Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
1.
J Obstet Gynaecol ; 30(6): 596-604, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20701510

RESUMEN

Postmenopausal women with localised, early breast cancer (n = 285) were enrolled in a prospective subprotocol of the 'arimidex, tamoxifen, alone or in combination' (ATAC) trial to assess gynaecological abnormalities arising during treatment with anastrozole (1 mg/day) or tamoxifen (20 mg/day). After 6 years' follow-up, there appeared to be non-significantly fewer endometrial abnormalities with anastrozole than with tamoxifen (12.4% vs 20.2%, odds ratio 0.52; 95% confidence intervals 0.20, 1.32; p = 0.17). The time to first endometrial abnormality was non-significantly longer for patients receiving anastrozole compared with tamoxifen (hazard ratio 0.57; 95% confidence intervals 0.26, 1.22; p = 0.15), with most abnormalities occurring within the first year of treatment. Fewer patients treated with anastrozole appeared to require medical intervention for endometrial abnormalities, compared with patients on tamoxifen. This study showed that there was no significant difference in endometrial pathology between anastrozole and tamoxifen treatment groups.


Asunto(s)
Antineoplásicos Hormonales/administración & dosificación , Endometrio/efectos de los fármacos , Endometrio/patología , Nitrilos/administración & dosificación , Tamoxifeno/administración & dosificación , Triazoles/administración & dosificación , Adulto , Anastrozol , Neoplasias de la Mama/tratamiento farmacológico , Quimioterapia Adyuvante , Femenino , Humanos , Persona de Mediana Edad , Posmenopausia , Resultado del Tratamiento
2.
J Natl Cancer Inst ; 88(23): 1770-6, 1996 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-8944008

RESUMEN

BACKGROUND: Epidermal growth factor (EGF)-related proteins, such as transforming growth factor-alpha (TGF-alpha), control cancer cell growth through hormonal pathways (i.e., autocrine [hormone acts on cell that produces it] and paracrine [hormone acts on nearby cells] pathways). Overexpression of TGF-alpha and/or its receptor (EGFR) has been detected in human cancers. The blockade of EGFR activation by the use of anti-EGFR monoclonal antibodies (MAbs) has been proposed as a potential anticancer therapy. The type I cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKAI) is generally overexpressed in human cancer cells and is involved in neoplastic transformation. Inhibition of PKAI by selective cAMP analogues, such as 8-chloro-cAMP (8-CI-cAMP), induces growth inhibition in various human cancer cell lines. PURPOSE: On the basis of our previous observations of a cooperative anti-proliferative effect of anti-EGFR Mab 528 and 8-Cl-cAMP in human cancer cell lines in vitro, we evaluated the anticancer activity in vivo of the combination of an anti-EGFR MAb (MAb C225) and 8-Cl-cAMP. METHODS: Athymic mice were injected subcutaneously with 10(7) human colon carcinoma GEO cells. After 7 days, when established tumor xenografts of 0.30-0.35 cm3 were detectable, 10-15 mice per group were treated intraperitoneally twice weekly with different doses of 8-Cl-cAMP and/or MAb C225. Cancer cell expression of various growth factors was evaluated by immunohistochemical analysis in tumors obtained from control and treated mice. Data were evaluated for statistical significance using the Student's t test and the Mantel-Cox logrank test. All P values represent two-sided tests of statistical significance. RESULTS: A 5-week treatment with low doses of 8-Cl-cAMP (0.5 mg/dose) and MAb C225 (0.25 mg/dose) blocked GEO tumor growth (compared with that in control mice; P < .00001) and suppressed cancer cell production of autocrine growth factors, such as TGF-alpha, amphiregulin, and CRIPTO, and of angiogenic (promotes new blood vessel formation) factors, such as vascular endothelial growth factor and basic fibroblast growth factor, with no signs of toxicity. Control and 8-Cl-cAMP (0.5 mg/dose)-treated mice died within 9-10 weeks after tumor cell injection. In MAb C225 (0.25 mg/dose)-treated mice, GEO tumors resumed a growth rate comparable to that in control animals within 3 weeks following the end of treatment and the mice died between 11 and 20 weeks after tumor cell injection. GEO tumor growth was significantly delayed in the MAb C225 plus 8-Cl-cAMP treatment group (P < .00001) and was accompanied by a prolonged survival of mice (P < .00001) as compared with the control group. CONCLUSIONS: Long-term treatment with a combination of agents that selectively inhibit two intracellular signal-transduction enzymes, such as the PKAI serine-threonine kinase and the EGFR tyrosine kinase, has anticancer activity in vivo, reflected by suppression of tumor proliferation and angiogenesis, with no signs of toxicity. IMPLICATIONS: Since these inhibitors of intracellular mitogenic (growth-stimulating) signaling have a different mechanism(s) of action and do not antagonize the effects of cytotoxic therapy, a combination of anti-EGFR MAb C225 and 8-Cl-cAMP should be investigated as a nontoxic, long-term treatment for cancer patients following chemotherapy.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Receptores ErbB/antagonistas & inhibidores , Factores Inmunológicos/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Desnudos
3.
J Natl Cancer Inst ; 90(14): 1087-94, 1998 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-9672257

RESUMEN

BACKGROUND: The expression of epidermal growth factor receptor (EGFR) and type I cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKAI) is associated with neoplastic transformation. By use of human renal cancer cell lines (i.e., 769-P, ACHN, A498, and SW839), we investigated the antiproliferative activity and the antitumor activity of an anti-EGFR humanized chimeric mouse monoclonal antibody, MAb C225, and a novel mixed backbone 18-mer antisense oligonucleotide, HYB 190, that targets expression of the RIalpha regulatory subunit of PKAI. METHODS: The antiproliferative activity of MAb C225 and oligonucleotide HYB 190, alone or in combination, on different renal cancer cell lines was determined by monitoring cell growth in soft agar. In addition, the induction of apoptosis by treatment with the anti-EGFR antibody and/or antisense PKAI oligonucleotides was evaluated by flow cytometric analysis of fragmented DNA. The antitumor activity of MAb C225 and oligonucleotide HYB 190 was determined in athymic mice bearing established ACHN tumor xenografts. Cell proliferation and tumor growth data were evaluated for statistical significance using Student's t test; reported P values are two-sided. RESULTS: MAb C225 and oligonucleotide HYB 190 inhibited colony formation in soft agar in a dose-dependent manner for all renal cancer cell lines tested. We observed a potentiation of growth inhibition and induction of apoptosis when 769-P cells and ACHN cells were treated with both agents. Combination treatment with MAb C225 and oligonucleotide HYB 190 caused regression of ACHN tumor xenografts, whereas single-agent treatment only delayed tumor growth. CONCLUSION: The combination of anti-EGFR MAb C225 and ited cooperative antiproliferative effects and cooperative antitumor effects on EGFR and PKAI-expressing human renal cancer cell lines.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Receptores ErbB/antagonistas & inhibidores , Terapia Genética/métodos , Inmunoterapia/métodos , Neoplasias Renales/terapia , Oligonucleótidos Antisentido/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados , Apoptosis , Cetuximab , Terapia Combinada , Citometría de Flujo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/genética , Neoplasias Renales/inmunología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Recombinantes de Fusión/uso terapéutico , Células Tumorales Cultivadas
4.
Cancer Res ; 51(4): 1294-9, 1991 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-1997168

RESUMEN

Unregulated or increased expression of epidermal growth factor receptor (EGF-R) is a common event in neoplastic transformation; modulation of such a receptor by physiological agents could be, therefore, of clinical interest. We have studied the binding ability, the availability at cell surface, and the synthesis of EGF-R in the A431 and KB human epidermoid cancer cell lines after treatment with recombinant alpha-interferon (IFN-alpha). After 48 h of treatment, IFN-alpha induces, in both cell lines, growth inhibition and enhances class I major histocompatibility HLA complex expression, which is a common marker of IFN action. [125I]EGF total binding assessed after 48 h of treatment with IFN-alpha shows a dose-dependent upregulation of EGF-R binding capacity. Saturation plots of the binding data show that IFN-alpha treatment does not dramatically alter the affinity of the EGF-R and indicate that IFN-alpha only increases the number of low affinity receptors. We show that this effect is due to a specific increase in the synthesis of the receptor protein, as assessed by immunoprecipitation of [35S]methionine-labeled cell extracts. Electron microscopy analysis has confirmed an increase of EGF-R proteins at cell surface without major changes in the morphology of the cells. Taken together, these results indicate that IFN-alpha consistently induces both the binding capacity and the synthesis of EGF-R in human epidermoid cancer cells and suggest the use of such a mechanism for new anticancer therapies.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Receptores ErbB/efectos de los fármacos , Interferón Tipo I/farmacología , Sitios de Unión , Carcinoma de Células Escamosas/metabolismo , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/biosíntesis , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Genes MHC Clase I/efectos de los fármacos , Antígenos HLA/biosíntesis , Humanos , Técnicas In Vitro , Microscopía Inmunoelectrónica , Células Tumorales Cultivadas , Regulación hacia Arriba
5.
Cancer Res ; 51(18): 4898-902, 1991 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-1893380

RESUMEN

Reversal of the drug resistance phenotype by the use of agents which induce cell differentiation offers an experimental approach to the study of chemoresistance. In numerous in vitro models, alpha-interferon (alpha-IFN) has been shown to induce phenotypical changes and to modulate the growth of cancer cells. The aim of the present study was to define the effect of alpha-IFN on the Adriamycin sensitivity of the human colon adenocarcinoma cell line, LoVo, and its Adriamycin-resistant variant, LoVo/DX. Pretreatment of LoVo/DX cells with 500 units/ml of alpha-IFN increased sensitivity to low doses of Adriamycin. Similar treatment conditions did not change the sensitivity of the parental cell line. Following treatment of the LoVo/DX cells with alpha-IFN plus 100 ng/ml Adriamycin for 1 h, 30% of the cells survived compared to 100% of untreated cells. This effect was not related to changes in cell cycle kinetics induced by alpha-IFN treatment and did not result from variations in the expression of P-glycoprotein at the cell surface, as assessed by flow cytometric analysis using monoclonal antibody MRK16. Adriamycin accumulation was increased by alpha-IFN as assessed by spectrofluorometric analysis. Thus, the data suggest that in LoVo/DX cells, alpha-IFN increased Adriamycin cytotoxicity through modulation of the multidrug resistance phenotype.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Doxorrubicina/farmacología , Interferón Tipo I/farmacología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Doxorrubicina/farmacocinética , Resistencia a Medicamentos/genética , Ensayos de Selección de Medicamentos Antitumorales , Expresión Génica , Humanos , Glicoproteínas de Membrana/genética , Fosfoproteínas/genética , Proteínas Recombinantes , Células Tumorales Cultivadas
6.
Cancer Res ; 54(15): 4123-8, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-7518350

RESUMEN

We have shown that a mutant derivative of Chinese hamster ovary CHO-K1 cells, ADR-5, which shows hypersensitivity to topoisomerase II (topo II)-inhibitory drugs, is cross-sensitive to the site-selective cyclic AMP analogue 8-chloro-cyclic AMP. We tested the hypothesis that overexpression of the type I alpha regulatory subunit of protein kinase A may represent a common element conferring hypersensitivity to both topo II inhibitors and 8-chloro-cyclic AMP in ADR-5 cells. We have demonstrated that ADR-5 cells overexpress RI alpha protein, compared to parental CHO-K1 cells. Moreover, retroviral vector-mediated transfer of the RI alpha gene into CHO-K1 cells was able to confer a drug-hypersensitive phenotype similar to that exhibited by ADR-5 cells. Analysis of topo II protein levels and activity revealed no differences between parental and infected cells, suggesting that protein kinase A may be involved in the downstream processing of topo II-mediated events.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Antineoplásicos/farmacología , Bleomicina/farmacología , Células CHO/efectos de los fármacos , Células CHO/enzimología , Cisplatino/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Etopósido/farmacología , Tenipósido/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Células CHO/patología , División Celular/efectos de los fármacos , Cricetinae , Ensayos de Selección de Medicamentos Antitumorales
7.
Cancer Res ; 57(22): 5107-11, 1997 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-9371510

RESUMEN

8-Chloro-cAMP (8-Cl-cAMP) is a novel agent that is able to inhibit the growth of a wide variety of cancer cell types in vitro and in vivo and, at doses devoid of toxicity, to achieve plasma concentrations in cancer patients in a range effective for cancer cell growth inhibition. In this study, we have demonstrated that 8-Cl-cAMP, at a dose causing mild or no growth inhibition, synergistically increased the growth-inhibitory effect of paclitaxel or cisplatin in a wide series of cell lines including human breast, lung, ovary, colon, and head carcinomas and melanoma. A similar effect was also observed with another taxane, docetaxel, and with the platinum-derivative carboplatin. 8-Cl-cAMP also markedly enhanced apoptotic cell death induced by each cytotoxic drug. A cooperative antitumor effect was also observed in vivo, because treatment with paclitaxel followed by 8-Cl-cAMP markedly inhibited the growth of GEO human colon cancer xenografts as compared to paclitaxel alone without signs of toxicity. These data demonstrate that 8-Cl-cAMP synergistically increases the antiproliferative activity of taxanes and platinum-derived compounds and provide a rationale to use 8-Cl-cAMP in combination with taxanes and platinum-derived compounds.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Paclitaxel/farmacología , Ensayo de Tumor de Célula Madre/métodos , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Apoptosis/genética , División Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Fase G2 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitosis , Trasplante de Neoplasias , Trasplante Heterólogo , Células Tumorales Cultivadas/efectos de los fármacos
8.
Cancer Res ; 49(13): 3689-91, 1989 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-2731182

RESUMEN

Although cytotoxic chemotherapy for human cancer has been reported to induce alterations in intestinal permeability, its effects on the absorptive process are still controversial. We have studied mediated and nonmediated absorption in 10 patients with metastatic breast cancer before and after treatment with Adriamycin by the use of specific test sugars given orally and their subsequent urinary recovery, as measured by chromatography. Mediated absorption was investigated by the use of D-xylose and 3-O-methylglucose, while lactulose and L-rhamnose were used to study nonmediated permeation. Lactulose is considered a marker of unmediated paracellular (tight junction) permeation, while L-rhamnose explores passage across cell membranes. The test was performed on patients before and on the second and the eighth days after Adriamycin administration, and only once in 22 age-matched healthy women. Under basal conditions, as well as 2 and 8 days after chemotherapy, D-xylose and 3-O-methylglucose absorption was 35% lower in patients than in controls (P less than 0.001). Lactulose absorption was significantly higher in patients than in controls under basal conditions (P less than 0.001); it reached levels three times higher the second day after chemotherapy, and returned to basal levels by the eighth day. The data suggest an early reversible effect of Adriamycin on cellular tight junctions with resulting increased permeabilization. This effect seems of a toxic nature rather than due to increased cell loss. It is interesting that both nonmediated absorption and mediated absorption were already altered before chemotherapy in cancer patients, suggesting a preexisting functional damage of the intestine. The significance of this alteration as a potential mechanism of cancer cachexia is discussed.


Asunto(s)
Neoplasias de la Mama/metabolismo , Metabolismo de los Hidratos de Carbono , Doxorrubicina/farmacología , Absorción Intestinal/efectos de los fármacos , 3-O-Metilglucosa , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Femenino , Humanos , Lactulosa/orina , Metilglucósidos/sangre , Metilglucósidos/metabolismo , Ramnosa/metabolismo , Xilosa/sangre , Xilosa/metabolismo
9.
Oncogene ; 9(11): 3233-40, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7936647

RESUMEN

MCF-10A, a nontransformed mammary epithelial cell line, requires a mixture of hormones and growth factors for optimal cell proliferation. In this report we show that when MCF-10A cells are cultured in serum free medium they become quiescent and accumulate in G0/G1 phases of the cell cycle. Following addition of complete medium to quiescent cells, MCF-10A cells enter into the S phase within 15-18 h and resume the cell cycle distribution of proliferating cells within 24 h. Measurement of RI alpha subunit of the cAMP-dependent protein kinase (PKA) shows a 10- to 15-fold increase in protein levels at 6 h following complete medium addition, thus preceding cell entry into the S phase. Retroviral vector-mediated overexpression of RI alpha, but not of RII beta or C alpha subunits of PKA, enables MCF-10A cells to grow in serum-free medium. In addition, RI alpha downregulation by specific antisense oligodeoxynucleotide treatment or following infection with a retroviral vector containing the RI alpha cDNA in antisense orientation determines growth arrest of proliferating MCF-10A cells and is able to partially block S phase entry of quiescent MCF-10A cells following complete medium addition. These results suggest that RI alpha/PKAI is involved in the control of cell cycle progression of mammary epithelial cells at a G1 to S transition border, and that its overexpression is able to overcome serum and growth factors requirement for cell proliferation.


Asunto(s)
Ciclo Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glándulas Mamarias Animales/citología , Animales , Sangre , Línea Celular , Clonación Molecular , Medio de Cultivo Libre de Suero , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/genética , ADN Complementario , Epitelio , Vectores Genéticos , Humanos , Retroviridae/genética
10.
Oncogene ; 14(8): 923-8, 1997 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-9050991

RESUMEN

Functional interactions between protein kinase A (PKA) and epidermal growth factor receptor (EGF-R) signalling pathways have been suggested. Unlike the type II isoform of PKA (PKAII), the type I (PKAI) and/or its regulatory subunit RIalpha are generally overexpressed in cancer cells and are induced following transforming growth factor alpha (TGF alpha)/EGF-R-dependent transformation. Downregulation of RIalpha/PKAI inhibits TGF alpha expression and EGF-R-dependent signalling. We have previously shown that addition of EGF to quiescent human normal epithelial MCF-10A cells determines PKAI expression and cell membrane translocation before cells enter S phase, while PKAI inhibition prevents S phase entry. Constitutive overexpression of PKAI confers the ability to grow in serum free medium, bypassing EGF requirement. Here we demonstrate a direct interaction of PKAI, but not of PKAII, with the activated EGF-R, that occurs within 5 min following EGF treatment of MCF-10A cells. Moreover, induction of mitogen-activated protein kinase (MAPK) activity following EGF-R activation is mimicked by PKAI overexpression and inhibited by downregulators of PKAI. Finally, the PKAI-EGF-R association occurs through the binding of RIalpha to the SH3 domain(s) of Grb2 adaptor protein, thus allowing the recruitment of the PKAI holoenzyme to the activated EGF-R. This is the first demonstration of a direct interaction of PKAI with the activated EGF-R macromolecular signalling complex.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptores ErbB/metabolismo , Proteínas/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteína Quinasa Tipo II Dependiente de AMP Cíclico , Activación Enzimática , Proteína Adaptadora GRB2 , Humanos , Unión Proteica , Transducción de Señal , Células Tumorales Cultivadas , Dominios Homologos src
11.
Cell Death Differ ; 10(2): 218-29, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12700650

RESUMEN

The mechanisms of tumor cell resistance to interferon-alpha (IFNalpha) are at present mostly unsolved. We have previously demonstrated that IFNalpha induces apoptosis on epidermoid cancer cells and EGF antagonizes this effect. We have also found that IFNalpha-induced apoptosis depends upon activation of the NH(2)-terminal Jun kinase-1 (Jnk-1) and p(38) mitogen-activated protein kinase, and that these effects are also antagonized by EGF. At the same time, IFNalpha increases the expression and function of the epidermal growth factor receptor (EGF-R). Here we report that the apoptosis induced by IFNalpha occurs together with activation of caspases 3, 6 and 8 and that EGF also antagonizes this effect. On the basis of these results, we have hypothesized that the increased EGF-R expression and function could represent an inducible survival response that might protect tumor cells from apoptosis caused by IFNalpha via extracellular signal regulated kinase 1 and 2 (Erk-1/2) cascades. We have found an increased activity of Ras and Raf-1 in IFNalpha-treated cells. Moreover, IFNalpha induces a 50% increase of the phosphorylated isoforms and enzymatic activity of Erk-1/2. We have also demonstrated that the inhibition of Ras activity induced by the transfection of the dominant negative Ras plasmid RASN17 and the inhibition of Mek-1 with PD098059 strongly potentiates the apoptosis induced by IFNalpha. Moreover, the selective inhibition of this pathway abrogates the counteracting effect of EGF on the IFNalpha-induced apoptosis. All these findings suggest that epidermoid tumor cells counteract the IFNalpha-induced apoptosis through a survival pathway that involves the hyperactivation of the EGF-dependent Ras->Erk signalling. The selective targeting of this pathway appears to be a promising approach in order to enhance the antitumor activity of IFNalpha.


Asunto(s)
Apoptosis/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Interferón-alfa/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas ras/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Caspasas/metabolismo , Supervivencia Celular , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/efectos de los fármacos , Receptores ErbB/metabolismo , Flavonoides/farmacología , Humanos , Interferón-alfa/metabolismo , Células KB , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/análisis , Proteínas Proto-Oncogénicas c-raf/efectos de los fármacos
12.
Cell Death Differ ; 6(8): 773-80, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10467351

RESUMEN

We have demonstrated that interferon-alpha2-recombinant (IFNalpha) at growth inhibitory concentrations enhances the expression and signalling activity of the epidermal growth factor receptor (EGF-R) in human epidermoid carcinoma KB cells. Here we report that KB cells exposed to IFNalpha underwent apoptotic cell death and this effect was antagonized by EGF. We have also found that IFNalpha enhanced the expression of heat shock proteins (HSP) HSP-70, HSP-90 and HSP-27 and activated the NH2-terminal Jun kinase-1 (JNK-1) and p38 mitogen activated protein kinase, the target enzymes of a stress-dependent intracellular transduction pathway. Moreover, the overexpression of the wild-type JNK-1, obtained through plasmid transfection of KB cells, induced apoptosis which was potentiated by the exposure of wild-type JNK-1 (JNK-1wt)-transfected cells to IFNalpha. All these effects were neutralized by the addition of EGF to parental and JNK-1wt-transfected KB cells exposed to IFNalpha. In conclusion, EGF has a protective effect on KB cells from apoptosis while antagonizing a stress response elicited by IFNalpha and targeted on the stress pathway terminal kinases.


Asunto(s)
Apoptosis , Factor de Crecimiento Epidérmico/metabolismo , Proteínas de Choque Térmico , Interferón-alfa/farmacología , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Proteínas de Choque Térmico HSP27 , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas HSP90 de Choque Térmico/biosíntesis , Humanos , Interferón-alfa/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Chaperonas Moleculares , Proteínas de Neoplasias/biosíntesis , Transfección , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos
13.
J Clin Oncol ; 14(10): 2702-8, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8874330

RESUMEN

PURPOSE: We studied retrospectively the interaction between c-erbB2 overexpression and adjuvant tomoxifen in node-negative breast cancer patients enrolled in the Gruppo Universitario Napoletano 1 (GUN-1) trial. PATIENTS AND METHODS: c-erbB2, evaluated by immunohistochemistry in 145 of 173 patients randomly assigned to 2-year adjuvant tamoxifen or no further therapy, was considered overexpressed if greater than 10% of the cells showed specific membrane staining. The role of each prognostic variable and their independent effect were studied using the Cox model. Disease-free (DFS) and overall (OAS) survival curves were estimated by the Kaplan-Meier method. RESULTS: As of November 30, 1994, the median follow-up period was 12 years. c-erbB2 was overexpressed in 43 of 145 patients (29.7%), which directly correlated with tumor size and inversely with estrogen receptor (ER) level. At univariate analysis, overexpression of c-erbB2 did not affect either DFS or OAS; tamoxifen had a greater effect on reducing the risk of recurrence than of death. Addition of c-erbB2 to a multivariate Cox model that contained menopausal status, tumor size, nuclear grade, and treatment as covariates did not affect the significance of the model for DSF or OAS, whereas addition of the first-order interaction between c-erbB2 and tamoxifen was statistically significant both for DFS and OAS. The same result was obtained when the model contained ER status and ER-tamoxifen interaction. Indeed, adjuvant tamoxifen significantly prolonged DFS and OAS in c-erbB2-negative cases, whereas it had no effect on DFS and OAS in c-erbB2-positive patients. CONCLUSION: In early-stage breast cancer patients, overexpression of c-erbB2 is a marker of lack of efficacy of adjuvant tamoxifen.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama , Proteínas de Neoplasias/metabolismo , Receptor ErbB-2/metabolismo , Tamoxifeno/uso terapéutico , Adulto , Anciano , Axila , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Persona de Mediana Edad
14.
J Clin Oncol ; 16(4): 1414-9, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9552045

RESUMEN

PURPOSE: To evaluate the activity and toxicity of the combination carboplatin plus vinorelbine in extensive small-cell lung cancer (SCLC). PATIENTS AND METHODS: A two-stage optimal Simon design was applied. To proceed after the first stage, responses from 8 of 11 treated patients were needed. Overall, 31 responses of 43 treated patients were required to comply with the design parameters. Inclusion criteria were cytohistologically proven SCLC; extensive disease; age of 70 years or less; Eastern Cooperative Oncology group performance status (ps ECOG) of 2 or less; normal cardiac, hepatic, renal, and bone marrow functions; and no previous chemotherapy. Patients were staged by physical examination; biochemistry; chest radiograph; brain, thoracic; and abdominal computed tomographic (CT) scans, and bone scan. All patients received carboplatin 300 mg/m2 intravenously (i.v.) day 1 and vinorelbine 25 mg/m2 i.v. on days 1 and 8 every 4 weeks up to six cycles. Of 43 enrolled patients, 36 were men and 7 women, with a median age of 63 years (range, 46 to 70 years). RESULTS: All patients were assessable for response and toxicity. We observed 32 (74%) objective responses, with 23% complete responses. Median time to progression was 25 weeks, and median survival was 37 weeks. The treatment was well tolerated. The reported main toxicities were leukopenia grade 3 in 21% of patients and grade 4 in 5% of patients, anemia grade 2 in 11% of patients and grade 3 in 2% of patients, and thrombocytopenia grade 3 in 7% of patients. CONCLUSION: These data show that carboplatin plus vinorelbine is an active and well-tolerated regimen in extensive SCLC. In view of the activity, low toxicity, and ease of administration, it may be a reasonable alternative to more toxic cisplatin-containing regimens.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Anciano , Antineoplásicos Fitogénicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carboplatino/administración & dosificación , Carcinoma de Células Pequeñas/mortalidad , Carcinoma de Células Pequeñas/patología , Femenino , Humanos , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Tasa de Supervivencia , Vinblastina/administración & dosificación , Vinblastina/análogos & derivados , Vinorelbina
15.
Artículo en Inglés | MEDLINE | ID: mdl-15655565

RESUMEN

OBJECTIVES: To determine whether COX-2, bcl-2 and neoangiogenesis are related to human prostate cancer relapse after definitive surgical treatment and progression toward androgen independence and to evaluate the association between the patterns of these tumoral biomarkers and other standard clinico-pathological parameters (such as Gleason score, PSA, TNM stage). MATERIALS AND METHODS: We retrospectively analyzed the records on 126 prostate cancer samples from patients treated at our University Hospital from 1995 to 2002. The 72 patients with clinically localized disease (group 1) had undergone radical prostatectomy. Another 54 patients (group 2) had metastatic androgen-independent disease. Archived material relating to the subjects was then immunostained for bcl-2, COX-2 and CD-31, using an anti-bcl-2 monoclonal primary antibody, an anti-COX-2 polyclonal rabbit antibody and an anti-CD-31 monoclonal mouse antibody to evaluate neoangiogenesis (MVD, microvessel density). RESULTS: We found that bcl-2, COX-2 and MVD expression increased from group 1 to group 2. The intergroup difference was significant only for high MVD (P < 0.05). On the other hand, high MVD, high bcl-2 and high COX-2 expression was correlated with a higher PSA level (P < 0.01), whereas only a high MVD was also related with Gleason score (P < 0.05). We used univariate analysis to evaluate the prognostic impact of biologic and clinico-pathologic parameters on the disease-free-survival of 72 patients treated by radical prostatectomy. A total of 30 patients (41.6%) experienced biochemical relapse; bcl-2, COX-2 and MVD significantly correlated with disease relapse in these patients. In fact, we observed disease relapse in 24/45 (53%) with high bcl-2 expression, in 15/21 (71%) with a high MVD count and finally, in 30/58 (52%) with high COX-2 expression. Finally, PSA value and Gleason score were the only two biologic markers significantly associated to disease relapse in a multivariate analysis. CONCLUSIONS: Our results strongly support a role for bcl-2, COX-2 and angiogenesis in the development and progression of prostate cancer. Of course, we are aware of the small sample size considered in our study. Further investigations would better clarify the prognostic and therapeutic implications of these findings.


Asunto(s)
Perfilación de la Expresión Génica , Recurrencia Local de Neoplasia , Neovascularización Patológica , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Neoplasias de la Próstata/fisiopatología , Neoplasias de la Próstata/cirugía , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ciclooxigenasa 2 , Progresión de la Enfermedad , Humanos , Masculino , Proteínas de la Membrana , Estadificación de Neoplasias , Pronóstico , Antígeno Prostático Específico/sangre , Prostatectomía , Neoplasias de la Próstata/patología , Estudios Retrospectivos
16.
Clin Cancer Res ; 6(6): 2506-12, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10873106

RESUMEN

Protein kinase A type I (PKAI) transduces mitogenic signals from different growth factors and oncogenes and is overexpressed in the majority of human cancers. We and other investigators previously have reported that different PKAI inhibitors, including antisense oligonucleotides, have antitumor activity. In this study, we used a novel hybrid DNA/RNA mixed-backbone oligonucleotide (MBO) targeting the PKAI subunit RIalpha. We demonstrated that after oral administration, the MBO antisense RIalpha inhibited the growth of human colon cancer xenografts in nude mice and showed a cooperative antitumor effect with Taxol, which outlasted treatment withdrawal and significantly prolonged survival of mice compared with untreated controls or to single-agent-treated mice. Immunohistochemical analysis of tumor specimens showed inhibition of target protein RIalpha and of growth factor expression along with a marked inhibition of angiogenesis and an increase in p27 expression. In conclusion, a novel MBO that targets PKAI, administered p.o., is effective and cooperates with the anticancer drug Taxol on both tumor growth and expression of factors involved in the control of cell proliferation, cell cycle, and angiogenesis. Because the MBO described has completed a phase I trial involving i.v. injection in cancer patients, these results provide the biological rationale of its activity after oral administration and may be translated into a therapeutic strategy in a clinical setting.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proteínas de Ciclo Celular , Neoplasias del Colon/tratamiento farmacológico , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Sustancias de Crecimiento/biosíntesis , Neovascularización Patológica , Oligonucleótidos Antisentido/uso terapéutico , Paclitaxel/farmacología , Proteínas Supresoras de Tumor , Administración Oral , Animales , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/biosíntesis , Trasplante de Neoplasias , Factores de Tiempo , Células Tumorales Cultivadas
17.
Clin Cancer Res ; 5(12): 4133-9, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10632351

RESUMEN

Tumor-node-metastasis (TNM) staging is the standard system for the estimation of prognosis of breast cancer patients. However, this system does not exploit information yielded by markers of the biological aggressiveness of breast cancer and is clearly unsatisfactory for optimal-treatment decision-making and for patient counseling. We have developed a prognostic model, based on a few routinely evaluated prognostic variables, that produces quantitative estimates for risk of relapse of individual breast cancer patients. We used data concerning 2441 of 2990 consecutive breast cancer patients to develop an artificial neural network (ANN) for the prediction of the probability of relapse over 5 years. The prognostic variables used were: patient age, tumor size, number of axillary metastases, estrogen and progesterone receptor levels, S-phase fraction, and tumor ploidy. Performances of the model were evaluated in terms of discrimination ability and quantitative precision. Predictions were validated on an independent series of 310 patients from an institution in another country. The ANN discriminated patients according to their risk of relapse better than the TNM classification (P = 0.0015). The quantitative precision of the model's estimates was accurate and was confirmed on the series from the second institution. The 5-year relapse risk yielded by the model varied greatly within the same TNM class, particularly for patients with four or more nodal metastases. The model discriminates prognosis better than the TNM classification and is able to identify patients with strikingly different risks of relapse within each TNM class.


Asunto(s)
Neoplasias de la Mama/patología , Modelos Estadísticos , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Supervivencia sin Enfermedad , Humanos , Persona de Mediana Edad , Modelos Biológicos , Estadificación de Neoplasias , Redes Neurales de la Computación , Valor Predictivo de las Pruebas , Pronóstico , Reproducibilidad de los Resultados , Factores de Riesgo
18.
Clin Cancer Res ; 6(9): 3739-47, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10999768

RESUMEN

Angiogenesis plays a key role in tumor growth and metastasis. The transforming growth factor alpha (TGF-alpha)-epidermal growth factor receptor (EGFR) autocrine pathway controls in part the production of angiogenic factors such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) in cancer cells. In this study, we have evaluated the antiangiogenic and antitumor activity of monoclonal antibody (MAb) C225, an anti-EGFR chimeric human-mouse MAb, alone and in combination with a human VEGF antisense (AS) 21-mer phosphorothioate oligonucleotide (VEGF-AS) in human GEO colon cancer cells. MAb C225 treatment determined a dose-dependent inhibition of VEGF, bFGF, and TGF-alpha production by GEO cells in vitro. Treatment with VEGF-AS caused a selective inhibition in VEGF expression by GEO cells in vitro. Treatment of immunodeficient mice bearing established, palpable GEO xenografts for 3 weeks with VEGF-AS or with MAb C225 determined a cytostatic reversible inhibition of tumor growth. In contrast, a prolonged inhibition of tumor growth was observed in all mice treated with the two agents, in combination with a significant improvement in mice survival compared with controls (P < .001), to MAb C225 (P < .001), or to VEGF-AS (P < .001) treated mice. All mice died within 4, 6, and 8 weeks after tumor cell injection in the control, VEGF-AS and MAb C225 groups, respectively. In contrast, 50% of mice treated with the combination of VEGF-AS and MAb C225 were alive at 13 weeks. Ten % of mice treated with VEGF-AS plus MAb C225 were alive at 20 weeks and had no histological evidence of GEO tumors. Immunohistochemical analysis of GEO tumor xenografts demonstrated a significant reduction of VEGF expression after treatment with VEGF-AS with a parallel reduction in microvessel count. MAb C225 treatment determined a reduction in the expression of VEGF, bFGF, and TGF-alpha with a reduction in microvessel count. Finally, a significant potentiation in inhibition of VEGF expression and little or no microvessels were observed in GEO tumors after the combined treatment with the two agents.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias del Colon/terapia , Factores de Crecimiento Endotelial/genética , Linfocinas/genética , Neovascularización Patológica/terapia , Oligonucleótidos Antisentido/farmacología , Tionucleótidos/farmacología , Animales , Western Blotting , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/metabolismo , Relación Dosis-Respuesta a Droga , Factores de Crecimiento Endotelial/biosíntesis , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Femenino , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Humanos , Inmunohistoquímica , Linfocinas/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/metabolismo , Factor de Crecimiento Transformador alfa/biosíntesis , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Clin Cancer Res ; 6(11): 4343-50, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11106252

RESUMEN

Recent studies have suggested that selective inhibition of mitogenic pathways may improve the antitumor activity of ionizing radiation. The epidermal growth factor receptor (EGFR) is overexpressed and is involved in autocrine growth control in the majority of human carcinomas. Protein kinase A type I (PKAI) plays a key role in neoplastic transformation and is overexpressed in cancer cells in which an EGFR autocrine pathway is activated. We used two specific inhibitors of EGFR and PKAI that are under clinical evaluation in cancer patients: C225, an anti-EGFR chimeric human-mouse monoclonal antibody (MAb); and a mixed-backbone antisense oligonucleotide targeting the PKAI RIalpha subunit (PKAI AS). We tested in human colon cancer (GEO) and ovarian cancer (OVCAR-3) cell lines the antiproliferative activity of MAb C225 and/or PKAI AS in combination with ionizing radiation. In vivo antitumor activity was evaluated in nude mice bearing established GEO xenografts. Dose-dependent inhibition of soft agar growth was observed in both cancer cell lines with ionizing radiation, C225, or PKAI AS oligonucleotide. A cooperative antiproliferative effect was obtained when cancer cells were treated with ionizing radiation followed by MAb C225 or PKAI AS oligonucleotide. This effect was observed at all doses tested in both GEO and OVCAR-3 cancer cell lines. A combination of the three treatments at the lowest doses produced an even greater effect than that observed when two modalities were combined. Treatment of mice bearing established human GEO colon cancer xenografts with radiotherapy (RT), MAb C225, or PKAI AS oligonucleotide produced dose-dependent tumor growth inhibition that was reversible upon treatment cessation. A potentiation of the antitumor activity was observed in all mice treated with RT in combination with MAb C225 or PKAI AS oligonucleotide. Long-term GEO tumor growth regression was obtained following treatment with ionizing radiation in combination with MAb C225 plus PKAI AS oligonucleotide, which produced a significant improvement in survival compared with controls (P < 0.001), the RT-treated group (P < 0.001), or the group treated with MAb C225 plus PKAI AS oligonucleotide (P < 0.001). All mice of the RT + MAb C225 + PKAI AS group were alive 26 weeks after tumor cell injection. Furthermore, 50% of mice in this group were alive and tumor-free after 35 weeks. This study provides a rationale for evaluating in cancer patients the combination of ionizing radiation and selective drugs that block EGFR and PKAI pathways.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Receptores ErbB/antagonistas & inhibidores , Neoplasias/terapia , Oligonucleótidos Antisentido/uso terapéutico , Animales , Terapia Combinada , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Células Tumorales Cultivadas
20.
Clin Cancer Res ; 1(2): 161-7, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9815969

RESUMEN

8-Chloro-cyclic AMP (8-Cl-cAMP), a site-selective cAMP analogue, is a specific inhibitor of type I cAMP-dependent protein kinase (PKAI) and induces growth inhibition in several human and rodent tumor cell lines. The anti-epidermal growth factor receptor (EGFR) mAb 528 is a blocking antibody able to inhibit the in vitro and in vivo growth of several human cancer cell lines that express functional EGFRs. Since enhanced levels of PKAI are generally found in tumor cells and an increase in PKAI expression is induced by transformation through a transforming growth factor alpha/EGFR autocrine pathway, we have evaluated whether treatment with mAb 528 in combination with 8-Cl-cAMP may have an additive or synergistic growth inhibitory effect on human cancer cells. A dose-dependent inhibition of monolayer cell growth was observed in two human colon cancer cell lines (GEO and CBS) and in a human breast cancer cell line (MDA-468) by treatment with either mAb 528 or 8-Cl-cAMP with 50% inhibitory concentration of 2-10 microgram/ml or 20-25 micrometer, respectively. The combined treatment with low noninhibitory doses of mAb 528 (0.25 microgram/ml) and with 8-Cl-cAMP had a more than additive growth inhibitory effect with a 3- to 5-fold reduction in the 8-Cl-cAMP 50% inhibitory concentration in all cell lines tested. This combined treatment was similarly effective in inhibiting the soft agar cloning efficiency of GEO cells. 8-Cl-cAMP treatment of GEO cells induced a dose-dependent increase in cell membrane-associated EGFRs with a maximum 3- to 4-fold increase within 48-72 h of treatment. These results suggest that a double blockade of the PKAI serine-threonine kinase-dependent and of the EGFR tyrosine kinase-dependent pathways is potentially useful in cancer therapy.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Anticuerpos Monoclonales/toxicidad , Antineoplásicos/toxicidad , Neoplasias de la Mama/patología , Ciclo Celular/fisiología , Fragmentación del ADN/fisiología , Receptores ErbB/inmunología , 8-Bromo Monofosfato de Adenosina Cíclica/toxicidad , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/análisis , Receptores ErbB/fisiología , Femenino , Humanos , Cinética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda