Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Psychol ; 14: 1136983, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38022985

RESUMEN

Conventional Buddhist texts illustrate meditation as a condition of relaxed alertness that must fend against extreme hypoarousal (sleep, drowsiness) and extreme hyperarousal (restlessness). Theoretical, neurophysiological, and neuroimaging investigations of meditation have highlighted the relaxing effects and hypoarousing without emphasizing the alertness-promoting effects. Here we performed a systematic review supported by an activation-likelihood estimate (ALE) meta-analysis in an effort to counterbalance the surfeit of scholarship emphasizing the hypoarousing and relaxing effects of different forms of Buddhist meditation. Specifically, the current systematic review-cum-meta-analytical review seeks to highlight more support for meditation's wake-promoting effects by drawing from neuroimaging research during wakefulness and meditation. In this systematic review and meta-analysis of 22 fMRI studies, we aim to highlight support for Buddhist meditation's wake-promoting or arousing effects by identifying brain regions associated with alertness during meditation. The most significant peaks were localized medial frontal gyrus (MFG) and precuneus. We failed to determine areas ostensibly common to alertness-related meditation such as the medial prefrontal cortex (mPFC), superior parietal lobule, basal ganglia, thalamus, most likely due to the relatively fewer fMRI investigations that used wakefulness-promoting meditation techniques. Also, we argue that forthcoming research on meditation, related to alertness or wakefulness, continues to adopt a multi-modal method to investigate the correlation between actual behaviors and neural networks connected to Buddhist meditation. Moreover, we recommend the implementation of fMRI paradigms on Buddhist meditation with clinically diagnosed participants to complement recent trends in psychotherapy such as mindfulness-based cognitive therapy (MBCT).

2.
Paediatr Drugs ; 22(5): 561-570, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32776159

RESUMEN

OBJECTIVE: This was a single-dose, one-period, multicenter, pharmacokinetic (PK) study to evaluate the PK of methylphenidate (MPH) hydrochloride multilayer extended-release capsules (MPH-MLR) in preschool children aged 4 to < 6 years, previously diagnosed with attention-deficit/hyperactivity disorder (ADHD), and on a stable dose of MPH. METHODS: Preschool-aged children (N = 10) received a single oral dose of MPH-MLR (10, 15, or 20 mg) sprinkled over applesauce; a dose equivalent to their pre-enrollment daily dose of MPH. Blood samples for the measurement of MPH concentrations were obtained pre-dose and at 0.5, 1, 2, 3, 4, 6, 8, 10, 12, and 24 h post-dose. No structural model was assumed in the derivation of PK values for analysis. Maximum plasma concentration (Cmax), area under the concentration-time curve (AUC), elimination half-life, clearance (CL), and volume of distribution (Vd) data were compared with a historical group of older children aged 6-11 years (N = 11) and analyzed by bodyweight. Safety (adverse event monitoring, vital signs, electrocardiogram, clinical laboratory testing, physical examination) was assessed. RESULTS: Mean dose-normalized Cmax and area under the curve to the last measurable observation (AUC0-t) values were similar across dose groups, ranging from 0.67 ng/mL/mg (MPH 15 mg) to 0.81 ng/mL/mg (MPH 10 mg) for Cmax/dose, and from 7.80 h × ng/mL/mg (MPH 20 mg) to 8.92 h × ng/mL/mg (MPH 10 mg) for AUC0-t/dose. PK results were integrated into a previously described pharmacostatistical population PK model. Visual predictive check plots showed greater variability in the 6- to 11-year-old group than the 4- to < 6-year-old group, and CL increased with increasing body weight in a greater than dose-proportional manner. Mean CL, normalized for body weight, was constant for all dose groups, ranging from 4.88 L/h/kg to 5.80 L/h/kg. Median time to Cmax ranged from 2.00 to 3.00 h post-dose, and overall, dose-normalized Cmax concentrations indicated greater systemic exposures of MPH-MLR in preschool children aged 4 to < 6 years compared with children aged 6-11 years. Children aged 4 to < 6 years had a lower Vd than children aged 6-11 years. There were no unexpected safety signals. CONCLUSION: The PK of MPH-MLR in preschool children demonstrated the biphasic absorption profile described earlier in older children, and the PK profile in children with ADHD aged 4 to < 6 years was similar to the profile in those aged 6-11 years, apart from a lower Vd and relatively higher systemic MPH levels for children in the preschool group. TRIAL REGISTRATION: Clinicaltrials.gov Identifier: NCT02470234.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/sangre , Estimulantes del Sistema Nervioso Central/farmacocinética , Metilfenidato/farmacocinética , Adulto , Peso Corporal , Cápsulas , Estimulantes del Sistema Nervioso Central/efectos adversos , Estimulantes del Sistema Nervioso Central/sangre , Niño , Preescolar , Preparaciones de Acción Retardada/efectos adversos , Preparaciones de Acción Retardada/farmacocinética , Femenino , Humanos , Masculino , Metilfenidato/efectos adversos , Metilfenidato/sangre , Modelos Biológicos
3.
J Pharmacol Exp Ther ; 339(2): 421-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21835932

RESUMEN

The mammalian target of rapamycin (mTOR) has proven to be a valid therapeutic target in a number of human cancers, and it is a candidate for clinical trials in human breast cancer. We report on a biomarker-based translational medicine approach to assess the efficacy and mechanism of action for the mTOR inhibitor temsirolimus (CCI-779) in a mammary carcinoma OncoMouse model [polyomavirus middle T antigen (PyMT)]. The mTOR signaling pathway biomarkers were assessed using a reverse-phase protein array. Pharmacokinetics studies were conducted in both the tumor and plasma compartments. Pharmacodynamic biomarkers for compound-target engagement of tumor phospho-S6 proteins were assayed by Western blot. Temsirolimus (intravenously once a week for 2 weeks) was administered in both early and advanced stages of tumors. Biomarkers for temsirolimus effects on tumor progression were assessed by three-dimensional ultrasound imaging in combination with immunohistochemistry to assess vascular density (Texas red-dextran and CD31 immunostaining) and macrophage burden (F4/80 antigen). Tumor growth was significantly arrested in temsirolimus (25 ± 14% from 8 to 10 weeks, p < 0.05, and 26 ± 17% from 11 to 13 weeks, p < 0.01), compared with 493 ± 160 and 376 ± 50% increases, respectively, in vehicle-treated groups. Temsirolimus reduced tumor vascular density, 36 to 48 and 58 to 60%, p < 0.05, by the Texas red-dextran method or CD31-positive vessel count, respectively. Temsirolimus reduced tumor macrophage burden by 46% at 13 weeks (p < 0.05). Temsirolimus inhibited (p < 0.05) the phosphoproteins S6 pS235/236 and S6 pS240/244 up to 81 and 87%, respectively. We conclude that the multimodal biomarkers of temsirolimus efficacy and mechanism of action (phosphoproteins) strongly suggest that it might translate to therapeutic efficacy in human tumors that bear congruency to features present in the mammary carcinoma of PyMT tumors.


Asunto(s)
Biomarcadores Farmacológicos/análisis , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Carga Tumoral/efectos de los fármacos , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Infusiones Intravenosas , Ratones , Ratones Transgénicos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacocinética , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Sirolimus/farmacocinética , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/análisis , Investigación Biomédica Traslacional/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Clin Cancer Res ; 17(10): 3193-203, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21325073

RESUMEN

PURPOSE: The aim of this study was to show preclinical efficacy and clinical development potential of PKI-587, a dual phosphoinositide 3-kinase (PI3K)/mTOR inhibitor. EXPERIMENTAL DESIGN: In vitro class 1 PI3K enzyme and human tumor cell growth inhibition assays and in vivo five tumor xenograft models were used to show efficacy. RESULTS: In vitro, PKI-587 potently inhibited class I PI3Ks (IC(50) vs. PI3K-α = 0.4 nmol/L), PI3K-α mutants, and mTOR. PKI-587 inhibited growth of 50 diverse human tumor cell lines at IC(50) values of less than 100 nmol/L. PKI-587 suppressed phosphorylation of PI3K/mTOR effectors (e.g., Akt), and induced apoptosis in human tumor cell lines with elevated PI3K/mTOR signaling. MDA-MB-361 [breast; HER2(+), PIK3CA mutant (E545K)] was particularly sensitive to this effect, with cleaved PARP, an apoptosis marker, induced by 30 nmol/L PKI-587 at 4 hours. In vivo, PKI-587 inhibited tumor growth in breast (MDA-MB-361, BT474), colon (HCT116), lung (H1975), and glioma (U87MG) xenograft models. In MDA-MB-361 tumors, PKI-587 (25 mg/kg, single dose i.v.) suppressed Akt phosphorylation [at threonine(T)308 and serine(S)473] for up to 36 hours, with cleaved PARP (cPARP) evident up to 18 hours. PKI-587 at 25 mg/kg (once weekly) shrank large (∼1,000 mm(3)) MDA-MB-361 tumors and suppressed tumor regrowth. Tumor regression correlated with suppression of phosphorylated Akt in the MDA-MB-361 model. PKI-587 also caused regression in other tumor models, and efficacy was enhanced when given in combination with PD0325901 (MEK 1/2 inhibitor), irinotecan (topoisomerase I inhibitor), or HKI-272 (neratinib, HER2 inhibitor). CONCLUSION: Significant antitumor efficacy and a favorable pharmacokinetic/safety profile justified phase 1 clinical evaluation of PKI-587.


Asunto(s)
Morfolinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Triazinas/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Femenino , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Morfolinas/farmacología , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Resultado del Tratamiento , Triazinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Drug Metab Dispos ; 39(1): 106-16, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20952552

RESUMEN

It is important to gain an understanding of the pharmacological activities of metabolite(s) of compounds in development, especially if they are found in systemic circulation in humans. Pharmacological evaluation of metabolites is normally conducted with synthetic standards, which become available during various stages of drug development. However, the synthesis of metabolite standards may be protracted, taking anywhere from several weeks to months to be completed. This often slows down early pharmacological evaluation of metabolites. Once a metabolite(s) is found to possess comparable (or greater) pharmacological activity than the parent compound, additional studies are performed to better understand the implications of circulating pharmacologically active metabolite(s). To conduct some of these studies as early as possible without slowing the progression of a compound in development is important, especially if critical go or no-go decisions impinge on the outcomes from these studies. Early pharmacological evaluation of significant metabolites is hereby proposed to be conducted in the drug discovery stage so that all pertinent studies and information can be gathered in a timely manner for decision-making. It is suggested that these major metabolites be isolated, either from biological or chemical sources, and quantified appropriately. For biologically generated metabolites, NMR is proposed as the tool of choice to quantitate these metabolites before their evaluation in pharmacological assays. For metabolites that have the same UV characteristics as the parent compound, quantitation can be conducted using UV spectroscopy instead of NMR. In this article, we propose a strategy that could be used to determine the pharmacological activities of metabolites isolated in submilligram quantities.


Asunto(s)
Descubrimiento de Drogas , Microsomas Hepáticos/metabolismo , Preparaciones Farmacéuticas/metabolismo , Animales , Evaluación Preclínica de Medicamentos , Humanos , Macaca fascicularis , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Masculino , Preparaciones Farmacéuticas/química , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad
6.
Bioorg Med Chem Lett ; 20(19): 5869-73, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20797855

RESUMEN

A series of mono-morpholino 1,3,5-triazine derivatives (8a-8q) bearing a 3-oxa-8-azabicyclo[3.2.1]octane were prepared and evaluated for PI3-kinase/mTOR activity. Replacement of one of the bis-morpholines in lead compound 1 (PKI-587) with 3-oxa-8-azabicyclo[3.2.1]octane and reduction of the molecular weight yielded 8m (PKI-179), an orally efficacious dual PI3-kinase/mTOR inhibitor. The in vitro activity, in vivo efficacy, and PK properties of 8m are discussed.


Asunto(s)
Morfolinas/química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/química , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Triazinas/química , Urea/análogos & derivados , Administración Oral , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Desnudos , Morfolinas/síntesis química , Morfolinas/farmacocinética , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacocinética , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR/metabolismo , Triazinas/síntesis química , Triazinas/farmacocinética , Tropanos/química , Urea/síntesis química , Urea/química , Urea/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Med Chem ; 53(6): 2636-45, 2010 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-20166697

RESUMEN

The PI3K/Akt signaling pathway is a key pathway in cell proliferation, growth, survival, protein synthesis, and glucose metabolism. It has been recognized recently that inhibiting this pathway might provide a viable therapy for cancer. A series of bis(morpholino-1,3,5-triazine) derivatives were prepared and optimized to provide the highly efficacious PI3K/mTOR inhibitor 1-(4-{[4-(dimethylamino)piperidin-1-yl]carbonyl}phenyl)-3-[4-(4,6-dimorpholin-4-yl-1,3,5-triazin-2-yl)phenyl]urea 26 (PKI-587). Compound 26 has shown excellent activity in vitro and in vivo, with antitumor efficacy in both subcutaneous and orthotopic xenograft tumor models when administered intravenously. The structure-activity relationships and the in vitro and in vivo activity of analogues in this series are described.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Triazinas/farmacología , Adenosina Trifosfato/química , Adenosina Trifosfato/farmacología , Animales , Área Bajo la Curva , Unión Competitiva , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Concentración 50 Inhibidora , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Desnudos , Modelos Químicos , Modelos Moleculares , Estructura Molecular , Morfolinas/química , Morfolinas/farmacocinética , Morfolinas/farmacología , Mutación , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/prevención & control , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Análisis de Supervivencia , Serina-Treonina Quinasas TOR , Triazinas/química , Triazinas/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Med Chem ; 53(1): 452-9, 2010 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-19928864

RESUMEN

Hyperactivation of the PI3K/AKT/mTOR signaling pathway is common in cancer, and PI3K and mTOR act synergistically in promoting tumor growth, survival, and resistance to chemotherapy. Thus, combined targeting of PI3K and mTOR presents an opportunity for robust and synergistic anticancer efficacy. 17-Hydroxywortmannin (2a) analogues conjugated to rapamycin (3a) analogues via a prodrug linker are uniquely positioned for this approach. Our efforts led to the discovery of diester-linked conjugates that, upon in vivo hydrolysis, released two highly potent inhibitors. Conjugate 7c provided enhanced solubility relative to 3a and to an equivalent mixture of 3a and 9a and demonstrated profound activity in U87MG mouse xenografts, achieving an MED of 1.5 mg/kg, following weekly intravenous dosing. At 15 mg/kg, 7c completely inhibited the growth of HT29 tumors, whereas an equivalent mixture of the inhibitors was poorly tolerated. In the A498 renal tumor model, 7c exhibited superior efficacy over 3a or 9a when administered as a single agent or in combination with bevacizumab. Thus, we have uncovered a novel approach to target both PI3K and mTOR via hybrid inhibitors, leading to a broader and more robust anticancer efficacy.


Asunto(s)
Androstadienos/farmacología , Antineoplásicos/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Renales/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Sirolimus/farmacología , Androstadienos/síntesis química , Androstadienos/química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Diseño de Fármacos , Estabilidad de Medicamentos , Humanos , Ratones , Ratones Desnudos , Conformación Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Ratas , Sirolimus/síntesis química , Sirolimus/química , Estereoisomerismo , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR , Células Tumorales Cultivadas , Wortmanina , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Med Chem ; 53(2): 897-910, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-20025292

RESUMEN

We are introducing a novel series of 2,4-diaminoquinazolines as beta-catenin/Tcf4 inhibitors which were identified by ligand-based design. Here we elucidate the SAR of this series and explain how we were able to improve key molecular properties such as solubility and cLogP leading to compound 9. Analogue 9 exhibited better biological activity and improved physical and pharmacological properties relative to the HTS hit 49. Furthermore, 9 demonstrated good cell growth inhibition against several human colorectal cancer lines such as LoVo and HT29. In addition, treatment with compound 9 led to gene expression changes that overlapped significantly with the transcriptional profile resulting from the pathway inhibition by siRNA knockdown of beta-catenin or Tcf4. Subsequently, 9 was tested for efficacy in a beta-catenin/RKE-mouse xenograft, where it led to more then 50% decrease in tumor volume.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Quinazolinas/síntesis química , Factores de Transcripción TCF/efectos de los fármacos , beta Catenina/efectos de los fármacos , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Diseño de Fármacos , Humanos , Ratones , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Relación Estructura-Actividad , Factor de Transcripción 4 , Factores de Transcripción/efectos de los fármacos , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Med Chem ; 53(2): 798-810, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19968288

RESUMEN

Herein we describe the identification and lead optimization of triazolopyrimidines as a novel class of potent dual PI3K/mTOR inhibitors, resulting in the discovery of 3 (PKI-402). Compound 3 exhibits good physical properties and PK parameters, low nanomolar potency against PI3Kalpha and mTOR, and excellent inhibition of cell proliferation in several human cancer cell lines. Furthermore, in vitro and in vivo biomarker studies demonstrated the ability of 3 to shut down the PI3K/Akt pathway and induce apoptosis in cancer cells. In addition, 3 showed excellent in vivo efficacy in various human cancer xenografts, validating suppression of PI3K/mTOR signaling as a potential anticancer therapy.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/síntesis química , Triazoles/síntesis química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase Ib , Humanos , Isoenzimas/antagonistas & inhibidores , Pirimidinas/farmacología , Ratas , Serina-Treonina Quinasas TOR , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Bioorg Med Chem Lett ; 19(17): 4980-3, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19640711

RESUMEN

The synthesis and SAR of a series of 2,4-diamino-quinazoline derivatives as beta-catenin/Tcf-4 inhibitors are described. This series was developed by modifying the initial lead 1, which was identified by screening of our compound library and found to inhibit the beta-catenin/Tcf-4 pathway. Replacement of the biphenyl moiety in compound 1 with the N-phenylpiperidine-4-carboxamide chain as in 2, resulted in a number of new analogues, which are potent inhibitors of the beta-catenin/Tcf-4 pathway. Compound such as 16k exhibited good cellular potency, solubility, metabolic stability and oral bioavailability.


Asunto(s)
Anilidas/química , Antineoplásicos/química , Neoplasias Colorrectales/tratamiento farmacológico , Quinazolinas/química , Factores de Transcripción TCF/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Anilidas/síntesis química , Anilidas/farmacocinética , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos , Quinazolinas/síntesis química , Quinazolinas/farmacocinética , Relación Estructura-Actividad , Factores de Transcripción TCF/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
13.
Hepatology ; 49(3): 745-52, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19072827

RESUMEN

UNLABELLED: Anti-hepatitis C virus (HCV) drug development has been challenged by a lack of experience with inhibitors inclusive of in vitro, animal model, and clinical study. This manuscript outlines activity and correlation across such a spectrum of models and into clinical trials with a novel selective nonstructural protein 5B (NS5B) polymerase inhibitor, HCV796. Enzyme assays yielded median inhibitory concentration (IC(50)) values of 0.01 to 0.14 microM for genotype 1, with half maximal effective concentration (EC(50)s) of 5 nM and 9 nM against genotype 1a and 1b replicons. In the chimeric mouse model, a 2.02 +/- 0.55 log reduction in HCV titer was seen with monotherapy, whereas a suboptimal dose of 30 mg/kg three times per day in combination with interferon demonstrated a 2.44 log reduction (P = 0.001 versus interferon alone) Clinical outcomes in combination with pegylated interferon and ribavirin have revealed additive efficacy in treatment naïve patients. Abnormal liver function test results were observed in 8% of HCV-796 patients treated for over 8 weeks, resulting in suspension of further trial activity. CONCLUSION: The RNA-dependent RNA polymerase inhibitor HCV796 demonstrated potent anti-HCV activity consistently through enzyme inhibition assays, subgenomic replicon, and chimeric mouse studies. Strong correlations of outcomes in the mouse model were seen with subsequent clinical trials, including a plateau in dose-related antiviral activity and additive impact from combination therapy with interferon. These outcomes demonstrate the utility of the range of in vitro and in vivo models now available for anti-HCV drug development and support the potential utility of polymerase inhibitors in future combination therapies for HCV treatment.


Asunto(s)
Antivirales/uso terapéutico , Benzofuranos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Hígado/efectos de los fármacos , Hígado/virología , Sulfonamidas/uso terapéutico , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Antivirales/farmacología , Benzofuranos/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Quimioterapia Combinada , Inhibidores Enzimáticos/farmacología , Hepacivirus/fisiología , Hepatocitos/trasplante , Humanos , Interferón alfa-2 , Interferón-alfa/uso terapéutico , Hígado/patología , Ratones , Ratones SCID , Polietilenglicoles , Proteínas Recombinantes , Replicón/efectos de los fármacos , Ribavirina/uso terapéutico , Sulfonamidas/farmacología
14.
Bioorg Med Chem ; 16(20): 9202-11, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18815050

RESUMEN

A series of substituted 7-alkenyl 4[3-chloro-4-(1-methyl-1H-imidazol-2-ylsulfanyl)]anilino-3-quinolinecarbonitrile analogs were synthesized and evaluated as MEK1 kinase inhibitors. The synthetic details, structure-activity relationships, biological activity, and selected oral exposure studies of these analogs are described. From these studies, compound 5m was chosen as a strong candidate for further evaluation. The selectivity of 5m was ascertained against a panel of 17 kinases, where activity was observed against EGFR, Src, Lyn, and IR kinases. Western blot studies in WM-266 cells demonstrated that 5m inhibited phosphorylation of ERK, while additional kinase pathways tested showed no inhibition at up to 10 microM of 5 m. PK studies, as well as a xenograft and in vivo biomarker studies are described for 5m.


Asunto(s)
Alquenos/química , Compuestos de Anilina/química , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Nitrilos/síntesis química , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Animales , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Isoquinolinas/química , MAP Quinasa Quinasa 1/metabolismo , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias/enzimología , Nitrilos/química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Especificidad por Sustrato , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Chemother Pharmacol ; 61(6): 1027-35, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-17668210

RESUMEN

PURPOSE: The present study aims to establish a method that provides fast, precise and reproducible pharmacokinetic (PK) parameters of antibody-calicheamicin conjugates. The method should discriminate between PK of the antibody moiety and PK of the conjugated calicheamicin (CM). METHODS: The conjugates gemtuzumab ozogamicin (CMA-676, Mylotarg) or inotuzumab ozogamicin (CMC-544) were injected in the tail vein of nude mice. At regular time intervals, 5 mul whole blood samples were taken from the tail artery. Concentrations of conjugated CMA-676 or CMC-544 as well as concentrations of their respective antibody moiety were determined by sandwich plasmon resonance. This detection system measures changes in the plasma resonance angle caused by the interaction of macromolecules on biosensor chips. We determined as a first measure the binding of CMA-676 or CMC-544 to their respective antigens, CD33 or CD22. As a second measure we determined the amount of CM on the antigen-bound conjugates. This was done by determination of changes in plasma resonance angle after binding of an anti-CM antibody. RESULTS: Sandwich plasmon resonance allowed detection of both conjugates in blood of mice in a range of 100-1,000 ng/ml protein. Due to the precision of the sampling and detection methods, PK values of each conjugate were determined in individual mice. Calicheamicin bound to antibody was eliminated faster than the antibody alone. The presence of a CD22-expressing tumour in mice reduced the plasma levels of the CD22-targeting conjugate but not of the CD33-targeting one. CONCLUSIONS: Using small blood samples from a mouse, the sandwich plasmon resonance method provided PK-values of CM-conjugates and information about the stability of the linkage in vivo. Comparison between the PK-values of CM-conjugates in tumour-bearing and tumour-free mice suggested that retention of the conjugate in tumour tissue due to antigen targeting could be deduced from the plasma levels.


Asunto(s)
Aminoglicósidos/farmacocinética , Anticuerpos Monoclonales/farmacocinética , Aminoglicósidos/administración & dosificación , Aminoglicósidos/sangre , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales Humanizados , Área Bajo la Curva , Línea Celular Tumoral , Gemtuzumab , Semivida , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Inotuzumab Ozogamicina , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Conejos , Resonancia por Plasmón de Superficie
16.
J Med Chem ; 49(4): 1373-8, 2006 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-16480272

RESUMEN

Phosphoinositide 3-kinase (PI3K) is an important target for cancer chemotherapy due to the deregulation of its signaling pathway in a wide spectrum of human tumors. Wortmannin and its analogues are potent PI3K inhibitors whose therapeutic use has been impeded by inherent defects such as instability and toxicity. Pegylation of wortmannin and 17-hydroxywortmannin gives rise to conjugates with improved properties, including a higher therapeutic index. Pegylated 17-hydroxywortmannin (8, PWT-458) has been selected for further development.


Asunto(s)
Androstadienos/síntesis química , Antineoplásicos/síntesis química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Polietilenglicoles/química , Androstadienos/química , Androstadienos/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Humanos , Ratones , Ratones Desnudos , Polietilenglicoles/síntesis química , Polietilenglicoles/farmacología , Relación Estructura-Actividad , Wortmanina , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Res ; 65(12): 5358-64, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15958584

RESUMEN

Src up-regulation is a common event in human cancers. In colorectal cancer, increased Src levels are an indicator of poor prognosis, and progression to metastatic disease is associated with substantial increases in Src activity. Therefore, we examined the activity of SKI-606, a potent inhibitor of Src and Abl kinases, against colon tumor lines in vitro and in s.c. tumor xenograft models. SKI-606 inhibited Src autophosphorylation with an IC(50) of approximately 0.25 micromol/L in HT29 cells. Phosphorylation of Tyr(925) of focal adhesion kinase, a Src substrate, was reduced by similar concentrations of inhibitor. Antiproliferative activity on plastic did not correlate with Src inhibition in either HT29 or Colo205 cells (IC(50)s, 1.5 and 2.5 micromol/L, respectively), although submicromolar concentrations of SKI-606 inhibited HT29 cell colony formation in soft agar. SKI-606 also caused loosely aggregated Colo205 spheroids to condense into compact spheroids. On oral administration to nude mice at the lowest efficacious dose, peak plasma concentrations of approximately 3 micromol/L, an oral bioavailability of 18%, and a t(1/2) of 8.6 hours were observed. SKI-606 was orally active in s.c. colon tumor xenograft models and caused substantial reductions in Src autophosphorylation on Tyr(418) in HT29 and Colo205 tumors. SKI-606 inhibited HT29 tumor growth on once daily administration, whereas twice daily administration was necessary to inhibit Colo205, HCT116, and DLD1 tumor growth. These results support development of SKI-606 as a therapeutic agent for treatment of colorectal cancer.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Administración Oral , Compuestos de Anilina/farmacocinética , Animales , Antineoplásicos/farmacocinética , Neoplasias del Colon/enzimología , Neoplasias del Colon/metabolismo , Femenino , Células HCT116 , Células HT29 , Humanos , Ratones , Ratones Desnudos , Nitrilos/farmacocinética , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacocinética , Quinolinas/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/antagonistas & inhibidores
18.
Cancer Biol Ther ; 4(5): 538-45, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15846106

RESUMEN

Deregulated phosphatidylinositol 3-kinase (PI3K) signaling pathway is widely implicated in tumor growth and resistance to chemotherapy. While a strong rationale exists for pharmacological targeting of PI3K, only a few proof-of-principle in vivo efficacy studies are currently available. PWT-458, pegylated-17-hydroxywortmannin, is a novel and highly potent inhibitor of PI3K in animal models. Upon in vivo cleavage of its poly(ethyleneglycol) (PEG), PWT-458 releases its active moiety 17-hydroxywortmannin (17-HWT), the most potent inhibitor in its class. Here we show that a single intravenous injection of PWT-458 rapidly inhibited PI3K signaling, as measured by a complete loss of AKT (Ser-473) phosphorylation in xenograft tumors grown in nude mice. Following a daily X5 dosing regimen, PWT-458 demonstrated single-agent antitumor activity in nude mouse xenograft models of U87MG glioma, nonsmall cell lung cancer (NSCLC) A549, and renal cell carcinoma (RCC) A498. Efficacious doses ranged from 0.5 mg/kg to 10 mg/kg, achieving a superior therapeutic index over 17-HWT. PWT-458 augmented anticancer efficacy of a suboptimal dose of paclitaxel against A549 and U87MG tumors. Combination treatment of PWT-458 and an mTOR inhibitor, Pegylated-Rapamycin (Peg-Rapa), resulted in an enhanced antitumor efficacy in U87MG. Finally, PWT-458 in combination with interferon-alpha (Intron-A) caused a dramatic regression of RCC A498, which was not achieved by either agent alone. These studies identify PWT-458 as an effective anticancer agent and provide strong proof-of-principle for targeting the PI3K pathway as novel anticancer therapy.


Asunto(s)
Androstadienos/farmacología , Neoplasias/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transducción de Señal/efectos de los fármacos , Androstadienos/química , Animales , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos Fitogénicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Glioma/tratamiento farmacológico , Glioma/metabolismo , Glioma/patología , Humanos , Interferón-alfa/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Estructura Molecular , Peso Molecular , Trasplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patología , Paclitaxel/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirolimus/farmacología , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA