (rat and human), and 32 did not inhibit CYP3A4 nor CYP2C9. These 4-(aminomethyl)benzamides are suitable for further optimization as inhibitors of filovirus entry, with the potential to be developed as therapeutic agents for the treatment and control of Ebola virus infections.


Assuntos
Antivirais/farmacologia , Benzamidas/farmacologia , Doença pelo Vírus Ebola/virologia , Doença do Vírus de Marburg/virologia , Internalização do Vírus/efeitos dos fármacos , Células A549 , Animais , Antivirais/química , Benzamidas/química , Chlorocebus aethiops , Inibidores do Citocromo P-450 CYP3A/química , Inibidores do Citocromo P-450 CYP3A/farmacologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Microssomos Hepáticos/efeitos dos fármacos , Simulação de Acoplamento Molecular , Relação Estrutura-Atividade , Toremifeno/química , Toremifeno/metabolismo , Toremifeno/farmacologia , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
4.
J Pharm Biomed Anal ; 156: 147-152, 2018 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-29704771

RESUMO

Antiestrogenic compounds such as tamoxifen, toremifen and chlomifen are used illegally by athletes to minimize physical impacts such as gynecomastia resulting from the secondary effects of anabolic androgenic steroids, used to increase athletic efficiency unlawfully. The use of these compounds is banned by the World Anti-Doping Agency (WADA) and controls are made through analytical methodologies such as HPLC-MS/MS, which do not fulfil the sample throughput requirements. Moreover, compounds such as tamoxifen are also used to treat hormone receptor-positive breast cancer (ER + ).Therapeutic drug monitoring (TDM) of tamoxifen may also be clinically useful for guiding treatment decisions. An accurate determination of these drugs requires a solid phase extraction of patient serum followed by HPLC-MS/MS. In the context of an unmet need of high-throughput screening (HTS) and quantitative methods for antiestrogenic substances we have approached the development of antibodies and an immunochemical assay for the determination of these antiestrogenic compounds. The strategy applied has taken into consideration that these drugs are metabolized and excreted in urine as the corresponding 4-hydroxylated compounds. A microplate-based ELISA procedure has been developed for the analysis of these metabolites in urine with a LOD of 0.15, 0.16 and 0.63 µg/L for 4OH-tamoxifen, 4OH-toremifen and 4OH-clomifen, respectively, much lower than the MRPL established by WADA (20 µg/L).


Assuntos
Dopagem Esportivo/prevenção & controle , Monitoramento de Medicamentos/métodos , Moduladores Seletivos de Receptor Estrogênico/urina , Congêneres da Testosterona/urina , Neoplasias da Mama/tratamento farmacológico , Cromatografia Líquida de Alta Pressão , Clomifeno/metabolismo , Clomifeno/uso terapêutico , Clomifeno/urina , Ensaio de Imunoadsorção Enzimática , Feminino , Voluntários Saudáveis , Ensaios de Triagem em Larga Escala/métodos , Humanos , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Extração em Fase Sólida , Tamoxifeno/análogos & derivados , Tamoxifeno/metabolismo , Tamoxifeno/uso terapêutico , Tamoxifeno/urina , Espectrometria de Massas em Tandem , Toremifeno/metabolismo , Toremifeno/uso terapêutico , Toremifeno/urina
5.
Nature ; 535(7610): 169-172, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27362232

RESUMO

Ebola viruses (EBOVs) are responsible for repeated outbreaks of fatal infections, including the recent deadly epidemic in West Africa. There are currently no approved therapeutic drugs or vaccines for the disease. EBOV has a membrane envelope decorated by trimers of a glycoprotein (GP, cleaved by furin to form GP1 and GP2 subunits), which is solely responsible for host cell attachment, endosomal entry and membrane fusion. GP is thus a primary target for the development of antiviral drugs. Here we report the first, to our knowledge, unliganded structure of EBOV GP, and high-resolution complexes of GP with the anticancer drug toremifene and the painkiller ibuprofen. The high-resolution apo structure gives a more complete and accurate picture of the molecule, and allows conformational changes introduced by antibody and receptor binding to be deciphered. Unexpectedly, both toremifene and ibuprofen bind in a cavity between the attachment (GP1) and fusion (GP2) subunits at the entrance to a large tunnel that links with equivalent tunnels from the other monomers of the trimer at the three-fold axis. Protein­drug interactions with both GP1 and GP2 are predominately hydrophobic. Residues lining the binding site are highly conserved among filoviruses except Marburg virus (MARV), suggesting that MARV may not bind these drugs. Thermal shift assays show up to a 14 °C decrease in the protein melting temperature after toremifene binding, while ibuprofen has only a marginal effect and is a less potent inhibitor. These results suggest that inhibitor binding destabilizes GP and triggers premature release of GP2, thereby preventing fusion between the viral and endosome membranes. Thus, these complex structures reveal the mechanism of inhibition and may guide the development of more powerful anti-EBOV drugs.


Assuntos
Antivirais/química , Antivirais/metabolismo , Ebolavirus/química , Toremifeno/química , Toremifeno/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Antivirais/farmacologia , Sítios de Ligação , Linhagem Celular , Sequência Conservada , Ebolavirus/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ibuprofeno/química , Ibuprofeno/metabolismo , Ibuprofeno/farmacologia , Ligantes , Marburgvirus/química , Fusão de Membrana/efeitos dos fármacos , Modelos Moleculares , Ligação Proteica , Estabilidade Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína/efeitos dos fármacos , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Temperatura , Toremifeno/farmacologia , Proteínas do Envelope Viral/antagonistas & inibidores , Ligação Viral/efeitos dos fármacos
6.
Drug Metab Pharmacokinet ; 30(5): 325-33, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26423799

RESUMO

Toremifene (TOR) and Tamoxifen (TAM) are widely used as endocrine therapy for estrogen receptor positive breast cancer. Poor metabolizers of TAM are likely to have worse clinical outcomes than patients who exhibit normal TAM metabolism due to lower plasma level of its active metabolite, 4-hydroxy-N-desmethyl (4OH-NDM) tamoxifen (endoxifen). In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Similar levels of NDM metabolites were formed for both TOR and TAM, and N-demethylation of both compounds was primarily carried out by CYP3A4. We found that the formation of 4OH-NDM-TOR was catalyzed both by CYP2C9 and CYP2D6, whereas the formation of 4OH-TAM and endoxifen was specifically catalyzed by CYP2D6 in HLMs. Our results suggest that the potential contribution of CYP2D6 in the bioactivation pathway of TOR may be lower compared to TAM, and may have a different impact on clinical outcome than CYP2D6 polymorphisms.


Assuntos
Microssomos Hepáticos/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/metabolismo , Toremifeno/metabolismo , Biotransformação , Linhagem Celular Tumoral , Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Inibidores das Enzimas do Citocromo P-450/farmacologia , Feminino , Humanos , Técnicas In Vitro , Microssomos Hepáticos/efeitos dos fármacos , Tamoxifeno/análogos & derivados
7.
Drug Metab Dispos ; 42(9): 1392-400, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24917585

RESUMO

Tamoxifen (Tam) is a selective estrogen receptor modulator used to inhibit breast tumor growth. Tam can be directly N-glucuronidated via the tertiary amine group or O-glucuronidated after cytochrome P450-mediated hydroxylation. In this study, the glucuronidation of Tam and its hydroxylated and/or chlorinated derivatives [4-hydroxytamoxifen (4OHTam), toremifene (Tor), and 4-hydroxytoremifene (4OHTor)] was examined using recombinant human UDP-glucuronosyltransferases (UGTs) from the 1A subfamily and human hepatic microsomes. Recombinant UGT1A4 catalyzed the formation of N-glucuronides of Tam and its derivatives and was the most active UGT enzyme toward these compounds. Therefore, it was hypothesized that single nucleotide polymorphisms (SNPs) in the promoter region of UGT1A4 have the ability to significantly decrease the glucuronidation rates of Tam metabolites in the human liver. In vitro activity of 64 genotyped human liver microsomes was used to determine the association between the UGT1A4 promoter and coding region SNPs and the glucuronidation rates of Tam, 4OHTam, Tor, and 4OHTor. Significant decreases in enzymatic activity were observed in microsomes for individuals heterozygous for -163G/A and -217T/G. These alterations in glucuronidation may lead to prolonged circulating half-lives and may potentially modify the effectiveness of these drugs in the treatment of breast cancer.


Assuntos
Glucuronosiltransferase/genética , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Tamoxifeno/metabolismo , Genótipo , Humanos , Hidroxilação/genética , Microssomos Hepáticos/metabolismo , Farmacogenética/métodos , Tamoxifeno/análogos & derivados , Toremifeno/metabolismo
8.
Drug Test Anal ; 6(5): 482-91, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24431005

RESUMO

The present study was designed to provide preliminary information on the potential impact of metabolic drug-drug interaction on the effectiveness of doping control strategies currently followed by the anti-doping laboratories to detect the intake of banned agents. In vitro assays based on the use of human liver microsomes and recombinant CYP isoforms were designed and performed to characterize the phase I metabolic profile of the prohibited agent toremifene, selected as a prototype drug of the class of selective oestrogen receptor modulators, both in the absence and in the presence of medicaments (fluconazole, ketoconazole, itraconazole, miconazole, cimetidine, ranitidine, fluoxetine, paroxetine, nefazodone) not included in the World Anti-Doping Agency list of prohibited substances and methods and frequently administered to athletes. The results show that the in vitro model developed in this study was adequate to simulate the in vivo metabolism of toremifene, confirming the results obtained in previous studies. Furthermore, our data also show that ketoconazole, itraconazole, miconazole and nefazodone cause a marked modification in the production of the metabolic products (i.e. hydroxylated and carboxylated metabolites) normally selected by the anti-doping laboratories as target analytes to detect toremifene intake; moderate variations were registered in the presence of fluconazole, paroxetine and fluoxetine; while no significant modifications were measured in the presence of ranitidine and cimetidine. This evidence imposes that the potential effect of drug-drug interactions is duly taken into account in anti-doping analysis, also for a broader significance of the analytical results.


Assuntos
Dopagem Esportivo/métodos , Interações Medicamentosas , Desintoxicação Metabólica Fase I , Toremifeno/metabolismo , Toremifeno/farmacocinética , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Técnicas In Vitro , Isoenzimas/genética , Isoenzimas/metabolismo , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacocinética , Detecção do Abuso de Substâncias
9.
Anal Bioanal Chem ; 405(16): 5467-87, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23604525

RESUMO

The use of selective oestrogen receptor modulators has been prohibited since 2005 by the World Anti-Doping Agency regulations. As they are extensively cleared by hepatic and intestinal metabolism via oxidative and conjugating enzymes, a complete investigation of their biotransformation pathways and kinetics of excretion is essential for the anti-doping laboratories to select the right marker(s) of misuse. This work was designed to characterize the chemical reactions and the metabolizing enzymes involved in the metabolic routes of clomiphene, tamoxifen and toremifene. To determine the biotransformation pathways of the substrates under investigation, urine samples were collected from six subjects (three females and three males) after oral administration of 50 mg of clomiphene citrate or 40 mg of tamoxifen or 60 mg of toremifene, whereas the metabolizing enzymes were characterized in vitro, using expressed cytochrome P450s and uridine diphosphoglucuronosyltransferases. The separation, identification and determination of the compounds formed in the in vivo and in vitro experiments were carried out by liquid chromatography coupled with mass spectrometry techniques using different acquisition modes. Clomiphene, tamoxifen and toremifene were biotransformed to 22, 23 and 18 metabolites respectively, these phase I reactions being catalyzed mainly by CYP3A4 and CYP2D6 isoforms and, to a lesser degree, by CYP3A5, CYP2B6, CYP2C9, CYP2C19 isoforms. The phase I metabolic reactions include hydroxylation in different positions, N-oxidation, dehalogenation, carboxylation, hydrogenation, methoxylation, N-dealkylation and combinations of them. In turn, most of the phase I metabolites underwent conjugation reaction to form the corresponding glucuro-conjugated mainly by UGT1A1, UGT1A3, UGT1A4, UGT2B7, UGT2B15 and UGT2B17 isoenzymes.


Assuntos
Cromatografia Líquida/métodos , Clomifeno/farmacocinética , Tamoxifeno/farmacocinética , Espectrometria de Massas em Tandem/métodos , Toremifeno/farmacocinética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Biotransformação , Clomifeno/metabolismo , Clomifeno/urina , Citocromo P-450 CYP2B6 , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A/metabolismo , Remoção de Radical Alquila , Dopagem Esportivo , Feminino , Glucuronosiltransferase/metabolismo , Humanos , Hidroxilação , Masculino , Microssomos Hepáticos/metabolismo , Oxirredução , Oxirredutases N-Desmetilantes/metabolismo , Tamoxifeno/metabolismo , Tamoxifeno/urina , Toremifeno/metabolismo , Toremifeno/urina
10.
Int J Cancer ; 132(6): 1475-85, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22915089

RESUMO

We investigated the in vitro metabolism and estrogenic and antiestrogenic activity of toremifene (TOR), tamoxifen (TAM) and their metabolites to better understand the potential effects of cytochrome P-450 2D6 (CYP2D6) status on the activity of these drugs in women with breast cancer. The plasma concentrations of TOR and its N-desmethyl (NDM) and 4-hydroxy (4-OH) metabolites during steady-state dosing with TOR were also determined. Unlike TOR, TAM and its NDM metabolite were extensively oxidized to 4-OH TAM and 4-OH-NDM TAM by CYP2D6, and the rate of metabolism was affected by CYP2D6 status. 4-OH-NDM TOR concentrations were not measurable at steady state in plasma of subjects taking 80 mg of TOR. Molecular modeling provided insight into the lack of 4-hydroxylation of TOR by CYP2D6. The 4-OH and 4-OH-NDM metabolites of TOR and TAM bound to estrogen receptor (ER) subtypes with fourfold to 30-fold greater affinity were 35- to 187-fold more efficient at antagonizing ER transactivation and had antiestrogenic potency that was up to 360-fold greater than their parent drugs. Our findings suggest that variations in CYP2D6 metabolic capacity may cause significant differences in plasma concentrations of active TAM metabolites (i.e., 4-OH TAM and 4-OH-NDM TAM) and contribute to variable pharmacologic activity. Unlike TAM, the clinical benefits in subjects taking TOR to treat metastatic breast cancer would not likely be subject to allelic variation in CYP2D6 status or affected by coadministration of CYP2D6-inhibiting medications.


Assuntos
Citocromo P-450 CYP2D6/fisiologia , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/metabolismo , Toremifeno/metabolismo , Adulto , Humanos , Masculino , Oxirredução , Relação Estrutura-Atividade , Tamoxifeno/análogos & derivados , Tamoxifeno/sangue
11.
Drug Metab Dispos ; 40(6): 1210-5, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22434874

RESUMO

Toremifene (TOR) is a selective estrogen receptor modulator used in adjuvant therapy for breast cancer and in clinical trials for prostate cancer prevention. The chemical structure of TOR differs from that of tamoxifen (TAM) by the presence of a chlorine atom in the ethyl side chain, resulting in a more favorable toxicity spectrum with TOR. In addition, some patients who fail on TAM therapy benefit from high-dose TOR therapy. Several studies have indicated that functional genetic variants in the TAM metabolic pathway influence response to therapy, but pharmacogenomic studies of patients treated with TOR are lacking. In this study, we examined individual variability in sulfation of 4-hydroxy TOR (4-OH TOR) (the active metabolite of TOR) in human liver cytosols from 104 subjects and found approximately 30-fold variation in activity. 4-OH TOR sulfation was significantly correlated (r = 0.98, P < 0.0001) with ß-naphthol sulfation (diagnostic for SULT1A1) but not with 17ß estradiol sulfation, a diagnostic substrate for SULT1E1(r = 0.09, P = 0.34). Examination of recombinant sulfotransferases (SULTs) revealed that SULT1A1 and SULT1E1 catalyzed 4-OH TOR sulfation, with apparent Km values of 2.6 and 6.4 µM and Vmax values of 8.5 and 5.5 nmol x min(-1) x mg protein(-1), respectively. 4-OH TOR sulfation was inhibited by 2,6-dichloro-4-nitrophenol (IC50 = 2.34 ± 0.19 µM), a specific inhibitor of SULT1A1. There was also a significant association between SULT1A1 genotypes and copy number and 4-OH TOR sulfation in human liver cytosols. These results indicate that variability in sulfation could contribute to response to TOR in the treatment of breast and prostate cancer.


Assuntos
Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Variação Genética/fisiologia , Farmacogenética , Tamoxifeno/análogos & derivados , Toremifeno/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Feminino , Humanos , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Farmacogenética/métodos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/metabolismo , Adulto Jovem
12.
Xenobiotica ; 41(10): 851-62, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21726172

RESUMO

Toremifene is an effective agent for the treatment of breast cancer in postmenopausal women and is being evaluated for its ability to prevent bone fractures in men with prostate cancer taking androgen deprivation therapy. Due to the potential for drug-drug interactions, the ability of toremifene and its primary circulating metabolite N-desmethyltoremifene (NDMT) to inhibit nine human cytochrome P450 (CYP) enzymes was determined using human liver microsomes. Induction of CYP1A2 and 3A4 by toremifene was also investigated in human hepatocytes. Toremifene did not significantly inhibit CYP1A2 or 2D6. However, toremifene is a competitive inhibitor of CYP3A4, non-competitive inhibitor of CYP2A6, 2C8, 2C9, 2C19 and 2E1 and mixed-type inhibitor of CYP2B6. CYP inhibition by NDMT was similar in magnitude to toremifene. Toremifene did not induce CYP1A2 but increased CYP3A4 monooxygenase activity and gene expression in drug-exposed human primary hepatocytes. Although clinical doses of toremifene produce steady state exposures to toremifene and NDMT that may be sufficient to cause pharmacokinetic drug-drug interactions with other drugs metabolised by CYP2B6, CYP2C8, CYP3A4, CYP2C9 and CYP2C19, these data indicate that toremifene is unlikely to play a role in clinical drug-drug interactions with substrate drugs of CYP1A2 and CYP2D6.


Assuntos
Inibidores das Enzimas do Citocromo P-450 , Tamoxifeno/análogos & derivados , Toremifeno/farmacologia , Células Cultivadas , Sistema Enzimático do Citocromo P-450/química , Interações Medicamentosas , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Humanos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Tamoxifeno/química , Tamoxifeno/farmacologia , Toremifeno/química , Toremifeno/metabolismo
13.
J Chromatogr A ; 1218(29): 4727-37, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21683367

RESUMO

Toremifene is a selective estrogen receptor modulator included in the list of prohibited substances in sport by the World Anti-doping Agency. The aim of the present study was to investigate toremifene metabolism in humans in order to elucidate the structures of the most abundant urinary metabolites and to define the best marker to detect toremifene administration through the analysis of urine samples. Toremifene (Fareston) was administered to healthy volunteers and the urine samples were subjected to different preparation methods to detect free metabolites as well as metabolites conjugated with glucuronic acid or sulphate. Urinary extracts were analyzed by LC-MS/MS with triple quadrupole analyzer using selected reaction monitoring mode. Transitions for potential metabolites were selected by using the theoretical [M+H](+) as precursor ion and m/z 72 or m/z 58 as product ions for N,N-dimethyl and N-desmethyl metabolites, respectively. Toremifene and 20 metabolites were detected in excretion study samples, excreted free or conjugated with glucuronic acid or sulphate. Structures for most abundant phase I metabolites were proposed using accurate mass measurements performed by QTOF MS, based on fragmentation pattern observed for those metabolites available as reference standards. Several metabolic pathways including mono- and di-hydroxylation, N-desmethylation, hydroxymethylation, oxidation, dehalogenation and combinations were proposed. All metabolites were detected up to one month after toremifene administration; the most abundant metabolites were detected in the free fraction and they were metabolites resulting from dehalogenation. Several of the metabolites elucidated in this work have not been reported until now in the scientific literature.


Assuntos
Dopagem Esportivo , Espectrometria de Massas em Tandem/métodos , Toremifeno/urina , Cromatografia Líquida , Ácido Glucurônico , Humanos , Redes e Vias Metabólicas , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/urina , Toremifeno/metabolismo
14.
Anal Bioanal Chem ; 401(2): 529-41, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21380606

RESUMO

In the present study, toremifene urinary excretion studies were evaluated in order to examine main metabolic reactions and to select target metabolites in doping control analysis. Urine samples from three female subjects were collected every 3 h for at least 15 days after the oral administration of a single dose of Fareston® (60 mg). The elemental compositions of the compounds detected were determined by liquid chromatography-mass spectrometry using a time-of-flight system with accurate mass measurement. More detailed structure elucidation was obtained by monitoring the presence or absence of structure-specific ions, using product ion scan and neutral loss acquisition modes, whereas the metabolites urinary profiles were evaluated in selected reaction monitoring acquisition mode. The results showed that the main routes of phase-I modifications involved carboxylation of the chlorinated side chain, N-demethylation and hydroxylation in different positions. Fifteen metabolites were found in all subjects studied, most of them were detected for more than 10 days in the free, glucuronide and sulphate fractions, with a maximum of excretion generally after 9-22 and 34-47 h from drug administration. These metabolites can be divided in two groups: metabolites with the characteristic chlorine isotope pattern and metabolites without the characteristic chlorine isotope pattern. The most abundant and long-term compounds were the carboxylated metabolites followed by the hydroxylated metabolites. Their product ions originating after collision-induced dissociation were observed to occur prevalently in the dimethylaminoethoxy and in the chlorinated side chains. These structure-specific ions were used to design screening and confirmation procedures to positively identify toremifene administration in doping control analysis.


Assuntos
Dopagem Esportivo , Substâncias para Melhoria do Desempenho/metabolismo , Substâncias para Melhoria do Desempenho/urina , Detecção do Abuso de Substâncias/métodos , Toremifeno/metabolismo , Toremifeno/urina , Adulto , Cromatografia Líquida , Feminino , Humanos , Espectrometria de Massas , Substâncias para Melhoria do Desempenho/química , Valores de Referência , Sensibilidade e Especificidade , Toremifeno/química
15.
Analyst ; 136(3): 467-72, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-21113546

RESUMO

The metabolism and excretion of toremifene were investigated in one healthy male volunteer after a single oral administration of 120 mg toremifene citrate. Different liquid chromatographic/tandem mass spectrometric (LC/MS/MS) scanning techniques were carried out for the characterization of the metabolites in human urine for doping control purposes. The potential characteristic fragmentation pathways of toremifene and its major metabolites were presented. An approach for the metabolism study of toremifene and its analogs by liquid chromatography-tandem mass spectrometry was established. Five different LC/MS/MS scanning methods based on precursor ion scan (precursor ion scan of m/z 72.2, 58.2, 44.2, 45.2, 88.2 relative to five metabolic pathways) in positive ion mode were assessed to recognize the metabolites. Based on product ion scan and precursor ion scan techniques, the metabolites were proposed to be identified as 4-hydroxy-toremifene (m/z 422.4), 4'-hydroxy-toremifene (m/z 422.4), α-hydroxy-toremifene (m/z 422.4), 3,4-dihydroxy-toremifene (m/z 404.2), toremifene acid (m/z 402.2), 3-hydroxy-4-methoxy-toremifene (m/z 456.2), dihydroxy-dehydro-toremifene (m/z 440.2), 3,4-dihydroxy-toremifene (m/z 438.2), N-demethyl-4-hydroxy-toremifene (m/z 408.3), N-demethyl-3-hydroxy-4-methoxy-toremifene (m/z 438.3). In addition, a new metabolite with a protonated molecule at m/z 390.3 was detected in all urine samples. The compound was identified by LC/MS/MS as N-demethyl-4,4'-dihydroxy-tamoxifene. The results indicated that 3,4-dihydroxy-toremifene (m/z 404.2), toremifene acid (m/z 402.2) and N-demethyl-4,4'-dihydroxy-tamoxifene (m/z 390.3) were major metabolites in human urine.


Assuntos
Cromatografia Líquida/métodos , Monitoramento de Medicamentos/métodos , Moduladores Seletivos de Receptor Estrogênico , Espectrometria de Massas em Tandem/métodos , Toremifeno , Administração Oral , Adulto , Biotransformação , Cromatografia Líquida/instrumentação , Monitoramento de Medicamentos/instrumentação , Humanos , Masculino , Estrutura Molecular , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacocinética , Moduladores Seletivos de Receptor Estrogênico/urina , Espectrometria de Massas em Tandem/instrumentação , Toremifeno/metabolismo , Toremifeno/farmacocinética , Toremifeno/urina
16.
Anal Bioanal Chem ; 394(5): 1341-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19139854

RESUMO

The metabolism of the selective estrogen receptor modulator toremifene was simulated in an on-line electrochemistry/enzyme reactor/liquid chromatography/mass spectrometry system. To simulate the oxidative phase I metabolism, toremifene was oxidized in an electrochemical (EC) flow-through cell at 1,500 mV vs. Pd/H2 to its phase I metabolites, some of which are reactive quinoid species. In the presence of glutathione-S-transferase (GST), these quinoid compounds react with glutathione, which is also the common detoxification mechanism in the body. While reacting with glutathione, the chlorine atom is eliminated from the toremifene moiety. Due to higher conversion rates, GST supplied in continuous flow proved to be more efficient than using immobilized GST on magnetic microparticles. In the absence of GST, not all GSH adducts are formed, proving the necessity of a phase II enzyme to simulate the complete metabolic pathway of xenobiotics in an on-line EC/LC/MS system.


Assuntos
Técnicas Eletroquímicas/instrumentação , Glutationa Transferase/metabolismo , Sistemas On-Line/instrumentação , Toremifeno/análise , Toremifeno/metabolismo , Técnicas Eletroquímicas/métodos , Espectrometria de Massas , Estrutura Molecular , Oxirredução , Moduladores Seletivos de Receptor Estrogênico/química , Toremifeno/química
17.
Cancer Sci ; 97(6): 468-77, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16734724

RESUMO

Effects of hepatocarcinogenic TAM and non-hepatocarcinogenic TOR on the formation of hepatic DNA adducts and on the gene expression of hepatic drug-metabolizing enzymes and DNA repair enzymes/proteins were comparatively examined in female Sprague-Dawley rats treated with TAM (20 or 40 mg/kg/day, i.g.) or TOR (40 mg/kg/day, i.g.) for 1, 2 or 8 weeks. Hepatic TAM-DNA adducts were formed even after 1 week of treatment with TAM at either dose, and the adduct levels increased in a dose- and treatment period-dependent manner, whereas no DNA adducts were detected in any of the TOR-treated rats. Conversely, TAM and TOR showed almost the same capacity for increasing the gene expression of drug-metabolizing enzymes responsible for metabolic activation and detoxification, at least up to the 2-week treatment mark. Accordingly, differences in DNA adduct formation between TAM- and TOR-treated rats would not be primarily dependent on the capacity for inducing hepatic drug-metabolizing enzymes. In addition, a drastic increase in the gene expression of cytochrome P4503A2 (CYP3A2), an activation enzyme of TAM, by the 8-week treatment with TAM might have contributed to the increased formation of DNA adducts. Gene expressions of DNA repair enzymes/proteins responsible for a nucleotide excision repair system were not significantly changed in any of the rats treated with either drug. The present findings suggest that the difference between TAM and TOR in hepatocarcinogenic potency is dependent on the capacity to form DNA adducts rather than modulating the expression of drug-metabolizing enzymes and DNA repair enzymes/proteins.


Assuntos
Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Fígado/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Tamoxifeno/toxicidade , Toremifeno/toxicidade , Animais , Cromatografia Líquida de Alta Pressão , Citocromos/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/metabolismo , Toremifeno/metabolismo
18.
Chem Res Toxicol ; 16(9): 1138-44, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12971802

RESUMO

An increased risk of developing endometrial cancer is observed in breast cancer patients treated with tamoxifen (TAM) and in healthy women undergoing TAM chemoprevention therapy. TAM-DNA adducts were detected in the endometrium of women taking TAM (Shibutani, S., et al. (2000) Carcinogenesis 21, 1461-1467) and are formed primarily through O-sulfonation of alpha-hydroxytamoxifen (alpha-OHTAM). To explore the genotoxicic mechanisms of TAM, TAM was incubated with one of multiple human cytochrome P450 enzymes, i.e., P450 1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5, 3A7, 4A11, 4F2, 4F3A, or 4F3B, in a NADPH regenerating system, and the metabolites were identified using HPLC/UV analysis with authentic standards. Among the 18 human P450 enzymes, P450 3A4 generated a significant amount of alpha-OHTAM. When some rat P450 enzymes were examined, P450 3A2 also catalyzed alpha-hydroxylation of TAM. Similarly, human P450 3A4 and rat P450 3A1 and 3A2 converted toremifene (TOR, a chlorinated TAM analogue) to alpha-hydroxytoremifene (alpha-OHTOR). The formation of alpha-OHTAM and alpha-OHTOR by these P450 enzymes was confirmed by tandem mass spectroscopy. Only the P450 3A subfamily enzymes are able to alpha-hydroxylate TAM and TOR. Although the formation of alpha-OHTOR by these enzymes was much higher than that of alpha-OHTAM, TOR is known to be much less genotoxic than TAM. The results support our proposed mechanism that the lower genotoxicity of TOR is due to limited O-sulfonation of alpha-OHTOR by hydroxysteroid sulfotransferases, resulting in the poor formation of DNA adducts (Shibutani, S., et al. (2001) Cancer Res. 61, 3925-3931).


Assuntos
Hidrocarboneto de Aril Hidroxilases/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/metabolismo , Toremifeno/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP3A , Nucleotídeos de Desoxiguanina/metabolismo , Feminino , Humanos , Hidroxilação , Espectrometria de Massas , NADPH Oxidases/metabolismo , Ratos , Tamoxifeno/síntese química
19.
Chem Res Toxicol ; 16(7): 832-7, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12870885

RESUMO

The antiestrogen, tamoxifen, has been extensively used in the treatment and prevention of breast cancer. Although tamoxifen showed benefits in the chemotherapy and chemoprevention of breast cancer, epidemiological studies in both tamoxifen-treated breast cancer patients and healthy women indicated that treatment caused an increased risk of developing endometrial cancer. These troubling side effects lead to concerns over long-term safety of the drug. Therefore, it is important to fully understand the relationship between the antiestrogenic and the genotoxic mechanisms of tamoxifen, other antiestrogens, and their metabolites. Previously, we have shown that o-quinone formation from tamoxifen and its analogues, droloxifene and 4-hydroxytoremifene, may not contribute to the cytotoxic effects of these antiestrogens; however, these o-quinones can form adducts with deoxynucleosides and this implies that the o-quinone pathway could contribute to the genotoxicity of the antiestrogens in vivo. To further investigate this potential genotoxic pathway, we were interested in the role of estrogen receptor (ER)(1) alpha and beta since work with catechol estrogens has shown that ERs seem to enhance DNA damage in breast cancer cell lines. As a result, we investigated the binding affinities of 4-hydroxy and 3,4-dihydroxy derivatives of tamoxifen and toremifene to ER alpha and beta. The antiestrogenic activities of the metabolites using the Ishikawa cells were also investigated as well as their activity in ERalpha and ERbeta breast cancer cells using the transient transfection reporter, estrogen response element-dependent luciferase assay. The data showed that the antiestrogenic activities of these compounds in the biological assays mimicked their activities in the ER binding assay. To determine if the compounds were toxic and if ERs played a role in this process, the cytotoxicity of these compounds in ERbeta41(2) (ERbeta), S30 (ERalpha), and MDA-MB-231 (ER(-)) cell lines was compared. The results showed that the cytotoxicity differences between the metabolites were modest. In addition, all of the metabolites showed similar toxicity patterns in both ER positive and negative cell lines, which means that the ER may not contribute to the cytotoxicity pathway. Finally, we compared the amount of DNA damage induced by these metabolites in these cell lines using the comet assay. The catechols 3,4-dihydroxytoremifene and 3,4-dihydroxytamoxifen induced a greater amount of cellular single strand DNA cleavage as compared with the phenols in all cell lines. The different amounts of DNA damage in ER positive and negative cell lines suggested that the ERs might play a role in this process. These data suggest that the formation of catechols represents a minor role in cytotoxic and antiestrogenic effects in cells as compared with their phenol analogues. However, catechols induced more DNA damage at nontoxic doses in breast cancer cells, which implies that o-quinones formed from catechols could contribute to genotoxicity in vivo, which is ER-dependent.


Assuntos
Dano ao DNA , Moduladores de Receptor Estrogênico/farmacologia , Receptores de Estrogênio/efeitos dos fármacos , Tamoxifeno/farmacologia , Toremifeno/farmacologia , Ligação Competitiva , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Catecóis/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaio Cometa , Ensaios de Seleção de Medicamentos Antitumorais , Estradiol/metabolismo , Estradiol/farmacologia , Moduladores de Receptor Estrogênico/metabolismo , Feminino , Humanos , Indolquinonas/biossíntese , Indolquinonas/química , Quinonas/química , Quinonas/metabolismo , Receptores de Estrogênio/metabolismo , Tamoxifeno/metabolismo , Toremifeno/metabolismo
20.
Biomed Chromatogr ; 16(5): 361-3, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12210510

RESUMO

The in vitro metabolism of toremifene has been studied in liver microsomal preparations from rat, mouse and human sources using high-performance liquid chromatography-electrospray ionisation mass spectrometry (HPLC-ESIMS). The metabolites detected were N-desmethyltoremifene (m/z 392), 4-hydroxytoremifene (m/z 422), 4'-hydroxytoremifene (m/z 422) and toremifene N-oxide m/z 422). In addition, a new polar metabolite with a protonated molecule at m/z 422 has been detected in all three species. The compound was identified by tandem MS-MS as alpha-hydroxytoremifene, an analogue of alpha-hydroxytamoxifen. The results showed that alpha-hydroxylation is a common feature of tamoxifen and toremifene metabolism and that alpha-hydroxytamoxifen is unlikely to be the reactive metabolite responsible for the hepatocarcinogenesis in rat, as widely believed.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Microssomos Hepáticos/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Espectrometria de Massas por Ionização por Electrospray/métodos , Toremifeno/metabolismo , Animais , Humanos , Camundongos , Ratos , Toremifeno/análogos & derivados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...