Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Oncotarget ; 8(23): 37061-37079, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28427157

RESUMEN

Ovarian cancer is the leading cause of death in women with gynecological cancers and despite recent advances, new and more efficient therapies are crucially needed. Müllerian Inhibiting Substance type II Receptor (MISRII, also named AMHRII) is expressed in most ovarian cancer subtypes and is a novel potential target for ovarian cancer immunotherapy. We previously developed and tested 12G4, the first murine monoclonal antibody (MAb) against human MISRII. Here, we report the humanization, affinity maturation and glyco-engineering steps of 12G4 to generate the Fc-optimized 3C23K MAb, and the evaluation of its in vivo anti-tumor activity. The epitopes of 3C23K and 12G4 were strictly identical and 3C23K affinity for MISRII was enhanced by a factor of about 14 (KD = 5.5 × 10-11 M vs 7.9 × 10-10 M), while the use of the EMABling® platform allowed the production of a low-fucosylated 3C23K antibody with a 30-fold KD improvement of its affinity to FcγRIIIa. In COV434-MISRII tumor-bearing mice, 3C23K reduced tumor growth more efficiently than 12G4 and its combination with carboplatin was more efficient than each monotherapy with a mean tumor size of 500, 1100 and 100 mm3 at the end of treatment with 3C23K (10 mg/kg, Q3-4D12), carboplatin (60 mg/kg, Q7D4) and 3C23K+carboplatin, respectively. Conversely, 3C23K-FcKO, a 3C23K form without affinity for the FcγRIIIa receptor, did not display any anti-tumor effect in vivo. These results strongly suggested that 3C23K mechanisms of action are mainly Fc-related. In vitro, antibody-dependent cytotoxicity (ADCC) and antibody-dependent cell phagocytosis (ADCP) were induced by 3C23K, as demonstrated with human effector cells. Using human NK cells, 50% of the maximal lysis was obtained with a 46-fold lower concentration of low-fucosylated 3C23K (2.9 ng/ml) than of 3C23K expressed in CHO cells (133.35 ng/ml). As 3C23K induced strong ADCC with human PBMC but almost none with murine PBMC, antibody-dependent cell phagocytosis (ADCP) was then investigated. 3C23K-dependent (100 ng/ml) ADCP was more active with murine than human macrophages (only 10% of living target cells vs. about 25%). These in vitro results suggest that the reduced ADCC with murine effectors could be partially balanced by ADCP activity in in vivo experiments. Taken together, these preclinical data indicate that 3C23K is a new promising therapeutic candidate for ovarian cancer immunotherapy and justify its recent introduction in a phase I clinical trial.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Receptores de Péptidos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Femenino , Glicosilación , Humanos , Ratones Desnudos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Ingeniería de Proteínas
2.
MAbs ; 6(5): 1314-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25517316

RESUMEN

Ovarian cancer has the highest mortality rate among gynecologic malignancies. The monoclonal antibody 12G4 specifically recognizes the human Müllerian inhibiting substance type II receptor (MISRII) that is strongly expressed in human granulosa cell tumors (GCT) and in the majority of human epithelial ovarian cancers (EOC). To determine whether MISRII represents an attractive target for antibody-based tumor therapy, we first confirmed by immunohistochemistry with 12G4 its expression in all tested GCT samples (4/4) and all, but one, EOC human tissue specimens (13/14). We then demonstrated in vitro the internalization of 12G4 in MISRII(high)COV434 cells after binding to MISRII and its ability to increase the apoptosis rate (FACS, DNA fragmentation) in MISRII(high)COV434 (GCT) and MISRII(medium)NIH-OVCAR-3 (EOC) cells that express different levels of MISRII. A standard (51)Cr release assay showed that 12G4 mediates antibody-dependent cell-meditated cytotoxicity. Finally, in vivo assessment of 12G4 anti-tumor effects showed a significant reduction of tumor growth and an increase of the median survival time in mice xenografted with MISRII(high)COV434 or MISRII(medium)NIH-OVCAR-3 cells and treated with 12G4 in comparison to controls treated with an irrelevant antibody. Altogether, our data indicate that MISRII is a new promising target for the control of ovarian GCTs and EOCs. A humanized version of the 12G4 antibody, named 3C23K, is in development for the targeted therapy of MISRII-positive gynecologic cancers.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Neoplasias Ováricas/inmunología , Receptores de Péptidos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Femenino , Tumor de Células de la Granulosa/inmunología , Tumor de Células de la Granulosa/metabolismo , Tumor de Células de la Granulosa/terapia , Humanos , Inmunohistoquímica , Inmunoterapia/métodos , Ratones Desnudos , Microscopía Fluorescente , Neoplasias Glandulares y Epiteliales/inmunología , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/terapia , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Receptores de Péptidos/antagonistas & inhibidores , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Nucl Med Biol ; 40(4): 471-80, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23522976

RESUMEN

INTRODUCTION: (125)I-labeled monoclonal antibodies ((125)I-mAbs) can efficiently treat small solid tumors. Here, we investigated the role of apoptosis, autophagy and mitotic catastrophe in (125)I-mAb toxicity in p53(-/-) and p53(+/+) cancer cells. METHODS: We exposed p53(-/-) and p53(+/+) HCT116 cells to increasing activities of internalizing (cytoplasmic location) anti-HER1 (125)I-mAbs, or non-internalizing (cell surface location) anti-CEA (125)I-mAbs. For each targeting model we established the relationship between survival and mean nucleus absorbed dose using the MIRD formalism. RESULTS: In both p53(-/-) and p53(+/+) HCT116 cells, anti-CEA (125)I-mAbs were more cytotoxic per Gy than anti-HER1 (125)I-mAbs. Sensitivity to anti-CEA (125)I-mAbs was p53-independent, while sensitivity to anti-HER1 (125)I-mAbs was higher in p53(-/-) HCT 116 cells, suggesting that they act through different signaling pathways. Apoptosis was only induced in p53(+/+) HCT116 cells and could not explain cell membrane radiation sensitivity. Inhibition of autophagy did not modify the cell response to (125)I-mAbs. By contrast, mitotic death was similarly induced in both p53(-/-) and p53(+/+) HCT116 cells by the two types of (125)I-mAbs. We also showed using medium transfer experiments that γ-H2AX foci were produced in bystander cells. CONCLUSION: Cell membrane sensitivity to (125)I-mAbs is not mediated by apoptosis and is p53-independent. Bystander effects-mediated mitotic death could be involved in the efficacy of (125)I-mAbs binding cell surface receptors.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Terapia Molecular Dirigida , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Efecto Espectador/efectos de la radiación , Membrana Celular/efectos de la radiación , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Citoplasma/efectos de la radiación , Receptores ErbB/metabolismo , Células HCT116 , Humanos , Radioisótopos de Yodo/uso terapéutico , Mitosis/efectos de los fármacos , Mitosis/efectos de la radiación , Transporte de Proteínas/efectos de la radiación , Radioinmunoterapia , Proteína p53 Supresora de Tumor/metabolismo
4.
J Immunol Methods ; 387(1-2): 11-20, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22910001

RESUMEN

Prescription of therapeutic antibodies has radically modified the prognosis of some important diseases. However, the very high cost of these new drugs is a problem for public health organizations, which require assessment of the effectiveness of the antibody for each patient before beginning or during the treatment. In vivo immunoimaging is particularly well adapted to meet this demand. However, full-length antibodies are unsuitable for in vivo imaging due to their persistence in the serum and must be engineered in smaller formats to improve their pharmacokinetic properties without modifying their affinity and specificity. The small bivalent antibody fragment called diabody perfectly meets these in vivo imaging requirements. However, obtaining diabodies is laborious, time-consuming and sometimes unsuccessful. Using a diabody derived from a monoclonal antibody (12G4) directed against the human anti-Müllerian hormone receptor, a biomarker of ovarian cancers for which therapeutic antibodies are already undergoing clinical trials, we describe here a new diabody refolding protocol with various reducing conditions. Diabody functionality was checked in vitro and ex vivo with, respectively, a new immunoassay involving the epitopic peptide as a tracer and flow cytometry experiments with cells expressing recombinant anti-Müllerian hormone receptors. Our optimized protocol allows us to find the best refolding conditions for each diabody and to obtain large amounts of functional diabodies.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/inmunología , Neoplasias Ováricas/inmunología , Receptores de Péptidos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Secuencia de Aminoácidos , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Especificidad de Anticuerpos/inmunología , Secuencia de Bases , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática/métodos , Epítopos/inmunología , Epítopos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Citometría de Flujo , Expresión Génica , Humanos , Cuerpos de Inclusión/metabolismo , Datos de Secuencia Molecular , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/terapia , Unión Proteica/inmunología , Replegamiento Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo
5.
J Biol Chem ; 282(39): 28328-28334, 2007 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-17631492

RESUMEN

ICI182,780 (Fulvestrant) is a pure anti-estrogen used in adjuvant therapies of breast cancer. This compound not only inhibits the transcriptional activities of the estrogen receptor-alpha (ER alpha) but also induces its proteasome-dependent degradation. The latter activity is believed to be required for the antiproliferative effects of ICI182,780. Estrogen receptor-related receptor-alpha (ERR alpha) is an orphan member of the nuclear receptor superfamily that is expressed in a wide range of tissues including breast tumors, in which its high expression correlates with poor prognosis. Although not regulated by any natural ligand, ERR alpha can be deactivated by the synthetic molecule XCT790. Here we demonstrate that this compound also induces a proteasome degradation of ERR alpha. We also show that although it does not act directly on the steady-state level of ER alpha, XCT790 potentiates the ICI182,780-induced ER alpha degradation. We suggest that treatment with XCT790 could thus enhance the efficacy of ICI182,780 in ER alpha-dependent pathologies such as breast cancer.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/metabolismo , Estradiol/análogos & derivados , Nitrilos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Tiazoles/farmacología , Antineoplásicos Hormonales/agonistas , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Sinergismo Farmacológico , Estradiol/agonistas , Estradiol/farmacología , Estradiol/uso terapéutico , Receptor alfa de Estrógeno/agonistas , Femenino , Fulvestrant , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Nitrilos/agonistas , Nitrilos/uso terapéutico , Pronóstico , Receptores de Estrógenos/agonistas , Tiazoles/agonistas , Tiazoles/uso terapéutico , Receptor Relacionado con Estrógeno ERRalfa
6.
Nat Cell Biol ; 9(4): 470-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17369819

RESUMEN

Aberrant constitutive expression of c-Rel, p65 and p50 NF-kappaB subunits has been reported in over 90% of breast cancers. Recently, we characterized a de novo RelB NF-kappaB subunit synthesis pathway, induced by the cytomegalovirus (CMV) IE1 protein, in which binding of p50-p65 NF-kappaB and c-Jun-Fra-2 AP-1 complexes to the RELB promoter work in synergy to potently activate transcription. Although RelB complexes were observed in mouse mammary tumours induced by either ectopic c-Rel expression or carcinogen exposure, little is known about RelB in human breast disease. Here, we demonstrate constitutive de novo RelB synthesis is selectively active in invasive oestrogen receptor alpha (ERalpha)-negative breast cancer cells. ERalpha signalling reduced levels of functional NF-kappaB and Fra-2 AP-1 and inhibited de novo RelB synthesis, leading to an inverse correlation between RELB and ERalpha gene expression in human breast cancer tissues and cell lines. Induction of Bcl-2 by RelB promoted the more invasive phenotype of ERalpha-negative cancer cells. Thus, inhibition of de novo RelB synthesis represents a new mechanism whereby ERalpha controls epithelial to mesenchymal transition (EMT).


Asunto(s)
Estrógenos/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transducción de Señal/fisiología , Factor de Transcripción ReIB/genética , Animales , Northern Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Transformación Celular Neoplásica/genética , Ensayo de Cambio de Movilidad Electroforética , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/fisiología , Femenino , Antígeno 2 Relacionado con Fos/genética , Antígeno 2 Relacionado con Fos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Immunoblotting , Ratones , Microscopía Fluorescente , FN-kappa B/genética , FN-kappa B/metabolismo , Fenotipo , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción ReIB/metabolismo
7.
Oncogene ; 24(8): 1434-44, 2005 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-15608675

RESUMEN

Breast cancer progression is likely a multistep process involving the activation and inactivation of a number of genes. Previously, we showed that the mRNA coding for Fra-1, a FOS family member and an AP-1 transcription factor component, was highly expressed in the more invasive estrogen receptor negative (ER-) breast cancer cell lines. We used a tet-off system to stably overexpress Fra-1 in MCF7 ER+ cells and evaluate the impact of Fra-1 on this aggressive phenotype. Conversely, Fra-1 was silenced in highly invasive ER-MDA-MB231 cells using RNA interference. We report that in both systems the Fra-1 expression level was positively associated with cell proliferation, cell motility and invasiveness assessed in vitro. In addition, Fra-1 inhibition in fibroblastoid ER- cells, which formed colonies with large stellate projections in Matrigel, resulted in morphological changes. Cells acquired an epithelioid shape and had a spherical appearance in Matrigel. Fra-1 regulated several genes, implicated in invasion, angiogenesis and cell proliferation independently of beta1-integrin activation, and directly induced MMP-1 and MMP-9 promoter activity. These overall results show that high Fra-1 expression is associated with a more malignant cell phenotype and suggest that Fra-1 could have a pivotal role in breast cancer progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colágeno/metabolismo , Doxiciclina/farmacología , Combinación de Medicamentos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Integrina beta1/fisiología , Laminina/metabolismo , Metaloproteinasa 1 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Invasividad Neoplásica/genética , Proteoglicanos/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/fisiología , ARN Interferente Pequeño/genética , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Factor de Transcripción AP-1/fisiología , Factor A de Crecimiento Endotelial Vascular/biosíntesis
8.
Oncogene ; 23(36): 6105-14, 2004 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-15208657

RESUMEN

We have recently reported that interleukin-8 (IL-8) expression was inversely correlated to estrogen receptor (ER) status and was overexpressed in invasive breast cancer cells. In the present study, we show that IL-8 overexpression in breast cancer cells involves a higher transcriptional activity of IL-8 gene promoter. Cloning of IL-8 promoter from MDA-MB-231 and MCF-7 cells expressing high and low levels of IL-8, respectively, shows the integrity of the promoter in both cell lines. Deletion and site-directed mutagenesis of the promoter demonstrate that NF-kappaB and AP-1 and to a lesser extent C/EBP binding sites play a crucial role in the control of IL-8 promoter activity in MDA-MB-231 cells. Knockdown of NF-kappaB and AP-1 activities by adenovirus-mediated expression of an NF-kappaB super-repressor and RNA interference, respectively, decreased IL-8 expression in MDA-MB-231 cells. On the contrary, restoration of Fra-1, Fra-2, c-Jun, p50, p65, C/EBPalpha and C/EBPbeta expression levels in MCF-7 cells led to a promoter activity comparable to that observed in MDA-MB-231 cells. Our data constitute the first extensive study of IL-8 gene overexpression in breast cancer cells and suggest that the high expression of IL-8 in invasive cancer cells requires a complex cooperation between NF-kappaB, AP-1 and C/EBP transcription factors.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Interleucina-8/genética , Sitios de Unión , Neoplasias de la Mama/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Interleucina-8/biosíntesis , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción AP-1/metabolismo , Transcripción Genética , Activación Transcripcional , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA