Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Gut ; 62(1): 112-20, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22466618

RESUMEN

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDA) is characterised by stromal desmoplasia and vascular dysfunction, which critically impair drug delivery. This study examines the role of an abundant extracellular matrix component, the megadalton glycosaminoglycan hyaluronan (HA), as a novel therapeutic target in PDA. METHODS: Using a genetically engineered mouse model of PDA, the authors enzymatically depleted HA by a clinically formulated PEGylated human recombinant PH20 hyaluronidase (PEGPH20) and examined tumour perfusion, vascular permeability and drug delivery. The preclinical utility of PEGPH20 in combination with gemcitabine was assessed by short-term and survival studies. RESULTS: PEGPH20 rapidly and sustainably depleted HA, inducing the re-expansion of PDA blood vessels and increasing the intratumoral delivery of two chemotherapeutic agents, doxorubicin and gemcitabine. Moreover, PEGPH20 triggered fenestrations and interendothelial junctional gaps in PDA tumour endothelia and promoted a tumour-specific increase in macromolecular permeability. Finally, combination therapy with PEGPH20 and gemcitabine led to inhibition of PDA tumour growth and prolonged survival over gemcitabine monotherapy, suggesting immediate clinical utility. CONCLUSIONS: The authors demonstrate that HA impedes the intratumoral vasculature in PDA and propose that its enzymatic depletion be explored as a means to improve drug delivery and response in patients with pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/fisiología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/fisiología , Ácido Hialurónico/fisiología , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Ductal Pancreático/irrigación sanguínea , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/fisiopatología , Moléculas de Adhesión Celular/administración & dosificación , Moléculas de Adhesión Celular/farmacología , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Doxorrubicina/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Hialuronoglucosaminidasa/administración & dosificación , Hialuronoglucosaminidasa/farmacología , Inmunohistoquímica , Estimación de Kaplan-Meier , Ratones , Ratones Transgénicos , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/fisiopatología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Análisis de Matrices Tisulares , Resultado del Tratamiento , Gemcitabina
2.
J Exp Med ; 166(2): 571-6, 1987 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-3110354

RESUMEN

We investigated the ability of cyclosporin A (CsA) and transforming growth factor beta (TGF-beta) to modulate the production of TNF-alpha and TNF-beta and IFN-gamma by unseparated, nonadherent, and adherent PBMC. Treatment of unseparated PBMC with CsA resulted in a significant dose-dependent inhibition of all three cytokines ranging from greater than 90% inhibition for IFN-gamma and TNF-beta, to approximately 70% for TNF-alpha. Pretreatment of unseparated or nonadherent PBMC with TGF-beta inhibited the production of IFN-gamma by 60-70%. However, the inhibition of TNF-alpha and TNF-beta production by these cells was only minimally affected, and at 0.1-1 ng/ml TGF-beta could enhance TNF-alpha production by unseparated PBMC. In contrast, pretreatment of adherent PBMC with TGF-beta inhibited the production of TNF-alpha by approximately 60%. TGF-beta also inhibited both TNF-alpha production and tumor cell cytotoxicity mediated by murine peritoneal-derived macrophages. These observations indicate that the biological effects of CsA and TGF-beta on immune functions are of a wider range than previously reported.


Asunto(s)
Productos Biológicos/biosíntesis , Ciclosporinas/farmacología , Sustancias de Crecimiento/farmacología , Péptidos/farmacología , Animales , Citocinas , Relación Dosis-Respuesta a Droga , Femenino , Glicoproteínas/biosíntesis , Humanos , Interferón gamma/biosíntesis , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Factores de Crecimiento Transformadores , Factor de Necrosis Tumoral alfa
3.
J Exp Med ; 175(1): 217-25, 1992 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-1346155

RESUMEN

The HER2 protooncogene encodes a 185-kD transmembrane phosphoglycoproteins, human epidermal growth factor receptor 2 (p185HER2), whose amplified expression on the cell surface can lead to malignant transformation. Overexpression of HER2/p185HER2 is strongly correlated with progression of human ovarian and breast carcinomas. Recent studies have shown that human T cells can be targeted with bispecific antibody to react against human tumor cells in vitro. We have developed a bispecific F(ab')2 antibody molecule consisting of a humanized arm with a specificity to p185HER2 linked to another arm derived from a murine anti-CD3 monoclonal antibody that we have cloned from UCHT1 hybridoma. The antigen-binding loops for the anti-CD3 were installed in the context of human variable region framework residues, thus forming a fully humanized BsF(ab')2 fragment. Additional variants were produced by replacement of amino acid residues located in light chain complementarity determining region 2 and heavy chain framework region 3 of the humanized anti-CD3 arm. Flow cytometry analysis showed that the bispecific F(ab')2 molecules can bind specifically to cells overexpressing p185HER2 and to normal human peripheral blood mononuclear cells bearing the CD3 surface marker. In additional experiments, the presence of bispecific F(ab')2 caused up to fourfold enhancement in the cytotoxic activities of human T cells against tumor cells overexpressing p185HER2 as determined by a 51Cr release assay. These bispecific molecules have a potential use as therapeutic agents for the treatment of cancer.


Asunto(s)
Anticuerpos Monoclonales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/genética , Neoplasias de la Mama/inmunología , Proteínas Proto-Oncogénicas/genética , Proto-Oncogenes , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Secuencia de Bases , Complejo CD3 , Clonación Molecular , Citotoxicidad Inmunológica , Femenino , Citometría de Flujo , Variación Genética , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos , Oligonucleótidos Antisentido , Reacción en Cadena de la Polimerasa , Conformación Proteica , Receptor ErbB-2 , Homología de Secuencia de Ácido Nucleico
4.
Science ; 241(4872): 1501-3, 1988 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-3138754

RESUMEN

A hybrid protein between interferon-gamma and tumor necrosis factor-beta was made by ligating the respective genes and expressing the fused genes under the control of the trp promoter in Escherichia coli. The antiproliferative activity of the hybrid protein in vitro was greatly increased compared with either interferon-gamma or tumor necrosis factor-beta alone, and both antiviral activity and cytotoxic effect were retained in the hybrid protein. The hybrid protein may have potential clinical application.


Asunto(s)
Antivirales , Inhibidores de Crecimiento , Interferón gamma , Factor de Necrosis Tumoral alfa , Clonación Molecular , Técnicas de Inmunoadsorción , Proteínas Recombinantes de Fusión/farmacología , Relación Estructura-Actividad
5.
Science ; 230(4728): 943-5, 1985 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-3933111

RESUMEN

Modulation of the growth of human and murine cell lines in vitro by recombinant human tumor necrosis factor-alpha (rTNF-alpha) and recombinant human interferon-gamma (rIFN-gamma) was investigated. rTNF-alpha had cytostatic or cytolytic effects on only some tumor cell lines. When administered together with rIFN-gamma, rTNF-alpha showed enhanced antiproliferative effects on a subset of the cell lines tested. In contrast to its effects on sensitive tumor cells, rTNF-alpha augmented the growth of normal diploid fibroblasts. Variations in the proliferative response induced by rTNF-alpha were apparently not due to differences in either the number of binding sites per cell or their affinity for rTNF-alpha. These observations indicate that the effects of rTNF-alpha on cell growth are not limited to tumor cells, but rather that this protein may have a broad spectrum of activities in vivo.


Asunto(s)
División Celular/efectos de los fármacos , Glicoproteínas/farmacología , Proteínas Recombinantes/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/patología , Sinergismo Farmacológico , Humanos , Interferón gamma/farmacología , Ratones , Factor de Necrosis Tumoral alfa
6.
Handb Exp Pharmacol ; (181): 183-219, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18071947

RESUMEN

The biology of the human epidermal growth factor (EGF) receptor-2 (HER2) has been reviewed numerous times and provides an excellent example for developing a targeted cancer therapeutic. Herceptin, the FDA-approved therapeutic monoclonal antibody against HER2, has been used to treat over 150,000 women with breast cancer. However, the developmental history of Herceptin, the key events within the program that created pivotal decision points, and the reasons why decisions were made to pursue the monoclonal antibody approach have never been adequately described. The history of Herceptin is reviewed in a way which allows the experience to be shared for the purposes of understanding the drug discovery and development process. It is the objective of this review to describe the pivotal events and explain why critical decisions were made that resulted in the first therapeutic to successfully target tyrosine kinases in cancer. New approaches and future prospects for therapeutics targeting the HER family are also discussed.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Inmunoterapia/métodos , Receptor ErbB-2/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/historia , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antineoplásicos/química , Antineoplásicos/historia , Antineoplásicos/farmacología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Diseño de Fármacos , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Inmunoterapia/historia , Modelos Moleculares , Conformación Proteica , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos , Trastuzumab , Resultado del Tratamiento
7.
Mol Cell Biol ; 11(2): 979-86, 1991 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1671297

RESUMEN

Monoclonal antibody (MAb) 4D5 was used to analyze the phosphorylation of p185HER2, the gene product of c-erbB-2/HER2, in SK-BR-3 cells. Culture in the continuous presence of 4D5 reduced the in vivo steady-state levels of p185HER2 phosphorylation by 80% in a dose-dependent manner, suggesting that MAb 4D5 may have interfered with the activation of phosphorylation of p185HER2. The observed MAb-mediated reduction of p185HER2 phosphorylation could not be completely accounted for by down-regulation. When cultures were grown under serum-free conditions, the steady-state levels of p185HER2 phosphorylation were reduced by 56%, and addition of 4D5 further inhibited phosphorylation to 20% of steady-state levels. With continuous exposure to increasing concentrations of newborn calf serum in these cultures, there was a linear increase in tyrosine-specific phosphorylation of p185HER2, reaching a 5.4-fold increase with 10% newborn calf serum. Phosphorylation of p185HER2 in the presence of newborn calf serum was not attributable to stimulation of the epidermal growth factor receptor by epidermal growth factor or by transforming growth factor-alpha. Extension of these observations to two other mammary carcinoma cell lines. MDA-MB-453 and BT-474, also demonstrated a significant capacity of serum to induce p185HER2 phosphorylation. The demonstration of antibody-mediated partial inhibition of phosphorylation under serum-free conditions suggests that mammary carcinoma cells may also produce and secrete a factor or factors which may activate p185HER2. Our observation that growth-inhibitory MAb 4D5 is able to reduce the phosphorylation of p185HER2 by newborn calf serum and by a cellular-derived factor(s) suggests the existence of a growth factor(s) which uses phosphorylation of p185HER2 as a signal transduction pathway to regulate cell proliferation.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Sustancias de Crecimiento/farmacología , Proteínas Proto-Oncogénicas/biosíntesis , Proto-Oncogenes , Animales , Animales Recién Nacidos , Sangre , Neoplasias de la Mama , Bovinos , Medios de Cultivo , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Cinética , Fosforilación , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Receptor ErbB-2
8.
Mol Cell Biol ; 5(4): 768-79, 1985 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2985969

RESUMEN

The classical human interferon-alpha (HuIFN-alpha) gene family is estimated to consist of 15 or more nonallelic members which encode proteins sharing greater than 77% amino acid sequence homology. Low-stringency hybridization with a HuIFN-alpha cDNA probe permitted the isolation of two distinct classes of bovine IFN-alpha genes. The first subfamily (class I) is more closely related to the known HuIFN-alpha genes than to the second subfamily (class II) of bovine IFN-alpha genes. Extensive analysis of the human genome has revealed a HuIFN-alpha gene subfamily corresponding to the class II bovine IFN-alpha genes. The class I human and bovine IFN-alpha genes encode mature IFN polypeptides of 165 to 166 amino acids, whereas the class II IFN-alpha genes encode 172 amino acid proteins. Expression in Escherichia coli of members of both gene subfamilies results in polypeptides having potent antiviral activity. In contrast to previous studies which found no evidence of class II IFN-alpha protein or mRNA expression, we demonstrate that the class I and class II IFN-alpha genes are coordinately induced in response to viral infection.


Asunto(s)
Interferón Tipo I/genética , Animales , Bovinos , Clonación Molecular , Regulación de la Expresión Génica , Genes , Ligamiento Genético , Humanos , Interferón Tipo I/clasificación , Virus de la Parainfluenza 1 Humana/genética , Interferencia Viral
9.
Mol Cell Biol ; 9(3): 1165-72, 1989 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2566907

RESUMEN

The HER2/c-erbB-2 gene encodes the epidermal growth factor receptorlike human homolog of the rat neu oncogene. Amplification of this gene in primary breast carcinomas has been show to correlate with poor clinical prognosis for certain cancer patients. We show here that a monoclonal antibody directed against the extracellular domain of p185HER2 specifically inhibits the growth of breast tumor-derived cell lines overexpressing the HER2/c-erbB-2 gene product and prevents HER2/c-erbB-2-transformed NIH 3T3 cells from forming colonies in soft agar. Furthermore, resistance to the cytotoxic effect of tumor necrosis factor alpha, which has been shown to be a consequence of HER2/c-erbB-2 overexpression, is significantly reduced in the presence of this antibody.


Asunto(s)
Neoplasias de la Mama/terapia , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Anticuerpos Monoclonales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , División Celular , Femenino , Regulación de la Expresión Génica , Humanos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Receptor ErbB-2 , Células Tumorales Cultivadas
10.
J Control Release ; 114(2): 230-41, 2006 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-16876899

RESUMEN

Subcutaneously injected therapeutics must pass through the interstitial matrix of the skin in order to reach their intended targets. This complex, three-dimensional structure limits the type and quantity of drugs that can be administered by local injection. Here we found that depolymerization of the viscoelastic component of the interstitial matrix in animal models with a highly purified recombinant human hyaluronidase enzyme (rHuPH20) increased the dispersion of locally injected drugs, across a broad range of molecular weights without tissue distortion. rHuPH20 increased infusion rates and the pattern and extent of appearance of locally injected drugs in systemic blood. In particular, rHuPH20 changed the pharmacokinetic profiles and significantly augmented the absolute bioavailability of locally injected large protein therapeutics. Importantly, within 24 h of injection, the interstitial viscoelastic barriers were restored without histologic alterations or signs of inflammation. rHuPH20 may function as an interstitial delivery enhancing agent capable of increasing the dispersion and bioavailability of coinjected drugs that may enable subcutaneous administration of therapeutics and replace intravenous delivery.


Asunto(s)
Hialuronoglucosaminidasa/farmacología , Preparaciones Farmacéuticas/metabolismo , Adenoviridae/genética , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Formación de Anticuerpos/efectos de los fármacos , Disponibilidad Biológica , Transporte Biológico Activo/efectos de los fármacos , Capilares/citología , Capilares/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Citocinas/administración & dosificación , Citocinas/farmacocinética , Sistemas de Liberación de Medicamentos , Quimioterapia , Células Endoteliales/metabolismo , Femenino , Terapia Genética , Humanos , Hialuronoglucosaminidasa/administración & dosificación , Inyecciones Subcutáneas , Interferón Tipo I/administración & dosificación , Interferón Tipo I/farmacocinética , Interferón Tipo I/uso terapéutico , Macaca mulatta , Masculino , Ratones , Ratones Desnudos , Peso Molecular , Tamaño de la Partícula , Polietilenglicoles , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología
11.
Cancer Res ; 47(3): 780-6, 1987 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-3802082

RESUMEN

Tumor necrosis factors (TNFs) are a class of cytokines secreted by activated effector cells involved in host defense against tumor progression. Epidermal growth factor (EGF) and recombinant human transforming growth factor-alpha (rHuTGF-alpha) were shown to interfere with the in vitro antiproliferative effects of recombinant human tumor necrosis factor-alpha (rHuTNF-alpha) and -beta on a human cervical carcinoma cell line, ME-180. The inhibitory effect could be observed at EGF or rHuTGF-alpha concentrations of 100 pg/ml, and was maximal between 1 and 10 ng/ml. This response was not due to down regulation of the TNF receptor or to alteration of the affinity of TNF-alpha for its receptor. Since the antiproliferative effect of recombinant human interferon-gamma was not significantly affected by the presence of EGF or rHuTGF-alpha, the inhibition was specific for recombinant TNFs and was not due solely to enhanced proliferation induced by the growth factors. Neither growth factor had a substantial protective effect on the synergistic cytotoxicity observed when tumor cells were exposed simultaneously to rHuTNF-alpha and recombinant human interferon-gamma. TGF-beta can also interfere with the antiproliferative effects of rHuTNF-alpha in vitro. At concentrations of less than 1 ng/ml, TGF-beta significantly antagonized the cytotoxic effects of rHuTNF-alpha on NIH 3T3 fibroblasts. Since EGF, platelet-derived growth factor, and TGF-beta all enhanced NIH 3T3 cell proliferation, but only TGF-beta interfered with rHuTNF-alpha cytotoxicity, the protective effects of TGF-beta were not related in a simple manner to enhanced cell proliferation. rHuTGF-alpha and TGF-beta did not have a significant protective effect against rHuTNF-alpha-mediated cytotoxicity on two other tumor cell lines, BT-20 and L-929 cells. Based upon these observations we suggest that growth factors might enhance tumor growth in vivo by a combination of distinct mechanisms: (a) by autocrine stimulation tumor cell growth; and/or (b) by interfering with normal effector mechanisms of host defense.


Asunto(s)
Glicoproteínas/farmacología , Sustancias de Crecimiento/farmacología , Animales , Neoplasias de la Mama , Carcinoma de Células Escamosas , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Cinética , Células L/citología , Células L/efectos de los fármacos , Ratones , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa , Neoplasias del Cuello Uterino
12.
Cancer Res ; 47(20): 5382-5, 1987 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-2820567

RESUMEN

Recombinant human tumor necrosis factor-alpha (rHuTNF-alpha) inhibited growth of the cervical carcinoma cell line, ME-180neo, at doses greater than 50 units/ml, but stimulated the growth of these cells at low doses (0.1-10 units/ml). ME-180neo variants selected for resistance to the cytotoxic effects of rHuTNF-alpha retained the ability to be growth stimulated at all concentrations tested. ME-180neo cells and the rHuTNF-alpha-resistant ME-180neo variants possessed equivalent steady state numbers of TNF-alpha receptors with similar Kd values. Recombinant human interferon-gamma (rHuIFN-gamma) augmented the rHuTNF-alpha-induced cytotoxic response of ME-180neo cells and overcame the resistance of the ME-180neo variants to rHuTNF-alpha cytotoxicity. In separate experiments we were able to show that the number of TNF-alpha binding sites on both rHuTNF-alpha-sensitive and -resistant ME-180neo cells was similar and was increased by treatment with rHuIFN-gamma. These results suggest that the growth stimulation of tumor cells mediated by rHuTNF-alpha can be dissociated from the cytotoxic response and that these responses are not related to the number or affinity of TNF-alpha binding sites.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Interferón gamma/farmacología , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Neoplasias del Cuello Uterino/patología , División Celular/efectos de los fármacos , Línea Celular , Femenino , Humanos , Cinética , Receptores de Superficie Celular/metabolismo , Receptores del Factor de Necrosis Tumoral
13.
Oncogene ; 10(4): 697-704, 1995 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-7862447

RESUMEN

Reconstitution of retinoblastoma gene (RB) deficient tumor cells with RB generally leads to growth suppression in vitro and/or reduced tumorigenicity in nude mice. An alternate approach to gene replacement is the delivery of the RB gene product (p110RB) into cells lacking its expression. In this report we demonstrate that exogenously added p110RB is taken up by and localized to the nucleus of cultured cells and has growth suppression properties similar to endogenous RB. RB-negative (RBneg) tumor cells are preferentially growth inhibited while most RB-positive (RBpos) tumor cells and normal cells are much less sensitive. We have extended these studies to relevant nude mouse xenograft models for human lung cancer. Local or systemic administration of p110RB inhibits tumor growth in treated animals. These results represent the first use of a tumor suppressor protein as a potential cancer therapeutic.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , División Celular/efectos de los fármacos , Neoplasias Experimentales/patología , Proteína de Retinoblastoma/farmacología , Animales , Núcleo Celular/metabolismo , Inhibidores de Crecimiento , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Trasplante Heterólogo , Células Tumorales Cultivadas
14.
Oncogene ; 15(3): 337-45, 1997 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-9233768

RESUMEN

This study analyses whether the inability of p53 to induce G1 arrest after the restriction point relates to an inability to modulate pRb phosphorylation. Transient p53 overexpression in normal human diploid fibroblasts and p53-deficient cancer cells led to increased levels of the cyclin-dependent kinase inhibitor p21 cip1/Waf1/Sdi1 and an accumulation of hypophosphorylated pRb in cells growing asynchronously and in cells synchronized in late G1 or M. Similarly, gamma-irradiation of asynchronous, late-G1, or S phase fibroblasts led to an increase in hypophosphorylated pRb. Experiments with fibroblasts expressing the HPV16 E6 protein indicated that accumulation of hypophosphorylated pRb required functional p53. Progression into and through S phase was not altered by the presence of hypophosphorylated pRb in late G1, consistent with the failure of p53 to mediate G1 arrest in cells that are past the restriction point. These data indicate that accumulation of hypophosphorylated pRb has significantly different effects on cell cycle progression in early G1 versus late G1 or S phase.


Asunto(s)
Ciclo Celular/fisiología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ciclinas/biosíntesis , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas , Ciclo Celular/efectos de la radiación , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Diploidia , Inhibidores Enzimáticos/metabolismo , Fibroblastos , Fase G1 , Rayos gamma , Humanos , Neoplasias Pulmonares , Proteínas Oncogénicas Virales/biosíntesis , Papillomaviridae/genética , Fosforilación , Proteínas Recombinantes/biosíntesis , Proteínas Represoras/biosíntesis , Fase S , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis
15.
Clin Cancer Res ; 4(7): 1649-59, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9676839

RESUMEN

p53 tumor suppressor gene therapy has been proposed for cancers characterized by inactivation of p53 function, and successful therapy will require efficient strategies for gene delivery. To maximize transgene expression in tumors, a clinical strategy has been proposed to treat neoplasms in the liver via hepatic artery administration of a recombinant adenovirus encoding wild-type p53 (rAd-p53). We have developed a syngeneic rat model using a p53mut hepatocellular carcinoma cell line (McA-RH7777) that results in multifocal liver tumor nodules to provide experimental support for this strategy. Treatment of McA-RH7777 cells with rAd-p53 in vitro resulted in efficient transgene expression, growth suppression, and apoptosis. Intrahepatic artery dosing with rAd-p53 or an adenovirus encoding beta-galactosidase (rAd-betagal) increased transgene expression in tumor tissue and decreased systemic exposure when compared with i.v. dosing. Daily hepatic artery dosing of rAd-p53 suppressed tumor growth when compared with untreated rats or animals treated with rAd-betagal. These data demonstrate the potential for arterial gene delivery to tumors using recombinant adenoviruses, and support continued investigation of rAd-p53 gene therapy for liver malignancies.


Asunto(s)
Adenoviridae , Carcinoma Hepatocelular/terapia , Genes p53 , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Neoplasias Hepáticas Experimentales/terapia , Proteínas de Neoplasias/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Carcinoma Hepatocelular/metabolismo , Virus Defectuosos , Femenino , Expresión Génica , Arteria Hepática , Humanos , Infusiones Intraarteriales , Infusiones Intravenosas , Neoplasias Hepáticas Experimentales/metabolismo , Proteínas de Neoplasias/genética , Ratas , Transgenes , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
16.
J Leukoc Biol ; 41(3): 196-204, 1987 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3031189

RESUMEN

The interaction of highly purified recombinant human tumor necrosis factor-alpha (rTNF-alpha) with human polymorphonuclear neutrophils (PMNs) was investigated. Binding of 125I-rTNF-alpha to PMN reached maximum levels in 30 min at 37 degrees C and in 2 h at 4 degrees C. Scatchard analysis of competitive binding data indicated approximately 6000 receptor sites per cell and a Kd of 1.37 nM. Binding data at 37 degrees C indicated a rapid internalization of rTNF-alpha. Following this receptor-mediated interaction, recombinant TNF-alpha was found to inhibit the migration of PMNs under agarose and to enhance PMN production of superoxide anion (O-2) in a dose-dependent manner. Furthermore, rTNF-alpha-activated PMNs caused a marked disruption of human umbilical-vein-derived endothelial cell monolayers and caused inhibition of their proliferative activities. These data substantiate the role of TNF-alpha as an activator of PMN functions and indicate that PMN/TNF-alpha/endothelial cell interactions may play a major role in inflammatory reactions.


Asunto(s)
Glicoproteínas/metabolismo , Neutrófilos/fisiología , Receptores de Superficie Celular/metabolismo , Unión Competitiva , División Celular , Movimiento Celular , Endotelio/fisiología , Humanos , Interferón gamma/metabolismo , Linfocitos/metabolismo , Monocitos/metabolismo , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes/metabolismo , Superóxidos/metabolismo , Factor de Necrosis Tumoral alfa
17.
Hum Gene Ther ; 6(11): 1403-16, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8573613

RESUMEN

We have investigated the use of column chromatography for the purification of ACN53, a recombinant adenovirus type 5 encoding the human p53 tumor suppressor protein. Anion exchange, size exclusion, hydrophobic interaction, and metal chelating resins were tested; each was found to have distinct advantages and disadvantages. Based on these data, a rapid method was devised for the purification of ACN53. The resultant product was characterized and compared to cesium chloride density-gradient purified virus by SDS-PAGE, Western blot analysis, absorbance spectrum, total particle-to-infectious particle ratio, expression of p53 gene product in Saos-2 cells, growth inhibition of Saos-2 cells, and contamination by ATCC-293 host cell proteins. The results show that column chromatography offers an alternative to ultracentrifugation for the purification of recombinant adenoviruses for use in human gene therapy trials and other research applications.


Asunto(s)
Adenovirus Humanos/aislamiento & purificación , Cromatografía/métodos , Vectores Genéticos/aislamiento & purificación , Proteína p53 Supresora de Tumor/genética , Adenovirus Humanos/genética , Resinas de Intercambio Aniónico/química , Fraccionamiento Celular , Línea Celular , Cesio/química , Cloruros/química , Cromatografía de Afinidad/métodos , Cromatografía en Gel/métodos , Endonucleasas/metabolismo , Etanolaminas/química , Vectores Genéticos/genética , Humanos , Zinc/química
18.
Cancer Gene Ther ; 3(2): 121-30, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8729911

RESUMEN

Human malignancies are often characterized by mutations of the p53 tumor suppressor gene. In a large proportion of cases, the mutation results in production of an altered protein that can bind and inactivate the wild-type gene product. This "dominant-negative" activity of mutant p53 molecules may limit the utility of p53 gene therapy of cancer. Using replication-deficient recombinant adenoviruses (rAd-p53) as a p53 gene delivery system, we evaluated the effects of p53 reintroduction on a series of 45 human cell lines containing wild-type, mutated, or no p53 protein. Results indicate a p53-specific, dose-dependent, and promoter-specific growth inhibition of a majority of p53-altered cell lines that correlates with the degree of adenovirus transgene expression. Similar effects were not observed on cells containing wild-type p53. rAd-p53 inhibited the growth of cells expressing various mutant p53 proteins including those characterized as "dominant negative mutants", and the antiproliferative effects were not abrogated by high levels of endogenous mutated p53 protein. In vivo, rAd-p53 also suppressed tumor growth and increased survival of nude mice bearing tumors that express mutant p53. These results support a role for p53 gene therapy of cancer, including malignancies harboring mutations in this tumor suppressor gene.


Asunto(s)
Adenoviridae/genética , Neoplasias Colorrectales/terapia , Genes p53/genética , Terapia Genética/métodos , Proteína p53 Supresora de Tumor/biosíntesis , Animales , División Celular/efectos de los fármacos , División Celular/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/fisiopatología , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Recombinación Genética , Tasa de Supervivencia , Transducción Genética , Células Tumorales Cultivadas
19.
J Immunol Methods ; 169(2): 231-40, 1994 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-7510760

RESUMEN

We have developed a family of monoclonal antibodies directed against the retinoblastoma gene product (p110RB). One of these monoclonal antibodies, 3C8, binds p110RB near the C-terminal end of the protein (aa886-aa905). It was characterized by immunoblotting, ELISA, fluorescence-activated flow cytometry and immunohistostaining. It was shown to be useful for the detection of p110RB in formalin-fixed and paraffin-embedded tissue sections. Because 3C8 binds outside of regions shown to be involved in p110RB interactions with other cellular proteins, it may be an especially useful reagent for the reliable detection of p110RB in tumor cells, and for the isolation by affinity chromatography of p110RB complexes with other cellular proteins.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Proteína de Retinoblastoma/inmunología , Animales , Anticuerpos Monoclonales/biosíntesis , Biomarcadores de Tumor , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Citometría de Flujo , Genes de Retinoblastoma/inmunología , Células HeLa , Humanos , Immunoblotting , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos BALB C , Retinoblastoma/diagnóstico , Células Tumorales Cultivadas
20.
Biochem Pharmacol ; 61(2): 179-89, 2001 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11163332

RESUMEN

The in vivo administration of enzyme-inhibiting drugs for cancer and infectious disease often results in overexpression of the targeted enzyme. We have developed an enzyme-catalyzed therapeutic agent (ECTA) approach in which an enzyme overexpressed within the resistant cells is recruited as an intracellular catalyst for converting a relatively non-toxic substrate to a toxic product. We have investigated the potential of the ECTA approach to circumvent the thymidylate synthase (TS) overexpression-based resistance of tumor cells to conventional fluoropyrimidine [i.e. 5-fluorouracil (5-FU)] cancer chemotherapy. (E)-5-(2-Bromovinyl)-2'-deoxy-5'-uridyl phenyl L-methoxyalaninylphosphoramidate (NB1011) is a pronucleotide analogue of (E)-5-(2-bromovinyl)-2'-deoxyuridine (BVdU), an antiviral agent known to be a substrate for TS when in the 5'-monophosphorylated form. NB1011 was synthesized and found to be at least 10-fold more cytotoxic to 5-FU-resistant, TS-overexpressing colorectal tumor cells than to normal cells. This finding demonstrates that the ECTA approach to the design of novel chemotherapeutics results in compounds that are selectively cytotoxic to tumor cell lines that overexpress the target enzyme, TS, and therefore may be useful in the treatment of fluoropyrimidine-resistant cancer.


Asunto(s)
Antineoplásicos/metabolismo , Bromodesoxiuridina/análogos & derivados , Bromodesoxiuridina/metabolismo , Diseño de Fármacos , Profármacos/metabolismo , Timidilato Sintasa/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Bromodesoxiuridina/administración & dosificación , Bromodesoxiuridina/farmacología , Nucleótidos de Desoxiuracil/química , Nucleótidos de Desoxiuracil/metabolismo , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Ratones Desnudos , Profármacos/administración & dosificación , Profármacos/farmacología , Timidilato Sintasa/administración & dosificación , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA