Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Drug Metab Dispos ; 48(9): 819-829, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32616543

RESUMEN

After oral administration to monkeys of [14C]GDC-0810, an α,ß-unsaturated carboxylic acid, unchanged parent and its acyl glucuronide metabolite, M6, were the major circulating drug-related components. In addition, greater than 50% of circulating radioactivity in plasma was found to be nonextractable 12 hours post-dose, suggesting possible covalent binding to plasma proteins. In the same study, one of the minor metabolites was a cysteine conjugate of M6 (M11) that was detected in plasma and excreta (urine and bile). The potential mechanism for the covalent binding to proteins was further investigated using in vitro methods. In incubations with glutathione (GSH) or cysteine (5 mM), GSH and cysteine conjugates of M6 were identified, respectively. The cysteine reaction was efficient with a half-life of 58.6 minutes (k react = 0.04 1/M per second). Loss of 176 Da (glucuronic acid) followed by 129 Da (glutamate) in mass fragmentation analysis of the GSH adduct of M6 (M13) suggested the glucuronic acid moiety was not modified. The conjugation of N-glucuronide M4 with cysteine in buffer was >1000-fold slower than with M6. Incubations of GDC-0810, M4, or M6 with monkey or human liver microsomes in the presence of NADPH and GSH did not produce any oxidative GSH adducts, and the respective substrates were qualitatively recovered. In silico analysis quantified the inherent reactivity differences between the glucuronide and its acid precursor. Collectively, these results show that acyl glucuronidation of α,ß-unsaturated carboxylic acids can activate the compound toward reactivity with GSH, cysteine, or other biologically occurring thiols and should be considered during the course of drug discovery. SIGNIFICANCE STATEMENT: Acyl glucuronidation of the α,ß-unsaturated carboxylic acid in GDC-0810 activates the conjugated alkene toward nucleophilic addition by glutathione or other reactive thiols. This is the first example that a bioactivation mechanism could lead to protein covalent binding to α,ß-unsaturated carboxylic acid compounds.


Asunto(s)
Antineoplásicos Hormonales/farmacocinética , Ácidos Carboxílicos/farmacocinética , Cinamatos/farmacocinética , Glucurónidos/metabolismo , Indazoles/farmacocinética , Administración Oral , Animales , Antineoplásicos Hormonales/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ácidos Carboxílicos/administración & dosificación , Cinamatos/administración & dosificación , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Indazoles/administración & dosificación , Macaca fascicularis , Microsomas Hepáticos , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/metabolismo
2.
Contrast Media Mol Imaging ; 2019: 1760184, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31787861

RESUMEN

Pictilisib (GDC-0941) is an inhibitor of phosphatidylinositol 3-kinase (PI3K), part of a signaling cascade involved in breast cancer development. The purpose of this study was to evaluate the pharmacokinetics of pictilisib noninvasively by radiolabeling it with 11C and to assess the usability of the resulting [11C]-pictilisib as a positron-emission tomography (PET) tracer to screen for pictilisib-sensitive tumors. In this study, pictilisib was radiolabeled with [11C]-methyl iodide to obtain 11C-methylated pictilisib ([11C]-pictilisib) using an automated synthesis module with a high radiolabeling yield. Considerably higher uptake ratios were observed in MCF-7 (PIK3CA mutation, pictilisib-sensitive) cells than those in MDA-MB-231 (PIK3CA wild-type, pictilisib-insensitive) cells at all evaluated time points, indicating good in vitro binding of [11C]-pictilisib. Dynamic micro-PET scans in mice and biodistribution results showed that [11C]-pictilisib was mainly excreted via the hepatobiliary tract into the intestines. MCF-7 xenografts could be clearly visualized on the static micro-PET scans, while MDA-MB-231 tumors could not. Biodistribution results of two xenograft models showed significantly higher uptake and tumor-to-muscle ratios in the MCF-7 xenografts than those in MDA-MB-231 xenografts, exhibiting high in vivo targeting specificity. In conclusion, [11C]-pictilisib was first successfully prepared, and it exhibited good potential to identify pictilisib-sensitive tumors noninvasively, which may have a great impact in the treatment of cancers with an overactive PI3K/Akt/mTOR signal pathway. However, the high activity in hepatobiliary system and intestines needs to be addressed.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Radioisótopos de Carbono , Indazoles , Proteínas de Neoplasias/análisis , Fosfatidilinositol 3-Quinasas/análisis , Tomografía de Emisión de Positrones , Radiofármacos , Sulfonamidas , Animales , Neoplasias de la Mama/patología , Radioisótopos de Carbono/farmacocinética , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Femenino , Eliminación Hepatobiliar , Xenoinjertos , Humanos , Indazoles/síntesis química , Indazoles/farmacocinética , Indazoles/farmacología , Concentración 50 Inhibidora , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Trasplante de Neoplasias , Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Transducción de Señal , Sulfonamidas/síntesis química , Sulfonamidas/farmacocinética , Sulfonamidas/farmacología , Distribución Tisular
3.
Cancer Chemother Pharmacol ; 83(1): 151-160, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30386887

RESUMEN

PURPOSE: H3B-6545, a novel selective estrogen receptor (ER)α covalent antagonist (SERCA) which inactivates both wild-type and mutant ERα, is in clinical development for the treatment of metastatic breast cancer. Preclinical studies were conducted to characterize the pharmacokinetics and metabolism of H3B-6545 in rat and monkeys. METHODS: The clearance and metabolic profiles of H3B-6545 were studied using rat, monkey and human hepatocytes, and reaction phenotyping was done using recombinant human cytochrome P450 enzymes. Blood stability, protein binding, and permeability were also determined in vitro. Pharmacokinetics of H3B-6545 was assessed after both intravenous and oral dosing. A nonclinical PBPK model was developed to assess in vitro-in vivo correlation of clearance. RESULTS: H3B-6545 had a terminal elimination half-life of 2.4 h in rats and 4.0 h in monkeys and showed low to moderate bioavailability, in line with the in vitro permeability assessment. Plasma protein binding was similar across species, at 99.5-99.8%. Nine metabolites of H3B-6545 were identified in hepatocyte incubations, none of which were unique to humans. Formation of glutathione-related conjugate of H3B-6545 was minimal in vitro. H3B-6545, a CYP3A substrate, is expected to be mostly cleared via hepatic phase 1 metabolism. Hepatocyte clearance values were used to adequately model the time-concentration profiles in rat and monkey. CONCLUSIONS: We report on the absorption and metabolic fate and disposition of H3B-6545 in rats and dogs and illustrate that in vitro-in vivo correlation of clearance is possible for targeted covalent inhibitors, provided reactivity is not a predominant mechanism of clearance.


Asunto(s)
Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Antagonistas del Receptor de Estrógeno/farmacología , Antagonistas del Receptor de Estrógeno/farmacocinética , Receptor alfa de Estrógeno/antagonistas & inhibidores , Hepatocitos/metabolismo , Indazoles/farmacología , Indazoles/farmacocinética , Microsomas Hepáticos/metabolismo , Piridinas/farmacología , Piridinas/farmacocinética , Animales , Disponibilidad Biológica , Células Cultivadas , Inhibidores Enzimáticos del Citocromo P-450/farmacocinética , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Perros , Evaluación Preclínica de Medicamentos , Femenino , Hepatocitos/efectos de los fármacos , Humanos , Técnicas In Vitro , Macaca fascicularis , Tasa de Depuración Metabólica , Microsomas Hepáticos/efectos de los fármacos , Unión Proteica , Ratas Sprague-Dawley , Especificidad de la Especie , Distribución Tisular
4.
Int J Cancer ; 137(8): 2007-18, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25868794

RESUMEN

Enhancer of Zeste Homolog 2 (EZH2) has emerged as a promising therapeutic target for treatment of a broad spectrum of tumors including gliomas. We explored the interactions of five novel, structurally similar EZH2 inhibitors (EPZ005687, EPZ-6438, UNC1999, GSK343 and GSK126) with P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2). The compounds were screened by in vitro transwell assays and EPZ005687, EPZ-6438 and GSK126 were further tested in vivo using wild-type (WT), Abcb1 and/or Abcg2 knockout mice. All EZH2 inhibitors are transported by P-gp and BCRP, although in vitro the transporter affinity of GSK126 was obscured by very low membrane permeability. Both P-gp and Bcrp1 restrict the brain penetration of EPZ005687 and GSK126, whereas the brain accumulation of EPZ-6438 is limited by P-gp only and efflux of EPZ-6438 was completely abrogated by elacridar. Intriguingly, an unknown factor present in all knockout mouse strains causes EPZ005687 and EPZ-6438 retention in plasma relative to WT mice, a phenomenon not seen with GSK126. In WT mice, the GSK126 tissue-to-plasma ratio for all tissues is lower than for EPZ005687 or EPZ-6438. Moreover, the oral bioavailability of GSK126 is only 0.2% in WT mice, which increases to 14.4% in Abcb1;Abcg2 knockout mice. These results are likely due to poor membrane permeability and question the clinical usefulness of GSK126. Although all tested EZH2 inhibitors are substrates of P-gp and BCRP, restricting the brain penetration and potential utility for treatment of glioma, EPZ-6438 would be the most suitable candidate of this series.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Inhibidores Enzimáticos/farmacocinética , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Administración Oral , Animales , Benzamidas/administración & dosificación , Benzamidas/farmacocinética , Disponibilidad Biológica , Compuestos de Bifenilo , Línea Celular , Perros , Evaluación Preclínica de Medicamentos , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos/administración & dosificación , Humanos , Indazoles/administración & dosificación , Indazoles/farmacocinética , Indoles/administración & dosificación , Indoles/farmacocinética , Células de Riñón Canino Madin Darby , Ratones , Ratones Noqueados , Morfolinas , Piridonas/administración & dosificación , Piridonas/farmacocinética
5.
J Med Chem ; 57(18): 7804-10, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25147929

RESUMEN

A functional high throughput screen identified a novel chemotype for the positive allosteric modulation (PAM) of the muscarinic acetylcholine receptor (mAChR) subtype 5 (M5). Application of rapid analog, iterative parallel synthesis efficiently optimized M5 potency to arrive at the most potent M5 PAMs prepared to date and provided tool compound 8n (ML380) demonstrating modest CNS penetration (human M5 EC50 = 190 nM, rat M5 EC50 = 610 nM, brain to plasma ratio (Kp) of 0.36).


Asunto(s)
Sistema Nervioso Central/metabolismo , Descubrimiento de Drogas , Indazoles/metabolismo , Indazoles/farmacología , Piperidinas/metabolismo , Piperidinas/farmacología , Receptor Muscarínico M5/química , Receptor Muscarínico M5/metabolismo , Sulfonamidas/metabolismo , Sulfonamidas/farmacología , Regulación Alostérica/efectos de los fármacos , Animales , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Indazoles/química , Indazoles/farmacocinética , Masculino , Piperidinas/química , Piperidinas/farmacocinética , Ratas , Especificidad por Sustrato , Sulfonamidas/química , Sulfonamidas/farmacocinética
6.
J Med Chem ; 57(12): 5129-40, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24884675

RESUMEN

A high throughput screening campaign identified 5-(2-chlorophenyl)indazole compound 4 as an antagonist of the transient receptor potential A1 (TRPA1) ion channel with IC50 = 1.23 µM. Hit to lead medicinal chemistry optimization established the SAR around the indazole ring system, demonstrating that a trifluoromethyl group at the 2-position of the phenyl ring in combination with various substituents at the 6-position of the indazole ring greatly contributed to improvements in vitro activity. Further lead optimization resulted in the identification of compound 31, a potent and selective antagonist of TRPA1 in vitro (IC50 = 0.015 µM), which has moderate oral bioavailability in rodents and demonstrates robust activity in vivo in several rodent models of inflammatory pain.


Asunto(s)
Indazoles/química , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Administración Oral , Analgésicos/química , Analgésicos/farmacocinética , Analgésicos/farmacología , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Disponibilidad Biológica , Células CHO , Canales de Calcio , Cricetulus , Adyuvante de Freund , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Indazoles/farmacocinética , Indazoles/farmacología , Masculino , Ratones Endogámicos C57BL , Planta de la Mostaza , Aceites de Plantas , Ratas Wistar , Especificidad de la Especie , Relación Estructura-Actividad , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/antagonistas & inhibidores
7.
J Med Chem ; 55(2): 903-13, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22148921

RESUMEN

A substantial body of evidence supports the utility of antiangiogenesis inhibitors as a strategy to block or attenuate tumor-induced angiogenesis and inhibition of primary and metastatic tumor growth in a variety of solid and hematopoietic tumors. Given the requirement of tumors for different cytokine and growth factors at distinct stages of their growth and dissemination, optimal antiangiogenic therapy necessitates inhibition of multiple, complementary, and nonredundant angiogenic targets. 11-(2-Methylpropyl)-12,13-dihydro-2-methyl-8-(pyrimidin-2-ylamino)-4H-indazolo[5,4-a]pyrrolo[3,4-c]carbazol-4-one (11b, CEP-11981) is a potent orally active inhibitor of multiple targets (TIE-2, VEGF-R1, 2, and 3, and FGF-R1) having essential and nonredundant roles in tumor angiogenesis and vascular maintenance. Outlined in this article are the design strategy, synthesis, and biochemical and pharmacological profile for 11b, which completed Phase I clinical assessing safety and pharmacokinetics allowing for the initiation of proof of concept studies.


Asunto(s)
Inhibidores de la Angiogénesis/síntesis química , Carbazoles/síntesis química , Indazoles/síntesis química , Receptor TIE-2/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Administración Oral , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/farmacología , Animales , Disponibilidad Biológica , Carbazoles/farmacocinética , Carbazoles/farmacología , Humanos , Indazoles/farmacocinética , Indazoles/farmacología , Macaca fascicularis , Masculino , Ratones , Ratones Desnudos , Modelos Moleculares , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor TIE-2/química , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Relación Estructura-Actividad , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Xenobiotica ; 41(12): 1088-99, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21838594

RESUMEN

The phosphatidylinositol 3-kinase (PI3K) pathway is a major determinant of cell cycling and proliferation. Its deregulation is associated with the development of many cancers. GDC-0941, a potent and selective inhibitor of PI3K, was characterised preclinically in in vitro and in vivo studies. Plasma protein binding was extensive, with free fraction less than 7%, and blood-to-plasma ratio ranged from 0.6 to 1.2 among the species tested. GDC-0941 human hepatic clearance was predicted to be moderate by liver microsomal incubations. GDC-0941 had high permeability in Madin-Darby canine kidney cells. The clearance of GDC-0941 was high in mouse (63.7 mL/min/kg), rat (49.3 mL/min/kg) and cynomolgus monkey (58.6 mL/min/kg), and moderate in dog (11.9 mL/min/kg). The volume of distribution ranged from 2.52 L/kg in rat to 2.94 L/kg in monkey. Oral bioavailability ranged from 18.6% in monkey to 77.9% in mouse. Predicted human clearance and volume of distribution using allometry were 6 mL/min/kg and 2.9 L/kg, respectively. The human efficacious doses were predicted based on results from preclinical pharmacokinetic studies and xenograft models. GDC-0941 preclinical characterisation and predictions of its properties in human supported its progression towards clinical development. GDC-0941 is currently in phase II clinical trials.


Asunto(s)
Indazoles/farmacocinética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacocinética , Sulfonamidas/farmacocinética , Administración Oral , Animales , Área Bajo la Curva , Autorradiografía , Radioisótopos de Carbono , Línea Celular , Permeabilidad de la Membrana Celular , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Indazoles/administración & dosificación , Indazoles/sangre , Indazoles/química , Masculino , Microsomas Hepáticos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/química , Especificidad de la Especie , Sulfonamidas/administración & dosificación , Sulfonamidas/sangre , Sulfonamidas/química , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Pharm Sci ; 99(3): 1278-87, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19743500

RESUMEN

NF-kappaB activation is clearly linked to the pathogenesis of multiple inflammatory diseases including arthritis. The prominent role of IkappaB kinase-2 (IKK-2) in regulating NF-kappaB signaling in response to proinflammatory stimuli has made IKK-2 a primary anti-inflammation therapeutic target. PHA-408, a potent and selective IKK-2 inhibitor, was identified internally and used for our studies to assess this target. In early in vivo studies, PHA-408 demonstrated efficacy at high doses; however, the correlation between PHA-408 exposure and efficacy could not be established using standard dosing paradigms for the rat disease models. Similar concerns arose from early in vivo safety studies where appropriate NOAEL margins were not achieved. Following a full investigation of the physicochemical properties of the molecule and pharmacokinetic modeling, an oral steady-state delivery strategy was designed to administer PHA-408 to the rat for both efficacy and safety studies. Using this steady-state delivery, a clear dose-response relationship was established between plasma concentrations of PHA-408 and efficacy in the rat arthritis model. The same steady-state delivery approach was used to demonstrate the target safety. In summary, a combination of pharmacokinetic modeling with a steady-state delivery approach allowed us to establish confidence in both the mechanism and safety of the target.


Asunto(s)
Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Sistemas de Liberación de Medicamentos/métodos , Quinasa I-kappa B/antagonistas & inhibidores , Indazoles/administración & dosificación , Ácidos Isonicotínicos/administración & dosificación , Administración Oral , Animales , Antiinflamatorios/efectos adversos , Antiinflamatorios/farmacocinética , Artritis Experimental/tratamiento farmacológico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Indazoles/efectos adversos , Indazoles/farmacocinética , Indazoles/farmacología , Ácidos Isonicotínicos/efectos adversos , Ácidos Isonicotínicos/farmacocinética , Ácidos Isonicotínicos/farmacología , Masculino , Modelos Biológicos , Ratas , Ratas Endogámicas Lew , Ratas Wistar , Factor de Necrosis Tumoral alfa/efectos de los fármacos
11.
Int J Hyperthermia ; 11(4): 545-59, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7594808

RESUMEN

Six cycles of the maximum tolerable intravenous doses of lonidamine (400 mg/m2) and doxorubicin (30 mg/m2) were administered to three normothermic dogs and three dogs undergoing whole-body hyperthermia (WBH) (42 degrees C X 90 min), at 3-week intervals. Lonidamine pharmacokinetics was unaltered by WBH. WBH increased doxorubicin clearance 1.6-fold, however this trend was not statistically significant. WBH resulted in a 2.4-fold increase in the volume of distribution (Vdss) of doxorubicin relative to dogs treated under euthermic conditions (p < 0.001). This finding suggests tissue extraction of doxorubicin was increased by WBH. The specific tissues in which this occurred is unknown, but myelosuppression and cardiotoxicity were only minimally increased. Therefore, doxorubicin uptake in critical normal tissues was probably unaffected. The biochemical and haematologic toxicities observed 6 h and 1 week after each treatment did not appear to differ in character or severity from that reported in dogs receiving lonidamine +/- WBH or doxorubicin +/- WBH. These results suggest WBH did not decrease the maximum tolerable dose of doxorubicin when given with lonidamine, and that the antitumour activity of this combination should be assessed.


Asunto(s)
Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Doxorrubicina/toxicidad , Hipertermia Inducida , Indazoles/farmacocinética , Indazoles/toxicidad , Animales , Antineoplásicos/sangre , Antineoplásicos/toxicidad , Ácidos y Sales Biliares/sangre , Análisis Químico de la Sangre , Perros , Doxorrubicina/sangre , Indazoles/sangre , Hígado/efectos de los fármacos , Hígado/ultraestructura , Tasa de Depuración Metabólica , Microscopía Electrónica , Contracción Miocárdica/efectos de los fármacos , Miocardio/ultraestructura , Vacuolas/efectos de los fármacos , Vacuolas/ultraestructura
12.
Int J Hyperthermia ; 11(4): 531-44, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7594807

RESUMEN

The pharmacokinetics and toxicity of intravenous lonidamine were investigated in dogs receiving four cycles of lonidamine (400 or 800 mg/m2) +/- whole-body hyperthermia (WBH). Clearance and volume of distribution in dogs receiving lonidamine during WBH increased 1.6-2.3 and 1.9-3.5-fold respectively, relative to dogs receiving lonidamine under euthermic conditions (p < 0.02). In dogs receiving lonidamine under euthermic conditions or 400 mg/m2 + WBH, the area under the lonidamine concentration versus time curve (AUC) measured during the fourth treatment was 21-58% lower than the first treatment AUC. However, in dogs receiving 800 mg/m2 + WBH, the fourth treatment AUC was four-fold higher than the first treatment AUC (p < 0.02). This suggests repeated exposure to 800 mg/m2 lonidamine and WBH impairs lonidamine metabolism. Weakness, hypoglycaemia, and elevations in amylase, alanine aminotransferase, alkaline phosphatase and bilirubin were more severe or occurred exclusively in dogs receiving 800 mg/m2 + WBH. Since these changes were attributable to marked AUC increases, which occurred secondary to repeated exposure to 800 mg/m2 lonidamine during WBH, 400 mg/m2 was identified as the maximum tolerable dose to be administered intravenously to dogs during WBH.


Asunto(s)
Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Hipertermia Inducida , Indazoles/farmacocinética , Indazoles/toxicidad , Alanina Transaminasa/sangre , Fosfatasa Alcalina/sangre , Amilasas/sangre , Animales , Antineoplásicos/administración & dosificación , Bilirrubina/sangre , Glucemia/metabolismo , Dióxido de Carbono/sangre , Creatina Quinasa/sangre , Perros , Hipopotasemia/etiología , Indazoles/administración & dosificación , Indazoles/sangre , Hígado/efectos de los fármacos , Hígado/patología , Sialorrea/etiología , Urea/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA