Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Biopharm Drug Dispos ; 44(5): 380-384, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37534716

RESUMO

Trastuzumab deruxtecan (T-DXd, DS-8201a) is an antibody-drug conjugate, comprising an anti-HER2 antibody at a drug-to-antibody ratio of 7-8 with the topoisomerase I inhibitor DXd. In this study, the concentrations of antibody-conjugated DXd and total antibody were determined and observed to decrease over time following intravenous administration of T-DXd to monkeys. The drug-to-antibody ratio of T-DXd also decreased in a time-dependent manner, which reached approximately 2.5 in 21 days after administration. It was suggested that antibody-conjugated DXd of T-DXd was relatively stable in vivo compared with that of other reported antibody-drug conjugates.

2.
Xenobiotica ; 50(10): 1242-1250, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32306807

RESUMO

Trastuzumab deruxtecan (T-DXd, DS-8201a) is an antibody-drug conjugate (ADC), comprising an anti-HER2 antibody (Ab) at a drug-to-Ab ratio of 7-8 with the topoisomerase I inhibitor DXd. In this study, we investigated the pharmacokinetics (PK), biodistribution, catabolism, and excretion profiles of T-DXd in HER2-positive tumour-bearing mice.Following intravenous (iv) administration of T-DXd, the PK profiles of T-DXd and total Ab (the sum of conjugated and unconjugated Ab) were almost similar, indicating that the linker is stable during circulation. Biodistribution studies using radiolabelled T-DXd demonstrated tumour-specific distribution and long-term retention. DXd was the main catabolite released from T-DXd in tumours, with exposure levels at least five times higher than those in normal tissues and seven times higher than those achieved by non-targeted control ADC. Following iv administration of DXd, it was rapidly cleared from the circulation (T1/2; 1.35 h) and excreted mainly through faeces as its intact form.The PK profiles reveal that T-DXd effectively delivers the expected payload, DXd, to tumours, while minimising payload exposure to the systemic circulation and normal tissues. The released DXd is rapidly cleared from systemic circulation, presumably via the bile with negligible metabolism, and excreted through the faeces.


Assuntos
Camptotecina/análogos & derivados , Imunoconjugados/farmacocinética , Trastuzumab/farmacocinética , Ado-Trastuzumab Emtansina , Animais , Camptotecina/farmacocinética , Linhagem Celular Tumoral , Camundongos , Inibidores da Topoisomerase I
3.
Xenobiotica ; 49(9): 1086-1096, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30351177

RESUMO

Trastuzumab deruxtecan (DS-8201a) is an antibody-drug conjugate (ADC) composed of a monoclonal antibody targeting human epidermal growth factor receptor 2 (HER2) conjugated to a topoisomerase I inhibitor (DXd) at a drug-to-antibody ratio (DAR) of 7-8. Here, we examined the pharmacokinetic (PK) profiles of DS-8201a and DXd in cynomolgus monkeys, a cross-reactive species. Following intravenous (iv) administration of DS-8201a, the linker was stable in plasma, and systemic DXd exposure was low. DXd was rapidly cleared following iv dosing. Biodistribution studies revealed that intact DS-8201a was present mostly in the blood without tissue-specific retention. The major pathway of excretion for DXd was the faecal route following iv administration of radiolabelled DS-8201a. The only detectable metabolite in the urine and faeces was unmetabolized DXd. DXd is a substrate of organic anion transporting polypeptides, P-gp, and breast cancer resistance protein. In conclusion, the stable linker in circulation and the high clearance of DXd upon release resulted in the low systemic exposure to DXd. Furthermore, the minimal tissue-specific retention and rapid excretion of DXd into faeces as its unmetabolized form with potentially limited impact on drug - drug interaction as a victim were also critical elements of the PK profile of DS-8201a.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Camptotecina/análogos & derivados , Imunoconjugados/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Ductos Biliares/cirurgia , Células CACO-2 , Camptotecina/farmacocinética , Radioisótopos de Carbono/farmacocinética , Humanos , Inativação Metabólica , Transportador 1 de Ânion Orgânico Específico do Fígado/metabolismo , Macaca fascicularis , Masculino , Proteínas de Neoplasias/metabolismo , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/metabolismo , Distribuição Tecidual , Inibidores da Topoisomerase I/farmacocinética , Trastuzumab
4.
Cancer Sci ; 107(7): 1039-46, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27166974

RESUMO

Antibody-drug conjugates deliver anticancer agents selectively and efficiently to tumor tissue and have significant antitumor efficacy with a wide therapeutic window. DS-8201a is a human epidermal growth factor receptor 2 (HER2)-targeting antibody-drug conjugate prepared using a novel linker-payload system with a potent topoisomerase I inhibitor, exatecan derivative (DX-8951 derivative, DXd). It was effective against trastuzumab emtansine (T-DM1)-insensitive patient-derived xenograft models with both high and low HER2 expression. In this study, the bystander killing effect of DS-8201a was evaluated and compared with that of T-DM1. We confirmed that the payload of DS-8201a, DXd (1), was highly membrane-permeable whereas that of T-DM1, Lys-SMCC-DM1, had a low level of permeability. Under a coculture condition of HER2-positive KPL-4 cells and negative MDA-MB-468 cells in vitro, DS-8201a killed both cells, whereas T-DM1 and an antibody-drug conjugate with a low permeable payload, anti-HER2-DXd (2), did not. In vivo evaluation was carried out using mice inoculated with a mixture of HER2-positive NCI-N87 cells and HER2-negative MDA-MB-468-Luc cells by using an in vivo imaging system. In vivo, DS-8201a reduced the luciferase signal of the mice, indicating suppression of the MDA-MB-468-Luc population; however, T-DM1 and anti-HER2-DXd (2) did not. Furthermore, it was confirmed that DS-8201a was not effective against MDA-MB-468-Luc tumors inoculated at the opposite side of the NCI-N87 tumor, suggesting that the bystander killing effect of DS-8201a is observed only in cells neighboring HER2-positive cells, indicating low concern in terms of systemic toxicity. These results indicated that DS-8201a has a potent bystander effect due to a highly membrane-permeable payload and is beneficial in treating tumors with HER2 heterogeneity that are unresponsive to T-DM1.


Assuntos
Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Efeito Espectador/efeitos dos fármacos , Camptotecina/análogos & derivados , Imunoconjugados/imunologia , Imunoconjugados/farmacologia , Neoplasias/genética , Neoplasias/patologia , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Ado-Trastuzumab Emtansina , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Camptotecina/imunologia , Camptotecina/farmacologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Maitansina/análogos & derivados , Maitansina/imunologia , Maitansina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/enzimologia , Neoplasias/imunologia , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/patologia , Inibidores da Topoisomerase I/farmacologia , Trastuzumab
5.
Bioorg Med Chem Lett ; 26(20): 5069-5072, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27599744

RESUMO

To establish a novel and widely applicable payload-linker technology for antibody-drug conjugates (ADCs), we have focused our research on applying exatecan mesylate (DX-8951f), a potent topoisomerase I inhibitor, which exhibits extensive antitumor activity as well as significant myelotoxicity, as the payload part. Through this study, we discovered a promising exatecan derivative (DX-8951 derivative, DXd), that has the characteristics of low membrane permeability and shows considerably less myelotoxicity than that shown by exatecan mesylate in an in vitro human colony forming unit-granulocyte macrophage assay. DXd was further used for drug conjugation by using commercially or clinically useful monoclonal antibodies to evaluate the potency of the ADC. The result revealed that the DXd-ADCs targeting CD30, CD33, and CD70 were effective against each of their respective target-expressing tumor cell lines. Moreover, a novel DXd-ADC targeting B7-H3, which is a new target for ADCs, also showed potent antitumor efficacy both in vitro and in vivo. In conclusion, this study showed that this novel topoisomerase I inhibitor-based ADC technology is widely applicable to a diverse number of antibodies and is expected to mitigate myelotoxicity, thereby possibly resulting in better safety profiles than that of existing ADC technologies.


Assuntos
Imunoconjugados/farmacologia , Inibidores da Topoisomerase I/farmacologia , Desenho de Fármacos , Humanos
6.
Drug Metab Pharmacokinet ; 56: 101001, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38643548

RESUMO

Trastuzumab deruxtecan (T-DXd) is an antibody-drug conjugate used for cancer treatment comprising an anti-human epidermal growth factor receptor type 2 (HER2) antibody and the topoisomerase I inhibitor DXd. The present study investigated the intratumor fate of T-DXd. Fluorescence-labeled T-DXd was found to accumulate in tumors of HER2-positive tumor xenograft mice and was observed to be distributed within lysosomes of in vitro tumor cells in accordance with their HER2 expression. DXd was released by cysteine proteases, including cathepsins, in lysosomal fractions in vitro in response to the pH. Tumor slices obtained from HER2-positive tumor xenograft mice treated with T-DXd were examined by semi-quantitative and three-dimensional immunohistochemical assays using phosphor-integrated dots, which visualized DXd-related signals in the nucleus, the site of topoisomerase I inhibition. In addition, based on the data showing the antibody component of T-DXd barely distributed in the nucleus, it was suggested that the DXd-related signals detected in the nucleus were predominantly derived from free DXd. These observations help support the mode of action of T-DXd from the perspective of drug disposition.


Assuntos
Camptotecina , Imunoconjugados , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto , Trastuzumab/farmacologia , Animais , Humanos , Camundongos , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Camptotecina/metabolismo , Imunoconjugados/farmacologia , Feminino , Camundongos Nus , Inibidores da Topoisomerase I/farmacologia , Inibidores da Topoisomerase I/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Linhagem Celular Tumoral , Camundongos Endogâmicos BALB C , Antineoplásicos Imunológicos/farmacologia
7.
Eur J Drug Metab Pharmacokinet ; 48(5): 541-552, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37530974

RESUMO

BACKGROUND AND OBJECTIVE: Fc fusion is an effective strategy for extending the half-lives of therapeutic proteins. This study aimed to evaluate the applicability of a human pharmacokinetics prediction method for Fc-fusion proteins by extending on reported methods for monoclonal antibodies (mAbs). METHODS: To predict human pharmacokinetic profiles following intravenous (IV) dosing, the pharmacokinetic data for 11 Fc-fusion proteins in monkeys were analysed by two approaches: a species-invariant time method with a range of allometric exponents in clearance (CL, 0.7-1.0) and a two-compartment model reported for mAbs. The pharmacokinetic profiles following subcutaneous (SC) dosing were predicted by simple dose normalisation from monkeys or using the geometric means of the absorption rate constant (Ka) and bioavailability (BA) for mAbs or Fc-fusion proteins in humans and compared. RESULTS: In the case of IV administration, the area under the curve could be predicted for more than 85% of Fc-fusion proteins within a twofold difference from the observed value using the species-invariant time method (scaling exponent for CL, 0.95). For SC dosing, incorporating the geometric means of absorption parameters for both mAbs (BA 68.2%, Ka 0.287 day-1) and Fc-fusion proteins (BA 63.0%, Ka 0.209 day-1) in humans provided better accuracy than simple normalisation from monkeys. CONCLUSION: We have successfully predicted the human pharmacokinetic profiles of Fc-fusion proteins for both IV and SC administration within twofold of the observed value from monkey pharmacokinetic data by extending on reported methods for mAbs. This method will facilitate drug discovery and development of Fc-fusion proteins.


Assuntos
Anticorpos Monoclonais , Modelos Biológicos , Humanos , Animais , Anticorpos Monoclonais/farmacocinética , Disponibilidade Biológica , Administração Intravenosa , Haplorrinos , Farmacocinética
8.
J Clin Invest ; 133(23)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37788110

RESUMO

Glycogen storage disease type 1a (GSD1a) is caused by a congenital deficiency of glucose-6-phosphatase-α (G6Pase-α, encoded by G6PC), which is primarily associated with life-threatening hypoglycemia. Although strict dietary management substantially improves life expectancy, patients still experience intermittent hypoglycemia and develop hepatic complications. Emerging therapies utilizing new modalities such as adeno-associated virus and mRNA with lipid nanoparticles are under development for GSD1a but potentially require complicated glycemic management throughout life. Here, we present an oligonucleotide-based therapy to produce intact G6Pase-α from a pathogenic human variant, G6PC c.648G>T, the most prevalent variant in East Asia causing aberrant splicing of G6PC. DS-4108b, a splice-switching oligonucleotide, was designed to correct this aberrant splicing, especially in liver. We generated a mouse strain with homozygous knockin of this variant that well reflected the pathophysiology of patients with GSD1a. DS-4108b recovered hepatic G6Pase activity through splicing correction and prevented hypoglycemia and various hepatic abnormalities in the mice. Moreover, DS-4108b had long-lasting efficacy of more than 12 weeks in mice that received a single dose and had favorable pharmacokinetics and tolerability in mice and monkeys. These findings together indicate that this oligonucleotide-based therapy could provide a sustainable and curative therapeutic option under easy disease management for GSD1a patients with G6PC c.648G>T.


Assuntos
Doença de Depósito de Glicogênio Tipo I , Hipoglicemia , Humanos , Camundongos , Animais , Oligonucleotídeos/genética , Camundongos Knockout , Doença de Depósito de Glicogênio Tipo I/genética , Doença de Depósito de Glicogênio Tipo I/terapia , Doença de Depósito de Glicogênio Tipo I/complicações , Fígado/patologia , Glucose-6-Fosfatase/genética , Hipoglicemia/genética , Hipoglicemia/prevenção & controle
9.
Antimicrob Agents Chemother ; 53(11): 4845-51, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19687241

RESUMO

CS-8958 is a prodrug of the pharmacologically active form R-125489, a selective neuraminidase inhibitor, and has long-acting anti-influenza virus activity in vivo. In this study, the tissue distribution profiles after a single intranasal administration of CS-8958 (0.5 micromol/kg of body weight) to mice were investigated, focusing especially on the retention of CS-8958 in the respiratory tract by comparing it with R-125489 and a marketed drug, zanamivir. After administration of [(14)C]CS-8958, radioactivity was retained in the respiratory tract over long periods. At 24 h postdose, the radioactivity concentrations after administration of [(14)C]CS-8958 were approximately 10-fold higher in both the trachea and the lung than those of [(14)C]R-125489 and [(14)C]zanamivir. The [(14)C]CS-8958-derived radioactivity present in these two tissues consisted both of unchanged CS-8958 and of R-125489 at 1 h postdose, while only R-125489, and no other metabolites, was detected at 24 h postdose. After administration of unlabeled CS-8958, CS-8958 was rapidly eliminated from the lungs, whereas the lung R-125489 concentration reached a maximum at 3 h postdose and gradually declined, with an elimination half-life of 41.4 h. The conversion of CS-8958 to R-125489 was observed in mouse trachea and lung S9 fractions and was inhibited by esterase inhibitors, such as diisopropylfluorophosphate and bis-p-nitrophenylphosphate. These results demonstrated that CS-8958 administered intranasally to mice was efficiently converted to R-125489 by a hydrolase(s) such as carboxylesterase, and then R-125489 was slowly eliminated from the respiratory tract. These data support the finding that CS-8958 has potential as a long-acting neuraminidase inhibitor, leading to significant efficacy as an anti-influenza drug by a single treatment.


Assuntos
Antivirais/farmacocinética , Inibidores Enzimáticos/farmacocinética , Pulmão/metabolismo , Neuraminidase/antagonistas & inibidores , Orthomyxoviridae/efeitos dos fármacos , Pró-Fármacos/farmacocinética , Traqueia/metabolismo , Zanamivir/análogos & derivados , Animais , Autorradiografia , Esterases/antagonistas & inibidores , Feminino , Guanidinas , Camundongos , Camundongos Endogâmicos BALB C , Piranos , Ácidos Siálicos , Zanamivir/farmacocinética
10.
Clin Cancer Res ; 22(20): 5097-5108, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27026201

RESUMO

PURPOSE: An anti-HER2 antibody-drug conjugate with a novel topoisomerase I inhibitor, DS-8201a, was generated as a new antitumor drug candidate, and its preclinical pharmacologic profile was assessed. EXPERIMENTAL DESIGN: In vitro and in vivo pharmacologic activities of DS-8201a were evaluated and compared with T-DM1 in several HER2-positive cell lines and patient-derived xenograft (PDX) models. The mechanism of action for the efficacy was also evaluated. Pharmacokinetics in cynomolgus monkeys and the safety profiles in rats and cynomolgus monkeys were assessed. RESULTS: DS-8201a exhibited a HER2 expression-dependent cell growth-inhibitory activity and induced tumor regression with a single dosing at more than 1 mg/kg in a HER2-positive gastric cancer NCI-N87 model. Binding activity to HER2 and ADCC activity of DS-8201a were comparable with unconjugated anti-HER2 antibody. DS-8201a also showed an inhibitory activity to Akt phosphorylation. DS-8201a induced phosphorylation of Chk1 and Histone H2A.X, the markers of DNA damage. Pharmacokinetics and safety profiles of DS-8201a were favorable and the highest non-severely toxic dose was 30 mg/kg in cynomolgus monkeys, supporting DS-8201a as being well tolerated in humans. DS-8201a was effective in a T-DM1-insensitive PDX model with high HER2 expression. DS-8201a, but not T-DM1, demonstrated antitumor efficacy against several breast cancer PDX models with low HER2 expression. CONCLUSIONS: DS-8201a exhibited a potent antitumor activity in a broad selection of HER2-positive models and favorable pharmacokinetics and safety profiles. The results demonstrate that DS-8201a will be a valuable therapy with a great potential to respond to T-DM1-insensitive HER2-positive cancers and low HER2-expressing cancers. Clin Cancer Res; 22(20); 5097-108. ©2016 AACR.


Assuntos
Anticorpos Monoclonais Humanizados , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Camptotecina/análogos & derivados , Imunoconjugados , Neoplasias Pancreáticas/tratamento farmacológico , Receptor ErbB-2/antagonistas & inibidores , Inibidores da Topoisomerase I/farmacologia , Ado-Trastuzumab Emtansina , Animais , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais Humanizados/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Camptotecina/efeitos adversos , Camptotecina/farmacocinética , Camptotecina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 1 do Ponto de Checagem/metabolismo , Feminino , Histonas/metabolismo , Humanos , Imunoconjugados/efeitos adversos , Imunoconjugados/farmacocinética , Imunoconjugados/farmacologia , Macaca fascicularis , Maitansina/análogos & derivados , Maitansina/farmacologia , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptor ErbB-2/imunologia , Trastuzumab/farmacologia
11.
Drug Metab Pharmacokinet ; 27(3): 354-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22146109

RESUMO

We have previously reported that human total body clearance (CL) and steady-state volume of distribution (Vss) of monoclonal antibodies (mAbs) could be predicted reasonably well from monkey data alone using simple allometry with scaling exponents of 0.79 and 1.12 (for soluble targets), and 0.96 and 1.00 (for membrane-bound targets). In the present study, to predict the plasma concentration-time profiles of mAbs in humans, we employed simple dose-normalization and species-invariant time methods (elementary Dedrick plot and complex Dedrick plot), based on the monkey data and the scaling exponents we previously determined. The results demonstrated that the species-invariant time methods were able to provide higher accuracy of prediction than simple dose-normalization, regardless of the type of target antigens (soluble or membrane-bound). The accuracy between elementary Dedrick plot and complex Dedrick plot was nearly equivalent. The predicted human CL and Vss using species-invariant time methods were within mostly 2-fold differences from the observed values. The prediction not only of pharmacokinetic (PK) parameters but also of the plasma concentration-time profile in humans can serve as guidelines for better planning of clinical studies on mAbs.


Assuntos
Anticorpos Monoclonais/farmacocinética , Modelos Biológicos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/sangue , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais Murinos/administração & dosagem , Anticorpos Monoclonais Murinos/sangue , Anticorpos Monoclonais Murinos/farmacocinética , Antígenos/química , Relação Dose-Resposta a Droga , Meia-Vida , Humanos , Macaca fascicularis , Macaca mulatta , Taxa de Depuração Metabólica , Pan troglodytes , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/sangue , Proteínas Recombinantes de Fusão/farmacocinética , Solubilidade , Especificidade da Espécie
12.
Drug Metab Pharmacokinet ; 26(4): 423-30, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21606605

RESUMO

Interspecies allometric scaling is a useful tool for calculating human pharmacokinetic (PK) parameters from data in animals. In this study, in order to determine the scaling exponent in a simple allometric equation that can predict human clearance (CL) and distribution volume at steady state (Vss) of monoclonal antibodies (mAbs) from monkey data alone, PK data of 24 mAbs were collected and analyzed according to the types of targeted antigens (soluble or membrane-bound antigens). Based on the observed PK data in humans (at clinical doses) and monkeys (at >1 mg/kg), where the PK is expected to be linear, the mean scaling exponents in the allometric equation for CL and Vss, respectively, against body weight were calculated to be 0.79 and 1.12 [95% confidence intervals (CIs): 0.69-0.89 and 0.96-1.28] for soluble antigens, and 0.96 and 1.00 (95% CIs: 0.83-1.09 and 0.87-1.13) for membrane-bound antigens. Using these exponents and monkey PK data (at >1 mg/kg) alone, both human CL and Vss of mAbs can be predicted with reasonable accuracy, i.e., within 2-fold of the observed values. Compared with traditional allometric scaling using PK data from three or more preclinical species, this approach is simple, quick, resource-saving, and useful in drug discovery and development.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais/farmacocinética , Adalimumab , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais Humanizados/sangue , Tamanho Corporal , Peso Corporal , Cetuximab , Bases de Dados Factuais , Relação Dose-Resposta a Droga , Descoberta de Drogas , Haplorrinos , Humanos , Modelos Animais , Especificidade da Espécie
13.
Drug Metab Dispos ; 35(10): 1846-52, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17620346

RESUMO

In rats, it has been reported that rofecoxib, a cyclooxygenase-2 (COX-2) inhibitor, reacts with the aldehyde group of allysine in elastin to give a condensation covalent adduct, thereby preventing the formation of cross-linkages in the elastin and causing degradation of the elastic fibers in aortas in vivo. Acid, organic solvent, and proteolytic enzyme treatments of human aortic homogenate after incubation with [(14)C]rofecoxib demonstrated that most of the radioactivity is covalently bound to elastin. The in vitro covalent binding was inhibited in the presence of beta-aminopropionitrile, D-penicillamine, and hydralazine, which suggested that the aldehyde group of allysine in human elastin was relevant to the covalent binding. The in vitro covalent binding of [(14)C]rofecoxib was significantly decreased by the addition of only nonradiolabeled rofecoxib but not the other COX-2 inhibitors, celecoxib, valdecoxib, etoricoxib, and CS-706 [2-(4-ethoxyphenyl)-4-methyl 1-(4-sulfamoylphenyl)-1H-pyrrole], a novel selective COX-2 inhibitor. All the above COX-2 inhibitors except for rofecoxib had no reactivity with the aldehyde group of benzaldehyde used as a model compound of allysine aldehyde under a physiological pH condition. On the other hand, no retention of the radioactivity of [(14)C]rofecoxib was observed in human aortic endothelial cells in vitro, suggesting that rofecoxib is not retained in aortic endothelial cells in vivo. These results suggest that rofecoxib, but not other COX-2 inhibitors, is capable of covalently binding to the aldehyde group of allysine in human elastin. This might be one of the main causes of cardiovascular events by rofecoxib in clinical situations.


Assuntos
Ácido 2-Aminoadípico/análogos & derivados , Aldeídos/metabolismo , Inibidores de Ciclo-Oxigenase 2/metabolismo , Elastina/metabolismo , Lactonas/metabolismo , Sulfonas/metabolismo , Ácido 2-Aminoadípico/metabolismo , Idoso , Animais , Aorta Abdominal/metabolismo , Aorta Torácica/metabolismo , Linhagem Celular , Feminino , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
14.
J Pharmacol Exp Ther ; 320(3): 1195-203, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17164475

RESUMO

We have previously reported that oral administration of [(14)C]rofecoxib to rats resulted in the long retention of radioactivity by the aorta as a consequence of covalent binding to elastin. Treatment of rats with alpha-phenyl-alpha-propylbenzeneacetic acid 2-[diethylamino]-ethyl ester hydrochloride (SKF-525A), a cytochrome P450 inhibitor, significantly decreased the systemic exposure of 5-hydroxyrofecoxib, one of the main metabolites of rofecoxib, whereas there was no statistically significant change in the retention of radioactivity from [(14)C]rofecoxib in the aorta. On the other hand, the aortic retention of radioactivity closely correlated to the systemic exposure of unchanged rofecoxib in the dose range between 2 and 10 mg/kg. A covalent binding study of [(14)C]rofecoxib in vitro using rat aorta homogenate in the presence of d-penicillamine, hydralazine, beta-aminopropionitrile, and sodium borohydride suggested that the aldehyde group of allysine in elastin was relevant to the covalent binding. In a model reaction using benzaldehyde, rofecoxib but not 5-hydroxyrofecoxib reacted with the aldehyde group of benzaldehyde in a manner of condensation reaction under a physiological pH condition. A histopathological examination using an electron microscope demonstrated that multiple oral administration of rofecoxib to rats caused marked degradation of the elastic fiber system of the aorta. These results suggested that rofecoxib as such is reactive in vivo, undergoing a condensation reaction with allysine, thereby preventing the formation of cross-linkages in elastin, i.e., desmosine and isodesmosine, and causing the degradation of the elastic fibers.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Aorta Torácica/efeitos dos fármacos , Elastina/metabolismo , Lactonas/farmacologia , Sulfonas/farmacologia , Animais , Anti-Inflamatórios não Esteroides/sangue , Anti-Inflamatórios não Esteroides/farmacocinética , Aorta Torácica/metabolismo , Aorta Torácica/ultraestrutura , Cromatografia Líquida de Alta Pressão , Técnicas In Vitro , Lactonas/sangue , Lactonas/farmacocinética , Masculino , Microscopia Eletrônica de Transmissão , Estrutura Molecular , Ligação Proteica , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Sulfonas/sangue , Sulfonas/farmacocinética , Espectrometria de Massas em Tandem
15.
Drug Metab Dispos ; 34(8): 1417-22, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16679386

RESUMO

Rofecoxib is a cyclooxygenase-2 (COX-2) inhibitor that has been withdrawn from the market because of an increased risk of cardiovascular (CV) events. With a special focus on the arteries, the distribution profiles of radioactivity in rats orally administered [14C]rofecoxib were investigated in comparison with two other COX-2 inhibitors, [14C]celecoxib and [14C]CS-706 (2-(4-ethoxyphenyl)-4-methyl 1-(4-sulfamoylphenyl)-1H-pyrrole), a novel selective COX-2 inhibitor. Whole-body autoradioluminography and quantitative determination of the tissue concentrations showed that considerable radioactivity is retained by and accumulated in the thoracic aorta of rats after oral administration of [14C]rofecoxib, but not [14C]celecoxib or [14C]CS-706. Acid, organic solvent, and proteolytic enzyme treatments of aorta retaining high levels of radioactivity from [14C]rofecoxib demonstrated that most of the radioactivity is covalently bound to elastin. In agreement with this result, the radioactivity was found to be highly localized on the elastic fibers in the aorta by microautoradiography. The retention of radioactivity on the elastic fibers was also observed in the aortic arch and the coronary artery. These findings indicate that [14C]rofecoxib and/or its metabolite(s) are covalently bound to elastin in the arteries. These data are consistent with the suggestion of modified arterial elasticity leading to an increased risk of CV events after long-term treatment with rofecoxib.


Assuntos
Aorta Torácica/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacocinética , Elastina/metabolismo , Lactonas/farmacocinética , Pirazóis/farmacocinética , Pirróis/farmacocinética , Sulfonamidas/farmacocinética , Sulfonas/farmacocinética , Animais , Radioisótopos de Carbono , Celecoxib , Inibidores de Ciclo-Oxigenase 2/sangue , Lactonas/sangue , Masculino , Ligação Proteica , Pirazóis/sangue , Pirróis/sangue , Ratos , Ratos Sprague-Dawley , Sulfonamidas/sangue , Sulfonas/sangue , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA