Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Methods Enzymol ; 586: 247-274, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28137566

RESUMEN

Colorectal cancer (CRC) is the third leading cause of cancer mortality for both men and women, and the second leading cause of cancer death for men and women combined. If detected early, before metastasis has occurred, survival following surgical resection of the tumor is >90%. Early detection is therefore critical for effective disease surveillance. Unfortunately, current biomarker assays lack the necessary sensitivity and specificity for reliable early disease detection. Development of new robust, non- or minimally invasive specific and sensitive biomarkers or panels with improved compliance and performance is therefore urgently required. The use of fecal samples offers several advantages over other clinical biospecimens (e.g., plasma or serum) as a source of CRC biomarkers, including: collection is noninvasive, the test can be performed at home, one is not sample limited, and the stool effectively samples the entire length of the inner bowel wall contents (including tumor) as it passes down the gastrointestinal tract. Recent advances in mass spectrometry now facilitate both the targeted discovery and validation of potential CRC biomarkers. We describe, herein, detailed protocols that can be used to mine deeply into the fecal proteome to reveal candidate proteins, identify proteotypic/unitypic peptides (i.e., peptides found in only a single known human protein that serve to identify that protein) suitable for sensitive and specific quantitative multiplexed analysis, and undertake high-throughput analysis of clinical samples. Finally, we discuss future directions that may further position this technology to support the current switch in translation research toward personalized medicine.


Asunto(s)
Biomarcadores de Tumor/aislamiento & purificación , Neoplasias Colorrectales/diagnóstico , Proteoma/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Biomarcadores de Tumor/química , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Cromatografía de Fase Inversa , Detección Precoz del Cáncer , Heces , Humanos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/aislamiento & purificación , Proteoma/química , Proteómica/métodos , Sensibilidad y Especificidad , Análisis de Secuencia de Proteína , Espectrometría de Masas en Tándem
2.
Cell Death Differ ; 23(4): 616-27, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26794444

RESUMEN

Interleukin-6 (IL-6) has been demonstrated to be involved in Hepatitis B virus (HBV)-associated hepatocarcinogenesis through activation of the STAT3 pathway. The sustained activation of the IL-6/STAT3 pathway is frequently associated with repression of SOCS3, which is both a target gene and a negative regulator of STAT3. However, the silencing mechanism of SOCS3 in hepatocellular carcinoma (HCC) remains to be elucidated. Here, we showed that the repression of SOCS3 and sustained activation of IL-6/STAT3 pathway in HBV-producing HCC cells were caused by HBV-induced mitochondrial ROS accumulation. Mechanistic studies revealed that ROS-mediated DNA methylation resulted in the silencing of SOCS3. Decreased SOCS3 expression significantly promoted the proliferation of HCC cells and growth of tumor xenografts in mice. Further studies revealed that HBV-induced ROS accumulation upregulated the expression of the transcription factor, Snail, which bound to the E-boxes of SOCS3 promoter and mediated the epigenetic silencing of SOCS3 in association with DNMT1 and HDAC1. In addition, we found that the expression of Snail and SOCS3 were inversely correlated in HBV-associated HCC patients, suggesting that SOCS3 and/or Snail could be used as prognostic markers in HCC pathogenesis. Taken together, our data show that HBV-induced mitochondrial ROS production represses SOCS3 expression through Snail-mediated epigenetic silencing, leading to the sustained activation of IL-6/STAT3 pathway and ultimately contributing to hepatocarcinogenesis.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Transformación Celular Viral , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Virus de la Hepatitis B/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Células Hep G2 , Virus de la Hepatitis B/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/genética , Factores de Transcripción de la Familia Snail/genética , Proteína 3 Supresora de la Señalización de Citocinas/genética
3.
Cancer Metastasis Rev ; 34(4): 715-34, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26337740

RESUMEN

The ß6 subunit of the αvß6 integrin heterodimer has long been an enigma in cancer biology though recent research has provided many new insights into its biology. Collectively, these findings include discovery of the transcriptional, translational and cell biological mechanisms by which ß6 acts, the identification of the cellular influences ß6 exerts upon the cell proteome, the characterisation of multiple ß6-centric pro-metastatic signalling systems and the search for pharmacological therapies (industry and academia) targeted against ß6. Once expressional restriction is overcome in early colorectal cancer (CRC), epithelial cell surface restricted αvß6 can physically interact with, and activate, known oncoproteins, and has the potential to enable the cross-talk through non-canonical signal transduction pathways, resulting in the adoption of an invasive/metastatic phenotype. This recent research has identified numerous interconnections and potential feedback loops, highlighting the fact that the expression of the ß6 subunit may initiate a cascade of downstream effects on the CRC cell rather than acting through a single mechanism. We here review these recent studies and postulate that the existence of a cell surface uPAR/αvß6/TGFß "metastasome" interactome in/on a proportion of colorectal cancer cells, where ß6 expression sequesters and activates multiple systems at the invasive front of tumour lesions, promoting cancer metastasis and hence explaining why ß6 has been correlated with reduced patient survival in CRC.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Neoplasias Colorrectales/patología , Integrinas/metabolismo , Metástasis de la Neoplasia/patología , Movimiento Celular , Quimiocina CXCL12/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Humanos , Regiones Promotoras Genéticas/genética , Factor de Crecimiento Transformador beta/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
4.
Colorectal Dis ; 15(7): e373-81, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23489866

RESUMEN

AIM: Annexin A2 (ANXA2) is known to be a tumourigenic molecule and is highly expressed in colorectal cancer (CRC). Its diagnostic and prognostic value is not fully understood. This study was designed to investigate the relationship between ANXA2 expression, clinicopathological characteristics, tumour recurrence and survival. METHOD: Immunohistochemical staining was used to evaluate ANXA2 expression in 150 matched samples from patients with CRC. Overall survival and recurrence were determined by Kaplan-Meier analysis. The Cox proportional hazards model was used to determine independent factors contributing to survival and recurrence. Receiver operating characteristic (ROC) curve and liner correlation analysis were used to estimate the sensitivity and specificity of ANXA2 expression for clinical diagnosis. RESULTS: ANXA2 was found to be strongly expressed in poorly differentiated tumours (P < 0.001), late stage (P = 0.020) and lymph node positivity (P = 0.002). ANXA2 expression was significantly related to recurrence (P < 0.001) and survival (P = 0.002). The Cox proportional hazards model indicated that ANXA2 expression [P < 0.001, hazard ratio (HR) = 1.366, 95% CI 1.232-1.515] and tumour location (P = 0.039, HR = 1.891, 95% CI 1.034-3.456) were independent factors in predicting overall survival while ANXA2 expression (P < 0.001, HR = 1.445, 95% CI 1.222-1.709) were independent factors predicting recurrence. Receiver operating characteristic (ROC) (AUC = 0.768, 95% CI = 0.642-0.894) and liner correlation analysis suggested that ANXA2 was suitable for the clinical diagnosis of CRC. CONCLUSION: These results indicate that ANXA2 is a biomarker with diagnostic and prognostic potential for patients with CRC.


Asunto(s)
Anexina A2/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/mortalidad , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Modelos Lineales , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Curva ROC , Sensibilidad y Especificidad
5.
Biochem Pharmacol ; 71(10): 1422-34, 2006 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-16522318

RESUMEN

The tyrphostin 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478) is a potent and specific inhibitor of EGFR tyrosine kinase whose favourable preclinical profile supports progression towards clinical trials. Microphysiometric evaluation revealed a short (<24 min) effective inhibition of cellular receptor response to EGF challenge in BaF/ERX cells indicating a need to maintain sustained levels of inhibitor. Initial pharmacokinetic evaluation in mice of novel AG1478 formulations in a beta-cyclodextrin (Captisol) showed monoexponential elimination from plasma (half-life 30 min) following subcutaneous administration. A two-fold dose escalation gave a 2.4-fold increase in the total AUC. Bolus i.v. and 6 h continuous infusion were investigated in rats to mimic a more clinically relevant administration regimen. Drug elimination following bolus i.v. administration was biphasic (terminal elimination half-life 30-48 min). The linear relationship between dose and AUC(0-->infinity) (r2=0.979) enabled the prediction of infusion rates and doses for sustained delivery using continuous 6 h infusions, where steady state was reached in 120 min. Plasma levels of AG1478>10 microM were achieved over the duration of the infusion. At the lowest dose, plasma drug levels after the cessation of infusion declined with a half-life of approximately 43 min. EGFR activity, measured both by autophosphorylation and downstream signalling, was inhibited in a dose-dependent manner by injection of AG1478 in mice bearing xenografts of the human glioblastoma cell line U87MG.delta2-7, which expresses a constitutively active variant of the EGF receptor. Taken together, these experiments provide essential data to assess the anti-tumour efficacy of AG1478 and will assist in the rational design of dose regimens for clinical studies.


Asunto(s)
Inhibidores Enzimáticos/farmacocinética , Receptores ErbB/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Tirfostinos/farmacocinética , Animales , Área Bajo la Curva , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Inyecciones Intravenosas , Inyecciones Subcutáneas , Ratones , Estructura Molecular , Quinazolinas , Ratas , Timidina/metabolismo , Tirfostinos/química , Tirfostinos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Microsc ; 217(Pt 3): 265-74, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15725130

RESUMEN

Current optical methods to collect Nomarski differential interference contrast (DIC) or phase images with a transmitted light detector (TLD) in conjunction with confocal laser scanning microscopy (CLSM) can be technically challenging and inefficient. We describe for the first time a simple method that combines the use of the commercial product QPm (Iatia, Melbourne Australia) with brightfield images collected with the TLD of a CLSM, generating DIC, phase, Zernike phase, dark-field or Hoffman modulation contrast images. The brightfield images may be collected at the same time as the confocal images. This method also allows the calculation of contrast-enhanced images from archival data. The technique described here allows for the creation of contrast-enhanced images such as DIC or phase, without compromising the intensity or quality of confocal images collected simultaneously. Provided the confocal microscope is equipped with a motorized z-drive and a TLD, no hardware or optical modifications are required. The contrast-enhanced images are calculated with software using the quantitative phase-amplitude microscopy technique (Barone-Nugent et al., 2002). This technique, being far simpler during image collection, allows the microscopist to concentrate on their confocal imaging and experimental procedures. Unlike conventional DIC, this technique may be used to calculate DIC images when cells are imaged through plastic, and without the use of expensive strain-free objective lenses.


Asunto(s)
Microscopía Confocal/métodos , Microscopía de Interferencia/métodos , Animales , Línea Celular Tumoral , Embrión no Mamífero/anatomía & histología , Fibroblastos , Humanos , Leishmania mexicana , Mastocitos , Ratones , Células 3T3 NIH , Ratas , Pez Cebra/embriología
7.
Growth Factors ; 20(2): 71-80, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12148565

RESUMEN

Epidermal Growth Factor (EGF) is a small growth factor containing 53 amino acid residues capable of stimulating the proliferation of both mesenchymal and epithelial cells. Comparison of the amino acid sequences of EGF from several species, and related proteins that can bind to the EGF receptor (e.g. TGFalpha, VGF, heparin-binding EGF, and betacellulin), suggests that Leu47, which is highly conserved, is important for biological function. Additionally, we have shown previously, using a combination of trypsin and carboxypeptidase Y digestion of native murine EGF, that removal of Leu47 results in more than 100-fold decrease in both receptor binding and mitogenic activity. We now describe a micromethod for the rapid generation of C-terminally modified EGFs to investigate further the role of C-terminal residues in determining functional activity. These analogues have been generated by digesting native murine EGF with trypsin, purifying the biologically inactive, but structurally intact, EGF1-45 core by micropreparative RP-HPLC, and then reversing the action of trypsin to couple synthetic peptides (e.g. DL, DI, DF, EL, DLLW) onto the C-terminus of the EGF1-45 core. This enzymic semisynthesis method allows multiple derivatives to be generated rapidly from microgram quantities of EGF1-45 in sufficient quantities for sensitive biological and physicochemical analysis. We have validated the method by regenerating EGF1-47 from EGF1-45 with equivalent mitogenic and receptor binding activity to EGF1-47 generated from wild type EGF by digestion with trypsin and carboxypeptidase Y. We have also investigated the effect of substituting alternative normal or nonphysiological amino acids (e.g. allo-Ile) for Asp46, Leu47 or Arg48. Even small changes in these C-terminal residues reduce the mitogenic potency of the analogue.


Asunto(s)
Factor de Crecimiento Epidérmico/análogos & derivados , Factor de Crecimiento Epidérmico/síntesis química , Células 3T3 , Secuencia de Aminoácidos , Aminoácidos/química , Animales , Carboxipeptidasas/farmacología , Catepsina A , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Humanos , Cinética , Leucina/química , Ligandos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Péptidos/química , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Relación Estructura-Actividad , Factores de Tiempo , Tripsina/farmacología
8.
EMBO J ; 20(17): 4762-73, 2001 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-11532940

RESUMEN

Vascular endothelial growth factor receptor-3 (VEGFR-3/Flt4) binds two known members of the VEGF ligand family, VEGF-C and VEGF-D, and has a critical function in the remodelling of the primary capillary vasculature of midgestation embryos. Later during development, VEGFR-3 regulates the growth and maintenance of the lymphatic vessels. In the present study, we have isolated and cultured stable lineages of blood vascular and lymphatic endothelial cells from human primary microvascular endothelium by using antibodies against the extracellular domain of VEGFR-3. We show that VEGFR-3 stimulation alone protects the lymphatic endothelial cells from serum deprivation-induced apoptosis and induces their growth and migration. At least some of these signals are transduced via a protein kinase C-dependent activation of the p42/p44 MAPK signalling cascade and via a wortmannin-sensitive induction of Akt phosphorylation. These results define the critical role of VEGF-C/VEGFR-3 signalling in the growth and survival of lymphatic endothelial cells. The culture of isolated lymphatic endothelial cells should now allow further studies of the molecular properties of these cells.


Asunto(s)
Apoptosis/fisiología , Factores de Crecimiento Endotelial/farmacología , Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/fisiología , Endotelio/fisiología , Sistema Linfático/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento/fisiología , Apoptosis/efectos de los fármacos , Técnicas Biosensibles , Capilares/embriología , Capilares/fisiología , División Celular , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Endotelio/citología , Endotelio Vascular/citología , Activación Enzimática , Humanos , Cinética , Sistema Linfático/citología , Microcirculación/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Quinasa C/metabolismo , ARN Mensajero/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento Endotelial Vascular , Piel/irrigación sanguínea , Venas Umbilicales , Factor C de Crecimiento Endotelial Vascular , Factor D de Crecimiento Endotelial Vascular , Receptor 3 de Factores de Crecimiento Endotelial Vascular
9.
Biochemistry ; 40(30): 8930-9, 2001 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-11467954

RESUMEN

Murine and human epidermal growth factor receptors (EGFRs) bind human EGF (hEGF), mouse EGF (mEGF), and human transforming growth factor alpha (hTGF-alpha) with high affinity despite the significant differences in the amino acid sequences of the ligands and the receptors. In contrast, the chicken EGFR can discriminate between mEGF (and hEGF) and hTGF-alpha and binds the EGFs with approximately 100-fold lower affinity. The regions responsible for this poor binding are known to be Arg(45) in hEGF and the L2 domain in the chicken EGFR. In this study we have produced a truncated form of the hEGFR ectodomain comprising residues 1-501 (sEGFR501), which, unlike the full-length hEGFR ectodomain (residues 1-621, sEGFR621), binds hEGF and hTGF-alpha with high affinity (K(D) = 13-21 and 35-40 nM, respectively). sEGFR501 was a competitive inhibitor of EGF-stimulated mitogenesis, being almost 10-fold more effective than the full-length EGFR ectodomain and three times more potent than the neutralizing anti-EGFR monoclonal antibody Mab528. Analytical ultracentrifugation showed that the primary EGF binding sites on sEGFR501 were saturated at an equimolar ratio of ligand and receptor, leading to the formation of a 2:2 EGF:sEGFR501 dimer complex. We have used sEGFR501 to generate three mutants with single position substitutions at Glu(367), Gly(441), or Glu(472) to Lys, the residue found in the corresponding positions in the chicken EGFR. All three mutants bound hTGF-alpha and were recognized by Mab528. However, mutant Gly(441)Lys showed markedly reduced binding to hEGF, implicating Gly(441), in the L2 domain, as part of the binding site that recognizes Arg(45) of hEGF.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Eliminación de Secuencia , Animales , Unión Competitiva/genética , Técnicas Biosensibles , Células CHO , Línea Celular , Pollos , Cricetinae , Dimerización , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Receptores ErbB/biosíntesis , Receptores ErbB/aislamiento & purificación , Inhibidores de Crecimiento/genética , Inhibidores de Crecimiento/metabolismo , Humanos , Ligandos , Ratones , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/aislamiento & purificación , Plásmidos/biosíntesis , Plásmidos/metabolismo , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Transfección , Factor de Crecimiento Transformador alfa/metabolismo
10.
Cancer Res ; 61(11): 4474-82, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11389078

RESUMEN

The chimeric monoclonal antibody KM871, directed against the G(D3) antigen, is under evaluation for its potential to target melanoma. To facilitate the in vivo evaluation of biodistribution properties and measurement of pharmacokinetics, KM871 was radiolabeled with (125)I via tyrosine residues and with (111)In via the bifunctional metal ion chelator C-functionalized trans-cyclohexyl diethylenetriaminepentaacetic acid (CHX-A"-DTPA) to lysine residues. Using antigen-positive SK-MEL-28 melanoma cells, immunoreactivities of 42 and 40% cell binding were obtained, respectively, for the two radioconjugates. Binding was enhanced in the presence of added unlabeled antibody. A humanized A33 antibody was similarly labeled with the two isotopes and used as a control. To determine and compare in vivo biodistribution characteristics of KM871 radiolabeled with (111)In or (125)I, mixtures of the radioconjugates were injected i.v. into BALB/c nude mice bearing G(D3)-positive-SK-MEL-28 melanoma xenografts. Gamma camera images were acquired; groups of five mice were sacrificed at various time intervals, and tumors, blood, and tissues were analyzed. (111)In-labeled CHX-A"-DTPA-KM871 showed a maximum tumor uptake of 41.9 +/- 7.0% injected dose/g at 72 h with prolonged retention over a 15-day period. The tumor:blood ratio was 3:1 by 72 h, and higher ratios were observed at later time points. No abnormal accumulation of (111)In-labeled conjugate was found in normal tissues. In contrast, there was little accumulation of (125)I-labeled KM871 in the same tumors. The specificity of antibody localization was confirmed by the low tumor uptake values for radiolabeled control antibody. Gamma camera imaging demonstrated excellent uptake of (111)In-labeled CHX-A"-DTPA-KM871 in the xenografts. Chromatographic analyses of xenograft cytosolic extracts demonstrated tumor internalization and catabolism of radiolabeled KM871 with the formation of small molecular weight metabolites. Laser scanning confocal microscopy demonstrated that the majority of intracellular KM871 is localized to lysosomes. Despite the catabolism of the radioconjugate, a dose-dependent increase in KM871 tumor localization was shown through immunohistochemical examination of xenograft biopsies. This study demonstrates for the first time the in vivo localization of a radiolabeled anti-G(D3) monoclonal antibody to G(D3)-expressing xenografts using gamma camera scanning techniques and tumor cell internalization of KM871 tagged with a green fluorescent dye, Alexa Fluor 488, through confocal microscopy. KM871 has potential for targeting tumors in patients with melanoma.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Gangliósidos/inmunología , Inmunoconjugados/farmacocinética , Melanoma/diagnóstico por imagen , Melanoma/metabolismo , Ácido Pentético/análogos & derivados , Radiofármacos/farmacocinética , Animales , Anticuerpos Monoclonales/inmunología , Quelantes/química , Quelantes/farmacocinética , Femenino , Cámaras gamma , Gangliósidos/biosíntesis , Humanos , Inmunohistoquímica , Radioisótopos de Indio/química , Radioisótopos de Yodo/química , Isotiocianatos/química , Isotiocianatos/farmacocinética , Marcaje Isotópico , Melanoma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ácido Pentético/química , Ácido Pentético/farmacocinética , Cintigrafía , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Immunol Immunother ; 50(2): 102-8, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11401024

RESUMEN

Bispecific antibodies are currently being used in clinical trials in increasing numbers in the areas of breast cancer, prostate cancer, non-Hodgkin's lymphoma and Hodgkin's lymphoma. We have previously performed two clinical trials in patients with Hodgkin's disease with an anti-CD30/anti-CD16 bispecific antibody and demonstrated a 30% response rate in a cohort of patients otherwise resistant to standard therapeutic modalities. However, no surrogate marker could be defined in these trials indicative of optimal antibody dosing/scheduling or predictive for favorable response. In order to evaluate accurately the potential biodistribution properties of bispecific antibody in patients, we have performed a detailed analysis of the binding properties and animal model in vivo characteristics of these constructs. For this purpose, the parental antibodies (anti-CD30 and anti-CD16) and the bispecific antibody (anti-CD30/anti-CD16) were radiolabeled with either 125I or 111In. Antibody integrity and binding properties after labeling were confirmed by Scatchard plot and Lindmo analysis. 111In-labeled antibodies revealed superior targeting properties in a standard SCID mouse tumor model. Both the bivalent parental anti-CD30 monoclonal antibody and the monovalent anti-CD30/anti-CD16 bispecific antibody showed excellent uptake in CD30+ tumors which did not differ significantly between the two (maximum uptake 16.5%+/-4.2% vs. 18.4%+/-3.8% injected dose/gram tissue). The equivalent targeting properties of the bispecific antibody compared with the parental anti-CD30 antibody encourages the further clinical development of this bispecific antibody, and might help to explain the clinical responses seen with this antibody so far in patients suffering from Hodgkin's disease.


Asunto(s)
Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Monoclonales/farmacocinética , Antígeno Ki-1/inmunología , Neoplasias Experimentales/terapia , Receptores de IgG/inmunología , Animales , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Femenino , Humanos , Radioisótopos de Indio , Radioisótopos de Yodo , Ratones , Ratones SCID , Neoplasias Experimentales/metabolismo , Distribución Tisular
12.
Exp Cell Res ; 266(1): 1-10, 2001 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11339819

RESUMEN

Signals from the epidermal growth factor (EGF) receptor and integrin-dependent adhesion to laminin contribute to the progression and metastasis of colonic tumors. However, little is know about the mechanisms by which these signals cooperate. Recently, we have reported that the colon cancer cell line LIM1215 secretes and adhere to autocrine laminin-10 via multiple integrin receptors and that EGF stimulates spreading of these cells on the same substrate. In this report, we investigate the effect of EGF and laminin-10 on colon cancer cell migration in vitro. EGF stimulates migration of LIM1215 cells in a wound healing assay. The response to EGF is inhibited by anti-EGF receptor antibody 528, the EGF receptor kinase inhibitor AG-1478, or the MAP kinase kinase inhibitor PD98059 but not the PI3-K inhibitor wortmannin. Using Transwell migration chambers, we demonstrate that laminin-10 but not collagen-I, collagen-IV, or a commercial preparation of human placental laminin is a potent motility factor for LIM1215 cells. The migration response to laminin-10 is increased upon stimulation of the cells with EGF and correlates with the up-regulation of alpha(6)beta(4) integrin expression as measured by analysis of Triton X-100-soluble cellular extracts. The results from integrin inhibition experiments indicate that basal migration on laminin-10 is mediated by alpha(3)beta(1) but not alpha(2)beta(1) nor alpha(6)beta(4) integrins. Alpha(3) blocking antibodies also inhibited EGF-stimulated chemokinetic migration of LIM1215 cells on laminin-10. However, in contrast to unstimulated cells, alpha(6) or beta(4) integrin-blocking antibodies inhibited the migration of EGF-stimulated cells by up to 50%. Taken together, these results support the cooperative role of EGF receptor and laminin-10 on colon cancer cell motility and suggest a critical role for both the alpha(3)beta(1) and the alpha(6)beta(4) integrins in this process.


Asunto(s)
Antígenos de Superficie/metabolismo , Carcinoma/metabolismo , Movimiento Celular/fisiología , Neoplasias del Colon/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Integrinas/metabolismo , Laminina/metabolismo , Laminina/farmacología , Metástasis de la Neoplasia/fisiopatología , Células Tumorales Cultivadas/metabolismo , Actinas/efectos de los fármacos , Actinas/metabolismo , Antígenos de Superficie/efectos de los fármacos , Bioensayo , Carcinoma/fisiopatología , Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/fisiopatología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Humanos , Integrina alfa3beta1 , Integrina alfa6beta4 , Integrinas/efectos de los fármacos , Factores de Tiempo , Células Tumorales Cultivadas/efectos de los fármacos
13.
J Chromatogr B Biomed Sci Appl ; 754(1): 193-9, 2001 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11318415

RESUMEN

The tyrphostin 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478) is undergoing evaluation as a potential new anticancer agent. We have developed a specific and sensitive reversed-phase HPLC assay for AG1478 in mouse plasma. The method involves a rapid and simple extraction process followed by separation on a Symmetry C8 stationary phase with a gradient of acetonitrile in ammonium acetate buffer. A linear response was achieved over the concentration range of 0.2-100 microM using multilevel calibration with internal standard method of calculation. Inter- and intra-assay accuracy and precision were better than +/-10%. The limit of quantitation was 0.2 microM. We have used this method to study the preclinical pharmacokinetics of this new agent in mice.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Receptores ErbB/antagonistas & inhibidores , Tirfostinos/sangre , Animales , Inhibidores Enzimáticos/sangre , Estabilidad de Enzimas , Congelación , Ratones , Quinazolinas , Estándares de Referencia , Sensibilidad y Especificidad , Tirfostinos/farmacocinética
14.
J Biol Chem ; 276(22): 19166-71, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11279005

RESUMEN

Human vascular endothelial growth factor-D (VEGF-D) binds and activates VEGFR-2 and VEGFR-3, receptors expressed on vascular and lymphatic endothelial cells. As VEGFR-2 signals for angiogenesis and VEGFR-3 is thought to signal for lymphangiogenesis, it was proposed that VEGF-D stimulates growth of blood vessels and lymphatic vessels into regions of embryos and tumors. Here we report the unexpected finding that mouse VEGF-D fails to bind mouse VEGFR-2 but binds and cross-links VEGFR-3 as demonstrated by biosensor analysis with immobilized receptor domains and bioassays of VEGFR-2 and VEGFR-3 cross-linking. Mutation of amino acids in mouse VEGF-D to those in the human homologue indicated that residues important for the VEGFR-2 interaction are clustered at, or are near, the predicted receptor-binding surface. Coordinated expression of VEGF-D and VEGFR-3 in mouse embryos was detected in the developing skin where the VEGF-D gene was expressed in a layer of cells beneath the developing epidermis and VEGFR-3 was localized on a network of vessels immediately beneath the VEGF-D-positive cells. This suggests that VEGF-D and VEGFR-3 may play a role in establishing vessels of the skin by a paracrine mechanism. Our study of receptor specificity suggests that VEGF-D may have different biological functions in mouse and man.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Bioensayo , Técnicas Biosensibles , Western Blotting , Reactivos de Enlaces Cruzados/farmacología , Electroforesis en Gel de Poliacrilamida , Embrión de Mamíferos/metabolismo , Factores de Crecimiento Endotelial/biosíntesis , Endotelio Vascular/metabolismo , Epidermis/metabolismo , Humanos , Inmunohistoquímica , Hibridación in Situ , Cinética , Ligandos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Homología de Secuencia de Aminoácido , Piel/embriología , Piel/metabolismo , Factores de Tiempo , Factor D de Crecimiento Endotelial Vascular , Receptor 3 de Factores de Crecimiento Endotelial Vascular
15.
J Pept Res ; 58(6): 493-503, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12005419

RESUMEN

The coiled coil is a common structural motif found both as the dominant structure in fibrous proteins and as an oligomerization domain in a variety of cytoskeletal and extracellular matrix proteins. Coiled-coils typically consist of two to four helices that are supercoiled around one another in either parallel or antiparallel orientations. In the past few years our knowledge of the structure and specificity of coiled coil interactions has increased, allowing the de novo design and preparation of coiled-coils with well-defined structure and specificity. Indeed, the design and synthesis of a peptide that binds specifically to a single coiled-coil-containing protein, adenomatous polyposis coli (APC) has been reported. We have optimized solid-phase synthesis techniques to produce a modified form of the anti-APC peptide that contains a biotin moiety specifically placed so as to allow selective orientation onto the surface of a biosensor or affinity support. These peptide surfaces have been used to both monitor and purify APC and APC complexes from cellular extracts.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/química , Proteína de la Poliposis Adenomatosa del Colon/aislamiento & purificación , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Secuencia de Aminoácidos , Avidina/metabolismo , Técnicas Biosensibles , Humanos , Datos de Secuencia Molecular , Pruebas de Precipitina , Conformación Proteica
16.
Am J Physiol Gastrointest Liver Physiol ; 279(3): G500-10, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10960348

RESUMEN

The murine A33 antigen is emerging as a definitive marker of intestinal epithelial cells. Cloning and sequence determination of cDNAs encoding mA33 antigen predict a novel type 1 transmembrane protein of 298 amino acids, comprising an extracellular domain with two immunoglobulin-like domains, a single-span transmembrane domain, and a highly acidic cytoplasmic domain. On the basis of conservation of amino acid sequence and genomic structure, the mA33 antigen is a member of a growing subfamily within the immunoglobulin superfamily, which includes transmembrane proteins CTX/ChT1, CTM/CTH, and CAR. During embryonic development, mA33 antigen expression is first observed in the inner cell mass of blastocysts before implantation. Intestinal expression of mA33 antigen is initiated in the hindgut at E14.5 and increases steadily throughout late embryonic and postnatal life into adulthood. The protein is specifically expressed on the basolateral surfaces of intestinal epithelial cells of all lineages, independent of their position along the rostrocaudal and crypt-villus axes. Thus the mA33 antigen appears to be a novel marker for both proliferating and differentiating intestinal epithelial cells.


Asunto(s)
Células Epiteliales/química , Mucosa Intestinal/citología , Glicoproteínas de Membrana/análisis , Glicoproteínas de Membrana/genética , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/genética , Secuencia de Bases , Biomarcadores , Western Blotting , Carcinoma Embrionario , Moléculas de Adhesión Celular/genética , Clonación Molecular , ADN Complementario , Células Epiteliales/fisiología , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunoglobulinas/genética , Mucosa Intestinal/embriología , Moléculas de Adhesión de Unión , Proteínas de la Membrana/genética , Metaloendopeptidasas , Ratones , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas
17.
Growth Factors ; 18(1): 11-29, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10831070

RESUMEN

The kinetics, binding equilibria and stoichiometry of the interaction between epidermal growth factor and the soluble extracellular domain of the epidermal growth factor receptor (sEGFR), produced in CHO cells using a bioreactor, have been studied by three methods: analytical ultracentrifugation, biosensor analysis using surface plasmon resonance detection (BIAcore 2000) and fluorescence anisotropy. These studies were performed with an sEGFR preparation purified in the absence of detergent using a mild two step chromatographic procedure employing anion exchange and size exclusion HPLC. The fluorescence anisotropy and analytical ultracentrifugation data indicated a 1:1 molar binding ratio between EGF and the sEGFR. Analytical ultracentrifugation further indicated that the complex comprised 2EGF:2sEGFR, consistent with the model proposed recently by Lemmon et al. (1997). Global analysis of the BIAcore binding data showed that a simple Langmuirian interaction does not adequately describe the EGF:sEGFR interaction and that more complex interaction mechanisms are operative. Furthermore, analysis of solution binding data using either fluorescence anisotropy or the biosensor, to determine directly the concentration of free sEGFR in solution competition experiments, yielded Scatchard plots which were biphasic and Hill coefficients of less than unity. Taken together our data indicate that in solution there are two sEGFR populations; one which binds EGF with a KD of 2-20 nM and the other with a KD of 400-550 nM.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Secuencia de Aminoácidos , Animales , Técnicas Biosensibles , Células CHO , Células Cultivadas , Cromatografía Líquida de Alta Presión , Cricetinae , Receptores ErbB/química , Receptores ErbB/aislamiento & purificación , Fibroblastos , Polarización de Fluorescencia , Cinética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Resonancia por Plasmón de Superficie , Ultracentrifugación
18.
Cancer Res ; 60(12): 3254-61, 2000 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10866319

RESUMEN

The Lewis Y (Ley) antigen is a blood group-related antigen that is expressed in a high proportion of epithelial cancers (including breast, colon, ovary, and lung cancer) and is an attractive target for monoclonal antibody-directed therapy. The murine monoclonal 3S193 (IgG3) was generated in BALB/c mice by immunization with Ley-expressing cells of the MCF-7 breast carcinoma cell-line. The murine 3S193 showed high specificity for Ley in ELISA tests with synthetic Ley and Ley-containing glycoproteins and glycolipids and also reacted strongly in rosetting assays and cytotoxic tests with Ley-expressing cells. We generated a humanized form of the murine 3S193 antibody by linking cDNA sequences encoding the variable region of murine 3S913 with frameworks of the human KOL heavy chain and REI K chain. The genes for the humanized 3S193 monoclonal antibody IgG1 were transfected into mouse myeloma NS0 cells and cloned for the establishment of high antibody-producing colonies. Humanized 3S193 antibody was subsequently produced through in vitro culture and under good manufacturing practice conditions using hollow-fiber bioreactors. The purified humanized 3S193 (hu3S193) was subsequently characterized and validated for use in preliminary immunotherapy investigations. hu3S193 reacted specifically with Ley antigen, with similar avidity to the murine form. hu3S193 demonstrated potent immune effector function, with higher antibody-dependent cell-mediated cytotoxicity than its murine counterpart and potent complement-dependent cytotoxicity (ED50, 1.0 microg/ml). The in vivo immunotherapeutic potential of hu3S193 was assessed in a human breast xenograft model using MCF-7, Ley-positive cells. Six i.v. doses of up to 1 mg of hu3S193 were administered to animals bearing established tumors (120-130 mm3) with no significant effect on tumor growth. In contrast, in an MCF-7 xenograft preventive model, a 1-mg hu3S193 dosage schedule was able to significantly slow tumor growth compared with placebo and isotype-matched control IgG1 antibody. hu3S193 has promise for immunotherapy of Ley-positive tumors and is currently entering Phase I clinical trials.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígenos del Grupo Sanguíneo de Lewis/inmunología , Neoplasias/terapia , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Técnicas Biosensibles , Neoplasias de la Mama/terapia , Clonación Molecular , ADN Complementario/metabolismo , Relación Dosis-Respuesta Inmunológica , Ensayo de Inmunoadsorción Enzimática , Humanos , Hibridomas/inmunología , Cinética , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Trasplante de Neoplasias , Homología de Secuencia de Aminoácido , Factores de Tiempo , Células Tumorales Cultivadas
19.
Eur J Biochem ; 267(9): 2505-15, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10785369

RESUMEN

Vascular endothelial growth factor-D (VEGF-D), the most recently discovered mammalian member of the VEGF family, is an angiogenic protein that activates VEGF receptor-2 (VEGFR-2/Flk1/KDR) and VEGFR-3 (Flt4). These receptor tyrosine kinases, localized on vascular and lymphatic endothelial cells, signal for angiogenesis and lymphangiogenesis. VEGF-D consists of a central receptor-binding VEGF homology domain (VHD) and N-terminal and C-terminal propeptides that are cleaved from the VHD to generate a mature, bioactive form consisting of dimers of the VHD. Here we report characterization of mAbs raised to the VHD of human VEGF-D in order to generate VEGF-D antagonists. The mAbs bind the fully processed VHD with high affinity and also bind unprocessed VEGF-D. We demonstrate, using bioassays for the binding and cross-linking of VEGFR-2 and VEGFR-3 and biosensor analysis with immobilized receptors, that one of the mAbs, designated VD1, is able to compete potently with mature VEGF-D for binding to both VEGFR-2 and VEGFR-3 for binding to mature VEGF-D. This indicates that the binding epitopes on VEGF-D for these two receptors may be in close proximity. Furthermore, VD1 blocks the mitogenic response of human microvascular endothelial cells to VEGF-D. The anti-(VEGF-D) mAbs raised to the bioactive region of this growth factor will be powerful tools for analysis of the biological functions of VEGF-D.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Factores de Crecimiento Endotelial/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Secuencia de Bases , División Celular/inmunología , Células Cultivadas , Cartilla de ADN , Factores de Crecimiento Endotelial/inmunología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Mapeo Epitopo , Humanos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Factor D de Crecimiento Endotelial Vascular , Receptor 3 de Factores de Crecimiento Endotelial Vascular
20.
J Chromatogr A ; 869(1-2): 261-73, 2000 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-10720242

RESUMEN

We have previously demonstrated the role of an optical biosensor (BIAcore 2000) as a specific detector to monitor chromatographic fractions during the purification and characterisation of ligands for orphan biomolecules. We have now extended this application to perform micropreparative ligand fishing directly on the sensor surface using an automated cuvette-based optical biosensor (Iasys Auto+) equipped with a high-capacity carboxymethyldextran surface (surface area 16 mm2). Using a F(ab)2' fragment of the A33 monoclonal antibody as bait, we have recovered microgram quantities of essentially homogeneous A33 ligand from the sensor surface in a form suitable for subsequent sensitive and specific down stream analysis (micropreparative HPLC, sodium dodecyl sulphate-polyacrylamide gel electrophoresis and Western blotting). The design of the cuvette-based system facilitates recovery of desorbed material from the constrained workspace in small volumes at high concentration. The use of on-surface detection allows the surface viability to be continuously monitored and permits direct quantitation of both bound and recovered material.


Asunto(s)
Técnicas Biosensibles , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Ligandos , Óptica y Fotónica , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA