Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Sci ; 108(3): 322-330, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28012218

RESUMEN

The role of HGF/SF-MET signaling is important in cancer progression, but its relation with Helicobacter pylori-positive gastric cancers remains to be elucidated. In total, 201 patients with primary gastric carcinoma who underwent curative or debulking resection without preoperative chemotherapy were studied. MET4 and anti-HGF/SF mAbs were used for immunohistochemical analysis. Survival of gastric cancer patients was estimated by Kaplan-Meier method and compared with log-rank. Cox proportional hazards models were fit to determine the independent association of MET-staining status with outcome. The effect of live H. pylori bacteria on cell signaling and biological behaviors was evaluated using gastric cancer cell lines. MET4-positive gastric cancers showed poorer prognosis than MET4-negative cases (overall survival, P = 0.02; relapse-free survival, P = 0.06). Positive staining for MET4 was also a statistically significant factor to predict poor prognosis in H. pylori-positive cases (overall survival, P < 0.01; relapse-free survival, P = 0.01) but not in H. pylori-negative cases. Gastric cancers positively stained with both HGF/SF and MET4 showed a tendency of the worst prognosis. Stimulation of MET-positive gastric cancer cells with live H. pylori bacteria directly upregulated MET phosphorylation and activated MET downstream signals such as p44/42MAPK and Akt, conferring cell proliferation and anti-apoptotic activity. In conclusion, positive staining for MET4 was useful for predicting poor prognosis of gastric cancers with H. pylori infection. Helicobacter pylori stimulated MET-positive gastric cancers and activated downstream signaling, thereby promoting cancer proliferation and anti-apoptotic activity. These results support the importance of H. pylori elimination from gastric epithelial surface in clinical therapy.


Asunto(s)
Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Neoplasias Gástricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor , Femenino , Infecciones por Helicobacter/microbiología , Helicobacter pylori , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Neoplasias Gástricas/microbiología
2.
Kidney Int ; 88(5): 1057-69, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26083655

RESUMEN

The study of kidney cancer pathogenesis and its treatment has been limited by the scarcity of genetically defined animal models. The FLCN gene that codes for the protein folliculin, mutated in Birt-Hogg-Dubé syndrome, presents a new target for mouse modeling of kidney cancer. Here we developed a kidney-specific knockout model by disrupting the mouse Flcn in the proximal tubules, thus avoiding homozygous embryonic lethality or neonatal mortality, and eliminating the requirement of loss of heterozygosity for tumorigenesis. This knockout develops renal cysts and early onset (6 months) of multiple histological subtypes of renal neoplasms featuring high tumor penetrance. Although the majority of the tumors were chromophobe renal cell carcinomas in affected mice under 1 year of age, papillary renal cell carcinomas predominated in the kidneys of older knockout mice. This renal neoplasia from cystic hyperplasia at 4 months to high-grade renal tumors by 16 months represented the progression of tumorigenesis. The mTOR and TGF-ß signalings were upregulated in Flcn-deficient tumors, and these two activated pathways may synergetically cause renal tumorigenesis. Treatment of knockout mice with the mTOR inhibitor rapamycin for 10 months led to the suppression of tumor growth. Thus, our model recapitulates human Birt-Hogg-Dubé kidney tumorigenesis, provides a valuable tool for further study of Flcn-deficient renal tumorigenesis, and tests new drugs/approaches to their treatment.


Asunto(s)
Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Quistes/patología , Modelos Animales de Enfermedad , Neoplasias Renales/genética , Neoplasias Renales/patología , Túbulos Renales Proximales/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Supresoras de Tumor/genética , Animales , Antibióticos Antineoplásicos/uso terapéutico , Carcinogénesis/genética , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Quistes/genética , Hiperplasia/patología , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
3.
Clin Dev Immunol ; 2013: 475809, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861692

RESUMEN

Human anthrax infection caused by exposure to Bacillus anthracis cannot always be treated by antibiotics. This is mostly because of the effect of the remaining anthrax toxin in the body. Lethal factor (LF) is a component of lethal toxin (LeTx), which is the major virulence of anthrax toxin. A murine IgG monoclonal antibody (mAb) against LF with blocking activity (coded LF8) was produced in a previous study. In this report, a human/murine chimeric Fab mAb (coded LF8-Fab) was developed from LF8 by inserting murine variable regions into human constant regions using antibody engineering to reduce the incompatibility of the murine antibody for human use. The LF8-Fab expressed in Escherichia coli could specifically identify LF with an affinity of 3.46 × 10(7) L/mol and could neutralize LeTx with an EC50 of 85 µ g/mL. Even after LeTx challenge at various time points, the LF8-Fab demonstrated protection of J774A.1 cells in vitro. The results suggest that the LF8-Fab might be further characterized and potentially be used for clinical applications against anthrax infection.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Bacillus anthracis/efectos de los fármacos , Toxinas Bacterianas/antagonistas & inhibidores , Fragmentos Fab de Inmunoglobulinas/farmacología , Macrófagos/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Antígenos Bacterianos/inmunología , Bacillus anthracis/crecimiento & desarrollo , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , Secuencia de Bases , Línea Celular , Escherichia coli/genética , Expresión Génica , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Datos de Secuencia Molecular , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología
4.
Stem Cell Res Ther ; 13(1): 306, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35841112

RESUMEN

BACKGROUND: Current treatments for salivary gland (SG) hypofunction are palliative and do not address the underlying cause or progression of the disease. SG-derived stem cells have the potential to treat SG hypofunction, but their isolation is challenging, especially when the tissue has been damaged by disease or irradiation for head and neck cancer. In the current study, we test the hypothesis that multipotent bone marrow-derived mesenchymal stem cells (BM-MSCs) in a rat model are capable of trans-differentiating to the SG epithelial cell lineage when induced by a native SG-specific extracellular matrix (SG-ECM) and thus may be a viable substitute for repairing damaged SGs. METHODS: Rat BM-MSCs were treated with homogenates of decellularized rat SG-ECM for one hour in cell suspension and then cultured in tissue culture plates for 7 days in growth media. By day 7, the cultures contained cell aggregates and a cell monolayer. The cell aggregates were hand-selected under a dissecting microscope, transferred to a new tissue culture dish, and cultured for an additional 7 days in epithelial cell differentiation media. Cell aggregates and cells isolated from the monolayer were evaluated for expression of SG progenitor and epithelial cell specific markers, cell morphology and ultrastructure, and ability to form SG-like organoids in vivo. RESULTS: The results showed that this approach was very effective and guided the trans-differentiation of a subpopulation of CD133-positive BM-MSCs to the SG epithelial cell lineage. These cells expressed amylase, tight junction proteins (Cldn 3 and 10), and markers for SG acinar (Aqp5 and Mist 1) and ductal (Krt 14) cells at both the transcript and protein levels, produced intracellular secretory granules which were morphologically identical to those found in submandibular gland, and formed SG-like organoids when implanted in the renal capsule in vivo. CONCLUSIONS: The results of this study suggest the feasibility of using autologous BM-MSCs as an abundant source of stem cells for treating SG hypofunction and restoring the production of saliva in these patients.


Asunto(s)
Células Madre Mesenquimatosas , Organoides , Animales , Diferenciación Celular , Transdiferenciación Celular , Matriz Extracelular/metabolismo , Ratas , Glándulas Salivales
5.
Infect Immun ; 77(1): 162-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18981257

RESUMEN

The bipartite anthrax lethal toxin (LeTx) consisting of protective antigen (PA) and lethal factor (LF) is a major virulence factor contributing to death from systemic Bacillus anthracis infection. The current vaccine elicits antibodies directed primarily to PA; however, in experimental settings serologic responses to LF can neutralize LeTx and contribute to protection against infection. The goals of the present study were to identify sequential B-cell epitopes of LF and to determine the capacity of these determinants to bind neutralizing antibodies. Sera of recombinant LF-immunized A/J mice exhibited high titers of immunoglobulin G anti-LF reactivity that neutralized LeTx in vitro 78 days after the final booster immunization and protected the mice from in vivo challenge with 3 50% lethal doses of LeTx. These sera bound multiple discontinuous epitopes, and there were major clusters of reactivity on native LF. Strikingly, all three neutralizing, LF-specific monoclonal antibodies tested bound specific peptide sequences that coincided with sequential epitopes identified in polyclonal antisera from recombinant LF-immunized mice. This study confirms that LF induces high-titer protective antibodies in vitro and in vivo. Moreover, the binding of short LF peptides by LF-specific neutralizing monoclonal antibodies suggests that generation of protective antibodies by peptide vaccination may be feasible for this antigen. This study paves the way for a more effective anthrax vaccine by identifying discontinuous peptide epitopes of LF.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , Epítopos de Linfocito B/inmunología , Animales , Carbunco/prevención & control , Vacunas contra el Carbunco/inmunología , Anticuerpos Antibacterianos/sangre , Anticuerpos Monoclonales/inmunología , Línea Celular , Mapeo Epitopo , Femenino , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos A , Pruebas de Neutralización
6.
Biochem Biophys Res Commun ; 380(2): 243-8, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19167346

RESUMEN

It has been reported that the plasma levels of VEGF in tumor patients decreased during dendritic cell (DC)-based immunotherapy, but the underlying mechanism remains unclear. Our current report demonstrates that VEGF levels were significantly decreased in the supernatants of DCs incubated with rhVEGF or tumor conditioned medium (TCM) while the intracellular VEGF in DCs was increased. The increased intracellular VEGF was not due to the de novo VEGF synthesis by DCs because exogenous VEGF inhibited the mRNA expression of VEGF in DCs. More direct evidence was provided to demonstrate that Cy3-labeled VEGF could be internalized by DCs specifically and efficiently. In addition, the activity of DCs to internalize VEGF was abolished by neutralizing antibody against VEGF receptor-1 (Flt-1) and inhibitors of endocytosis such as carbonyl cyanide m-chlorophenyl hydrazone (CCCP) and genistein. This study highlights a novel function of DCs and allows a better understanding of the DC-VEGF interaction.


Asunto(s)
Células Dendríticas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Línea Celular Tumoral , Resinas Compuestas/farmacología , Medios de Cultivo Condicionados/metabolismo , Células Dendríticas/efectos de los fármacos , Endocitosis/efectos de los fármacos , Humanos , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
7.
Methods Mol Biol ; 525: 161-74, xv, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19252831

RESUMEN

Phage display is a powerful tool in identifying specific antibody fragment binding to the target. Several strategies can be used to screen out a specific Fab from the phage library that binds to an antigen; it may be performed on immobilized targeted molecule, on the intact cells, or by other strategies. Antibodies with properties of recognizing tumor cell surface receptors in native conformation and ability to internalize to tumor cells through receptor binding are ideal carriers for targeted immuno-chemo and/or immuno-radiation therapy. This chapter describes a unique bio-panning method to screen out a Fab fragment against human epithelial growth factor receptor (EGFR) from a naive human Fab phage library for potential targeted therapeutic application on EGFR overexpressed cancers.


Asunto(s)
Endocitosis/inmunología , Fragmentos Fab de Inmunoglobulinas/inmunología , Biología Molecular/métodos , Biblioteca de Péptidos , Western Blotting , Línea Celular Tumoral , Células Clonales , Ensayo de Inmunoadsorción Enzimática , Receptores ErbB/química , Receptores ErbB/metabolismo , Escherichia coli , Células Eucariotas/metabolismo , Virus Helper , Humanos , Proteínas Inmovilizadas , Estructura Terciaria de Proteína , Volumetría
8.
Monoclon Antib Immunodiagn Immunother ; 38(5): 185-189, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31486711

RESUMEN

The separation of plasma from blood cells is critical for the accuracy of blood tests because cellular fractions can create discrepancies in analysis. The most common method to separate blood cells from the liquid part of the blood is centrifugation, which is not always applicable in resource-constrained areas and countries. In this study, we describe the generation of monoclonal antibodies (mAbs) against glycophorin A (GPA) of human erythrocytes. BALB/c mice were immunized with human erythrocytes followed by purified GPA. The splenocytes of the immunized mice were fused with Sp2/0 myeloma cells by hybridoma technique. Hybridoma clones were screened by hemagglutination assay and enzyme-linked immunosorbent assay (ELISA). Six hybridoma clones were obtained and subcloned. The characterization of the purified mAbs demonstrates that they are able to bind and retain erythrocytes in hemagglutination assay. Furthermore, one of the mAbs 1A9 recognizes purified GPA in ELISA, whereas the other mAb 1G7 is able to immunoprecipitate GPA from human erythrocyte lysates, and a band of 38 kDa is detected. In conclusion, the anti-GPA mAbs are useful tools in developing a quick and easy way to separate blood plasma from whole blood for clinical tests, and in developing bi-specific antibodies for other clinical applications.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Separación Celular/métodos , Eritrocitos/inmunología , Glicoforinas/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Especificidad de Anticuerpos , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/citología , Pruebas de Hemaglutinación , Humanos , Hibridomas/inmunología , Ratones
9.
J Transl Med ; 6: 77, 2008 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-19055779

RESUMEN

Animal models greatly facilitate understanding of cancer and importantly, serve pre-clinically for evaluating potential anti-cancer therapies. We developed an invasive orthotopic human glioblastoma multiforme (GBM) mouse model that enables real-time tumor ultrasound imaging and pre-clinical evaluation of anti-neoplastic drugs such as 17-(allylamino)-17-demethoxy geldanamycin (17AAG). Clinically, GBM metastasis rarely happen, but unexpectedly most human GBM tumor cell lines intrinsically possess metastatic potential. We used an experimental lung metastasis assay (ELM) to enrich for metastatic cells and three of four commonly used GBM lines were highly metastatic after repeated ELM selection (M2). These GBM-M2 lines grew more aggressively orthotopically and all showed dramatic multifold increases in IL6, IL8, MCP-1 and GM-CSF expression, cytokines and factors that are associated with GBM and poor prognosis. DBM2 cells, which were derived from the DBTRG-05MG cell line were used to test the efficacy of 17AAG for treatment of intracranial tumors. The DMB2 orthotopic xenografts form highly invasive tumors with areas of central necrosis, vascular hyperplasia and intracranial dissemination. In addition, the orthotopic tumors caused osteolysis and the skull opening correlated to the tumor size, permitting the use of real-time ultrasound imaging to evaluate antitumor drug activity. We show that 17AAG significantly inhibits DBM2 tumor growth with significant drug responses in subcutaneous, lung and orthotopic tumor locations. This model has multiple unique features for investigating the pathobiology of intracranial tumor growth and for monitoring systemic and intracranial responses to antitumor agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzoquinonas/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Lactamas Macrocíclicas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Glioblastoma/irrigación sanguínea , Humanos , Imagenología Tridimensional , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia
10.
Clin Cancer Res ; 13(20): 6049-55, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17947467

RESUMEN

PURPOSE: Aberrant c-Met expression has been implicated in most types of human cancer. We are developing Met-directed imaging and therapeutic agents. EXPERIMENTAL DESIGN: To seek peptides that bind specifically to receptor Met, the Met-expressing cell lines S114 and SK-LMS-1 were used for biopanning with a random peptide phage display library. Competition ELISA, fluorescence-activated cell sorting analysis, an internalization assay, and a cell proliferation assay were used to characterize a Met-binding peptide in vitro. To evaluate the utility of the peptide as a diagnostic agent in vivo, 125I-labeled peptide was injected i.v. into nude mice bearing s.c. xenografts of the Met-expressing and hepatocyte growth factor (HGF)/scatter factor-expressing SK-LMS-1/HGF, and total body scintigrams were obtained between 1 and 24 h postinjection. RESULTS: One Met-binding peptide (YLFSVHWPPLKA), designated Met-pep1, reacts with Met on the cell surface and competes with HGF/scatter factor binding to Met in a dose-dependent manner. Met-pep1 is internalized by Met-expressing cells after receptor binding. Met-pep1 inhibits human leiomyosarcoma SK-LMS-1 cell proliferation in vitro. In SK-LMS-1 mouse xenografts, tumor-associated activity was imaged as early as 1 h postinjection and remained visible in some animals as late as 24 h postinjection. CONCLUSIONS: Met-pep1 specifically interacts with Met: it is internalized by Met-expressing cells and inhibits tumor cell proliferation in vitro; it is a potential diagnostic agent for tumor imaging.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Biblioteca de Péptidos , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Proliferación Celular , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Ratones , Ratones Desnudos , Células 3T3 NIH , Trasplante de Neoplasias , Péptidos/química
11.
Oncogene ; 24(1): 101-6, 2005 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-15531925

RESUMEN

Downstream signaling that results from the interaction of hepatocyte growth factor/scatter factor (HGF/SF) with the receptor tyrosine kinase Met plays critical roles in tumor development, progression, and metastasis. This ligand-receptor pair is an attractive target for new diagnostic and therapeutic agents, preclinical development of which requires suitable animal models. The growth of heterotopic and orthotopic Met-expressing human tumor xenografts in conventional strains of immunocompromised mice inadequately replicates the paracrine stimulation by human HGF/SF (hHGF/SF) that occurs in humans with cancer. We have therefore generated a mouse strain transgenic for hHGF/SF (designated hHGF-Tg) on a severe combined immunodeficiency (SCID) background. We report here that the presence of ectopically expressed hHGF/SF ligand significantly enhances growth of heterotopic subcutaneous xenografts derived from human Met-expressing cancer cells, including the lines SK-LMS-1 (human leiomyosarcoma), U118 (human glioblastoma), and DU145 (human prostate carcinoma), but not that of M14-Mel xenografts (human melanoma that expresses insignificant levels of Met). Our results indicate that ectopic hHGF/SF can specifically activate Met in human tumor xenografts. This new hHGF-Tg strain of mice should provide a powerful tool for evaluating drugs and diagnostic agents that target the various pathways influenced by Met activity.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Trasplante Heterólogo , Animales , Factor de Crecimiento de Hepatocito/genética , Humanos , Huésped Inmunocomprometido/inmunología , Ratones , Ratones SCID , Ratones Transgénicos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-met , Receptores de Factores de Crecimiento/genética , Factores de Tiempo , Células Tumorales Cultivadas
12.
Clin Cancer Res ; 11(17): 6190-7, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16144920

RESUMEN

PURPOSE: Hepatocyte growth factor/scatter factor (HGF/SF) and its receptor, c-Met, play important roles in tumor development and progression. In this study, we measured the serum HGF levels in patients with esophageal squamous cell carcinoma (ESCC) to evaluate its relationships with clinicopathologic features and the role of HGF in ESCC. EXPERIMENTAL DESIGN: One hundred and forty-nine patients with ESCC were studied. Pretherapy serum was collected and ELISA was used to detect the concentrations of HGF, vascular endothelial growth factor (VEGF), and interleukin 8 (IL-8). The function of HGF was shown by invasion chamber assay. RESULTS: Pretherapy serum HGF was found to be significantly higher in patients with ESCC than in control subjects. The levels of HGF correlated significantly with advanced tumor metastasis stage and survival. Multivariate analyses showed that serum HGF level in cell migration was an independent prognostic factor. Increased HGF serum levels correlated positively with serum levels of VEGF and IL-8. Our results also showed that HGF was overexpressed in ESCC tissues and cell lines. In vitro study showed that HGF could stimulate ESCC cell to express VEGF and IL-8 and markedly enhance invasion and migration of ESCC cells. Furthermore, HGF-induced IL-8 and VEGF expression was dependent on extracellular signal-regulated kinase signaling pathways. The inhibition of extracellular signal-regulated kinase activation reduced HGF-mediated IL-8 and VEGF expression. CONCLUSIONS: Our results suggest that serum HGF may be a useful biomarker of tumor progression and a valuable independent prognostic factor in patients with ESCC. HGF may be involved in the progression of ESCC as an autocrine/paracrine factor via enhancing angiogenesis and tumor cell invasion and migration.


Asunto(s)
Biomarcadores de Tumor/sangre , Carcinoma de Células Escamosas/sangre , Movimiento Celular/fisiología , Neoplasias Esofágicas/sangre , Factor de Crecimiento de Hepatocito/sangre , Interleucina-8/sangre , Factor A de Crecimiento Endotelial Vascular/sangre , Anciano , Carcinoma de Células Escamosas/diagnóstico , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Neoplasias Esofágicas/diagnóstico , Esófago/metabolismo , Femenino , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Neovascularización Patológica , Pronóstico , Proteínas Proto-Oncogénicas c-met/metabolismo
13.
Clin Cancer Res ; 11(19 Pt 2): 7064s-7069s, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16203803

RESUMEN

PURPOSE: Met, an oncogene product and receptor tyrosine kinase, is a keystone molecule for malignant progression in solid human tumors. We are developing Met-directed imaging and therapeutic agents, including anti-Met monoclonal antibodies (MetSeek). In this study, we compared two antibodies, Met5 and Met3, for nuclear imaging of human and canine Met-expressing tumor xenografts in nude mice. EXPERIMENTAL DESIGN: Xenografts representing cancers of three different human tissue origins and metastatic canine prostate cancer were raised s.c. in host athymic nude mice. Animals were injected i.v. with I-125-Met5 or I-125-Met3, posterior total body gamma camera images were acquired for several days postinjection, and quantitative region-of-interest activity analysis was done. RESULTS: PC-3, SK-LMS-1/HGF, and CNE-2 xenografts imaged with I-125-Met5 were compared with PC-3, SK-LMS-1/HGF, and DU145 xenografts imaged with I-125-Met3. Nuclear imaging contrast was qualitatively similar for I-125-Met5 and I-125-Met3 in PC-3 and SK-LMS-1/HGF host mice. However, by region-of-interest analysis, the set of human tumors imaged with I-125-Met3 exhibited a pattern of rapid initial tumor uptake followed by a continuous decline in activity, whereas the set of human tumors imaged with I-125-Met5 showed slow initial uptake, peak tumor-associated activity at 1 day postinjection, and persistence of activity in xenografts for at least 5 days. GN4 canine prostate cancer xenografts were readily imaged with I-125-Met5. CONCLUSIONS: We conclude that radioiodinated Met3 and Met5 offer qualitatively similar nuclear images in xenograft-bearing mice, but quantitative considerations indicate that Met5 might be more useful for radioimmunotherapy. Moreover, canine prostate cancer seems to be a suitable model for second-stage preclinical evaluation of Met5.


Asunto(s)
Anticuerpos Monoclonales/química , Neoplasias/diagnóstico , Neoplasias/patología , Proteínas Proto-Oncogénicas c-met/biosíntesis , Animales , Perros , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Ratones , Ratones Desnudos , Neoplasias Nasofaríngeas/patología , Trasplante de Neoplasias , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-met/fisiología , ARN Interferente Pequeño/metabolismo , Radioinmunoterapia/métodos , Cintigrafía , Factores de Tiempo
14.
Cancer Res ; 62(2): 589-96, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11809714

RESUMEN

Met tyrosine kinase, the receptor for HGF/SF, is important in various cellular functions, including proliferation, mitogenesis, formation of branching tubules, angiogenesis, and tumor cell invasion and metastasis. However, the role of Met/HGF signaling pathway in nasopharyngeal carcinoma (NPC) has not been evaluated. In this study, we determined the expression profile and clinical correlation of Met/HGF in 66 cases of advanced NPC and the activation mechanisms of Met receptor in five NPC cell lines. Immunofluorescent staining and quantitative image analysis showed that the Met protein was expressed throughout the tumors and normal nasopharyngeal epithelia. Compared with NPC, the Met expression level was higher in columnar nasopharyngeal epithelium but lower in squamous nasopharyngeal epithelium. The normal interstitial stromal tissue expressed the lowest level of Met protein. HGF was detected mainly in the normal interstitial tissue surrounding the tumor. Met protein expression level was inversely correlated with patients' survival time; the correlation coefficient was -0.319 (P = 0.009). The mean survival time was 118 months in low Met expression group versus 52 months in high expression group (P = 0.0004). The cumulative 5-year survival rate was 77.68% in low expression group versus 38.24% in high expression group. The clinical stage was also significantly more advanced in high Met expression group. In the multivariate analysis, both clinical stage and Met protein expression level were independent prognostic indicators for patient survival. All of the five NPC cell lines tested did not express hgf mRNA but expressed met mRNA, and tyrosine phosphorylation of Met protein was mainly induced by exogenous HGF stimulation in these cells. No mutation was found in the tyrosine kinase and the juxtamembrane domains of Met receptor in the five NPC cell lines tested. These results indicate that: (a) high Met protein expression level correlates with poorer survival in late-stage NPC and serves as an independent prognostic indicator; and (b) the Met receptor in NPC is activated by its paracrine ligand HGF from the interstitial tissues rather than by an autocrine loop or its activating mutation.


Asunto(s)
Neoplasias Nasofaríngeas/enzimología , Proteínas Proto-Oncogénicas c-met/biosíntesis , Adulto , Anciano , Biopsia , Femenino , Factor de Crecimiento de Hepatocito/biosíntesis , Factor de Crecimiento de Hepatocito/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Pronóstico , Proteínas Proto-Oncogénicas c-met/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Tasa de Supervivencia
15.
Mol Biotechnol ; 31(1): 41-54, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16118414

RESUMEN

Inappropriate expression of the receptor tyrosine kinase Met and its ligand hepatocyte growth factor (HGF)/scatter factor (SF) is usually associated with an aggressive solid tumor phenotype (angiogenesis, invasiveness, and metastasis) and poor clinical prognosis. We report here the design and construction of a large, human naïve antigen-binding fragment (Fab) phage-display library with a diversity of 2.0 x 109, which allows rapid isolation of antigen-specific human antibody fragments. A Fab fragment specifically against Met (designated hFab-Met-1) was successively selected from this library by using biopanning on Met-transfected cell line S114. The specificity of hFab-Met-1 was characterized by immunoprecipitation, Western blotting, and flow cytometry. The results demonstrate that hFab-Met-1 reacts with the extracellular domain of Met in its native conformation. Moreover, functional analysis by Madine-Darby canine kidney cell scattering and urokinase-type plasminogen activator assays demonstrated that hFab-Met-1 is not an agonist to HGF/Met signaling compared with a murine intact monoclonal antibody (MAb) Met5. To confirm that hFab-Met-1 interacts with Met-expressing tumors in vivo, I-125-labeled hFab-Met-1 was nuclear-imaged in a mouse xenograft of Met- and HGF/SF-expressing human leiomyosarcoma. Total body scintigrams were obtained between 1 and 48 h postinjection (PI). Tumor-associated activity was imaged as early as 1 h PI, and remained visible in some animals as late as 24 h PI. As expected, activity was highest in the kidneys in early images, whereas thyroid activity became predominant in later images. In conclusion, hFab-Met-1 interacts with Met both in vitro and in vivo, and is a promising candidate for clinical diagnosis and therapeutics.


Asunto(s)
Anticuerpos Monoclonales , Fragmentos Fab de Inmunoglobulinas/genética , Biblioteca de Péptidos , Proteínas Proto-Oncogénicas c-met/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Western Blotting , Electroforesis en Gel de Poliacrilamida , Factor de Crecimiento de Hepatocito , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/uso terapéutico , Inmunoprecipitación , Leiomiosarcoma/diagnóstico por imagen , Leiomiosarcoma/tratamiento farmacológico , Invasividad Neoplásica , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-met/genética , Radioinmunodetección , Radiofármacos , Trasplante Heterólogo , Células Tumorales Cultivadas
16.
Clin Cancer Res ; 9(10 Pt 2): 3839S-44S, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14506181

RESUMEN

PURPOSE: Inappropriate expression of the receptor tyrosine kinase Met and its ligand is associated with an aggressive phenotype and poor clinical prognosis for a wide variety of solid human tumors. We are developing imaging and therapeutic agents that target this receptor-ligand complex. In this study, we evaluated the ability of radioiodinated anti-Met monoclonal antibodies from a single hybridoma clone to image human Met-expressing tumor xenografts. EXPERIMENTAL DESIGN: Xenografts of four different tissue origins were raised s.c. in host athymic nude mice. Animals received i.v. injections of I-125-Met3, posterior total body gamma camera images were acquired for several days after injection, and quantitative region-of-interest activity analysis was performed. RESULTS: The autocrine Met-expressing tumors S-114 and SK-LMS-1/HGF and the paracrine Met-expressing human prostate carcinoma PC-3 were satisfactorily imaged with I-125-Met3. By region-of-interest analysis, mean initial tumor-associated activities in S-114, SK-LMS-1/HGF, and PC-3 were 18.6 +/- 2.1, 7.2 +/- 2.2, and 5.4 +/- 2.6% estimated injected activity, and the mean ratios of tumor:total body activity at 3 days after injection were 0.32 +/- 0.13, 0.15 +/- 0.06, and 0.10 +/- 0.04, respectively. Human melanoma xenografts, however, accounted for < or =3% of injected or total body activity. We observed a direct rank order correlation between relative levels of Met3-derived radioactivity in xenografts and relative quantities of Met expressed by the respective cultured tumor cell lines. CONCLUSIONS: We conclude that I-125-Met3 is effective for imaging human Met-expressing xenografts of different tissue origins, and we infer that I-125-Met3 distinguishes human tumor xenografts according to their levels of Met expression.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Radioinmunodetección/métodos , Animales , Anticuerpos Monoclonales/química , Línea Celular Tumoral , Separación Celular , Citometría de Flujo , Humanos , Hibridomas , Inmunohistoquímica , Ligandos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fenotipo , Unión Proteica , Proteínas Proto-Oncogénicas c-met/química , Factores de Tiempo
17.
Clin Cancer Res ; 10(19): 6629-37, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15475453

RESUMEN

PURPOSE: A reliable method for diagnosing parathyroid carcinoma has remained elusive over the years, resulting in its under-recognition and suboptimal therapy. Obtaining an accurate diagnosis has become an even more pressing matter with recent evidence that germline HRPT2 gene mutations are found in patients with apparently sporadic parathyroid carcinoma. There is a high prevalence of HRPT2 gene mutations and biallelic inactivation in parathyroid carcinoma. We hypothesize that loss of parafibromin, the protein product of the HRPT2 gene, would distinguish carcinoma from benign tissue. EXPERIMENTAL DESIGN: We generated a novel antiparafibromin monoclonal antibody and performed immunostaining on 52 definite carcinoma specimens, 6 equivocal carcinoma specimens, 88 benign specimens, and 9 hyperparathyroidism-jaw tumor (HPT-JT) syndrome-related adenomas from patients with primary hyperparathyroidism from nine worldwide centers and one national database. RESULTS: We report that the loss of parafibromin nuclear immunoreactivity has 96% sensitivity [95% confidence interval (CI), 85-99%] and 99% specificity (95% CI, 92-100%) in diagnosing definite carcinoma. Inter-observer agreement for evaluation of parafibromin loss was excellent, with unweighted kappa of 0.89 (95% CI, 0.79-0.98). Two equivocal carcinomas misclassified as adenomas were highlighted by parafibromin immunostaining. One of these tumors has since recurred, satisfying criteria for a definite carcinoma. Similarly, eight of nine HPT-JT syndrome-related adenomas showed absent nuclear immunoreactivity. CONCLUSIONS: Parafibromin is a promising molecular marker for diagnosing parathyroid carcinoma. The similar loss of parafibromin immunoreactivity in HPT-JT syndrome-related adenomas suggests that this is a pivotal step in parathyroid tumorigenesis.


Asunto(s)
Neoplasias de las Paratiroides/patología , Proteínas/análisis , Anticuerpos Monoclonales/inmunología , Línea Celular , Diagnóstico Diferencial , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica/métodos , Neoplasias de las Paratiroides/metabolismo , Proteínas/genética , Proteínas/inmunología , Reproducibilidad de los Resultados , Análisis de Secuencia de Proteína , Proteínas Supresoras de Tumor
18.
Hum Antibodies ; 12(4): 129-35, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-15156101

RESUMEN

Anthrax toxin is the dominant virulence factor of Bacillus anthracis; drugs blocking its action could therefore have therapeutic benefit. We report here the production of a neutralizing monoclonal antibody (mAb) against anthrax lethal factor (LF) and the inhibition by the antibody of anthrax lethal toxin (LeTx) formation. The anti-LF monoclonal antibody LF8 neutralized the LeTx challenge both in vitro with macrophage J774A.1 cells and in vivo in nude mice. Our data suggested that LF8 binds LF at or near the PA binding domain. A set of dodecameric peptides was selected from a phage-displayed peptide library through their specific binding to anti-LF neutralizing mAb LF8. These small peptides compete with LF to bind LF8. Further investigation is undergoing to test the potential application of these peptides to the clinical treatment of anthrax infection by blocking LeTx formation.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos , Toxinas Bacterianas , Proteínas Portadoras/antagonistas & inhibidores , Animales , Proteínas Portadoras/inmunología , Proteínas Portadoras/toxicidad , Línea Celular , Mapeo Epitopo , Femenino , Ratones , Ratones Desnudos , Pruebas de Neutralización , Biblioteca de Péptidos , Relación Estructura-Actividad
19.
Methods Mol Biol ; 1105: 3-15, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24623215

RESUMEN

This chapter describes methods for the use of antibody microarrays with rolling-circle amplification (RCA). The methods are divided into three sections. The first section covers antibody preparation and microarray production, the second describes the method for using biological samples on antibody microarrays, and the third describes the method for RCA use on antibody microarrays. RCA can be used on antibody microarrays to increase the signal from each antibody spot and lower the detection limits of the assays. We also describe a practical method for running multiple, low-volume microarrays on a single microscope slide. These methods should be useful for researchers interested in rapidly developing and optimizing custom immunoassays for the analysis of low-abundance analytes using low sample volumes.


Asunto(s)
Técnicas de Amplificación de Ácido Nucleico , Animales , Anticuerpos Inmovilizados , Análisis Químico de la Sangre , Proteínas Sanguíneas/metabolismo , ADN de Cadena Simple/química , Humanos , Inmunoensayo , Análisis por Matrices de Proteínas
20.
Artículo en Inglés | MEDLINE | ID: mdl-24555932

RESUMEN

Pancreatic cancer remains a major challenge for research studies and clinical management. Monoclonal antibodies (MAbs) against pancreatic cancer biomarkers could be used in the early diagnosis of pancreatic cancer. The hippocalcin-like 1 protein (HPCAL1) is expressed in most pancreatic cancer cell lines. The goal of this work was to generate anti-HPCAL1 MAbs that have potential clinical applications. Recombinant HPCAL1 protein was successfully produced in an Escherichia coli expression system and was used as immunogen in mice. MAbs against HPCAL1 were generated using the standard hybridoma technique. Anti-HPCAL1 MAb 1E10 detects denatured HPCAL1 protein in Western blot analysis and immunoprecipitates native HPCAL1 protein from cell lysates. MAb 5A5 detects HPCAL1 in formalin-fixed, paraffin-embedded pancreatic cancer cell lines, making it a potentially useful reagent for pathologists. The MAb pair 1E10 and 5A5 had the optimal binding affinity for soluble HPCAL1 detection in a sandwich ELISA system, which is a potential clinical detection method for pancreatic cancer. In conclusion, multi-functional anti-HPCAL1 MAbs 1E10 and 5A5 have been generated and characterized, which may be used in early diagnosis of pancreatic cancer.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Neurocalcina/inmunología , Neoplasias Pancreáticas/inmunología , Animales , Biomarcadores de Tumor/inmunología , Línea Celular Tumoral , Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Pancreáticas/diagnóstico , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA