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.
J Vet Med Sci ; 80(1): 152-155, 2018 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-29142147

RESUMEN

Two dogs presented to the emergency service after accidental ingestion of afloqualone tablets, a muscle relaxant used for back pain in humans. Toxic effects of the drug in these dogs included vomiting, respiratory depression, seizures, ataxia, bradycardia, and hematuria. Treatment consisted of fluid diuresis, furosemide, and propofol. Flumazenil, a gamma-amino butyric acid antagonist, was administered intravenously; however, it was not effective in stopping the seizures in these dogs. Both dogs recovered with supportive treatment. To the authors' knowledge, this is the first documented report of afloqualone intoxication in dogs.


Asunto(s)
Enfermedades de los Perros/inducido químicamente , Quinazolinas/envenenamiento , Animales , Ataxia/inducido químicamente , Ataxia/veterinaria , Bradicardia/inducido químicamente , Bradicardia/veterinaria , Perros , Flumazenil/uso terapéutico , Masculino , Quinazolinas/antagonistas & inhibidores , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/veterinaria , Convulsiones/inducido químicamente , Convulsiones/veterinaria , Vómitos/inducido químicamente , Vómitos/veterinaria
2.
Cell Death Dis ; 7: e2307, 2016 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-27441659

RESUMEN

Ferroptosis is an iron-dependent, oxidative cell death, and is distinct from apoptosis, necrosis and autophagy. In this study, we demonstrated that lysosome disrupting agent, siramesine and a tyrosine kinase inhibitor, lapatinib synergistically induced cell death and reactive oxygen species (ROS) in MDA MB 231, MCF-7, ZR-75 and SKBr3 breast cancer cells over a 24 h time course. Furthermore, the iron chelator deferoxamine (DFO) significantly reduced cytosolic ROS and cell death following treatment with siramesine and lapatinib. Furthermore, we determined that FeCl3 levels were elevated in cells treated with siramesine and lapatinib indicating an iron-dependent cell death, ferroptosis. To confirm this, we treated cells with a potent inhibitor of ferroptosis, ferrastatin-1 that effectively inhibited cell death following siramesine and lapatinib treatment. The increase levels of iron could be due to changes in iron transport. We found that the expression of transferrin, which is responsible for the transport of iron into cells, is increased following treatment with lapatinib alone or in combination with siramesine. Knocking down of transferrin resulted in decreased cell death and ROS after treatment. In addition, ferroportin-1 (FPN) is an iron transport protein, responsible for removal of iron from cells. We found its expression is decreased after treatment with siramesine alone or in combination with lapatinib. Overexpression FPN resulted in decreased ROS and cell death whereas knockdown of FPN increased cell death after siramesine and lapatinib treatment. This indicates a novel induction of ferroptosis through altered iron regulation by treating breast cancer cells with a lysosome disruptor and a tyrosine kinase inhibitor.


Asunto(s)
Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Indoles/farmacología , Hierro/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Especies Reactivas de Oxígeno/agonistas , Compuestos de Espiro/farmacología , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Ciclohexilaminas/farmacología , Deferoxamina/farmacología , Sinergismo Farmacológico , Femenino , Ferritinas/genética , Ferritinas/metabolismo , Regulación de la Expresión Génica , Humanos , Imidazoles/farmacología , Indoles/antagonistas & inhibidores , Quelantes del Hierro/farmacología , Lapatinib , Lisosomas/efectos de los fármacos , Células MCF-7 , Fenilendiaminas/farmacología , Quinazolinas/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo , Transducción de Señal , Compuestos de Espiro/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transferrina/genética , Transferrina/metabolismo
3.
Tumour Biol ; 37(5): 5811-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26581908

RESUMEN

Development of new therapeutic strategies is becoming increasingly important to overcome tamoxifen resistance. Recently, much interest has been focused on anti-tumor effects of metformin commonly used to treat type II diabetes. Increased protein expression and signaling of epidermal growth factor receptor (EGFR) family is a possible mechanism involved in tamoxifen resistance. Since HER2/HER3 heterodimers are able to induce strong downstream signaling and activate various biological responses such as cellular proliferation and growth, we investigated the anti-cancer effect of metformin by inhibition of signaling pathway via downregulation of HER2 and HER3 using tamoxifen-resistant MCF-7 (TR MCF-7) cells. Compared to MCF-7 cells, TR MCF-7 cells showed increased expression of EGFR, HER2, and HER3, and metformin inhibited the expression of these proteins in a dose- and time-dependent manner. Metformin inhibited activation of HER2 (Tyr1248)/HER3 (Tyr1289)/Akt (Ser473) as well as cell proliferation and colony formation by estrogenic promotion in MCF-7 and TR MCF-7 cells. Known as a HER3 ligand, heregulin (HRG)-ß1-induced phosphorylation of HER2, HER3 and Akt, and protein interaction of HER2/HER3 and colony formation were inhibited by metformin in both cells. Consistent with the results in the two cell lines, we identified that metformin inhibited HER2/HER3/Akt signaling axis activated by HRG-ß1 using the HER2 and HER3-overexpressing breast cancer cell line SK-BR-3. Lastly, lapatinib-induced HER3 upregulation was significantly inhibited by treatment of metformin in HER3 siRNA-transfected TR MCF-7 cells. These data suggest that metformin might overcome tamoxifen resistance through the inhibition of expression and signaling of receptor tyrosine kinase HER2 and HER3.


Asunto(s)
Adenocarcinoma/patología , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Metformina/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Tamoxifeno/farmacología , Antineoplásicos Hormonales/farmacología , Resistencia a Antineoplásicos/genética , Inducción Enzimática/efectos de los fármacos , Receptores ErbB/biosíntesis , Estradiol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes erbB-1 , Genes erbB-2 , Humanos , Lapatinib , Células MCF-7 , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neurregulina-1/antagonistas & inhibidores , Neurregulina-1/fisiología , Quinazolinas/antagonistas & inhibidores , Quinazolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor ErbB-2/biosíntesis , Receptor ErbB-3/biosíntesis , Receptor ErbB-3/genética , Ensayo de Tumor de Célula Madre
4.
J Appl Microbiol ; 118(4): 864-72, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25604161

RESUMEN

AIMS: The interaction of quinolone and indoloquinazoline alkaloids concerning their antimycobacterial activity was studied. METHODS AND RESULTS: The antimycobacterial and modulating activity of evodiamine (1), rutaecarpine (2) and evocarpine (3) was tested on mycobacteria including three multidrug-resistant (MDR) clinical isolates of Mycobacterium tuberculosis. Antagonistic effects were concluded from fractional inhibitory concentration (FICI) values. Interaction energies of the compounds were calculated using GLUE docking module implemented in GRID. 1 and 2 exhibited weak inhibition of rapidly growing mycobacteria, however, 1 was active against Myco. tuberculosis H37Rv (MIC = 10 mg l(-1) ) while 2 was inactive. Both 1 and 2 showed a marked antagonistic effect on the susceptibility of different mycobacterial strains to 3 giving FICI values between 5 and 9. The interaction energies between compounds 1 and 2 with compound 3 suggested the possibility of complex formation in solution. CONCLUSIONS: Indoloquinazoline alkaloids markedly reduce the antimycobacterial effect of the quinolone alkaloid evocarpine. Complex formation may play a role in the attenuation of its antimycobacterial activity. SIGNIFICANCE AND IMPACT OF THE STUDY: This study gives a striking example of antagonism between compounds present in the same plant extract which should be considered in natural product based screening projects.


Asunto(s)
Alcaloides/antagonistas & inhibidores , Antibacterianos/farmacología , Antagonismo de Drogas , Mycobacterium tuberculosis/efectos de los fármacos , Quinazolinas/antagonistas & inhibidores , Quinolonas/antagonistas & inhibidores , Humanos , Mycobacterium tuberculosis/fisiología , Extractos Vegetales/antagonistas & inhibidores , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología
5.
Antimicrob Agents Chemother ; 58(3): 1516-22, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24366746

RESUMEN

Drugs that target both the liver and blood stages of malaria will be needed to reduce the disease's substantial worldwide morbidity and mortality. Evaluation of a 259-member library of compounds that block proliferation of the blood stage of malaria revealed several scaffolds--dihydroquinazolinones, phenyldiazenylpyridines, piperazinyl methyl quinolones, and bis-benzimidazoles--with promising activity against the liver stage. Focused structure-activity studies on the dihydroquinazolinone scaffold revealed several molecules with excellent potency against both blood and liver stages. One promising early lead with dual activity is 2-(p-bromophenyl)-3-(2-(diethylamino)ethyl)-2,3-dihydroquinazolin-4(1H)-one with 50% effective concentrations (EC50s) of 0.46 µM and 0.34 µM against liver stage Plasmodium berghei ANKA and blood stage Plasmodium falciparum 3D7 parasites, respectively. Structure-activity relationships revealed that liver stage activity for this compound class requires a 3-dialkyl amino ethyl group and is abolished by substitution at the ortho-position of the phenyl moiety. These compounds have minimal toxicity to mammalian cells and are thus attractive compounds for further development.


Asunto(s)
Antimaláricos/farmacología , Hígado/parasitología , Plasmodium/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Estadios del Ciclo de Vida/efectos de los fármacos , Malaria/sangre , Malaria/tratamiento farmacológico , Malaria/parasitología , Plasmodium/crecimiento & desarrollo , Plasmodium berghei/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Quinazolinas/antagonistas & inhibidores , Relación Estructura-Actividad
6.
J Clin Endocrinol Metab ; 98(7): E1163-72, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23633194

RESUMEN

CONTEXT: Inhibition of dipeptidyl peptidase-4 (DPP-4) is a potent strategy to increase glucose-dependent insulinotropic polypeptide and glucagon like peptide 1 (GLP-1) induced insulin secretion in diabetes. It is important to know whether new drugs approved for the treatment of type 2 diabetes have direct effects on the ß-cell. OBJECTIVE: Herein we investigated the effect of linagliptin, a novel DPP-4 inhibitor, on ß-cell function and survival. DESIGN: Human islets were exposed to a diabetic milieu (11.1-33.3 mM glucose, 0.5 mM palmitate, the mixture of 2 ng/mL IL-1ß+1000 U/mL interferon-γ, or 50 µM H2O2) with or without 500 ng/mL IL-1 receptor antagonist (IL-1Ra) or 30-50 nM linagliptin. RESULTS: Linagliptin restored ß-cell function and turnover, which was impaired when islets were exposed to elevated glucose, palmitate, cytokines, or H2O2. Pretreatment with IL-1Ra was similarly effective, except against H2O2 treatment. Nitrotyrosine concentrations in islet lysates, an indicator of oxidative stress, were highly elevated under diabetic conditions but not in islets treated with linagliptin or IL-1Ra. Linagliptin also reduced cytokine secretion and stabilized GLP-1 in islet supernatants. CONCLUSIONS: We show that the novel DPP-4 inhibitor linagliptin protected from gluco-, lipo-, and cytokine-toxicity and stabilized active GLP-1 secreted from human islets. This provides a direct GLP-1 mediated protective effect of linagliptin on ß-cell function and survival.


Asunto(s)
Antioxidantes/farmacología , Supervivencia Celular/efectos de los fármacos , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Purinas/farmacología , Quinazolinas/farmacología , Antiinflamatorios no Esteroideos/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/farmacología , Antioxidantes/química , Citocinas/metabolismo , Dipeptidil Peptidasa 4/química , Inhibidores de la Dipeptidil-Peptidasa IV/química , Agonismo Inverso de Drogas , Péptido 1 Similar al Glucagón/agonistas , Péptido 1 Similar al Glucagón/química , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/genética , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Linagliptina , Ácido Palmítico/metabolismo , Fragmentos de Péptidos/farmacología , Estabilidad Proteica/efectos de los fármacos , Purinas/antagonistas & inhibidores , Pirazinas/farmacología , Quinazolinas/antagonistas & inhibidores , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Fosfato de Sitagliptina , Técnicas de Cultivo de Tejidos , Triazoles/farmacología
7.
J Clin Oncol ; 30(26): 3234-41, 2012 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-22649126

RESUMEN

PURPOSE: To determine whether the antibody-drug conjugate trastuzumab emtansine (T-DM1), which combines human epidermal growth factor receptor 2 (HER2) -targeted delivery of the potent antimicrotubule agent DM1 with the antitumor activity of trastuzumab, is effective in patients with HER2-positive metastatic breast cancer (MBC) who have previously received all standard HER2-directed therapies. PATIENTS AND METHODS: In this single-arm phase II study, T-DM1 3.6 mg/kg was administered intravenously every 3 weeks to patients with HER2-positive MBC who had prior treatment with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. The primary objectives were overall response rate (ORR) by independent review and safety. RESULTS: Among 110 pretreated patients (median, seven prior agents for MBC; median follow-up, 17.4 months), the ORR was 34.5% (95% CI, 26.1% to 43.9%), clinical benefit rate was 48.2% (95% CI, 38.8% to 57.9%), median progression-free survival (PFS) was 6.9 months (95% CI, 4.2 to 8.4 months), and median duration of response was 7.2 months (95% CI, 4.6 months to not estimable). In patients with confirmed HER2-positive tumors (n = 80 by retrospective central testing), the response rate was 41.3% (95% CI, 30.4% to 52.8%), and median PFS was 7.3 months (95% CI, 4.6 to 12.3 months). Most adverse events were grades 1 to 2; the most frequent grade ≥ 3 events were thrombocytopenia (9.1%), fatigue (4.5%), and cellulitis (3.6%). CONCLUSION: T-DM1 is well tolerated and has single-agent activity in patients with HER2-positive MBC who have previously received both approved HER2-directed therapies and multiple chemotherapy agents. T-DM1 may be an effective new treatment for this patient population.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Inmunotoxinas/uso terapéutico , Maitansina/análogos & derivados , Terapia Molecular Dirigida/métodos , Quinazolinas/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Ado-Trastuzumab Emtansina , Adulto , Anciano , Antraciclinas/administración & dosificación , Anticuerpos Monoclonales Humanizados/administración & dosificación , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Hidrocarburos Aromáticos con Puentes/administración & dosificación , Capecitabina , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Supervivencia sin Enfermedad , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/análogos & derivados , Humanos , Lapatinib , Masculino , Maitansina/uso terapéutico , Persona de Mediana Edad , Metástasis de la Neoplasia , Quinazolinas/administración & dosificación , Taxoides/administración & dosificación , Trastuzumab
8.
Lung Cancer ; 71(2): 241-3, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21168933

RESUMEN

The fusion gene EML4-ALK (echinoderm microtubule-associated protein-like 4 gene and the anaplastic lymphoma kinase gene) was recently identified as a novel genetic alteration in non-small cell lung cancer (NSCLC). EML4-ALK translocations correlate with specific clinical and pathological features, in particular lack of EGFR and K-ras mutations, and may be associated with resistance to EGFR tyrosine-kinase inhibitors (TKIs). Here, we report a case of a patient with a concomitant EGFR mutation and ALK translocation resistant to erlotinib. Considering this report, ALK status should be investigated in unexplained cases of EGFR-TKI-resistance of EGFR mutated NSCLCs.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Proteínas de Ciclo Celular/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Proteínas Asociadas a Microtúbulos/genética , Mutación/genética , Proteínas Tirosina Quinasas Receptoras/genética , Serina Endopeptidasas/genética , Quinasa de Linfoma Anaplásico , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/patología , Resistencia a Antineoplásicos/genética , Clorhidrato de Erlotinib , Genes ras/genética , Humanos , Hibridación Fluorescente in Situ , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Quinazolinas/antagonistas & inhibidores , Quinazolinas/uso terapéutico , Translocación Genética
10.
J Pharmacol Exp Ther ; 330(3): 792-801, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19491325

RESUMEN

There is a pressing need for immunosuppressants with an improved safety profile. The search for novel approaches to blocking T-cell activation led to the development of the selective protein kinase C (PKC) inhibitor AEB071 (sotrastaurin). In cell-free kinase assays AEB071 inhibited PKC, with K(i) values in the subnanomolar to low nanomolar range. Upon T-cell stimulation, AEB071 markedly inhibited in situ PKC catalytic activity and selectively affected both the canonical nuclear factor-kappaB and nuclear factor of activated T cells (but not activator protein-1) transactivation pathways. In primary human and mouse T cells, AEB071 treatment effectively abrogated at low nanomolar concentration markers of early T-cell activation, such as interleukin-2 secretion and CD25 expression. Accordingly, the CD3/CD28 antibody- and alloantigen-induced T-cell proliferation responses were potently inhibited by AEB071 in the absence of nonspecific antiproliferative effects. Unlike former PKC inhibitors, AEB071 did not enhance apoptosis of murine T-cell blasts in a model of activation-induced cell death. Furthermore, AEB071 markedly inhibited lymphocyte function-associated antigen-1-mediated T-cell adhesion at nanomolar concentrations. The mode of action of AEB071 is different from that of calcineurin inhibitors, and AEB071 and cyclosporine A seem to have complementary effects on T-cell signaling pathways.


Asunto(s)
Inmunosupresores/farmacología , Activación de Macrófagos/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/antagonistas & inhibidores , Quinazolinas/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Animales , Antígenos CD28/metabolismo , Calcio/metabolismo , Adhesión Celular/efectos de los fármacos , Citocinas/biosíntesis , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Genes Reporteros , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Células Jurkat , Ratones , Ratones Noqueados , FN-kappa B/efectos de los fármacos , Factores de Transcripción NFATC/metabolismo , Proteína Quinasa C/genética , Receptores de Antígenos de Linfocitos T/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
11.
Biochemistry ; 48(4): 688-98, 2009 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-19128036

RESUMEN

The diverse inhibitors of bovine heart mitochondrial complex I (NADH-ubiquinone oxidoreductase) are believed to share a common large binding domain with partially overlapping sites, though it remains unclear how these binding sites relate to each other. To obtain new insight into the inhibitor binding domain in complex I, we synthesized a photoreactive azidoquinazoline {[(125)I]-6-azido-4-(4-iodophenethylamino)quinazoline, [(125)I]AzQ}, in which a photolabile azido group was introduced into the toxophoric quinazoline ring to allow specific cross-linking, and carried out a photoaffinity labeling study using bovine heart submitochondrial particles. Analysis of the photo-cross-linked proteins by peptide mass fingerprinting and immunoblotting revealed that [(125)I]AzQ specifically binds to the 49 kDa and ND1 subunits with a frequency of approximately 4:1. The cross-linking was completely blocked by excess amounts of other inhibitors such as acetogenin and fenpyroximate. Considerable cross-linking was also detected in the ADP/ATP carrier and 3-hydroxybutyrate dehydrogenase, though it was not associated with dysfunction of the two proteins. The partial proteolysis of the [(125)I]AzQ-labeled 49 kDa subunit by V8-protease and N-terminal sequencing of the resulting peptides revealed that the amino acid residue cross-linked by [(125)I]AzQ is within the sequence region Thr25-Glu143 (118 amino acids). Furthermore, examination of fragment patterns generated by exhaustive digestion of the [(125)I]AzQ-labeled 49 kDa subunit by V8-protease, lysylendopeptidase, or trypsin strongly suggested that the cross-linked residue is located within the region Asp41-Arg63 (23 amino acids). The present study has revealed, for the first time, the inhibitor binding site in complex I at the sub-subunit level.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Mitocondrias Cardíacas/enzimología , Etiquetas de Fotoafinidad/metabolismo , Quinazolinas/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Sitios de Unión/fisiología , Bovinos , Cristalografía por Rayos X , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Mitocondrias Cardíacas/genética , Datos de Secuencia Molecular , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Quinazolinas/metabolismo
12.
Invest New Drugs ; 27(1): 1-11, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18493719

RESUMEN

Treatment with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) has led to dramatic clinical improvement in selected patients with non-small cell lung cancer (NSCLC). However, intrinsic and acquired resistance to EGFR-TKI remains a common phenomenon. Novel EGFR-TKI, structurally different with erlotinib or gefitinib might be beneficial for patients with NSCLC. In this study, we examined the antitumor effect of a newly synthesized novel EGFR tyrosine kinase inhibitor (Zhao260054). In vitro studies in a panel of four different human lung cancer cell lines revealed that Zhao260054 inhibited cell proliferation with high potency and induced G(0)/G(1) arrest of cell cycle and apoptosis. Zhao260054 markedly reduced phosphorylation of EGFR and inhibited activation of ERK1/2 and AKT. Oral administration of Zhao260054 (200 mg/kg/day) to BALB/c nude mice bearing SPC-A1 xenografts significantly retarded tumor growth. In conclusion, Zhao260054 has potent antitumor activity on human lung cancer in vitro and in vivo.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Compuestos de Anilina/farmacología , Antineoplásicos/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Quinazolinas/farmacología , Compuestos de Anilina/síntesis química , Compuestos de Anilina/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinazolinas/antagonistas & inhibidores , Quinazolinas/síntesis química , Quinazolinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Thorac Oncol ; 3(4): 430-40, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18379366

RESUMEN

After failure of first-line chemotherapy for advanced non-small cell lung cancer, many patients remain candidates to receive further antitumor treatment. To guide clinical management of these patients and to suggest priorities for clinical research, an International Panel of Experts met in Naples (Italy) in April 2007. Results and evidence-based conclusions are presented in this article. Single-agent chemotherapy with docetaxel or pemetrexed is the recommended option for unselected patients with performance status 0 to 2 who are candidates for second-line chemotherapy for advanced non-small cell lung cancer. Docetaxel has demonstrated superiority compared with best supportive care. Pemetrexed has been shown to be noninferior to docetaxel, with a more favorable toxicity profile. Erlotinib is effective in pretreated patients, and can be given second-line in patients not suitable or intolerant to chemotherapy, and in all patients as third-line treatment after failure of second-line chemotherapy. Gefitinib failed to show superiority to placebo as second- or third-line treatment, but it has been shown to be noninferior to docetaxel. In selected patients such as lifetime nonsmokers or those of East-Asian ethnicity, erlotinib, or gefitinib (where licensed) may be considered as second-line treatment even if they are fit for chemotherapy. Best supportive care in addition to active treatment remains important for all patients, but may be the exclusive option for patients unsuitable for more aggressive therapy. Further research is mandatory, to find better treatments, and to identify clinical and molecular predictive markers of efficacy, both for chemotherapy and for novel biologic agents.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Recuperativa , Carcinoma de Pulmón de Células no Pequeñas/secundario , Docetaxel , Clorhidrato de Erlotinib , Gefitinib , Glutamatos/administración & dosificación , Guanina/administración & dosificación , Guanina/análogos & derivados , Humanos , Neoplasias Pulmonares/patología , Pemetrexed , Guías de Práctica Clínica como Asunto , Pautas de la Práctica en Medicina , Pronóstico , Quinazolinas/administración & dosificación , Quinazolinas/antagonistas & inhibidores , Ensayos Clínicos Controlados Aleatorios como Asunto , Estudios Retrospectivos , Tasa de Supervivencia , Taxoides/administración & dosificación
14.
Semin Thromb Hemost ; 32(4 Pt 2): 430-6, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16810619

RESUMEN

Patients with essential thrombocythemia carry a high risk for thromboembolic and bleeding events but they have an almost normal life expectancy. A careful evaluation of the medical history and an exact diagnosis is mandatory to estimate each patient's risk for morbidity and to choose the most appropriate treatment measure. In patients with the need of cytoreductive therapy, the benefits of therapy have to outweigh the potential risks of drug toxicities. Hydroxyurea is the most useful cytoreductive drug for elderly patients; in younger persons, interferon alpha or anagrelide may be the drugs of choice. The combination of anagrelide with acetylsalicylic acid may be contraindicated in patients with a history of bleeding.


Asunto(s)
Trombocitemia Esencial/diagnóstico , Trombocitemia Esencial/tratamiento farmacológico , Factores de Edad , Antineoplásicos/antagonistas & inhibidores , Antineoplásicos/uso terapéutico , Aspirina/antagonistas & inhibidores , Aspirina/uso terapéutico , Antagonismo de Drogas , Femenino , Fibrinolíticos/uso terapéutico , Hemorragia/etiología , Humanos , Hidroxiurea/uso terapéutico , Interferón-alfa/uso terapéutico , Esperanza de Vida , Masculino , Quinazolinas/antagonistas & inhibidores , Quinazolinas/uso terapéutico , Factores de Riesgo , Trombocitemia Esencial/complicaciones
15.
Biochem Pharmacol ; 70(10): 1458-68, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16191427

RESUMEN

Thymidylate synthase (TS) is an important target of several chemotherapeutic agents. During TS inhibition, dTTP levels decrease with a subsequent increase in dUTP. Uracil incorporated into the genome is removed by base excision repair (BER). BER has been hypothesized to play a role in the response to thymidylate deprivation, despite a lack of direct evidence. We previously found that beta-pol null murine fibroblasts were approximately six-fold more resistant than wild-type cells to raltitrexed, a folate-based inhibitor specific for TS. In this study, a number of endpoints were determined to understand the influence of BER and beta-pol during raltitrexed treatment. Raltitrexed induced apoptosis in wild-type cells to a greater extent than in beta-pol null cells. A PARP inhibitor decreased the sensitivity to raltitrexed, although the extent was not different between wild-type and beta-pol null cells. No evidence was seen for extensive strand break formation that preceded apoptosis, although raltitrexed induced more sister chromatid exchanges in wild-type cells. Increased levels of uracil in DNA were detected following treatment in wild-type and beta-pol null cells. However, uracil levels were only approximately two-fold higher in DNA from treated cells compared to untreated. Uracil DNA glycosylase activity was slightly higher in beta-pol null cells, although not sufficiently different to explain the difference in sensitivity to raltitrexed. Taken together, the data suggest that the sensitivity of the wild-type cells to raltitrexed is not associated with activation of PARP-1 dependent BER, extensive uracil incorporation into DNA and persistent strand breaks, but rather with changes suggestive of DNA recombination.


Asunto(s)
Apoptosis/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Reparación del ADN/fisiología , Intercambio de Cromátides Hermanas/efectos de los fármacos , Timidilato Sintasa/antagonistas & inhibidores , Uracilo/metabolismo , Animales , Apoptosis/fisiología , Línea Celular , Células Cultivadas , ADN Polimerasa beta/antagonistas & inhibidores , ADN Polimerasa beta/farmacología , Reparación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/patología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Ratones , Poli Adenosina Difosfato Ribosa/antagonistas & inhibidores , Poli Adenosina Difosfato Ribosa/farmacología , Quinazolinas/antagonistas & inhibidores , Quinazolinas/toxicidad , Intercambio de Cromátides Hermanas/fisiología , Tiofenos/antagonistas & inhibidores , Tiofenos/toxicidad , Timidilato Sintasa/efectos de los fármacos , Factores de Tiempo , Uracilo/química , Uracilo/farmacología , Uracil-ADN Glicosidasa/química , Uracil-ADN Glicosidasa/efectos de los fármacos , Uracil-ADN Glicosidasa/metabolismo
16.
Eur J Med Chem ; 39(4): 299-304, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15072839

RESUMEN

In order to study the influence of 3-substitution on the cytotoxic activity of 2-styrylquinazolinones, new 6-chloro-2-styryl-3-(heteroaryl)-4(3H)-quinazolinones were synthesized by refluxing equimolar amounts of 6-chloro-2-methyl-3-(heteroaryl)-4(3H)-quinazolinones and benzaldehyde in glacial acetic acid. At 1 microg ml(-1) concentration, almost all 2-styrylquinazolinones showed some cytotoxic activity against the L1210 and K562 leukemia cell lines. However, only 6-chloro-2-styryl-3-(pyrimidin-2yl)-4(3H)-quinazolinone inhibited the growth of these cells by over 50%. This last compound was also the only member of the series that inhibited tubulin polymerization, with an IC(50) value of 5.8 versus 3.2 microM for colchicine. It was also examined for effects on the growth of human MCF7 breast carcinoma cells and Burkitt lymphoma CA46 cells, which had IC(50) values of 0.34 and 1.0 microM, respectively. At 10 microM 6-chloro-2-styryl-3-(pyrimidin-2yl)-4(3H)-quinazolinone induced G2/M arrest (66%) in Burkitt cells, with a mitotic index of 20%. At 3.4 microM, it caused disruption of the cellular microtubule system of the MCF7 cells. Both these cellular effects are consistent with its mechanism of action resulting from its inhibitory effect on tubulin assembly.


Asunto(s)
Antineoplásicos/síntesis química , Supervivencia Celular/efectos de los fármacos , Compuestos Heterocíclicos/química , Quinazolinas/síntesis química , Tubulina (Proteína)/química , Animales , Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Estructura Molecular , Polímeros/química , Polímeros/metabolismo , Quinazolinas/antagonistas & inhibidores , Quinazolinas/farmacología , Ratas , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina , Células Tumorales Cultivadas
17.
Pancreas ; 25(4): 387-92, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12409834

RESUMEN

INTRODUCTION AND AIMS: Adenosine shows protective effects against cellular damage and dysfunction under several adverse conditions such as inflammation and ischemia. In the current study, we examined the effects of 3-[1-(6,7-diethoxy-2-morpholinoquinazolin-4-yl)piperidin-4-yl]-1,6-dimethyl-2,4(1,3 )-quinazolinedione hydrochloride (KF24345), an adenosine uptake inhibitor, on cerulein-induced acute pancreatitis in mice to investigate whether inhibition of adenosine uptake could ameliorate the severity of acute pancreatitis. METHODOLOGY: Acute pancreatitis was induced in mice with six intraperitoneal injections of cerulein (50 microg/kg each) at hourly intervals. RESULTS: The cerulein injection increased activities of serum amylase and lipase and caused pathologic changes such as interstitial edema, polymorphonuclear cell infiltration, and acinar cell necrosis in the pancreas. KF24345 (10 mg/kg p.o.) ameliorated all these changes observed in mice with acute pancreatitis, and the suppressing effect of KF24345 on the elevation in serum amylase activity was abolished by the treatment with 8-(p-sulfophenyl)theophylline, an adenosine receptor antagonist. In addition, 2-(aminocarbonyl)- -(4-amino-2,6-dichlorophenyl)-4-[5,5-bis-(4-fluorophenyl)pentyl]-1-piperazineacetamide (R75231) and dipyridamole, other adenosine uptake inhibitors, also decreased the elevated serum amylase activity. CONCLUSIONS: These are the first demonstrations that the adenosine uptake inhibitors ameliorate cerulein-induced acute pancreatitis in mice, and these data suggest that adenosine uptake inhibition could ameliorate the severity of acute pancreatitis in vivo.


Asunto(s)
Adenosina/antagonistas & inhibidores , Pancreatitis/tratamiento farmacológico , Pirimidinonas/uso terapéutico , Quinazolinas/uso terapéutico , Teofilina/análogos & derivados , Enfermedad Aguda , Adenosina/metabolismo , Amilasas/sangre , Animales , Transporte Biológico/efectos de los fármacos , Ceruletida , Dipiridamol/farmacología , Femenino , Lipasa/sangre , Ratones , Ratones Endogámicos BALB C , Modelos Químicos , Pancreatitis/inducido químicamente , Pancreatitis/enzimología , Pancreatitis/patología , Piperazinas/farmacología , Pirimidinonas/antagonistas & inhibidores , Pirimidinonas/química , Quinazolinas/antagonistas & inhibidores , Quinazolinas/química , Teofilina/farmacología
18.
Semin Radiat Oncol ; 12(3 Suppl 2): 21-6, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12174341

RESUMEN

Approximately two thirds of all human solid tumors derive from epithelial tissues. The epidermal growth factor receptor (EGFR) serves as an important regulator of cellular growth in epithelial tumors. A spectrum of new anticancer agents have been specifically designed to target the EGFR in an effort to inhibit malignant growth. Although several of these new drugs show single-agent activity in early clinical trials, the predominant growth effect of EGFR signaling inhibition is cytostatic. However, the interaction of EGFR inhibition combined with conventional cytotoxic therapies (radiation and chemotherapy) is more potent, and shows great promise in the treatment of a variety of solid tumors that overexpress this receptor. This report focuses primarily on the capacity of EGFR inhibitors to modulate cellular and overall tumor response to ionizing radiation.


Asunto(s)
Receptores ErbB/biosíntesis , Receptores ErbB/efectos de la radiación , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Ensayos Clínicos como Asunto , Terapia Combinada , Receptores ErbB/efectos de los fármacos , Humanos , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/secundario , Neoplasias Glandulares y Epiteliales/terapia , Quinazolinas/antagonistas & inhibidores , Quinazolinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/uso terapéutico , Resultado del Tratamiento
19.
Jpn J Cancer Res ; 92(1): 74-82, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11173547

RESUMEN

The folate-dependent enzymes are attractive targets for cancer chemotherapy. Methotrexate (MTX), which inhibits dihydrofolate reductase, has been widely used for the treatment of solid tumors and hematological cancers. Raltitrexed ("Tomudex") ), which inhibits thymidylate synthase, is a novel anticancer agent active against colorectal cancer and some other solid tumors. We studied the optimal schedule of raltitrexed and MTX in combination against four human colon cancer cell lines Colo201, Colo320, LoVo, and WiDr. These cells were simultaneously exposed to raltitrexed and MTX for 24 h, or sequentially exposed to raltitrexed for 24 h followed by MTX for 24 h, or vice versa. Cell growth inhibition after 5 days was determined by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The effects of drug combinations at the concentrations of drug that produced 80% and 50% cell growth inhibition (IC(80) and IC(50)) were analyzed by the isobologram method (Steel and Peckham, 1979). Cytotoxic interactions between raltitrexed and MTX were schedule-dependent. The simultaneous exposure to raltitrexed and MTX showed additive effects in Colo201, LoVo and WiDr cells and antagonistic effects in Colo320 cells. The sequential exposure to raltitrexed followed by MTX produced additive effects in all four cell lines. The sequential exposure to MTX followed by raltitrexed produced synergistic effects in Colo201, LoVo and WiDr cells and additive effects in Colo320 cells. These findings suggest that the sequential administration of MTX followed by raltitrexed produces more than the expected cytotoxicity and may be the optimal schedule at the cellular level. Further in vivo and clinical studies will be necessary to determine the toxicity and to test the antitumor effects of sequential administration of MTX followed by raltitrexed proposed on the basis of the in vitro synergism.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias del Colon/tratamiento farmacológico , Metotrexato/farmacología , Quinazolinas/farmacología , Tiofenos/farmacología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Metotrexato/administración & dosificación , Metotrexato/antagonistas & inhibidores , Quinazolinas/administración & dosificación , Quinazolinas/antagonistas & inhibidores , Tiofenos/administración & dosificación , Tiofenos/antagonistas & inhibidores , Células Tumorales Cultivadas/efectos de los fármacos
20.
Med Parazitol (Mosk) ; (1): 26-9, 1998.
Artículo en Ruso | MEDLINE | ID: mdl-9608206

RESUMEN

The reversing action of anthelminthic praziquantel (P) on the effect of chloroquine (C) and compound R-70-Zh (styrylquinazoline) was revealed on a Plasmodium berghei model (white inbred mice), using a LNK65 isolate with naturally reduced sensitivity to chloroquine and its polyresistant line LNK65CHLFR with acquired resistance to chloroquine/fansidar (selected in our laboratory). P (125 mg/kg) in combination with C showed a potentiating effect not only on the LNK65 isolate, but also on the LNK65CHLFR line, while investigated separately on this line, both drugs were not effective in tested doses. Moreover, the similar effect of C on the LNK65CHLFR line was achieved in the dose that was 4 times higher than that of P/C combination. P in a standard dose on the LNK65 isolate showed a more marked activation of compound R-70-Zh that on C. The potentiating effect was manifested in combination with R-70-Zh in the dose half as high as that of C; this phenomenon was also reflected by the efficiency index (5.0 against the 4.0) accepted in our laboratory and may be associated with the higher sensitivity of the LNK65 isolate to R-70-Zh. P showed some antimalarial action which manifested itself only by morphological changes on P. berghei parasites similar to those observed under the action of some dihydropholate reductase inhibitors, such as pyrimethamine.


Asunto(s)
Antimaláricos/antagonistas & inhibidores , Antiplatelmínticos/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Cloroquina/antagonistas & inhibidores , Resistencia a Múltiples Medicamentos , Plasmodium berghei/efectos de los fármacos , Praziquantel/farmacología , Quinazolinas/antagonistas & inhibidores , Estirenos/antagonistas & inhibidores , Animales , Antimaláricos/uso terapéutico , Antiplatelmínticos/uso terapéutico , Bloqueadores de los Canales de Calcio/uso terapéutico , Cloroquina/uso terapéutico , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Malaria/tratamiento farmacológico , Malaria/parasitología , Ratones , Plasmodium berghei/aislamiento & purificación , Praziquantel/uso terapéutico , Pirimetamina/antagonistas & inhibidores , Quinazolinas/uso terapéutico , Estirenos/uso terapéutico , Sulfadoxina/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA