Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Food Chem Toxicol ; 166: 113246, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35728726

RESUMEN

Irinotecan is a first-line treatment for colorectal cancer and the prodrug of 7-ethyl-10-hydroxy-camptothecin (SN-38). However, its fatal gastrointestinal (GI) toxicity raises serious concern. In liver, irinotecan generates its inactive metabolite, SN-38G via UDP-glucuronosyltransferase (UGT)1A1. Subsequently, SN-38G is excreted into GI tract where it is reactivated by microbiome to yield the toxic metabolite, SN-38. Activation of toll-like receptor (TLR)/myeloid differentiation primary response 88 (MyD88) by bacterial endotoxin decreases drug-metabolizing enzymes. In this study, we treated C57BL6/J mice with 50 mg/kg irinotecan once daily until observing grade 4 diarrhea. Mice were sacrificed on day0, day2 and day8. Based on the finding in C57BL6/J mice, we repeated the treatment in Tlr2-/-, Tlr4-/- and Myd88-/- mice to determine the impact of inflammation on UGT metabolism. Our toxicity study in C57BL6/J mice showed that mice started bloody diarrhea after 6 days' injection of irinotecan. Ugt1a1 expression in GI tract started decreasing after 24h since first dose, before the onset of diarrhea. In Tlr4-/- and Myd88-/- mice, no Ugt1a1 reduction was observed in distal GI tract after irinotecan injection. In Tlr2-/- mice, intestinal Ugt1a1 expression was down-regulated. Our results indicate that after two doses of irinotecan, mice started losing capability of detoxifying SN-38. TLR4 plays more important role in Ugt1a1 reduction than TLR2, despite that TLR2 and TLR4 share MyD88 as common adaptor protein. We concluded that irinotecan reduced intestinal Ugt1a1 via TLR4/MyD88 pathway, which eventually triggers the onset of diarrhea. Our finding unveils a novel mechanism underlying irinotecan-induced diarrhea and provides a new direction to prevent chemotherapy side effect.


Asunto(s)
Antineoplásicos Fitogénicos , Glucuronosiltransferasa , Irinotecán , Factor 88 de Diferenciación Mieloide , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Camptotecina/toxicidad , Diarrea/inducido químicamente , Diarrea/metabolismo , Glucuronosiltransferasa/metabolismo , Irinotecán/efectos adversos , Ratones , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Uridina Difosfato
3.
Chem Biol Interact ; 360: 109946, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35430260

RESUMEN

Irinotecan, a first-line chemotherapy for gastrointestinal (GI) cancers has been causing fatal toxicities like bloody diarrhea and steatohepatitis for years. Irinotecan goes through multiple-step drug metabolism after injection and one of its intermediates 7-ethyl-10-hydroxy-camptothecin (SN-38) is responsible for irinotecan side effect. However, it is unclear what is the disposition kinetics of SN-38 in the organs subjected to toxicity. No studies ever quantified the effect of each enzyme or transporter on SN-38 distribution. In current study, we established a new physiologically based pharmacokinetic (PBPK) model to predict the disposition kinetics of irinotecan. The PBPK model was calibrated with in-house mouse pharmacokinetic data and evaluated with external datasets from the literature. We separated the contribution of each parameters in irinotecan pharmacokinetics by calculating the normalized sensitivity coefficient (NSC). The model gave robust prediction of SN-38 distribution in GI tract, the site of injury. We identified that bile excretion and UDP-glucuronosyltransferases (UGT) played more important roles than fecal excretion and renal clearance in SN-38 pharmacokinetics. Our NSC showed that the impact of enzyme and transporter on irinotecan and SN-38 pharmacokinetics evolved when time continued. Additionally, we mapped out the effect of inflammation on irinotecan metabolic pathways with PBPK modelling. We discovered that inflammation significantly increased the blood and liver exposure of irinotecan and SN-38 in the mice receiving bacterial endotoxin. Inflammation suppressed UGT, microbial metabolism but increased fecal excretion. The present PBPK model can serve as an efficacious and versatile tool to quantitively assess the risk of irinotecan toxicity.


Asunto(s)
Antineoplásicos Fitogénicos , Camptotecina , Animales , Antineoplásicos Fitogénicos/toxicidad , Camptotecina/efectos adversos , Diarrea/inducido químicamente , Glucuronosiltransferasa/metabolismo , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Irinotecán , Ratones
4.
Toxicol Res (Camb) ; 11(1): 184-194, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35237423

RESUMEN

Neratinib is a pan-HER tyrosine kinase inhibitor newly approved by FDA in 2017 to treat HER2-positive breast cancer, but the phase III trial of neratinib showed that 96% of the patients taking neratinib experienced diarrhea. So far very few mechanistic studies explore neratinib-induced gastrointestinal (GI) toxicity. Hereby, we performed toxicity studies in mice to characterize the potential mechanism underlying this adverse effect. C57BL/6 J mice were separated into three groups A, B, C. Group A received vehicle; group B was orally dosed with 100 mg/kg neratinib once daily for 18 days. Group C was dosed with 100 mg/kg neratinib for 12 days and switched to vehicle for 6 days. Intestine and liver were collected for further analysis. Human intestine-derived cells were treated with neratinib in vitro. Our results showed that 12 days treatment of neratinib caused persistent histological damage in mouse GI tract. Both gene expression and activity of Cyp3a11, the major enzyme metabolizing neratinib in mice was reduced in small intestine. The gene expression of proinflammatory cytokines increased throughout the GI tract. Such damages were not recovered after 6 days without neratinib treatment. In addition, in vitro data showed that neratinib was potent in killing human intestine-derived cell lines. Based on such findings, we hypothesized that neratinib downregulates intestinal CYP3A enzyme to cause excessive drug disposition, eventually leading to gut injury.

5.
Expert Rev Vaccines ; 20(5): 623-634, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33666138

RESUMEN

OBJECTIVE: Infectious disease emergencies like the 2013-2016 Ebola epidemic and the 2009 influenza and current SARS-CoV-2 pandemics illustrate that vaccines are now given to diverse populations with preexisting pathologies requiring pharmacological management. Many natural biomolecules (steroid hormones, fatty acids, vitamins) and ~60% of prescribed medications are processed by hepatic cytochrome P450 (CYP) 3A4. The objective of this work was to determine the impact of infection and vaccines on drug metabolism. METHODS: The impact of an adenovirus-based vaccine expressing Ebola glycoprotein (AdEBO) and H1N1 and H3N2 influenza viruses on hepatic CYP 3A4 and associated nuclear receptors was evaluated in human hepatocytes (HC-04 cells) and in mice. RESULTS: CYP3A activity was suppressed by 55% in mice 24 h after administration of mouse-adapted H1N1, while ˂10% activity remained in HC-04 cells after infection with H1N1 and H3N2 due to global suppression of cellular translation capacity, indicated by reduction (70%, H1N1, 56%, H3N2) of phosphorylated eukaryotic translation initiation factor 4e (eIF4E). AdEBO suppressed CYP3A activity in vivo (44%) and in vitro (26%) 24 hours after infection. CONCLUSION: As the clinical evaluation of vaccines for SARS-CoV-2 and other global pathogens rise, studies to evaluate the impact of new vaccines and emerging pathogens on CYP3A4 and other metabolic enzymes are warranted to avoid therapeutic failures that could further compromise the public health during infectious disease emergencies.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Hepatocitos/enzimología , Hepatocitos/metabolismo , Hígado/enzimología , Hígado/metabolismo , Preparaciones Farmacéuticas/metabolismo , Animales , Células Cultivadas , Factor 4E Eucariótico de Iniciación , Humanos , Inmunización/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
6.
Expert Opin Drug Metab Toxicol ; 16(11): 1109-1124, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32841068

RESUMEN

INTRODUCTION: Toxicity of chemotherapy drugs is the leading cause of poor therapeutic outcome in many cancer patients. Gastrointestinal (GI) toxicity and hepatotoxicity are among the most common side effects of current chemotherapies. Emerging studies indicate that many chemotherapy-induced toxicities are driven by drug metabolism, but very few reviews summarize the role of drug metabolism in chemotherapy-induced GI toxicity and hepatotoxicity. In this review, we highlighted the importance of drug metabolizing enzymes (DMEs) in chemotherapy toxicity. AREAS COVERED: Our review demonstrated that altered activity of DMEs play important role in chemotherapy-induced GI toxicity and hepatotoxicity. Besides direct changes in catalytic activities, the transcription of DMEs is also affected by inflammation, cell-signaling pathways, and/or by drugs in cancer patients due to the disease etiology. EXPERT OPINION: More studies should focus on how DMEs are altered during chemotherapy treatment, and how such changes affect the metabolism of chemotherapy drug itself. This mutual interaction between chemotherapies and DMEs can lead to excessive exposure of parent drug or toxic metabolites which ultimately cause GI adverse effect.


Asunto(s)
Antineoplásicos/efectos adversos , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedades Gastrointestinales/inducido químicamente , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Enfermedades Gastrointestinales/fisiopatología , Humanos , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos
7.
Toxicol Appl Pharmacol ; 398: 115032, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32387182

RESUMEN

BACKGROUND AND PURPOSE: Irinotecan-induced diarrhea (IID) results from intestinal damages by its active metabolite SN-38. Alleviation of these damages has focused on lowering luminal SN-38 concentrations. However, it is unclear if the enteric bioavailability of SN-38 is mostly dependent on luminal SN-38 concentrations. EXPERIMENTAL APPROACH: Irinotecan (50 mg/kg, i.p. once daily for 6 days) was administered to female wildtype FVB, Mdr1a (-/-), Mrp2 (-/-) and Bcrp1 (-/-) mice for pharmacokinetic (PK), toxicokinetic (TK) and biodistribution studies. Plasma PK/TK profiles and tissues drug distribution were determined after first or sixth daily doses, along with activities of blood and gut esterases and intestinal Ugts. Caco-2 cells and bile-cannulate mice were used to further investigate intestinal and biliary disposition of irinotecan and its metabolites. KEY RESULTS: Significant differences in IID severity were observed with the susceptible rank of Bcrp1(-/-) > wildtype FVB > Mdr1a(-/-) > Mrp2(-/-). This rank order did not correlate with biliary excretion rates of SN-38/SN-38G. Rather, the severity was best correlated (R = 0.805) with the intestinal ratio of Css SN-38/SN-38G, a measure of gut Ugt activity. On the contrary, IID was poorly correlated with plasma AUC ratio of SN-38/SN-38G (R = 0.227). Increased intestinal esterase activities due to repeated dosing and gut efflux transporter functionality are the other key factors that determine SN-38 enteric exposures. CONCLUSION AND IMPLICATIONS: Intestinal SN-38 exposure is mainly affected by intestinal Ugt activities and blood esterase activities, and strongly correlated with severity of IID. Modulating intestinal SN-38 concentration and gut Ugt expression should be the focus of future studies to alleviate IID.


Asunto(s)
Diarrea/inducido químicamente , Glucuronosiltransferasa/metabolismo , Intestinos/efectos de los fármacos , Irinotecán/farmacología , Animales , Antineoplásicos Fitogénicos , Área Bajo la Curva , Bilis/metabolismo , Sistema Biliar/efectos de los fármacos , Sistema Biliar/metabolismo , Células CACO-2 , Línea Celular Tumoral , Diarrea/metabolismo , Esterasas/metabolismo , Femenino , Humanos , Ratones , Distribución Tisular/efectos de los fármacos
8.
Chem Biol Interact ; 316: 108933, 2020 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-31870839

RESUMEN

Irinotecan is a chemotherapeutic drug used in the treatment of advanced colorectal cancer and elevated blood concentrations of its active metabolite, SN-38 leads to increased gastrointestinal (GI) toxicity and diarrhea in patients. In this study, we investigated the effects of inflammation on the pharmacokinetics (PK) of irinotecan (CPT-11) and its active metabolite, SN-38. Mice were i.p.-injected with either saline or lipopolysaccharide (LPS) to induce inflammation. After 16 h, irinotecan was administered orally. Blood was collected from the tail vein of mice from 0 to 24 h after dosing. Concentrations of irinotecan, SN-38 and SN-38G were analyzed using LC-MS/MS. The AUC, Cmax, and tmax were derived using WinNonlin® 5.2. A PK model was developed using Phoenix NLME® to describe the PK of irinotecan and SN-38 during inflammation. Results indicated a significant increase in the blood concentrations of irinotecan and SN-38 in mice during inflammation. The AUC of irinotecan and SN-38 in LPS group were 2.6 and 2-folds, respectively, of those in control saline-treated mice. The Cmax of irinotecan and SN-38 in LPS treated mice were 2.4 and 2.3-folds of those in saline-treated mice. The PK model was successfully developed and validated. The best-fit plots of individual PK analysis showed a good correlation between observed and predicted concentrations of irinotecan and SN-38. Together, this study reveals that SN-38 concentrations are elevated during inflammation, which may increase the GI toxicity and diarrhea in patients who receive irinotecan; and the developed PK model can quantitatively describe the PK of irinotecan and SN-38 during inflammation.


Asunto(s)
Inflamación/etiología , Irinotecán/farmacocinética , Administración Oral , Animales , Área Bajo la Curva , Cromatografía Líquida de Alta Presión , Semivida , Inflamación/metabolismo , Irinotecán/administración & dosificación , Irinotecán/sangre , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Teóricos , Curva ROC , Espectrometría de Masas en Tándem
9.
Curr Cancer Drug Targets ; 19(7): 551-560, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31509102

RESUMEN

BACKGROUND: Diarrhea is a severe side effect of irinotecan, a pro-drug of SN-38 used for the treatment of many types of cancers. Pre-clinical and clinical studies showed that decreasing the colonic exposure of SN-38 can mitigate irinotecan-induced diarrhea. OBJECTIVE: The purpose of this study is to evaluate the anti-diarrhea potential of Xiao-Chai-Hu-Tang (XCHT), a traditional Chinese herbal formula, against irinotecan-induced diarrhea by determining if and how XCHT alters the disposition of SN-38. METHODS: LC-MS/MS was used to quantify the concentrations of irinotecan and its major metabolites (i.e., SN-38, SN-38G). An Intestinal perfusion model was used to determine the effect of XCHT on the biliary and intestinal secretions of irinotecan, SN-38, and SN-38G. Pharmacokinetic (PK) studies were performed to determine the impact of XCHT on the blood and fecal concentrations of irinotecan, SN-38, and SN-38G. RESULTS: The results showed that XCHT significantly inhibits both biliary and intestinal excretions of irinotecan, SN-38, and SN-38G (range: 35% to 95%). PK studies revealed that the fecal concentrations of irinotecan and SN-38 were significantly decreased from 818.35 ± 120.2 to 411.74 ± 138.83 µg/g or from 423.95 ± 76.44 to 245.63 ± 56.72 µg/g (p<0.05) by XCHT, respectively, suggesting the colonic exposure of SN-38 is significantly decreased by XCHT. PK studies also showed that the plasma concentrations of irinotecan, SN-38, and SN-38G were not affected by XCHT. CONCLUSION: In conclusion, XCHT significantly decreased the exposure of SN-38 in the gut without affecting its plasma level, thereby possessing the potential of alleviating irinotecan-induced diarrhea without negatively impacting its therapeutic efficacy.


Asunto(s)
Sistema Biliar/metabolismo , Diarrea/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Mucosa Intestinal/metabolismo , Irinotecán/toxicidad , Animales , Sistema Biliar/efectos de los fármacos , Diarrea/inducido químicamente , Diarrea/metabolismo , Diarrea/patología , Mucosa Intestinal/efectos de los fármacos , Irinotecán/farmacocinética , Masculino , Ratas , Ratas Wistar , Distribución Tisular
10.
Sci Rep ; 9(1): 6663, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31040347

RESUMEN

Cytochrome P450 (CYP)3A is the most abundant CYP enzyme in the human liver, and a functional impairment of this enzyme leads to unanticipated adverse reactions and therapeutic failures; these reactions result in the early termination of drug development or the withdrawal of drugs from the market. The transcriptional regulation mechanism of the Cyp3a gene is not fully understood and requires a thorough investigation. We mapped the transcriptome of the Cyp3a gene in a mouse model. The Cyp3a gene was induced using the mPXR activator pregnenolone-16alpha-carbonitrile (PCN) and was subsequently downregulated using lipopolysaccharide (LPS). Our objective was to identify the transcription factors (TFs), epigenetic modulators and molecular pathways that are enriched or repressed by PCN and LPS based on a gene set enrichment analysis. Our analysis shows that 113 genes were significantly upregulated (by at least 1.5-fold) with PCN treatment, and that 834 genes were significantly downregulated (by at least 1.5-fold) with LPS treatment. Additionally, the targets of the 536 transcription factors were enriched by a combined treatment of PCN and LPS, and among these, 285 were found to have binding sites on Cyp3a11. Moreover, the repressed targets of the epigenetic markers HDAC1, HDAC3 and EZH2 were further suppressed by LPS treatment and were enhanced by PCN treatment. By identifying and contrasting the transcriptional regulators that are altered by PCN and LPS, our study provides novel insights into the transcriptional regulation of CYP3A in the liver.


Asunto(s)
Citocromo P-450 CYP3A/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Transcriptoma , Animales , Biología Computacional/métodos , Citocromo P-450 CYP3A/metabolismo , Activación Enzimática , Epigénesis Genética , Proteínas de la Membrana/metabolismo , Ratones , Transducción de Señal , Factores de Transcripción
11.
Ann Pharmacother ; 53(6): 612-620, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30607980

RESUMEN

OBJECTIVE: To review the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of neratinib in human epidermal growth factor receptor (HER2)+ breast cancer (BC). DATA SOURCES: A PubMed search was performed using the term neratinib between September 12, 2018, and November 21, 2018. References of published articles and reviews were also assessed for additional information. STUDY SELECTION AND DATA EXTRACTION: English-language preclinical and clinical studies on the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of neratinib were evaluated. DATA SYNTHESIS: Neratinib, an irreversible inhibitor of HER1, HER2, and HER4, is Food and Drug Administration approved for the extended adjuvant treatment of stage I-III HER2+ BC to follow trastuzumab-based therapy. A phase III study has demonstrated statistically significant improvement in 5-year disease-free survival rate (90.2 vs 87.7; hazard ratio = 0.73, 95% CI = 0.57-0.92, P = 0.0083). Its most common adverse effect is diarrhea, observed in more than 90% of patients. The incidence of grade 3/4 diarrhea (~40%) is reduced by half with loperamide prophylaxis, which is recommended for the first 8 weeks of neratinib therapy. Other common adverse reactions are nausea and fatigue. The patients need to be monitored for liver function tests and drug interactions with acid-reducing agents, CYP3A4 inhibitors/inducers, and P-glycoprotein substrates with narrow therapeutic window. Relevance to Patient Care and Clinical Practice: American Society of Clinical Oncology and National Comprehensive Cancer Network clinical guidelines suggest the use of neratinib for extended adjuvant therapy following 1-year trastuzumab in stage I to III HER2+ BC. Diarrhea remains a clinically significant but manageable adverse event. CONCLUSION: Neratinib significantly improves treatment outcomes and has manageable toxicity in stage I to III HER2+ BC patients.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Quinolinas/uso terapéutico , Adulto , Femenino , Humanos , Persona de Mediana Edad , Quinolinas/farmacología , Resultado del Tratamiento
12.
Chem Biol Interact ; 291: 87-94, 2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-29913120

RESUMEN

Irinotecan is highly effective in the treatment of metastatic colorectal cancer as well as many other cancers. However, irinotecan is known to cause severe diarrhea, which pose significant problems in patients undergoing irinotecan based chemotherapy. Dietary and herbal components have shown promise in improving gastrointestinal health. Therefore, we compared the effect of grain-based chow diet containing phytoestrogens and corn/alfalfa as fat source to purified diets containing either animal-derived fat source (lard) or plant-derived fat source (soybean oil) on irinotecan-induced toxicities in mice. The concentration of the toxic metabolite, SN-38, was measured in the serum, and the activity of main enzyme, carboxylesterase (CEs) involved in biotransformation of irinotecan to SN-38 formation was measured in the liver. We found that the grain-based diet was protective against irinotecan-induced diarrhea. Interestingly, purified diet containing lard caused fatty liver in mice, while grain-based chow diet containing corn/alfa-alfa or purified diet with soybean oil did not cause fat deposition in the liver. Serum SN-38 concentration was significantly higher in the mice fed with purified diets compared to the chow-fed mice. Hepatic CEs activity was induced in the presence of irinotecan in mice on purified diets, but not chow diet. These results indicate that components of grain-based natural diet (presumably phytoestrogens and/or the macronutrients balance) compared to purified diets may have a beneficial effect by controlling the adverse effects of irinotecan in cancer patients.


Asunto(s)
Camptotecina/análogos & derivados , Dieta , Pruebas de Toxicidad , Animales , Peso Corporal/efectos de los fármacos , Camptotecina/efectos adversos , Camptotecina/sangre , Camptotecina/toxicidad , Carboxilesterasa/metabolismo , Diarrea/sangre , Diarrea/inducido químicamente , Hígado Graso/sangre , Hígado Graso/inducido químicamente , Hígado Graso/patología , Glucuronidasa/metabolismo , Irinotecán , Masculino , Metaboloma , Ratones Endogámicos C57BL
13.
Drug Metab Dispos ; 46(4): 397-404, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29440179

RESUMEN

Cytochrome P450 CYP3A4 is the most abundant drug-metabolizing enzyme and is responsible for the metabolism of ∼50% of clinically available drugs. Induction of CYP3A4 impacts the disposition of its substrates and leads to harmful clinical consequences, such as failure of therapy. To prevent such undesirable consequences, the molecular mechanisms of regulation of CYP3A4 need to be fully understood. CYP3A4 induction is regulated primarily by the xenobiotic nuclear receptor pregnane-X receptor (PXR). After ligand binding, PXR is translocated to the nucleus, where it binds to the CYP3A4 promoter and induces its gene expression. PXR function is modulated by phosphorylation(s) by multiple kinases. In this study, we determined the role of the c-Jun N-terminal kinase (JNK) in PXR-mediated induction of CYP3A4 enzyme in vitro. Human liver carcinoma cells (HepG2) were transfected with CYP3A4 luciferase and PXR plasmids, followed by treatment with JNK inhibitor (SP600125; SP) and PXR activators rifampicin (RIF) or hyperforin. Our results indicate that SP treatment significantly attenuated PXR-mediated induction of CYP3A4 reporter activity, as well as gene expression and enzyme activity. JNK knockdown by siRNA (targeting both JNK 1 and 2) also attenuated CYP3A4 induction by RIF. Interestingly, SP treatment attenuated JNK activation by RIF. Furthermore, treatment with RIF increased PXR nuclear levels and binding to the CYP3A4 promoter; SP attenuated these effects. This study shows that JNK is a novel mechanistic regulator of CYP3A4 induction by PXR.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Receptores de Esteroides/metabolismo , Línea Celular Tumoral , Inducción Enzimática/efectos de los fármacos , Células Hep G2 , Humanos , Inactivación Metabólica/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Receptor X de Pregnano , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Rifampin/farmacología
14.
Expert Opin Drug Metab Toxicol ; 13(9): 901-923, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28766962

RESUMEN

INTRODUCTION: Busulfan (Bu) is an alkylating agent with a limited therapeutic margin and exhibits inter-patient variability in pharmacokinetics (PK). Despite decades of use, mechanisms of Bu PK-based drug-drug interactions (DDIs), as well as the negative downstream effects of these DDIs, have not been fully characterized. Areas covered: This article provides an overview of Bu PK, with a primary focus on how known and potentially unknown drug metabolism pathways influence Bu-associated DDIs. In addition, pharmacogenomics of Bu chemotherapy and Bu-related DDIs observed in the stem cell transplant clinic (SCT) are summarized. Finally the increasing importance of Bu therapeutic drug monitoring is highlighted. Expert opinion: Mechanistic studies of Bu metabolism have shown that in addition to GST isoenzymes, other oxidative enzymes (CYP, FMO) and ABC/MDR drug transporters likely contribute to the overall clearance of Bu. Despite many insights, results from clinical studies, especially in polypharmacy settings and between pediatric and adult patients, remain conflicting. Further basic science and clinical investigative efforts are required to fully understand the key factors determining Bu PK characteristics and its effects on complications after SCT. Improved TDM strategies are promising components to further investigate, for instance DDI mechanisms and patient outcomes, in the highly complex SCT treatment setting.


Asunto(s)
Antineoplásicos Alquilantes/administración & dosificación , Busulfano/administración & dosificación , Monitoreo de Drogas/métodos , Adulto , Animales , Antineoplásicos Alquilantes/farmacocinética , Busulfano/farmacocinética , Niño , Interacciones Farmacológicas , Humanos , Farmacogenética , Trasplante de Células Madre/métodos
15.
Expert Opin Drug Metab Toxicol ; 13(6): 605-616, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28537216

RESUMEN

INTRODUCTION: Drug-metabolizing enzymes (DMEs) are primarily down-regulated during infectious and inflammatory diseases, leading to disruption in the metabolism of small molecule drugs (smds), which are increasingly being prescribed therapeutically in combination with biologics for a number of chronic diseases. The biologics may exert pro- or anti-inflammatory effect, which may in turn affect the expression/activity of DMEs. Thus, patients with infectious/inflammatory diseases undergoing biologic/smd treatment can have complex changes in DMEs due to combined effects of the disease and treatment. Areas covered: We will discuss clinical biologics-SMD interaction and regulation of DMEs during infection and inflammatory diseases. Mechanistic studies will be discussed and consequences on biologic-small molecule combination therapy on disease outcome due to changes in drug metabolism will be highlighted. Expert opinion: The involvement of immunomodulatory mediators in biologic-SMDs is well known. Regulatory guidelines recommend appropriate in vitro or in vivo assessments for possible interactions. The role of cytokines in biologic-SMDs has been documented. However, the mechanisms of drug-drug interactions is much more complex, and is probably multi-factorial. Studies aimed at understanding the mechanism by which biologics effect the DMEs during inflammation/infection are clinically important.


Asunto(s)
Productos Biológicos/farmacología , Enzimas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Animales , Productos Biológicos/administración & dosificación , Productos Biológicos/efectos adversos , Citocinas/metabolismo , Regulación hacia Abajo , Interacciones Farmacológicas , Humanos , Infecciones/enzimología , Inflamación/enzimología
16.
Toxicol In Vitro ; 41: 75-82, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28242239

RESUMEN

The bacterial receptor, Toll-like receptor (TLR) 4 mediates inflammatory responses and has been linked to a broad array of diseases. TLR4 agonists are being explored as potential treatments for cancer and other diseases. We have previously shown that activation of TLR4 by lipopolysaccharide (LPS) leads to down-regulation of drug metabolizing enzymes/transporters (DMETs), and altered pharmacokinetics/pharmacodynamics (PK/PD) of drugs. These changes can increase the risk of drug-drug interactions (DDIs) in patients on multiple medications. Clinically, DDI was observed for combination chemotherapy of paclitaxel (TLR4 ligand) and irinotecan. To determine the role of TLR4 in DDI between paclitaxel and irinotecan in vitro, primary hepatocytes from TLR4-wild-type (WT) and mutant mice were pre-treated with paclitaxel, followed by irinotecan. Gene expression of DMETs was determined. Paclitaxel treatment increased the levels of irinotecan metabolites, SN-38 and SN-38 glucuronide (SN-38G) in TLR4-dependent manner. Paclitaxel-mediated induction of genes involved in irinotecan metabolism such as Cyp3a11 and Ugt1a1 was TLR4-dependent, while induction of the transporter Mrp2 was TLR4-independent. These novel findings demonstrate that paclitaxel can affect irinotecan metabolism by a TLR4-dependent mechanism. This provides a new perspective towards evaluation of marketed drugs according to their potential to exert DDIs in TLR4-dependent manner.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Camptotecina/análogos & derivados , Paclitaxel/farmacología , Receptor Toll-Like 4/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Camptotecina/farmacología , Carboxilesterasa/metabolismo , Células Cultivadas , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Citocinas/genética , Interacciones Farmacológicas , Regulación de la Expresión Génica/efectos de los fármacos , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Irinotecán , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C3H , Ratones Mutantes , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Receptor Toll-Like 4/genética , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
17.
Methods Mol Biol ; 1395: 55-68, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26910068

RESUMEN

Cancer is the second leading cause of mortality worldwide; however the response rate to chemotherapy treatment remains slow, mainly due to narrow therapeutic index and multidrug resistance. Paclitaxel (taxol) has a superior outcome in terms of response rates and progression-free survival. However, numerous cancer patients are resistant to this drug. In this investigation, we tested the hypothesis that induction of cytochrome P450 (Cyp)3a11 gene by paclitaxel is downregulated by the inflammatory mediator, lipopolysaccharide (LPS), and that the pro-inflammatory cytokine, tumor necrosis factor (TNF)-α, attenuates human CYP3A4 gene induction by rifampicin. Primary mouse hepatocytes were pretreated with LPS (1 µg/ml) for 10 min, followed by paclitaxel (20 µM) or vehicle for 24 h. RNA was extracted from the cells by trizol method followed by cDNA synthesis and analysis by real-time PCR. Paclitaxel significantly induced gene expression of Cyp3a11 (~30-fold) and this induction was attenuated in LPS-treated samples. Induction and subsequent downregulation of CYP3A enzyme can impact paclitaxel treatment in cancer patients where inflammatory mediators are activated. It has been shown that the nuclear receptor, pregnane X receptor (PXR), plays a role in the induction of CYP enzymes. In order to understand the mechanisms of regulation of human CYP3A4 gene, we co-transfected HepG2 cells (human liver cell line) with CYP3A4-luciferase construct and a PXR expression plasmid. The cells were then treated with the pro-inflammatory cytokine, TNFα, followed by the prototype CYP3A inducer rifampicin. It is well established that rifampicin activates PXR, leading to CYP3A4 induction. We found that induction of CYP3A4-luciferase activity by rifampicin was significantly attenuated by TNFα. In conclusion, we describe herein several in vitro approaches entailing primary and cultured hepatocytes, real-time PCR, and transcriptional activation (transfection) assays to investigate the molecular regulation of CYP3A, which plays a pivotal role in the metabolism of numerous chemotherapeutic drugs. Genetic or drug-induced variation in CYP3A and/or PXR expression could contribute to drug resistance to chemotherapeutic agents in cancer patients.


Asunto(s)
Antineoplásicos/farmacología , Citocromo P-450 CYP3A/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Paclitaxel/farmacología , Rifampin/farmacología , Animales , Citocromo P-450 CYP3A/biosíntesis , ADN Complementario/biosíntesis , ADN Complementario/genética , Inducción Enzimática/efectos de los fármacos , Genes Reporteros/genética , Células Hep G2 , Humanos , Lipopolisacáridos/farmacología , Hígado/metabolismo , Luciferasas/genética , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Factor de Necrosis Tumoral alfa/farmacología
18.
Drug Metab Dispos ; 44(1): 61-7, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26470915

RESUMEN

The expressions and activities of hepatic drug-metabolizing enzymes and transporters (DMETs) are altered during infection and inflammation. Inflammatory responses in the liver are mediated primarily by Toll-like receptor (TLR)-signaling, which involves recruitment of Toll/interleukin (IL)-1 receptor (TIR) domain containing adaptor protein (TIRAP) and TIR domain containing adaptor inducing interferon (IFN)-ß (TRIF) that eventually leads to induction of proinflammatory cytokines and mitogen-activated protein kinases (MAPKs). Lipopolysaccharide (LPS) activates the Gram-negative bacterial receptor TLR4 and polyinosinic:polycytidylic acid (polyI:C) activates the viral receptor TLR3. TLR4 signaling involves TIRAP and TRIF, whereas TRIF is the only adaptor protein involved in the TLR3 pathway. We have shown previously that LPS-mediated downregulation of DMETs is independent of TIRAP. To determine the role of TRIF, we treated TRIF(+/+) and TRIF(-/-) mice with LPS or polyI:C. LPS downregulated (∼40%-60%) Cyp3a11, Cyp2a4, Ugt1a1, Mrp2 mRNA levels, whereas polyI:C downregulated (∼30%-60%) Cyp3a11, Cyp2a4, Cyp1a2, Cyp2b10, Ugt1a1, Mrp2, and Mrp3 mRNA levels in TRIF(+/+) mice. This downregulation was not attenuated in TRIF(-/-) mice. Induction of cytokines by LPS was observed in both TRIF(+/+) and TRIF(-/-) mice. Cytokine induction was delayed in polyI:C-treated TRIF(-/-) mice, indicating that multiple mechanisms mediating polyI:C signaling exist. To assess the role of MAPKs, primary hepatocytes were pretreated with specific inhibitors before treatment with LPS/polyI:C. We found that only the c-jun-N-terminal kinase (JNK) inhibitor attenuated the down-regulation of DMETs. These results show that TRIF-independent pathways can be involved in the downregulation of DMETs through TLR4 and 3. JNK-dependent mechanisms likely mediate this downregulation.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Glucuronosiltransferasa/metabolismo , Hepatocitos/enzimología , Hígado/enzimología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Células Cultivadas , Sistema Enzimático del Citocromo P-450/genética , Citocinas/genética , Citocinas/metabolismo , Regulación Enzimológica de la Expresión Génica , Glucuronosiltransferasa/genética , Hepatocitos/efectos de los fármacos , Isoenzimas , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Poli I-C/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 4/agonistas
19.
Life Sci ; 139: 132-8, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26334566

RESUMEN

AIM: Our aim is to investigate the impact of high fat diet-induced obesity on plasma concentrations of the toxic irinotecan metabolite, SN-38, in mice. MAIN METHODS: Diet-induced obese (DIO, 60% kcal fed) and lean mice (10% kcal fed) were treated orally with a single dose of 10mg/kg irinotecan to determine pharmacokinetic (PK) parameters. Feces and livers were collected for quantification of irinotecan and its metabolites (SN-38 & SN-38G). SN-38G formation by Ugt1a1 enzyme was analyzed in liver S9 fractions. Expression of the pro-inflammatory cytokine, TNF-α was determined in liver and plasma. Hepatic ß-glucuronidase and carboxylesterase enzymes (CES) were also determined. KEY FINDINGS: AUC0-8 and Cmax of SN-38 increased by 2-fold in DIO mice compared to their lean controls. This was accompanied by a~2-fold reduction in AUC0-8 and Cmax of SN-38G in DIO mice. There were no differences in the PK parameters of irinotecan in DIO or lean mice. Conversion of SN-38 to SN-38G by Ugt1a1 enzyme was reduced by ~2-fold in liver S9 fractions in DIO mice. Furthermore, in DIO mice, ß-glucuronidase activity increased by 2-fold, whereas there was no change in CES activity. TNF-α mRNA expression was 3 fold higher in DIO mice. SIGNIFICANCE: Our study demonstrates that reduced hepatic Ugt1a activity during obesity likely contributes to reduced glucuronidation, and results in higher levels of the toxic metabolite, SN-38. Thus, irinotecan dosage should be closely monitored for effective and safe chemotherapy in obese cancer patients who are at a higher risk of developing liver toxicity.


Asunto(s)
Antineoplásicos Fitogénicos/metabolismo , Camptotecina/análogos & derivados , Glucuronatos/metabolismo , Glucuronosiltransferasa/metabolismo , Obesidad/metabolismo , Inhibidores de Topoisomerasa I/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacocinética , Camptotecina/metabolismo , Camptotecina/farmacocinética , Carboxilesterasa/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Heces/química , Glucuronatos/farmacocinética , Glucuronidasa/metabolismo , Glucuronosiltransferasa/análisis , Humanos , Irinotecán , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/genética , ARN Mensajero/genética , Inhibidores de Topoisomerasa I/farmacocinética , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/genética
20.
Drug Metab Dispos ; 42(1): 172-81, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24194512

RESUMEN

Impairment of drug disposition in the liver during inflammation has been attributed to downregulation of gene expression of drug-metabolizing enzymes (DMEs) and drug transporters. Inflammatory responses in the liver are primarily mediated by Toll-like receptors (TLRs). We have recently shown that activation of TLR2 or TLR4 by lipoteichoic acid (LTA) and lipopolysaccharide (LPS), respectively, leads to the downregulation of gene expression of DMEs/transporters. However, the molecular mechanism underlying this downregulation is not fully understood. The xenobiotic nuclear receptors, pregnane X receptor (PXR) and constitutive androstane receptor (CAR), regulate the expression of DMEs/transporter genes. Downregulation of DMEs/transporters by LTA or LPS was associated with reduced expression of PXR and CAR genes. To determine the role of CAR, we injected CAR(+/+) and CAR(-/-) mice with LTA or LPS, which significantly downregulated (~40%-60%) RNA levels of the DMEs, cytochrome P450 (Cyp)3a11, Cyp2a4, Cyp2b10, uridine diphosphate glucuronosyltransferase 1a1, amine N-sulfotransferase, and the transporter, multidrug resistance-associated protein 2, in CAR(+/+) mice. Suppression of most of these genes was attenuated in LTA-treated CAR(-/-) mice. In contrast, LPS-mediated downregulation of these genes was not attenuated in CAR(-/-) mice. Induction of these genes by mouse CAR activator 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene was sustained in LTA- but not in LPS-treated mice. Similar observations were obtained in humanized CAR mice. We have replicated these results in primary hepatocytes as well. Thus, LPS can downregulate DME/transporter genes in the absence of CAR, whereas the effect of LTA on these genes is attenuated in the absence of CAR, indicating the potential involvement of CAR in LTA-mediated downregulation of DME/transporter genes.


Asunto(s)
Transporte Biológico/genética , Expresión Génica/genética , Inactivación Metabólica/genética , Hígado/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Receptores Toll-Like/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Receptor de Androstano Constitutivo , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Masculino , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Receptor X de Pregnano , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Ácidos Teicoicos/farmacología , Receptores Toll-Like/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA