Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Chem Pharm Bull (Tokyo) ; 69(11): 1054-1060, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34719586

RESUMEN

In the evaluation of the druggability of candidate compounds, it was vital to predict the oral bioavailability of compounds from apparent permeability (Papp) across Caco-2 cell-culture model of intestinal epithelium cultured on commercial transwell plate inserts. The study was to investigate the transport characteristics and permeability of FL118 (10, 11-Methylenedioxy-20(S)-camptothecin) derivatives 7-Q6 (7-(4-Ethylphenyl)-10, 11-methylenedioxy-20(S)-camptothecin) and 7-Q20 (7-(4-Trifluoromethylphenyl)-10, 11-methylenedioxy-20(S)-camptothecin). Transport characteristics and permeability of the tested compounds to the small intestine were assessed at different concentrations (0.5, 1 µM) via Caco-2 cell monolayers model in vitro. Uptake studies based on Caco-2 cells, including temperatures, concentrations, and the influence of efflux transporters, were combined to confirm the transport characteristics of the tested compounds. Furthermore, cytotoxicity results showed that the concentrations used in the experiments were non-toxic and harmless to cells. In addition, The Papp of 7-Q6 was (3.69 ± 1.07) × 10-6 cm/s with efflux ratio (ER) 0.98, while the Papp of 7-Q20 was (7.78 ± 0.89) × 10-6 cm/s with ER 1.05 for apical-to-basolateral (AP→BL) at 0.5 µM, suggesting that 7-Q20 might possess higher oral bioavailability in vivo. Furthermore, P-glycoprotein (P-gp) was proved to slightly affect the accumulations of 7-Q20, while the absorption of 7-Q6 was irrelevant with P-gp and breast cancer resistant protein (BCRP) based on the cellular uptake assays. Accordingly, 7-Q6 was completely absorbed by passive diffusion, and 7-Q20 was mainly dependent on passive diffusion with being effluxed by P-gp slightly. Meanwhile, both 7-Q6 and 7-Q20 were potential antitumor drugs that might exhibit high oral bioavailability in the body.


Asunto(s)
Antineoplásicos/química , Benzodioxoles/química , Membrana Celular/metabolismo , Indolizinas/química , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Antineoplásicos/administración & dosificación , Benzodioxoles/administración & dosificación , Disponibilidad Biológica , Transporte Biológico , Células CACO-2 , Camptotecina/química , Camptotecina/metabolismo , Membrana Celular/ultraestructura , Permeabilidad de la Membrana Celular , Supervivencia Celular/efectos de los fármacos , Absorción Gastrointestinal , Humanos , Indolizinas/administración & dosificación , Mucosa Intestinal/metabolismo
2.
Cell Death Dis ; 12(2): 179, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589591

RESUMEN

Human epidermal growth factor receptor 2 gene (HER2) is focally amplified in approximately 20% of breast cancers. HER2 inhibitors alone are not effective, and sensitizing agents will be necessary to move away from a reliance on heavily toxic chemotherapeutics. We recently demonstrated that the efficacy of HER2 inhibitors is mitigated by uniformly low levels of the myeloid cell leukemia 1 (MCL-1) endogenous inhibitor, NOXA. Emerging clinical data have demonstrated that clinically advanced cyclin-dependent kinase (CDK) inhibitors are effective MCL-1 inhibitors in patients, and, importantly, well tolerated. We, therefore, tested whether the CDK inhibitor, dinaciclib, could block MCL-1 in preclinical HER2-amplified breast cancer models and therefore sensitize these cancers to dual HER2/EGFR inhibitors neratinib and lapatinib, as well as to the novel selective HER2 inhibitor tucatinib. Indeed, we found dinaciclib suppresses MCL-1 RNA and is highly effective at sensitizing HER2 inhibitors both in vitro and in vivo. This combination was tolerable in vivo. Mechanistically, liberating the effector BCL-2 protein, BAK, from MCL-1 results in robust apoptosis. Thus, clinically advanced CDK inhibitors may effectively combine with HER2 inhibitors and present a chemotherapy-free therapeutic strategy in HER2-amplified breast cancer, which can be tested immediately in the clinic.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Óxidos N-Cíclicos/farmacología , Indolizinas/farmacología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Compuestos de Piridinio/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Óxidos N-Cíclicos/administración & dosificación , Sinergismo Farmacológico , Femenino , Amplificación de Genes , Humanos , Indoles/administración & dosificación , Indoles/farmacología , Indolizinas/administración & dosificación , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Oxazoles/administración & dosificación , Oxazoles/farmacología , Piridinas/administración & dosificación , Piridinas/farmacología , Compuestos de Piridinio/administración & dosificación , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Quinolinas/administración & dosificación , Quinolinas/farmacología , Distribución Aleatoria , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Clin Transl Sci ; 13(6): 1178-1188, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32738099

RESUMEN

The combination of drugs targeting Ral and PI3K/AKT signaling has antitumor efficacy in preclinical models of pancreatic cancer. We combined dinaciclib (small molecule cyclin dependent kinase inhibitor with MK-2206 (Akt inhibitor) in patients with previously treated/metastatic pancreatic cancer. Patients were treated with dinaciclib (6-12 mg/m2 i.v.) and MK-2206 (60-135 mg p.o.) weekly. Tumor biopsies were performed to measure pAKT, pERK, and Ki67 at baseline and after one completed cycle (dose level 2 and beyond). Thirty-nine patients participated in the study. The maximum tolerated doses were dinaciclib 9 mg/m2 and MK-2206 135 mg. Treatment-related grade 3 and 4 toxicities included neutropenia, lymphopenia, anemia, hyperglycemia, hyponatremia, and leukopenia. No objectives responses were observed. Four patients (10%) had stable disease as their best response. At the recommended dose, median survival was 2.2 months. Survival rates at 6 and 12 months were 11% and 5%, respectively. There was a nonsignificant reduction in pAKT composite scores between pretreatment and post-treatment biopsies (mean 0.76 vs. 0.63; P = 0.635). The combination of dinaciclib and MK-2206 was a safe regimen in patients with metastatic pancreatic cancer, although without clinical benefit, possibly due to not attaining biologically effective doses. Given the strong preclinical evidence of Ral and AKT inhibition, further studies with better tolerated agents should be considered.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Carcinoma Ductal Pancreático/tratamiento farmacológico , Óxidos N-Cíclicos/toxicidad , Compuestos Heterocíclicos con 3 Anillos/toxicidad , Indolizinas/toxicidad , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/toxicidad , Compuestos de Piridinio/toxicidad , Administración Oral , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Biopsia , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Óxidos N-Cíclicos/administración & dosificación , Femenino , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Humanos , Indolizinas/administración & dosificación , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Páncreas/efectos de los fármacos , Páncreas/patología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Inhibidores de Proteínas Quinasas/administración & dosificación , Compuestos de Piridinio/administración & dosificación , Tasa de Supervivencia , Resultado del Tratamiento
4.
Phytomedicine ; 60: 152938, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31078367

RESUMEN

BACKGROUND: Tylophorine (TYL) is an alkaloid with antiproliferative action in cancer cells. Vascular smooth muscle cell (VSMC) proliferation and neointima formation contribute to restenosis after percutaneous coronary interventions. HYPOTHESIS/PURPOSE: Our goal was to examine the potential of TYL to inhibit VSMC proliferation and migration, and to dissect underlying signaling pathways. STUDY DESIGN AND METHODS: TYL was administered to platelet-derived growth factor (PDGF-BB)-stimulated, serum-stimulated, quiescent and unsynchronized VSMC of rat and human origin. BrdU incorporation and resazurin conversion were used to assess cell proliferation. Cell cycle progression was analyzed by flow cytometry of propidium iodide-stained nuclei. Expression profiles of proteins and mRNAs were determined using western blot analysis and RT-qPCR. The Click-iT OPP Alexa Fluor 488 assay was used to monitor protein biosynthesis. RESULTS: TYL inhibited PDGF-BB-induced proliferation of rat aortic VSMCs by arresting cells in G1 phase of the cell cycle with an IC50 of 0.13 µmol/l. The lack of retinoblastoma protein phosphorylation and cyclin D1 downregulation corroborated a G1 arrest. Inhibition of proliferation and cyclin D1 downregulation were species- and stimulus-independent. TYL also decreased levels of p21 and p27 proteins, although at later time points than observed for cyclin D1. Co-treatment of VSMC with TYL and MG132 or cycloheximide (CHX) excluded proteasome activation by TYL as the mechanism of action. Comparable time-dependent downregulation of cyclin D1, p21 and p27 in TYL- or CHX-treated cells, together with decreased protein synthesis observed in the Click-iT assay, suggests that TYL is a protein synthesis inhibitor. Besides proliferation, TYL also suppressed migration of PDGF-activated VSMC. In a human saphenous vein organ culture model for graft disease, TYL potently inhibited intimal hyperplasia. CONCLUSION: This unique activity profile renders TYL an interesting lead for the treatment of vasculo-proliferative disorders, such as restenosis.


Asunto(s)
Alcaloides/farmacología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclina D1/efectos de los fármacos , Indolizinas/farmacología , Fenantrenos/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Alcaloides/administración & dosificación , Alcaloides/química , Animales , Becaplermina/administración & dosificación , Ciclina D1/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Indolizinas/administración & dosificación , Indolizinas/química , Miocitos del Músculo Liso/efectos de los fármacos , Técnicas de Cultivo de Órganos , Fenantrenos/administración & dosificación , Fenantrenos/química , Ratas , Ratas Sprague-Dawley , Venas Umbilicales
5.
Pharmacol Rep ; 71(2): 233-242, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30807980

RESUMEN

BACKGROUND: Diabetes mellitus is a deadly disorder in human which induce chronic complications. The streptozotocin (STZ)-induced diabetes in rat is the most common animal model of human diabetes. The present study investigated the effects of novel indolizine derivatives (1-16) on plasma blood glucose concentrations in STZ-diabetic rats. METHODS: In vitro experiments were performed on 1,1-diphenyl-2-picrylhydrazyl (DPPH), superoxide free radicals, α-glucosidase enzyme and in vivo studies on normal, oral glucose loaded and STZ-induced diabetic rats. RESULTS: Among all synthetic derivatives, compound 12 showed good inhibitory profile against DPPH, superoxide free radicals and α-glucosidase enzyme with half maximal inhibitory concentration (IC50) values of 56.2, 33.5 and 26.5 µg/mL, respectively. The lethal dosage of indolizine derivatives was found to be above 1000 mg/kg body weight (b.w.). From the in vivo studies, it can be determined that the compound 12 depicted pronounced protective hypoglycemic effects in normal, glucose-loaded and STZ-induced diabetic rats with respect to the standard. Furthermore, 21 days of successive treatment with compound 12 in diabetic rats exhibited better recovery of body weight and considerable variations in biochemical parameters as that of the standard drug. Moreover, the histopathological section of pancreas and testes justifies the rehabilitation and regeneration of islets, acini and Sertoli cells in animals treated with compound 12. CONCLUSION: Our data suggest that the indolizine derivatives can be a benchmarks for designing potent oral antidiabetic agents.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/farmacología , Indolizinas/farmacología , Administración Oral , Animales , Diabetes Mellitus Experimental/fisiopatología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/química , Indolizinas/administración & dosificación , Indolizinas/química , Concentración 50 Inhibidora , Masculino , Ratas , Ratas Wistar , Estreptozocina
6.
Int J Oncol ; 53(2): 801-814, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29901068

RESUMEN

Adult T­cell leukemia/lymphoma (ATLL) is a disorder involving human T-cell leukemia virus type 1 (HTLV­1)-infected T­cells characterized by increased clonal neoplastic proliferation. PDZ-binding kinase (PBK) [also known as T­lymphokine-activated killer cell-originated protein kinase (TOPK)] is a serine/threonine kinase expressed in proliferative cells and is phosphorylated during mitosis. In this study, the expression and phosphorylation of PBK/TOPK were examined by western blot analysis and RT­PCR. We found that PBK/TOPK was upregulated and phosphorylated in HTLV­1-transformed T­cell lines and ATLL­derived T­cell lines. Notably, CDK1/cyclin B1, which phosphorylates PBK/TOPK, was overexpressed in these cells. HTLV­1 infection upregulated PBK/TOPK expression in peripheral blood mononuclear cells (PBMCs) in co-culture assays. The potent PBK/TOPK inhibitors, HI­TOPK­032, and fucoidan from brown algae, decreased the proliferation and viability of these cell lines in a dose­dependent manner. By contrast, the effect of HI­TOPK­032 on PBMCs was less pronounced. Treatment with HI­TOPK­032 resulted in G1 cell cycle arrest, and decreased CDK6 expression and pRb phosphorylation, which are critical determinants of progression through the G1 phase. In addition, HI­TOPK­032 induced apoptosis, as evidenced by morphological changes, the cleavage of poly(ADP-ribose) polymerase with the activation of caspase­3, -8 and -9, and an increase in the sub­G1 cell population and APO2.7-positive cells. Moreover, HI­TOPK­032 inhibited the expression of cellular inhibitor of apoptosis 2 (c-IAP2), X-linked inhibitor of apoptosis protein (XIAP), survivin and myeloid cell leukemia­1 (Mcl­1), and induced the expression of Bak and interferon-induced protein with tetratricopeptide repeats (IFIT)1, 2 and 3. It is noteworthy that the use of this inhibitor led to the inhibition of the phosphorylation of IκB kinase (IKK)α, IKKß, IκBα, phosphatase and tensin homolog (PTEN) and Akt, and to the decreased protein expression of JunB and JunD, suggesting that PBK/TOPK affects the nuclear factor-κB, Akt and activator protein­1 signaling pathways. In vivo, the administration of HI­TOPK­032 suppressed tumor growth in an ATLL xenograft model. Thus, on the whole, this study on the identification and functional analysis of PBK/TOPK suggests that this kinase is a promising molecular target for ATLL treatment.


Asunto(s)
Proteína Quinasa CDC2/genética , Ciclina B1/genética , Leucemia-Linfoma de Células T del Adulto/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Regulación hacia Arriba , Animales , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Ciclina B1/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Indolizinas/administración & dosificación , Indolizinas/farmacología , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Leucemia-Linfoma de Células T del Adulto/genética , Leucocitos Mononucleares , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Trasplante de Neoplasias , Fosforilación , Quinoxalinas/administración & dosificación , Quinoxalinas/farmacología
7.
PLoS Negl Trop Dis ; 10(8): e0004851, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27509020

RESUMEN

UNLABELLED: CELADEN was a randomized placebo-controlled trial of 50 patients with confirmed dengue fever to evaluate the efficacy and safety of celgosivir (A study registered at ClinicalTrials.gov, number NCT01619969). Celgosivir was given as a 400 mg loading dose and 200 mg bid (twice a day) over 5 days. Replication competent virus was measured by plaque assay and compared to reverse transcription quantitative PCR (qPCR) of viral RNA. Pharmacokinetics (PK) correlations with viremia, immunological profiling, next generation sequence (NGS) analysis and hematological data were evaluated as exploratory endpoints here to identify possible signals of pharmacological activity. Viremia by plaque assay strongly correlated with qPCR during the first four days. Immunological profiling demonstrated a qualitative shift in T helper cell profile during the course of infection. NGS analysis did not reveal any prominent signature that could be associated with drug treatment; however the phylogenetic spread of patients' isolates underlines the importance of strain variability that may potentially confound interpretation of dengue drug trials conducted during different outbreaks and in different countries. Celgosivir rapidly converted to castanospermine (Cast) with mean peak and trough concentrations of 5727 ng/mL (30.2 µM) and 430 ng/mL (2.3 µM), respectively and cleared with a half-life of 2.5 (± 0.6) hr. Mean viral log reduction between day 2 and 4 (VLR2-4) was significantly greater in secondary dengue than primary dengue (p = 0.002). VLR2-4 did not correlate with drug AUC but showed a trend of greater response with increasing Cmin. PK modeling identified dosing regimens predicted to achieve 2.4 to 4.5 times higher Cmin. than in the CELADEN trial for only 13% to 33% increase in overall dose. A small, non-statistical trend towards better outcome on platelet nadir and difference between maximum and minimum hematocrit was observed in celgosivir-treated patients with secondary dengue infection. Optimization of the dosing regimen and patient stratification may enhance the ability of a clinical trial to demonstrate celgosivir activity in treating dengue fever based on hematological endpoints. A new clinical trial with a revised dosing regimen is slated to start in 2016 (NCT02569827). Furthermore celgosivir's potential value for treatment of other flaviruses such as Zika virus should be investigated urgently. TRIAL REGISTRATION: ClinicalTrials.gov NCT01619969.


Asunto(s)
Antivirales/administración & dosificación , Antivirales/farmacocinética , Virus del Dengue/efectos de los fármacos , Dengue/tratamiento farmacológico , Dengue/inmunología , Indolizinas/administración & dosificación , Indolizinas/farmacocinética , Carga Viral/efectos de los fármacos , Adulto , Antivirales/efectos adversos , Citocinas/sangre , Dengue/virología , Virus del Dengue/genética , Virus del Dengue/aislamiento & purificación , Virus del Dengue/fisiología , Femenino , Semivida , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Indolizinas/efectos adversos , Indolizinas/sangre , Masculino , Filogenia , Células TH1/inmunología , Viremia/tratamiento farmacológico , Replicación Viral/efectos de los fármacos
8.
Antiviral Res ; 127: 10-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26794905

RESUMEN

Although the antiviral drug celgosivir, an α-glucosidase I inhibitor, is highly protective when given twice daily to AG129 mice infected with dengue virus, a similar regimen of twice daily dosing did not significantly reduce serum viral loads in patients in a recent clinical trial. This failure presumably might reflect the initiation of treatment when patients were already viremic. To better mimic the clinical setting, we used viruses isolated from patients to develop new mouse models of DENV1 and DENV2 infection and employed the models to test the twice daily treatment, begun either on the day of infection or on the third day post-infection, when the mice had peak of viremia. We found that, although the treatment started on day 0 was effective on viral load reduction, it provided no benefit when begun on day 3, indicating that in vivo antiviral efficacy becomes less prominent once viremia reaches the peak level. To determine if the therapeutic regimen in humans could be improved, we tested regimen of four-times daily treatment and found that the treatment significantly reduced viremia, suggesting that a similar regimen may be effective in a human clinical trial. A new clinical trial to investigate an altered dosing regimen has been approved (NCT02569827).


Asunto(s)
Virus del Dengue/aislamiento & purificación , Dengue/tratamiento farmacológico , Indolizinas/administración & dosificación , Animales , Antivirales/administración & dosificación , Línea Celular , Chlorocebus aethiops , Cricetinae , Citocinas/sangre , Dengue/sangre , Dengue/virología , Virus del Dengue/clasificación , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Humanos , Técnicas In Vitro , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Serogrupo , Células Vero , Carga Viral/efectos de los fármacos , Viremia/tratamiento farmacológico , Viremia/virología , alfa-Glucosidasas/metabolismo
9.
Int J Oncol ; 47(1): 179-87, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25936609

RESUMEN

Neuroblastoma is the most common cancer in infants and the fourth most common cancer in children. Our previous study showed that PF403 had a potent antitumor ability. In the present study, we evaluated the anti-neuroblastoma property of PF403 and investigated the underlying mechanisms. MTT assay, colony formation assay and flow cytometry assay were used to assess cytotoxicity of PF403 on SH-SY5Y cells. Transwell assay was chosen to estimate the anti-invasion ability of PF403 on neuroblastoma cells. The protein expression was detected by western blot analysis. The SH-SY5Y brain xenograft model was used to assess in vivo antitumor activity of PF403. PF403-mediated SH-SY5Y cell death was found to be dose- and time-dependent, and PF403 was able to limit invasion and metastasis of neuroblastoma cells. MRI and pathology analysis proved that the pro-drug of PF403, CAT3, inhibited SH-SY5Y cells in vivo. PF403 decreased expression of phosphorylated FAK, MMP-2 and MMP-9 proteins, and downregulated the activity of PI3K/AKT and Raf/ERK pathways, followed by regulation of the proteins expression of Bcl-2 family, activated caspase-3, -9 and PARP and initiation of apoptosis of neuroblastoma cells. PF403 exerted cytotoxicity against SH-SY5Y neuroblastoma cell both in vitro and in vivo, and inhibited its invasion ability, suggesting PF403 has potential as a new anticancer drug for the treatment of neuroblastoma.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Indolicidinas/administración & dosificación , Indolizinas/administración & dosificación , Neuroblastoma/tratamiento farmacológico , Fenantrenos/administración & dosificación , Profármacos/administración & dosificación , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indolicidinas/farmacología , Indolizinas/farmacología , Ratones , Invasividad Neoplásica , Fenantrenos/farmacología , Profármacos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cancer Res ; 74(24): 7487-97, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25512388

RESUMEN

Anticancer agent FL118 was recently identified in screening of small-molecule inhibitors of human survivin expression. Although FL118 is a camptothecin analogue, its antitumor potency is much superior to other FDA-approved camptothecin analogues (irinotecan and topotecan). The mechanism of action (MOA) underlying the antitumor effects of FL118 remains to be fully elucidated. Here, we report that FL118 activates tumor suppressor p53 as a novel MOA in p53 wild-type cancer cells. Our studies show that this MOA involves an induction of proteasomal degradation of MdmX, a critical negative regulator of p53, in a manner largely independent of ATM-dependent DNA damage signaling pathway but dependent on E3-competent Mdm2. FL118 inhibits p53 polyubiquitination and monoubiquitination by Mdm2-MdmX E3 complex in cells and in cell-free systems. In contrast, FL118 stimulates Mdm2-mediated MdmX ubiquitination. Coimmunoprecipitation revealed that FL118 slightly decreases Mdm2-p53 interactions and moderately increases Mdm2-MdmX interactions, suggesting a change of targeting specificity of Mdm2-MdmX E3 complex from p53 to MdmX, resulting in accelerated MdmX degradation. As a result, p53 ubiquitination by Mdm2-MdmX E3 complex is reduced, which in turn activates p53 signaling. Activation of the p53 pathway by FL118 induces p53-dependent senescence in colorectal cancer cells. However, in the absence of p53 or in the presence of MdmX overexpression, FL118 promotes p53-independent apoptosis. These two distinct cellular consequences collectively contribute to the potent effects of FL118 to inhibit clonogenic potential of colon cancer cells. This study identifies a potential application of FL118 as an MdmX inhibitor for targeted therapies.


Asunto(s)
Benzodioxoles/administración & dosificación , Neoplasias Colorrectales/genética , Indolizinas/administración & dosificación , Proteínas Nucleares/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Proteína p53 Supresora de Tumor/genética , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular , Línea Celular Tumoral , Senescencia Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Daño del ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/efectos de los fármacos , Survivin , Proteína p53 Supresora de Tumor/biosíntesis , Ubiquitinación/efectos de los fármacos
11.
Mol Cancer ; 12: 82, 2013 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-23895055

RESUMEN

BACKGROUND: Anti-angiogenesis targeting VEGFR2 has been considered as an important strategy for cancer therapy. Tylophorine is known to possess anti-inflammatory and antitumor activity, but its roles in tumor angiogenesis, the key step involved in tumor growth and metastasis, and the involved molecular mechanism is still unknown. Therefore, we examined its anti-angiogenic effects and mechanisms in vitro and in vivo. METHODS: We used tylophorine and analyzed its inhibitory effects on human umbilical vein endothelial cells (HUVEC) in vitro and Ehrlich ascites carcinoma (EAC) tumor in vivo. RESULTS: Tylophorine significantly inhibited a series of VEGF-induced angiogenesis processes including proliferation, migration, and tube formation of endothelial cells. Besides, it directly inhibited VEGFR2 tyrosine kinase activity and its downstream signaling pathways including Akt, Erk and ROS in endothelial cells. Using HUVECs we demonstrated that tylophorine inhibited VEGF-stimulated inflammatory responses including IL-6, IL-8, TNF-α, IFN-γ, MMP-2 and NO secretion. Tylophorine significantly inhibited neovascularization in sponge implant angiogenesis assay and also inhibited tumor angiogenesis and tumor growth in vivo. Molecular docking simulation indicated that tylophorine could form hydrogen bonds and aromatic interactions within the ATP-binding region of the VEGFR2 kinase unit. CONCLUSION: Tylophorine exerts anti-angiogenesis effects via VEGFR2 signaling pathway thus, may be a viable drug candidate in anti-angiogenesis and anti-cancer therapies.


Asunto(s)
Alcaloides/farmacología , Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Fitogénicos/farmacología , Indolizinas/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Fenantrenos/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Alcaloides/administración & dosificación , Alcaloides/química , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/química , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Indolizinas/administración & dosificación , Indolizinas/química , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Conformación Molecular , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/mortalidad , Neoplasias/patología , Óxido Nítrico/metabolismo , Fenantrenos/administración & dosificación , Fenantrenos/química , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Tylophora/química , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Int J Mol Sci ; 14(4): 8496-516, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23595000

RESUMEN

The purpose of this study was to assess the in vitro antimicrobial activity of alkaloid-enriched extracts from Prosopis juliflora (Fabaceae) pods in order to evaluate them as feed additives for ruminants. As only the basic chloroformic extract (BCE), whose main constituents were juliprosopine (juliflorine), prosoflorine and juliprosine, showed Gram-positive antibacterial activity against Micrococcus luteus (MIC = 25 µg/mL), Staphylococcus aureus (MIC = 50 µg/mL) and Streptococcus mutans (MIC = 50 µg/mL), its influence on ruminal digestion was evaluated using a semi-automated in vitro gas production technique, with monensin as the positive control. Results showed that BCE has decreased gas production as efficiently as monensin after 36 h of fermentation, revealing its positive influence on gas production during ruminal digestion. Since P. juliflora is a very affordable plant, this study points out this alkaloid enriched extract from the pods of Prosopis juliflora as a potential feed additive to decrease gas production during ruminal digestion.


Asunto(s)
Alimentación Animal/análisis , Antibacterianos/química , Prosopis/química , Rumen/efectos de los fármacos , Rumen/fisiología , Alcaloides/administración & dosificación , Animales , Antibacterianos/administración & dosificación , Bovinos , Digestión , Fermentación , Aditivos Alimentarios/administración & dosificación , Aditivos Alimentarios/química , Técnicas In Vitro , Indolizinas/administración & dosificación , Metano/biosíntesis , Pruebas de Sensibilidad Microbiana , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Rumen/microbiología
13.
J Med Chem ; 55(23): 10363-77, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23167614

RESUMEN

A series of novel tylophorine-derived dibenzoquinolines has been synthesized and their biological activity evaluated. Three assays were conducted: inhibition of cancer cell proliferation, inhibition of TGEV replication for anticoronavirus activity, and suppression of nitric oxide production in RAW264.7 cells (a measure of anti-inflammation). The most potent compound from these assays, dibenzoquinoline 33b, showed improved solubility compared to tylophorine 9a, in vivo efficacies in a lung A549 xenografted tumor mouse model and a murine paw edema model, good bioavailability, and no significant neurotoxicity (as tested by a rota-rod test for motor coordination). This is the first study to explore in detail the role of the tylophorine E ring on biological activity and very strongly suggests that tylophorine-derived dibenzoquinolines merit further development into orally active agents.


Asunto(s)
Alcaloides/farmacología , Indolizinas/farmacología , Fenantrenos/farmacología , Administración Oral , Alcaloides/administración & dosificación , Alcaloides/química , Alcaloides/farmacocinética , Animales , Disponibilidad Biológica , Línea Celular , Línea Celular Tumoral , Coronavirus/efectos de los fármacos , Coronavirus/fisiología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indolizinas/administración & dosificación , Indolizinas/química , Indolizinas/farmacocinética , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Ratones , Fenantrenos/administración & dosificación , Fenantrenos/química , Fenantrenos/farmacocinética , Replicación Viral/efectos de los fármacos
14.
Antiviral Res ; 96(1): 32-5, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22867971

RESUMEN

Celgosivir (6-O-butanoyl castanospermine), a pro-drug of the naturally occurring castanospermine, is an inhibitor of α-glucosidase I and II that is found to be a potent inhibitor of several enveloped viruses including all four serotypes of dengue virus. We showed previously that the compound fully protected AG129 mice from lethal infection with a mouse adapted dengue virus at a dose of 50mg/kg twice daily (BID) for 5days and was effective even after 48h delayed treatment. Here we show that the protection by celgosivir is dose- and schedule-dependent and that a twice-a-day regimen of 50, 25 or 10mg/kg is more protective than a single daily dose of 100mg/kg. Treatment with 50mg/kg BID castanospermine had comparable efficacy as 25mg/kg BID celgosivir, suggesting that celgosivir is approximately twice as potent as castanospermine with respect to in vivo antiviral efficacy. Pharmacokinetics (PK) studies of celgosivir in mice showed that it rapidly metabolized to castanospermine. Simulation of the PK data with the survival data for the various doses of celgosivir tested suggests that the steady-state minimum concentration is a critical parameter to note in choosing dose and schedule. These results influenced the selection of the dose regimen for a proof-of-concept clinical trial of celgosivir as a treatment against dengue fever.


Asunto(s)
Antivirales/administración & dosificación , Dengue/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Indolizinas/administración & dosificación , Animales , Antivirales/farmacocinética , Virus del Dengue/efectos de los fármacos , Modelos Animales de Enfermedad , Quimioterapia/métodos , Indolizinas/farmacocinética , Ratones , Análisis de Supervivencia
15.
Bioorg Med Chem Lett ; 22(7): 2411-4, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22406151

RESUMEN

The structure-based design, synthesis, and biological evaluation of two novel series of potent and selective MEK kinase inhibitors are described herein. The elaboration of a lead pyrrole derivative to a bicyclic dihydroindolone core provided compounds with high potency and good drug-like pharmaceutical properties. Further scaffold modification afforded a series of dihydroindolizinone inhibitors, including an orally available advanced preclinical MEK inhibitor with potent in vivo antitumor efficacy.


Asunto(s)
Indolizinas/síntesis química , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/síntesis química , Pirroles/síntesis química , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Diseño de Fármacos , Células HT29 , Humanos , Indolizinas/administración & dosificación , Indolizinas/uso terapéutico , Quinasas Quinasa Quinasa PAM/metabolismo , Estructura Molecular , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirroles/administración & dosificación , Pirroles/uso terapéutico , Ratas , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Bioorg Med Chem Lett ; 22(2): 1095-8, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22189140

RESUMEN

A novel, potent, and orally bioavailable class of product-like inhibitors of the HCV NS3 protease was discovered by constraining the P2-P3 amide bond and the P3 hydrocarbon substituent to the protease-bound conformation. This preorganization was accomplished by incorporation of the P2-P3 amide into a six-membered ring attached to the P2-proline 5-position. Isothermal calorimetric characterization of the role of hydrocarbon substitution of this six-membered ring, upon binding the HCV NS3 protease, was found to be exclusively entropic in nature. The synthesis, preliminary SAR and pharmacokinetic profiling of this compact, indolizidinone-derived scaffold are described.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Indolizinas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Administración Oral , Disponibilidad Biológica , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Indolizinas/administración & dosificación , Indolizinas/química , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Proteínas no Estructurales Virales/metabolismo
17.
Antiviral Res ; 92(3): 453-60, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22020302

RESUMEN

Dengue virus (DENV) infections continue to spread aggressively around the world. Here we demonstrate that celgosivir (6-O-butanoyl castanospermine), strongly inhibits all four DENV serotypes. We show by fluorescence microscopy that the antiviral mechanism of celgosivir, is in part, due to misfolding and accumulation of DENV non-structural protein 1 (NS1) in the endoplasmic reticulum. Moreover, celgosivir modulates the host's unfolded protein response (UPR) for its antiviral action. Significantly, celgosivir is effective in lethal challenge mouse models that recapitulate primary or secondary antibody-dependent enhanced DENV infection. Celgosivir treated mice showed enhanced survival, reduced viremia and robust immune response, as reflected by serum cytokine analysis. Importantly, survival increased even after treatment was delayed till 2 days post-infection. Together the present study suggests that celgosivir, which has been clinically determined to be safe in humans, may be a valuable candidate for clinical testing in dengue patients.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Dengue/tratamiento farmacológico , Indolizinas/farmacología , Proteínas no Estructurales Virales/metabolismo , Animales , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Línea Celular , Cricetinae , Dengue/genética , Modelos Animales de Enfermedad , Humanos , Indolizinas/administración & dosificación , Indolizinas/uso terapéutico , Ratones , Ratones de la Cepa 129 , Pruebas de Sensibilidad Microbiana , Unión Proteica , Pliegue de Proteína/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/genética , Proteínas no Estructurales Virales/química
18.
Drug Metab Dispos ; 36(8): 1659-69, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18505789

RESUMEN

The disposition and metabolism of paraherquamide (PHQ), a potent and broad-spectrum anthelminthic, were examined in sheep, dogs, and gerbils. The metabolism of PHQ in these species was extensive and marked by significant species differences both in vitro and in vivo. In sheep and gerbils, PHQ metabolism occurs mainly at the pyrrolidine moiety, generating several metabolites that, for the most part, retained nematodicidal activity in vitro. In dogs, the dioxepene group was also extensively metabolized, ultimately resulting in formation of a catechol and loss of pharmacological activity. After oral administration of [3H]PHQ to intact sheep, gerbils, and dogs, the majority of the administered radioactivity was recovered in feces. Intact PHQ accounted for 0% (dogs) to approximately 30% (sheep and gerbils) of drug-related material in feces. A detailed investigation of the composition of the intestinal content of sheep indicated that a significant amount of the dose was still present in the rumen 24 h after dose and that PHQ underwent significant dehydration in the cecum. The oral pharmacokinetic parameters of PHQ in sheep and dogs suggest that its absorption is rapid in both species but that its apparent elimination rate is significantly higher in the dog (t(1/2) approximately 1.5 h) than it is in sheep (t(1/2) approximately 8.5 h). The short elimination half-life and the absence of PHQ or other active components in the dog gastrointestinal tract provide a potential explanation of the lack of efficacy of PHQ in this species.


Asunto(s)
Indolizinas/farmacocinética , Compuestos de Espiro/farmacocinética , Administración Oral , Animales , Perros , Heces/química , Gerbillinae , Semivida , Indolizinas/administración & dosificación , Ovinos , Compuestos de Espiro/administración & dosificación , Tritio
19.
Br J Pharmacol ; 153(7): 1399-408, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18264128

RESUMEN

BACKGROUND AND PURPOSE: Secretory phospholipase A2 (sPLA2) is implicated in atherosclerosis, although the effects of specific sPLA2 inhibitors have not been studied. We investigated the effects of the indole analogue indoxam on low-density lipoprotein (LDL) modification by sPLA2 enzymes of different types and on the associated inflammatory responses in human umbilical vein endothelial cells (HUVEC). EXPERIMENTAL APPROACH: LDL modification was assessed by measuring the contents of two major molecular species of lysophosphatidylcholine (LPC) using electrospray ionization-liquid chromatography/mass spectrometry. The proinflammatory activity of the modified LDL was evaluated by determining monocyte chemoattractant protein-1 (MCP-1) mRNA expression and transcriptional factor nuclear factor-kappaB (NF-kappaB) activity in HUVEC. KEY RESULTS: Indoxam dose-dependently inhibited palmitoyl- and stearoyl-LPC production in LDL incubated with snake venom sPLA2 (IC50 1.2 microM for palmitoyl-LPC, 0.8 microM for stearoyl-LPC). MCP-1 mRNA expression and NF-kappaB activity were enhanced by venom sPLA2-treated LDL, which was completely suppressed by indoxam but not by thioetheramide-PC, a competitive sPLA2 inhibitor. Indoxam also suppressed LPC production in LDL treated with human synovial type IIA sPLA2. Tumour necrosis factor alpha (TNFalpha) increased type V sPLA2 expression in HUVEC. Indoxam dose-dependently suppressed LPC production in native and glycoxidized LDL treated with TNFalpha-stimulated HUVEC. Indoxam suppressed MCP-1 mRNA expression and NF-kappaB activity in TNFalpha-stimulated HUVEC incubated with native or glycoxidized LDL. CONCLUSIONS AND IMPLICATIONS: Indoxam prevented sPLA2-induced LPC production in native and glycoxidized LDL as well as LDL-induced inflammatory activity in HUVEC. Our results suggest that indoxam may be a potentially useful anti-atherogenic agent.


Asunto(s)
Carbamatos/farmacología , Inhibidores Enzimáticos/farmacología , Indolizinas/farmacología , Inhibidores de Fosfolipasa A2 , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Carbamatos/administración & dosificación , Células Cultivadas , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/metabolismo , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Indolizinas/administración & dosificación , Lipoproteínas LDL/efectos de los fármacos , Lipoproteínas LDL/metabolismo , Lisofosfatidilcolinas/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Venas Umbilicales/citología , Venas Umbilicales/metabolismo
20.
J Pharmacokinet Pharmacodyn ; 33(2): 167-93, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16547797

RESUMEN

Many physiological factors are regulated by homeostatic mechanisms to maintain normal body function. Empirical lower Rl (Model I and IV) or upper Rh limits (Model II and III) were included in current basic indirect response (IDR) models to account for the additional role of physiological limits (IDRPL). Various characteristics of these models were evaluated with simulations and explicit equations. The simulations reveal that the expanded models exhibit most properties of basic indirect response models, such as slow response initiation, lag time between the kinetic and dynamic peaks, a large dose plateau, and shift in Tmax with dose. The proposed models always produce lesser net responses than predicted by basic IDR models. Simulations demonstrate that addition of a parameter limit which is close to the baseline has a great influence on the overall and maximum responses and fitted model parameters. Only stimulatory IDRPL Models III and IV allow resolution of all model parameters in the absence of clear indications or predetermined values of the lower or upper limits. However, all four models are able to resolve model parameters when subgroups with different baselines are simultaneously fitted. These models create new interpretations of the determinants of baseline conditions which can be important in assessing inter-subject variability in responses. The applicability of IDRPL models is demonstrated using several examples from the published literature. Indirect response models with physiological limits will be useful in characterizing drug responses for turnover systems which are maintained within a certain range.


Asunto(s)
Homeostasis/efectos de los fármacos , Modelos Biológicos , Preparaciones Farmacéuticas/administración & dosificación , Inhibidores de 5-alfa-Reductasa , Adulto , Aldehído Reductasa/antagonistas & inhibidores , Algoritmos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Homeostasis/fisiología , Humanos , Indolizinas/administración & dosificación , Indolizinas/farmacocinética , Indolizinas/farmacología , Inyecciones Intravenosas , Masculino , Persona de Mediana Edad , Naftalenos/administración & dosificación , Naftalenos/farmacocinética , Naftalenos/farmacología , Preparaciones Farmacéuticas/metabolismo , Sorbitol/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA