Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Cancer ; 139(8): 1851-63, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27144973

RESUMEN

c-Met is a prototypic member of a sub-family of RTKs. Inappropriate c-Met activation plays a crucial role in tumor formation, proliferation and metastasis. Using a key c-Met dimerization assay, a set of 12 murine whole IgG1 monoclonal antibodies was selected and a lead candidate, m224G11, was humanized by CDR-grafting and engineered to generate a divalent full antagonist humanized IgG1 antibody, hz224G11. Neither m224G11 nor hz224G11 bind to the murine c-Met receptor. Their antitumor activity was investigated in vitro in a set of experiments consistent with the reported pleiotropic effects mediated by c-Met and, in vivo, using several human tumor xenograft models. Both m224G11 and hz224G11 exhibited nanomolar affinities for the receptor and inhibited HGF binding, c-Met phosphorylation, and receptor dimerization in a similar fashion, resulting in a profound inhibition of all c-Met functions in vitro. These effects were presumably responsible for the inhibition of c-Met's major functions including cell proliferation, migration, invasion scattering, morphogenesis and angiogenesis. In addition to these in vitro properties, hz224G11 dramatically inhibits the growth of autocrine, partially autophosphorylated and c-Met amplified cell lines in vivo. Pharmacological studies performed on Hs746T gastric cancer xenografts demonstrate that hz224G11 strongly downregulates c-Met expression and phosphorylation. It also decreases the tumor mitotic index (Ki67) and induces apoptosis. Taken together, the in vitro and in vivo data suggest that hz224G11 is a promising candidate for the treatment of tumors. This antibody, now known as ABT-700 and currently in Phase I clinical trials, may provide a novel therapeutic approach to c-Met-expressing cancers.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales de Origen Murino/farmacología , Neoplasias/terapia , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/inmunología , Células A549 , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales de Origen Murino/inmunología , Células CHO , Línea Celular Tumoral , Cricetulus , Femenino , Factor de Crecimiento de Hepatocito/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunoglobulina G/inmunología , Ligandos , Células MCF-7 , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias/inmunología , Proteínas Proto-Oncogénicas c-met/biosíntesis , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Int J Cancer ; 132(6): 1463-74, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22886345

RESUMEN

To identify new potential targets in oncology, functional approaches were developed using tumor cells as immunogens to select monoclonal antibodies targeting membrane receptors involved in cell proliferation. For that purpose cancer cells were injected into mice and resulting hybridomas were screened for their ability to inhibit cell proliferation in vitro. Based on this functional approach coupled to proteomic analysis, a monoclonal antibody specifically recognizing the human junctional adhesion molecule-A (JAM-A) was defined. Interestingly, compared to both normal and tumor tissues, we observed that JAM-A was mainly overexpressed on breast, lung and kidney tumor tissues. In vivo experiments demonstrated that injections of anti-JAM-A antibody resulted in a significant tumor growth inhibition of xenograft human tumors. Treatment with monoclonal antibody induced a decrease of the Ki67 expression and downregulated JAM-A levels. All together, our results show for the first time that JAM-A can interfere with tumor proliferation and suggest that JAM-A is a potential novel target in oncology. The results also demonstrate that a functional approach coupled to a robust proteomic analysis can be successful to identify new antibody target molecules that lead to promising new antibody-based therapies against cancers.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Moléculas de Adhesión Celular/fisiología , Neoplasias/tratamiento farmacológico , Receptores de Superficie Celular/fisiología , Animales , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Antígeno Ki-67/análisis , Ratones , Ratones Endogámicos BALB C , Neoplasias/patología , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/antagonistas & inhibidores
3.
Mol Cancer Ther ; 19(1): 168-177, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31594825

RESUMEN

The insulin-like growth factor type 1 receptor (IGF-1R) is important in tumorigenesis, and its overexpression occurs in numerous tumor tissues. To date, therapeutic approaches based on mAbs and tyrosine kinase inhibitors targeting IGF-1R have only shown clinical benefit in specific patient populations. We report a unique IGF-1R-targeted antibody-drug conjugate (ADC), W0101, designed to deliver a highly potent cytotoxic auristatin derivative selectively to IGF-1R overexpressing tumor cells. The mAb (hz208F2-4) used to prepare the ADC was selected for its specific binding properties to IGF-1R compared with the insulin receptor, and for its internalization properties. Conjugation of a novel auristatin derivative drug linker to hz208F2-4 did not alter its binding and internalization properties. W0101 induced receptor-dependent cell cytotoxicity in vitro when applied to various cell lines overexpressing IGF-1R, but it did not affect normal cells. Efficacy studies were conducted in several mouse models expressing different levels of IGF-1R to determine the sensitivity of the tumors to W0101. W0101 induced potent tumor regression in certain mouse models. Interestingly, the potency of W0101 correlated with the expression level of IGF-1R evaluated by IHC. In an MCF-7 breast cancer model with high-level IGF-1R expression, a single injection of W0101 3 mg/kg led to strong inhibition of tumor growth. W0101 provides a potential new therapeutic option for patients overexpressing IGF-1R. A first-in-human trial of W0101 is currently ongoing to address clinical safety.


Asunto(s)
Inmunoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Receptor IGF Tipo 1/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/farmacología , Ratones , Ratones Desnudos , Neoplasias/patología
4.
Int J Cancer ; 124(10): 2281-93, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19165858

RESUMEN

IGF-1 receptor (IGF-1R) plays a key role in the development of numerous tumors. Blockade of IGF-1R axis using monoclonal antibodies constitutes an interesting approach to inhibit tumor growth. We have previously shown that h7C10, a humanized anti-IGF-1R Mab, exhibited potent antitumor activity in vivo. However, mechanisms of action of h7C10 are still unknown. Here, we showed that h7C10 inhibited IGF-1-induced IGF-1R phosphorylation in a dose-dependent manner. Also, h7C10 abolished IGF-1-induced activation of PI3K/AKT and MAPK pathways. Cell cycle progression and colony formation were affected in the presence of h7C10 probably because of the inhibition of IGF-1-induced cyclin D1 and E expression. In addition, we demonstrated that h7C10 induced a rapid IGF-1R internalization leading to an accumulation into cytoplasm resulting in receptor degradation. Using lysosome and proteasome inhibitors, we observed that the IGF-1R alpha- and beta-chains could follow different degradation routes. Thus, we demonstrated that antitumoral properties of h7C10 are the result of IGF-1-induced cell signaling inhibition and down-regulation of IGF-1R level suggesting that h7C10 could be a candidate for therapeutic applications.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Receptor IGF Tipo 1/inmunología , Animales , Secuencia de Bases , Ciclinas/metabolismo , Femenino , Citometría de Flujo , Humanos , Inmunoprecipitación , Lisosomas/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Microscopía Fluorescente , Fosforilación , ARN Interferente Pequeño , Receptor IGF Tipo 1/metabolismo , Proteína de Retinoblastoma/metabolismo , Trasplante Heterólogo , Ubiquitinación
5.
Mol Cancer Ther ; 15(8): 1890-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27297868

RESUMEN

The type IV C-X-C-motif chemokine receptor (CXCR4) is expressed in a large variety of human cancers, including hematologic malignancies, and this receptor and its ligand, stromal cell-derived factor-1 (SDF-1), play a crucial role in cancer progression. We generated a humanized immunoglobulin G1 mAb, hz515H7, which binds human CXCR4, efficiently competes for SDF-1 binding, and induces a conformational change in CXCR4 homodimers. Furthermore, it inhibits both CXCR4 receptor-mediated G-protein activation and ß-arrestin-2 recruitment following CXCR4 activation. The binding of the hz515H7 antibody to CXCR4 inhibits the SDF-1-induced signaling pathway, resulting in reduced phosphorylation of downstream effectors, such as Akt, Erk1/2, p38, and GSK3ß. Hz515H7 also strongly inhibits cell migration and proliferation and, while preserving normal blood cells, induces both antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity against neoplastic cells. In mouse xenograft models, hz515H7 displays antitumor activities with multiple hematologic tumor cell lines, with its Fc-mediated effector functions proving essential in this context. Furthermore, hz515H7 binds to primary tumor cells from acute myeloid leukemia and multiple myeloma patients. Collectively, our results demonstrate two major mechanisms of action, making hz515H7 unique in this regard. Its potential as a best-in-class molecule is currently under investigation in a phase I clinical trial. Mol Cancer Ther; 15(8); 1890-9. ©2016 AACR.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Unión Competitiva , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Proteínas del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Unión Proteica , Multimerización de Proteína , Receptores CXCR4/química , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Arrestina beta 2/metabolismo
6.
Thromb Haemost ; 88(1): 123-30, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12152652

RESUMEN

Tissue Factor (TF), an integral membrane glycoprotein that initiates the extrinsic pathway of blood coagulation, is thought to play a major part in coronary acute events. Adenosine, an endogenous nucleoside produced by the degradation of intracellular adenosine triphosphate, has been shown to exert many cardioprotective effects via an inhibition of platelets and neutrophils. This study was conducted to determine whether adenosine (ADO) could modulate the expression of TF by human monocytes. We found that ADO inhibited TF antigen and activity on endotoxin-stimulated monocytes in a dose-dependent manner. The mechanism was at least pre-translational since ADO caused a change in the TF mRNA level. Using ADO receptor-specific analogs, we showed that highly selective A3 agonist N6-(3-iodobenzyl)-adenosine-5'-N'-methyluronamide (IB-MECA) inhibited LPS-induced TF activity expression more potently than A1 agonist R-phenylisopropyladenosine (R-PIA) and A2 agonist CGS 2180. Furthermore, A1/A3 antagonist, xanthine amine congener (XAC) blocked the effect of ADO whereas A2a, A2b and A1 antagonists were ineffective. In addition, we observed that ADO agonists inhibited monocyte TF expression in LPS-stimulated whole blood. The rank order of agonist potency suggested that A2 and A3 receptors might be involved (2-Cado > CGS = IB-MECA > R-PIA). This was supported by the fact that A2 and A3 antagonists reversed the action of 2-Cado. We conclude that TF inhibition by ADO on human purified monocytes involved A3 receptors.


Asunto(s)
Adenosina/farmacología , Monocitos/efectos de los fármacos , Receptores Purinérgicos P1/fisiología , Tromboplastina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lipopolisacáridos/farmacología , Monocitos/metabolismo , ARN Mensajero/efectos de los fármacos , Receptor de Adenosina A3 , Tromboplastina/genética
7.
Methods Mol Biol ; 988: 305-17, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23475728

RESUMEN

In order to improve therapeutic antibodies efficacy in cancer patients, several strategies were developed. One of these strategies consists in the enhancement of effector functions. Antibody-dependent cellular cytotoxicity (ADCC) was shown to mediate the activity of several therapeutic antibodies through interaction of the constant fragment (Fc) with immune cells. The interactions of Fc fragment can be modulated by engineering through modifications of the carbohydrate moieties or through modifications of some critical amino acids for its binding. Such modifications have to be studied in an in vitro assay to evaluate their impact on the regulation of effector functions. Here, we described a method to evaluate ADCC using a nonradioactive assay based on the measurement of lactate dehydrogenase (LDH) release. NK cells were purified by negative immunomagnetic selection and used as effector cells to trigger ADCC against specific target tumor cells. The LDH release measurement from lysed cells is performed after 4 h incubation. This method can replace the (51)Cr release assay since it is less restrictive and highly sensitive.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Ensayos de Selección de Medicamentos Antitumorales/métodos , L-Lactato Deshidrogenasa/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Separación Celular/métodos , Pruebas de Enzimas/métodos , Citometría de Flujo/métodos , Humanos , Células Asesinas Naturales/enzimología , Células Asesinas Naturales/inmunología , L-Lactato Deshidrogenasa/química , Trastuzumab
8.
Methods Mol Biol ; 988: 319-29, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23475729

RESUMEN

One of the major issues for antibody treatment is enhancement of efficacy. Recent studies have highlighted the important role of effector functions in improvement of antibody therapy. Among effector functions, complement-dependent cytotoxicity (CDC), which induces cell lysis by a cascade of activation triggered by the binding of C1q subunits to the Fc regions of antibodies bound to the cell surface, is part of the mechanism of several antibody therapies. CDC can be modulated by either Fc isotype engineering or Fc genetic mutations or Fc glycosylation profile modifications. To evaluate the impact of such modifications on CDC, we describe a luminescence method based on ATP measurement to estimate tumor damaged cells and a flow cytometry method to evaluate the binding of C1q on the Fc region and the binding of C4b on cell surface. The luminescence method coupled with complement protein analysis by flow cytometry encompasses all needed methods to evaluate antibody ability to trigger CDC.


Asunto(s)
Adenosina Trifosfato/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos , Complemento C1q/metabolismo , Complemento C4b/metabolismo , Pruebas Inmunológicas de Citotoxicidad/métodos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Anticuerpos Monoclonales/farmacología , Antígenos CD20/inmunología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Citometría de Flujo/métodos , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Masculino , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA