Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
ESMO Open ; 7(5): 100585, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36156447

RESUMEN

BACKGROUND: The detection of homologous recombination deficiency (HRD) can identify patients who are more responsive to platinum and poly ADP ribose polymerase inhibitors (PARPi). MyChoice CDx (Myriad) is the most used HRD test in ovarian cancer (OC). However, some limitations of commercial tests exist, because of the high rate of inconclusive results, costs, and the impossibility of evaluating functional resistance mechanisms. PATIENTS AND METHODS: Two academic genomic tests and a functional assay, the RAD51 foci, were evaluated to detect HRD. One hundred patients with high-grade OC enrolled in the MITO16A/MaNGO-OV2 trial and treated with first-line therapy with carboplatin, paclitaxel, and bevacizumab were analyzed. RESULTS: The failure rate of the two genomic assays was 2%. The sensitivity in detecting HRD when compared with Myriad was 98.1% and 90.6%, respectively. The agreement rate with Myriad was 0.92 and 0.87, with a Cohen's κ coefficient corresponding to 0.84 and 0.74, respectively. For the RAD51 foci assay, the failure rate was 30%. When the test was successful, discordant results for deficient and proficient tumors were observed, and additional HRD patients were identified compared to Myriad; sensitivity was 82.9%, agreement rate was 0.65, and Cohen's κ coefficient was 0.18. The HRD detected by genomic assays and residual tumor at primary surgery and stage was correlated with progression-free survival at multivariate analysis. CONCLUSIONS: Results suggest the feasibility of academic tests for assessing HRD status that show robust concordance with Myriad and correlation with clinical outcome. The contribution of the functional information related to the RAD51 foci test to the genomic data needs further investigation.


Asunto(s)
Mangifera , Neoplasias Ováricas , Femenino , Humanos , Bevacizumab/uso terapéutico , Carboplatino/uso terapéutico , Recombinación Homóloga , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Paclitaxel/uso terapéutico , Platino (Metal)/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/uso terapéutico
2.
Eur J Pharm Biopharm ; 154: 246-258, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32682943

RESUMEN

In this work, we evaluated, for the first time, the antitumor effect of cannabidiol (CBD) as monotherapy and in combination with conventional chemotherapeutics in ovarian cancer and developed PLGA-microparticles as CBD carriers to optimize its anticancer activity. Spherical microparticles, with a mean particle size around 25 µm and high entrapment efficiency were obtained. Microparticles elaborated with a CBD:polymer ratio of 10:100 were selected due to the most suitable release profile with a zero-order CBD release (14.13 ± 0.17 µg/day/10 mg Mps) for 40 days. The single administration of this formulation showed an in vitro extended antitumor activity for at least 10 days and an in ovo antitumor efficacy comparable to that of CBD in solution after daily topical administration (≈1.5-fold reduction in tumor growth vs control). The use of CBD in combination with paclitaxel (PTX) was really effective. The best treatment schedule was the pre + co-administration of CBD (10 µM) with PTX. Using this protocol, the single administration of microparticles was even more effective than the daily administration of CBD in solution, achieving a ≈10- and 8- fold reduction in PTX IC50 respectively. This protocol was also effective in ovo. While PTX conducted to a 1.5-fold tumor growth inhibition, its combination with both CBD in solution (daily administered) and 10-Mps (single administration) showed a 2-fold decrease. These results show the promising potential of CBD-Mps administered in combination with PTX for ovarian cancer treatment, since it would allow to reduce the administered dose of this antineoplastic drug maintaining the same efficacy and, as a consequence, reducing PTX adverse effects.


Asunto(s)
Antineoplásicos Fitogénicos/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Cannabidiol/metabolismo , Microesferas , Neoplasias Ováricas/metabolismo , Paclitaxel/metabolismo , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Cannabidiol/administración & dosificación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Embrión de Pollo , Relación Dosis-Respuesta a Droga , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/metabolismo , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/metabolismo
3.
Biochem Pharmacol ; 144: 52-62, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28782526

RESUMEN

Trabectedin and its analogue lurbinectedin are effective drugs used in the treatment of ovarian cancer. Since the presence of ascites is a frequent event in advanced ovarian cancer we asked the question whether ascites could modify the activity of these compounds against ovarian cancer cells. The cytotoxicity induced by trabectedin or lurbinectedin against A2780, OVCAR-5 cell lines or primary culture of human ovarian cancer cells was compared by performing treatment in regular medium or in ascites taken from either nude mice or ovarian cancer patients. Ascites completely abolished the activity of lurbinectedin at up to 10nM (in regular medium corresponds to the IC90), strongly reduced that of trabectedin, inhibited the cellular uptake of lurbinectedin and, to a lesser extent, that of trabectedin. Since α1-acid glycoprotein (AGP) is present in ascites at relatively high concentrations, we tested if the binding of the drugs to this protein could be responsible for the reduction of their activity. Adding AGP to the medium at concentration range of those found in ascites, we reproduced the anticytotoxic effect of ascites. Erythromycin partially restored the activity of the drugs, presumably by displacing them from AGP. Equilibrium dialysis experiments showed that both drugs bind AGP, but the affinity of binding of lurbinectedin was much greater than that of trabectedin. KD values are 8±1.7 and 87±14nM for lurbinectedin and trabectedin, respectively. The studies intimate the possibility that AGP present in ascites might reduce the activity of lurbinectedin and to a lesser extent of trabectedin against ovarian cancer cells present in ascites. AGP plasma levels could influence the distribution of these drugs and thus they should be monitored in patients receiving these compounds.


Asunto(s)
Ascitis/metabolismo , Carbolinas/metabolismo , Dioxoles/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/metabolismo , Orosomucoide/metabolismo , Neoplasias Ováricas/metabolismo , Tetrahidroisoquinolinas/metabolismo , Animales , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Carbolinas/farmacología , Carbolinas/uso terapéutico , Línea Celular Tumoral , Dioxoles/farmacología , Dioxoles/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/cirugía , Unión Proteica/fisiología , Tetrahidroisoquinolinas/farmacología , Tetrahidroisoquinolinas/uso terapéutico , Trabectedina , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
Br J Cancer ; 110(2): 330-40, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24281000

RESUMEN

BACKGROUND: Aberrant choline metabolism has been proposed as a novel cancer hallmark. We recently showed that epithelial ovarian cancer (EOC) possesses an altered MRS-choline profile, characterised by increased phosphocholine (PCho) content to which mainly contribute over-expression and activation of choline kinase-alpha (ChoK-alpha). METHODS: To assess its biological relevance, ChoK-alpha expression was downmodulated by transient RNA interference in EOC in vitro models. Gene expression profiling by microarray analysis and functional analysis was performed to identify the pathway/functions perturbed in ChoK-alpha-silenced cells, then validated by in vitro experiments. RESULTS: In silenced cells, compared with control, we observed: (I) a significant reduction of both CHKA transcript and ChoK-alpha protein expression; (II) a dramatic, proportional drop in PCho content ranging from 60 to 71%, as revealed by (1)H-magnetic spectroscopy analysis; (III) a 35-36% of cell growth inhibition, with no evidences of apoptosis or modification of the main cellular survival signalling pathways; (IV) 476 differentially expressed genes, including genes related to lipid metabolism. Ingenuity pathway analysis identified cellular functions related to cell death and cellular proliferation and movement as the most perturbed. Accordingly, CHKA-silenced cells displayed a significant delay in wound repair, a reduced migration and invasion capability were also observed. Furthermore, although CHKA silencing did not directly induce cell death, a significant increase of sensitivity to platinum, paclitaxel and doxorubicin was observed even in a drug-resistant context. CONCLUSION: We showed for the first time in EOC that CHKA downregulation significantly decreased the aggressive EOC cell behaviour also affecting cells' sensitivity to drug treatment. These observations open the way to further analysis for ChoK-alpha validation as a new EOC therapeutic target to be used alone or in combination with conventional drugs.


Asunto(s)
Colina Quinasa/genética , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/enzimología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/enzimología , Carcinoma Epitelial de Ovario , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Colina/genética , Colina/metabolismo , Colina Quinasa/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Doxorrubicina/farmacología , Femenino , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Terapia Molecular Dirigida , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Paclitaxel/farmacología , Fosforilcolina/metabolismo , Platino (Metal)/farmacología , Interferencia de ARN/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transcriptoma
5.
Br J Cancer ; 110(3): 625-35, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24335926

RESUMEN

BACKGROUND: Limited knowledge is available on alterations induced by cytostatic drugs on magnetic resonance spectroscopy (MRS) and imaging (MRI) parameters of human cancers, in absence of apoptosis or cytotoxicity. We here investigated the effects of a cytostatic cisplatin (CDDP) treatment on (1)H MRS and MRI of HER2-overexpressing epithelial ovarian cancer (EOC) cells and in vivo xenografts. METHODS: High-resolution MRS analyses were performed on in vivo passaged SKOV3.ip cells and cell/tissue extracts (16.4 or 9.4 T). In vivo MRI/MRS quantitative analyses (4.7 T) were conducted on xenografts obtained by subcutaneous implantation of SKOV3.ip cells in SCID mice. The apparent diffusion coefficient (ADC) and metabolite levels were measured. RESULTS: CDDP-induced cytostatic effects were associated with a metabolic shift of cancer cells towards accumulation of MRS-detected neutral lipids, whereas the total choline profile failed to be perturbed in both cultured cells and xenografts. In vivo MRI examinations showed delayed tumour growth in the CDDP-treated group, associated with early reduction of the ADC mean value. CONCLUSION: This study provides an integrated set of information on cancer metabolism and physiology for monitoring the response of an EOC model to a cytostatic chemotherapy, as a basis for improving the interpretation of non-invasive MR examinations of EOC patients.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Espectroscopía de Resonancia Magnética/métodos , Neoplasias Ováricas/tratamiento farmacológico , Receptor ErbB-2/genética , Animales , Línea Celular Tumoral , Cisplatino/administración & dosificación , Citostáticos/administración & dosificación , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Radiografía , Receptor ErbB-2/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Biomed Mater Res B Appl Biomater ; 80(2): 297-303, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16767724

RESUMEN

This study evaluated the microtensile bond strength and the interfacial morphology of newer adhesives. The occlusal surfaces of extracted teeth were ground flat for random allocation to four equal groups. Resin composite was bonded to each surface using either Clearfil SE Bond [SEB], Clearfil Protect Bond [PB], G-Bond [GB], or an experimental adhesive, SSB-200 [SSB]. After storage for 24 h in water at 37 degrees C, they were sectioned into beams (cross-sectional area 1 mm(2)) for microtensile bond strength testing (muTBS) at a crosshead speed of 1 mm/min. The load at failure of each was recorded; the data were analyzed by one-way ANOVA and Games Howell tests. The surfaces of the fractured specimens were observed using SEM. For the ultra-morphology of the interface, the occlusal surfaces of four more teeth were prepared as before and a thin layer of flowable resin composite was bonded to each surface using one of the four adhesives. The mean muTBS ranged from 39.68 MPa (GB) to 64.97 MPa (SEB). There were no statistical differences between SEB and SSB, or between PB and GB (p > 0.05). The muTBS of SEB and SSB were significantly greater than that of PB and GB (p < 0.05). SEMs of the fractured surfaces revealed a mixed (cohesive/interfacial) failure. TEM examination highlighted differences in the hybrid layer; SEB had a thicker layer than the others. In conclusion, the newer all-in-one adhesives produced a thin hybrid layer but varied in their bond strengths. The 2-step self-etching adhesives do not necessarily produce higher bond strengths than that of the all-in-one systems.


Asunto(s)
Recubrimientos Dentinarios , Resinas Compuestas , Dentina/ultraestructura , Humanos , Técnicas In Vitro , Ensayo de Materiales , Metacrilatos , Microscopía Electrónica , Microscopía Electrónica de Rastreo , Cementos de Resina , Propiedades de Superficie , Resistencia a la Tracción
7.
Gene Ther ; 10(12): 1018-25, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12776159

RESUMEN

The alpha-folate receptor (FR) is selectively overexpressed in 90% of nonmucinous ovarian carcinomas, whereas no expression is detectable in normal ovarian surface epithelium (OSE). Indirect evidence suggests that FR expression is associated with tumor progression and affects cell proliferation. To evaluate better the role of FR, we developed an approach based on intracellular expression of single-chain (sc) antibodies (intrabody) to downmodulate membrane expression of FR in ovary cancer cells. IGROV-1 and SKOV3 ovarian carcinoma cell lines were transfected with an anti-FR intrabody. Transfectants and parental cells were tested for FR, integrins and anti-FR intrabody expression by fluorescence-activated cell sorting (FACS), reverse transcription and polymerase chain reaction (RT-PCR) and/or immunoblotting. Cell growth characteristics and adhesion properties were evaluated in liquid, semisolid and organotypic cultures. The anti-FR scFv inhibited FR expression from 60 to 99%. At physiological concentrations of folate, proliferation varied directly as a function of FR expression. FR downmodulation was accompanied by reduced colony-forming ability in soft agar, morphological change of the cells, significant enhanced adhesion to laminin or Matrigel, a two- to three-fold increase in alpha6beta4 integrin expression, and a marked reduction in laminin production. In three-dimensional organotypic cultures, anti-FR intrabody-transfected IGROV1 cells grew as a single-ordered layer, reminiscent of normal OSE growth in vivo. In conclusion, the anti-FR intrabody reverses the transformed phenotype in ovary cancer cells and may provide an efficient means to inhibit selectively the growth of these cells.


Asunto(s)
Anticuerpos Monoclonales/genética , Proteínas Portadoras/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Ováricas/metabolismo , Anticuerpos Monoclonales/metabolismo , Proteínas Portadoras/metabolismo , Adhesión Celular , División Celular , Transformación Celular Neoplásica , Regulación hacia Abajo , Femenino , Receptores de Folato Anclados a GPI , Ácido Fólico/metabolismo , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Humanos , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/patología , Fenotipo , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Transfección , Células Tumorales Cultivadas
9.
Cytokine ; 15(1): 27-38, 2001 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-11509006

RESUMEN

To study the role of cytokines that are relevant in cancer cachexia syndrome due to intracerebral tumours, mice were injected with human A431 epidermoid carcinoma, OVCAR3 ovarian carcinoma and GBLF glioma cells comparing intracerebral (i.c.) and systemic (i.p. or s.c.) routes of implantation. Anorexia and weight loss developed within 7-10 days in mice injected i.c. with A431 or OVCAR3 cells well before a large tumour developed, while i.c.-injected GBLF cells did not induce cachexia until day 20, when the tumour was large. By contrast, mice injected i.p. or s.c. developed tumours without evidence of anorexia. Thus, intracerebrally-growing A431 and OVCAR3 resulted in cancer cachexia independent of tumour mass, and we investigated their cytokine pattern. Serum levels of murine and human cytokines are not predictive of cancer cachexia development. Reverse-transcriptase polymerase chain reaction (RT-PCR) analysis revealed in the brain of i.c.-injected A431 tumour-bearing mice expression of human interleukin-(IL-)1alpha, IL-1beta and LIF in all samples and IL-6 in two of four samples while in i.c.-injected OVCAR3 tumour-bearing animals IL-6, and LIF were detected in all samples and tumour necrosis factor-alpha (TNFalpha) in two of four samples. Only LIF was expressed in brains of mice injected with GBLF cells. Murine IL-6 was increased only in the brains of A431-bearing mice. Only mice injected i.c. simultaneously with a monoclonal antibody (mAb) directed against the murine IL-6 receptor and OVCAR3 cells, but not those with mAb and A431 cells, showed a significant increase in survival time with a partial and temporary attenuation of cachexia symptoms. These results suggest that IL-6 in OVCAR3 model may be important cachectogenic factor when centrally released by even a limited number of tumour cells.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Caquexia/metabolismo , Citocinas/fisiología , Neoplasias/metabolismo , Animales , Anorexia/metabolismo , Peso Corporal , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Conducta Alimentaria , Femenino , Inhibidores de Crecimiento/sangre , Inhibidores de Crecimiento/metabolismo , Humanos , Interleucina-1/sangre , Interleucina-1/metabolismo , Interleucina-6/sangre , Interleucina-6/metabolismo , Factor Inhibidor de Leucemia , Linfocinas/sangre , Linfocinas/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Células Tumorales Cultivadas
10.
Cancer Immunol Immunother ; 49(12): 679-86, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11258794

RESUMEN

The HER-2 antigen, which is overexpressed in many breast carcinomas, is an ideal target for monoclonal antibodies due to its low expression in normal tissue and its homogeneous distribution in the tumor mass. We have developed and characterized the murine MAb MGR6 against HER-2, which is able to inhibit proliferation of tumor cells overexpressing HER-2. On the basis of these preclinical results, phase I studies in breast carcinoma patients were conducted and radiolocalization data indicated an antibody half life which directly paralleled that of other whole antibodies and thus resulting in a limited in vivo diagnostic capacity. To obtain a smaller reagent with possibly improved in vivo properties, a single chain variable fragment (scFv) of the original MGR6-producing hybridoma was generated by phage display technology. Biologically active MGR6 scFv was purified rapidly and at high yield by metal affinity chromatography. Competition FACS and ELISA analyses identified an epitope on the HER-2 extracellular domain that was shared by the scFv and the parental MAb. BlAcore analysis indicated a Koff of 9.3 x 10(-4) s(-1), similar to that of the intact MGR6 MAb. Distribution and elimination half-lives of MGR6 scFv, calculated from in vivo preclinical evaluations, were much faster (13 min and 6.2 h, respectively) than previously published results for the intact MAb (mean t1/2beta of 46 h). This represents a theoretical improvement in pharmacokinetics with respect to the parental murine MAb and points to the potential for utilizing this fragment in redirecting therapeutic agents, such as radioisotopes, to different human carcinomas overexpressing HER-2.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Región Variable de Inmunoglobulina/inmunología , Receptor ErbB-2/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Expresión Génica , Genes myc/genética , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Trasplante de Neoplasias , Biblioteca de Péptidos , Radioinmunoensayo , Distribución Tisular
11.
Cancer Immunol Immunother ; 49(6): 296-304, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10946811

RESUMEN

Aberrant glycosylation is one of the most constant traits of malignant cells. The CaMBr1 hexasaccharide antigen, originally defined on the human breast carcinoma cell line MCF7, is expressed on some normal tissues but overexpressed in a high percentage of human breast, ovary, prostate and lung carcinomas. CaMBr1 overexpression is associated with poor prognosis. The epitope consists of the tetrasaccharide Fuc(alpha1-2)Ga1(beta1-3)GalNAc(beta1-3)Ga1alpha-O-spacer, which has recently become available as a synthetic oligosaccharide. Here we report the CaMBr1 tetrasaccharide conjugation to two different carrier proteins (CRM197 and KLH) and the evaluation of conjugate immunogenicity in mice following their administration in various vaccine formulations with two adjuvants (MPL-SE and Detox-PC). Radioimmunoassay to determine the level and isotype of anti-tetrasaccharide antibodies in mouse sera, and cytofluorimetric analysis and 51Cr-release assay on human tumor cells, to evaluate specificity of binding and complement-dependent lysis respectively, identified CaMBr1-CRM197, in association with the MPL-SE adjuvant, as the best vaccine formulation. This combination induced (1) production of tetrasaccharide-specific antibodies, with negligible side-effects; (2) antibodies with complement-mediated cytotoxic activity on human CaMBr1-positive cells and (3) a high titer of IgG1 detected in sera obtained 3 months after the first injection, indicating that the anti-tetrasaccharide antibody response was mediated by T cell activation. The availability of CaMBr1-glycoconjugate in the minimal and functional antigenic structure and the identification of an efficacious vaccine formulation opens the way to exploring the activity of this glycoconjugate in a clinical setting.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/inmunología , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Vacunas contra el Cáncer/inmunología , Oligosacáridos/inmunología , Animales , Relación Dosis-Respuesta Inmunológica , Humanos , Inmunización , Inmunoglobulina G/sangre , Ratones , Células Tumorales Cultivadas
12.
Cancer Res ; 58(5): 991-6, 1998 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-9500461

RESUMEN

The display of repertoires of human antibody (Ab) fragments on filamentous phages and selection by binding of the phage to antigen (Ag) have provided a ready means of deriving human Ab against purified Ag. However, it has been more difficult to obtain phage Ab against an individual Ag of a complex mixture, such as cell surface Ag. Using the technique of "guided selection," we generated human Ab against the high-affinity folate-binding protein (FBP), a cell surface Ag that is overexpressed in many human ovarian carcinomas. The guiding Ab template was provided by the light chain of mouse monoclonal Ab Mov19 (K[aff], 10[8] M[-1]) directed against FBP; this was paired with repertoires of human heavy chains displayed on phages, and the hybrid Ab fragments were selected by binding to an ovarian carcinoma cell line (OVCAR3). The selected human heavy chains were then paired with repertoires of human light chains. Further panning led to the isolation of a human Fab fragment, C4, with a binding affinity of 0.2 x 10(8) M(-1). This was highly specific for FBP, as demonstrated by ELISA and flow cytometry data and by immunoprecipitation of the relevant molecule from the cell surface of ovarian carcinoma cells. Moreover, C4 targeted the same or a closely related epitope of the Ag, as did the template rodent monoclonal Ab Mov19. These results suggest the usefulness of guided selection as a simple means to deriving human Ab against cell surface Ag for which a rodent Ab is available.


Asunto(s)
Anticuerpos Antineoplásicos/genética , Carcinoma/inmunología , Biblioteca de Genes , Fragmentos Fab de Inmunoglobulinas/genética , Neoplasias Ováricas/inmunología , Biblioteca de Péptidos , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Antineoplásicos/inmunología , Bacteriófagos/genética , Femenino , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Ratones , Datos de Secuencia Molecular
13.
Cancer Gene Ther ; 5(6): 401-7, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9917095

RESUMEN

The antitumor specificity of T cells can be induced by gene transfer using a recently developed therapeutic approach (T body). In this work, we genetically conferred anticarbohydrate specificity onto T cells using the variable regions of monoclonal antibody MLuC1, which binds the Lewis(Y) (LeY) tumor-associated antigen that is overexpressed on several human carcinomas. The variable regions of MLuC1, which are in a single-chain Fv (ScFv) configuration, were cloned and spliced in a eukaryotic expression vector with both the gene encoding the signal-transducing gamma-chain of the human Fcgamma receptor and a flexible hinge domain. The chimeric ScFv-gamma gene was expressed in a murine cytotoxic T-cell hybridoma. Transfectants receiving vector only served as a negative control (mock). Screening for functional transfectants was carried out using a tumor growth inhibition assay. The soluble form of MLuC1 ScFv was recovered from bacteria periplasm and tested for binding to LeY-expressing cells by the fluorescence-activated cell sorter analysis. Despite the low binding ability of the soluble MLuC1 ScFv, 7 of 13 genetically engineered cytotoxic T lymphocyte clones inhibited the growth of LeY-positive cells and did not affect growth of LeY-negative cells. None of the mock clones tested specifically inhibited tumor growth. These data indicate that, by chimeric MLuC1 ScFv-gamma gene transfer, it is possible to confer anticarbohydrate specificity onto T cells and extend the applicability of the T-body approach to tumor-associated antigens that are naturally not recognized by T cells.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Técnicas de Transferencia de Gen , Antígenos del Grupo Sanguíneo de Lewis/inmunología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antineoplásicos , Especificidad de Anticuerpos , Secuencia de Bases , Neoplasias de la Mama/inmunología , Femenino , Citometría de Flujo , Humanos , Hibridomas , Región Variable de Inmunoglobulina , Inmunoterapia , Ratones , Datos de Secuencia Molecular , Neoplasias Ováricas/inmunología , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Cutáneas/inmunología , Células Tumorales Cultivadas
14.
Cancer Immunol Immunother ; 44(5): 257-64, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9247560

RESUMEN

T cell triggering can be achieved by monoclonal antibodies (mAbs) specific for the CD3/TcR complex. In the presence of appropriate costimulation and/or progression factors, such triggering permits the generation of effector cells for immunotherapy protocols involving the redirection of T cell lysis against tumor cells by mAbs bispecific for anti-CD3/anti-tumor cells (bs-mAbs). Focusing our analysis on the clinically relevant bs-mAb OC/TR, we found that bs-mAbs generated with the same anti tumor specificity, but two other anti-CD3 mAbs, TR66 and OKT3, have the same and a significantly lower lytic potential, respectively, compared with that of OC/TR. To evaluate the relevance of the anti-CD3 component, we examined several anti-CD3 mAbs with respect to binding parameters and the ability to trigger T lymphocytes. Competitive binding assays suggested that all anti-CD3 mAbs recognized the same or overlapping epitopes, although mAbs BMA030 and OC/TR bound with lower avidity than did alpha CD3 (the bivalent anti-CD3 mAb produced by the hybrid hybridoma OC/TR). TR66 and OKT3, as determined by measurement of the affinity constants. In all lymphocyte populations examined, which included resting peripheral blood mononuclear cells (PBMC), activated PBMC and T cell clones, OKT3, BMA033 and OC/TR failed to mobilize Ca2+ without cross-linking, whereas alpha CD3, in both murine and murine-human chimeric versions, TR66 and BMA030, did not require cross-linking. The ability to induce CD3 modulation was associated in part with the induction of Ca2+ fluxes. Despite the differences in the behavior of these mAbs in triggering the events that precede proliferation, all of them ultimately led to expression of the IL-2 receptor and to proliferation in T cells in the presence of accessory cells. Our data suggest that anti-CD3 mAbs that bind more rapidly (strong Ca2+ mobilizers) and more tightly under physiological conditions are good candidates for retargeting T cells in the bs-mAb clinical application.


Asunto(s)
Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Antineoplásicos/biosíntesis , Especificidad de Anticuerpos , Complejo CD3/inmunología , Neoplasias Ováricas/inmunología , Linfocitos T/inmunología , Anticuerpos Monoclonales/biosíntesis , Citotoxicidad Inmunológica , Femenino , Humanos , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/inmunología
16.
Cancer Res ; 56(23): 5443-9, 1996 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8968099

RESUMEN

One of the major limitations to the immunotherapy of ovarian carcinoma based on the use of anti-CD3/antitumor bispecific monoclonal antibodies (bi-mAb) is the need for preactivation of effector cells ex vivo, because cross-linking of the T cell receptor-CD3 complex per se may lead to T-cell unresponsiveness or even apoptosis. The bi-mAb OC/TR, which recognizes the folate-binding protein (FBP) overexpressed in 90% of ovarian carcinomas and the CD3 molecule on T cells, has demonstrated efficacy in a clinical setting. Here we investigated the possibility of delivering accessory signals to OC/TR-retargeted peripheral blood mononuclear cells (PBMCs) via an anti-CD28 mAb or an anti-FBP/anti-CD28 bi-mAb. Coculture of resting PBMCs from healthy donors with OC/TR, anti-FBP/anti-CD28 bi-mAb, and FBP+ tumor cell lines resulted in a highly activated phenotype of effector cells and in a dramatic in vitro growth inhibition of the target cells without an increase in OC/TR-redirected lysis. Whereas both the CD4 and CD8 T cell subsets were involved in the growth inhibition, only the CD8 subpopulation accounted for the cytotoxic activity. The in vitro tumor growth inhibition was mediated mainly by soluble factors, which were active on both FBP+ and FBP- ("bystander effect") cell lines. Activation and antitumor activity were also observed, albeit to a lesser extent, using OC/TR and monospecific bivalent anti-CD28 mAb. In vitro analysis demonstrated that cross-linking between tumor and effector cells for at least 24 h was needed to achieve T-cell activation and development of antitumor activities. Thus, ex vivo CD3-CD28 costimulation on resting PBMCs might be of therapeutic utility for local treatment of minimal residual disease.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Proteínas Portadoras/inmunología , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos , Proteínas de Neoplasias/inmunología , Neoplasias Ováricas/terapia , Receptores de Superficie Celular , Subgrupos de Linfocitos T/inmunología , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Especificidad de Anticuerpos , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Técnicas de Cocultivo , Femenino , Receptores de Folato Anclados a GPI , Humanos , Linfocinas/metabolismo , Neoplasia Residual , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Tumorales Cultivadas
17.
J Hematother ; 4(5): 423-7, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8581379

RESUMEN

The high frequency of relapse after induction chemotherapy in advanced ovarian carcinoma patients calls for new therapeutic modalities. Retargeted T cell-mediated lysis can be achieved using the bispecific antibody (BsmAb) OCTR, directed to CD3 on T cells and to the folate receptor on ovarian carcinoma cells. Twenty-eight patients with limited intraperitoneal disease after first-line therapy entered a phase II study. They received two i.p. 5 day cycles of activated PBMC retargeted with OCTR. Despite unfavorable tumor characteristics, 7 of 26 patients (27%) showed complete or partial intraperitoneal responses with strict surgicopathologic evaluation. In most cases, the disease relapsed outside the peritoneal cavity, and in 1 case complete intraperitoneal response was accompanied by progression in retroperitoneal lymph nodes. The morbidity was mild to moderate and transient. Combination of i.v. and i.p. administration of OCTR-retargeted lymphocytes will possibly lead to extraperitoneal cure. Ongoing clinical studies indicate that the i.v. infusion of up to 8 x 10(8) OCTR-retargeted T lymphocytes does not induce a higher toxicity than the i.p. treatment. To avoid PBMC preactivation, new approaches for delivering accessory signals are under investigation. Preliminary results indicate that nonactivated PBMC retargeted by OCTR in the presence of an anti-CD28 monoclonal antibody (mAb) are able to significantly inhibit tumor growth.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Proteínas Portadoras/inmunología , Activación de Linfocitos , Muromonab-CD3/uso terapéutico , Proteínas de Neoplasias/inmunología , Neoplasias Ováricas/terapia , Neoplasias Peritoneales/secundario , Receptores de Superficie Celular , Linfocitos T/inmunología , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/inmunología , Especificidad de Anticuerpos , Terapia Combinada , Citotoxicidad Inmunológica , Femenino , Receptores de Folato Anclados a GPI , Humanos , Inmunización Pasiva , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Metástasis Linfática , Muromonab-CD3/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/cirugía , Neoplasias Peritoneales/inmunología , Neoplasias Peritoneales/terapia , Neoplasias Retroperitoneales/secundario , Resultado del Tratamiento , Células Tumorales Cultivadas
18.
J Cell Biochem ; 58(1): 47-55, 1995 May.
Artículo en Inglés | MEDLINE | ID: mdl-7642722

RESUMEN

The monoclonal antibody MOv19 directed to a folate binding protein shows temperature-dependent potentiation of binding of the noncompeting monoclonal antibody MOv18 to the relevant antigen, but the mechanism involved in this phenomenon had remained unclear. Use of chimeric versions of both monoclonal antibodies and the F(ab')2 and Fab fragments of MOv19 revealed an increment in MOv18 binding in all combinations irrespective of the origin of the Fc portion of the monoclonal antibody. The potentiating effect of bivalent MOv19 fragments on 125I-MOv18 binding was similar to that of the entire monoclonal antibody and occurred at saturating concentrations of both reagents at which monovalent binding prevails. Similarly, the monovalent fragment also induced a significant increase in MOv18 binding. However, the potentiation occurred only at very high concentrations of antibody fragment. Homologous inhibition was drastically reduced using MOv19 Fab fragment, suggesting a low binding stability of the monovalent reagent. Immunoblotting analysis and binding in the presence of exogenous purified folate binding protein indicated a cross-linking between soluble and cell surface molecules mediated by the bivalent monoclonal antibodies. The extent of the increase in MOv18 binding at 0 degrees C with high amounts of exogenous folate binding protein was lower than that obtained at 37 degrees C in the absence of added molecule. Release of 125I-MOv18 from the cell surface was significantly higher in the absence of MOv19 than in its presence.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Anticuerpos Monoclonales/inmunología , Reacciones Antígeno-Anticuerpo , Proteínas Portadoras/inmunología , Receptores de Superficie Celular/inmunología , Anticuerpos Monoclonales/química , Afinidad de Anticuerpos , Sitios de Unión de Anticuerpos , Unión Competitiva , Carcinoma , Femenino , Receptores de Folato Anclados a GPI , Humanos , Immunoblotting , Fragmentos Fab de Inmunoglobulinas/inmunología , Cinética , Neoplasias Ováricas , Conformación Proteica , Proteínas Recombinantes de Fusión/inmunología , Factores de Tiempo , Células Tumorales Cultivadas
19.
Cancer Res ; 54(9): 2448-55, 1994 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-7512887

RESUMEN

The MOv18 (gamma 1, kappa) and MOv19 (gamma 2a, kappa) murine monoclonal antibodies (MAbs) recognize different epitopes on the human folate binding receptor which is overexpressed on 90% of nonmucinous epithelial ovarian tumors. A chimeric murine-human (human gamma 1, kappa) version of both antibodies was constructed and expressed. The genes encoding the murine heavy and light chain variable regions of the MOv18 and MOv19 MAbs were cloned from the parental hybridomas, fused with genes encoding the human heavy (gamma 1) and light (kappa) chain constant regions, respectively, and expressed in the SP2/0 murine myeloma cell line. Using human peripheral blood mononuclear cells as effector cells and conditions that provide for maximum lysis (effector target = 50:1, saturating antibody concentration), the murine MOv18 MAb (IgG1) mediated variable levels of specific cytolysis of the target ovarian cancer cell line IGROV1. In contrast, the chimeric MOv18 MAb mediated higher and more consistent lysis even at a 10-100-fold lower antibody concentration. The murine MOv19 MAb (IgG2a) mediated specific lysis of IGROV1 cells, and the chimeric version of this antibody mediated an amount of lysis at least equal to that mediated by its murine counterpart. A comparison of the ED50 values obtained for the murine MOv19 and chimeric MOv19 antibodies indicates that the chimeric MOv19 MAb was 3 to 10 times more potent than the murine MOv19 antibody. In addition, the ED50 values obtained for the chimeric MOv18 and chimeric MOv19 MAbs were similar, indicating that these MAbs are equally potent. The level of maximal lysis obtained was dependent on the number of target molecules/cell; the same high level of lysis mediated by cMOv18, MOv19, and cMOv19 was observed with both IGROV1 and OvCA432 target cells. However, only low levels of lysis were obtained when the SW626 cell line, which expresses 1 x 10(4) folate binding protein sites/cell, was used as a target. An equimolar mixture of the chimeric MOv18 and MOv19 MAbs was no more effective in the mediation of lysis than an equivalent amount of either chimeric MAb alone. These data suggest that the folate binding receptor is expressed on IGROV1 cells at a density sufficient to provide for optimal levels of antibody-mediated lysis using a single chimeric antibody directed at the folate binding receptor.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Epítopos/genética , Ácido Fólico , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Neoplasias Ováricas/inmunología , Receptores de Superficie Celular/inmunología , Animales , Anticuerpos Monoclonales/genética , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Femenino , Humanos , Ratones , Receptores de Superficie Celular/genética , Células Tumorales Cultivadas
20.
Int J Cancer ; 55(6): 931-7, 1993 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-8253530

RESUMEN

Targeting of T lymphocytes against epidermal growth-factor-receptor (EGF-R)+ tumor cells was achieved by constructing a hybrid hybridoma which secretes an anti-EGF-R/anti-CD3 bispecific monoclonal antibody (biMAb) of hybrid isotype (IgG1/IgG2a). Purification of biMAb molecules from parental anti-EGF-R and anti-CD3 MAbs was performed by protein-A chromatography. The purified biMAb was able to trigger the lysis of EGF-R+ tumor cell lines (A431, IGROV-1, MDA-468 and U-87) and of NIH-3T3 transfectants expressing human EGF-R by cytolytic T lymphocytes, but it was ineffective in the case of EGF-R-negative tumor targets. Normal EGF-R+ cells (keratinocytes and endometrial cells) were also susceptible to biMAb-targeted cytolysis. However, the amount of biMAb required to induce half-maximal cytolysis of tumor cells over-expressing the EGF-R molecule (A431) was considerably lower than that required to induce lysis of EGF-R+ tumor or normal cells which express EGF-R at considerably lower density. The ability of such biMAbs to deliver activation signals to T cells was evaluated by Ca++ mobilization and lymphokine production experiments. The soluble anti-EGF-R/anti-CD3 biMAb failed to induce intracellular Ca++ increases, which occurred only after cross-linking induced by an anti-mouse IgG antibody. Secretion of lymphokines (IFN-gamma, TNF-alpha and GM-CSF) was induced by contact of the biMAb-coated effector cells with the relevant tumor target, whereas the soluble biMAb was virtually ineffective. In addition, biMAb-coated effector cells retained the ability to recognize and to lyse EGF-R+ tumor cells for a prolonged period of time. Our data indicate that activation of effector cells targeted by biMAbs can only occur at the tumor site, where cross-linking of surface CD3 molecules is induced by contact with the tumor cells.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Complejo CD3/inmunología , Receptores ErbB/inmunología , Inmunoglobulina G/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Relación Dosis-Respuesta Inmunológica , Receptores ErbB/metabolismo , Humanos , Hibridomas/inmunología , Linfocinas/metabolismo , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...