Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Cancer Res ; 21(6): 1281-8, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25398449

RESUMEN

PURPOSE: Endosialin (TEM-1, CD248) is a protein expressed on the surface of activated mesenchymal cells, including certain subsets of tumors. Preclinical models suppressing endosialin function have shown antitumor activity. A humanized monoclonal antibody, MORAb-004, was engineered to target endosialin and is the first agent in clinical development for this mesenchymal cell target. EXPERIMENTAL DESIGN: This first-in-human, open-label, phase I study recruited patients with treatment-refractory solid tumors. MORAb-004 was administered intravenously once weekly in 4-week cycles. Objectives included determination of the safety of multiple infusions of MORAb-004, identification of the maximum tolerated dose (MTD), pharmacokinetic modeling, detection of any anti-human antibody response, and assessment of objective radiographic response to therapy. RESULTS: Thirty-six patients were treated at 10 dose levels of MORAb-004, ranging from 0.0625 to 16 mg/kg. Drug-related adverse events were primarily grade 1-2 infusion toxicities. Dose-limiting toxicity of grade 3 vomiting was observed at 16 mg/kg. Eighteen of 32 evaluable patients across all doses achieved disease stability, with minor radiographic responses observed in 4 patients (pancreatic neuroendocrine, hepatocellular, and sarcoma tumor types). Pharmacokinetics showed MORAb-004 accumulation beginning at 4 mg/kg and saturable elimination beginning at 0.25 mg/kg. Exposure increased in a greater-than-dose-proportional manner with terminal half-life increasing proportionally with dose. The MTD was identified as 12 mg/kg. CONCLUSIONS: Preliminary antitumor activity was observed. Safety profile, pharmacokinetics, and early antitumor activity suggest that MORAb-004 is safe at doses up to 12 mg/kg and should be studied further for efficacy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Anciano , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Infusiones Intravenosas , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad
2.
Cancer Immun ; 7: 20, 2007 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-18088084

RESUMEN

Novel therapeutic agents that are safe and effective are needed for the treatment of pancreatic, ovarian, lung adenocarcinomas and mesotheliomas. Mesothelin is a glycosyl-phosphatidyl inositol (GPI)-linked membrane protein of 40 kDa over-expressed in all pancreatic adenocarcinoma and mesothelioma, in >70% of ovarian adenocarcinoma, and in non-small cell lung and colorectal cancers. The biological functions of mesothelin are not known, although it appears to be involved in cell adhesion via its interaction with MUC16. We have recently developed MORAb-009, a mouse-human chimeric IgG1kappa monoclonal antibody with an affinity of 1.5 nM for human mesothelin. Here we provide evidence that MORAb-009 prevents adhesion of mesothelin-bearing tumor cells to MUC16 positive cells and can elicit cell-mediated cytotoxicity on mesothelin-bearing tumor cells. Treatment that included MORAb-009 in combination with chemotherapy led to a marked reduction in tumor growth of mesothelin-expressing tumors in nude mice compared to chemotherapy or MORAb-009 treatment alone. No adverse effects of MORAb-009 were noted during toxicology studies conducted in non-human primates. The preclinical data obtained from our studies warrants pursuing clinical testing of MORAb-009. We have in fact initiated a Phase I clinical study enrolling patients with mesothelin-positive pancreatic, mesothelioma, non-small cell lung and ovarian cancers.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/uso terapéutico , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/inmunología , Neoplasias/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Endocitosis/efectos de los fármacos , Proteínas Ligadas a GPI , Humanos , Mesotelina , Ratones , Ratones Desnudos , Neoplasias/inmunología
3.
Proc Natl Acad Sci U S A ; 104(46): 17965-70, 2007 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-17986615

RESUMEN

Endosialin/TEM1 was originally discovered as a human embryonic fibroblast-specific antigen and was later found to be differentially expressed in tumor stroma and endothelium. Endosialin/TEM1 overexpression has been observed in many cancers of various tissue origin, including colon, breast, pancreatic, and lung. The knockout (KO) mouse model showed the absence of endosialin/TEM1 expression reduced growth, invasion, and metastasis of human tumor xenografts. In addition, lack of endosialin/TEM1 led to an increase in small immature blood vessels and decreased numbers of medium and large tumor vessels. This abnormal angiogenic response could be responsible for the reduced tumor growth and invasion observed in endosialin/TEM1 KO mice, suggesting a role for endosialin/TEM1 in controlling the interaction among tumor cells, endothelia, and stromal matrix. Here we report the identification of fibronectin (FN) and collagen types I and IV as specific ligands for endosialin/TEM1. More importantly, cells expressing endosialin/TEM1 exhibit enhanced adhesion to FN as well as enhanced migration through matrigel, although these properties could be blocked by a humanized antibody directed against human endosialin/TEM1. Our results pinpoint to a molecular mechanism by which expression of endosialin/TEM1 in the tumor stroma and endothelium may support tumor progression and invasion.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Adhesión Celular , Movimiento Celular , Proteínas de la Matriz Extracelular/metabolismo , Animales , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/fisiología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/fisiología , Western Blotting , Células CHO , Cricetinae , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Hidrólisis , Ratones , Ratones Noqueados , Unión Proteica
4.
Cancer Immun ; 7: 6, 2007 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-17346028

RESUMEN

The highly restricted distribution of human folate receptor-alpha (FRalpha) in normal tissues and its high expression in some tumors, along with its putative role in tumor cell transformation, make this antigen a suitable target for antigen-specific, monoclonal antibody-based immunotherapy for oncology indications. We have developed a therapeutic humanized monoclonal antibody with high affinity for FRalpha, named MORAb-003, which was derived from the optimization of the LK26 antibody using a whole cell genetic evolution platform. Here we show that MORAb-003 possesses novel, growth-inhibitory functions on cells overexpressing FRalpha. In addition, MORAb-003 elicited robust antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) in vitro, and inhibited growth of human ovarian tumor xenografts in nude mice. Because of its multimodal activity in vitro and its safe toxicology profile in non-human primates, MORAb-003 development has recently been advanced to clinical trials involving ovarian cancer patients.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/inmunología , Inmunoterapia/métodos , Neoplasias Ováricas/tratamiento farmacológico , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/inmunología , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos , Femenino , Receptores de Folato Anclados a GPI , Humanos , Cinética , Ratones , Ratones Desnudos , Proteínas de Neoplasias/inmunología , Primates/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Expert Opin Drug Discov ; 2(12): 1619-29, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23488905

RESUMEN

The genomics era has provided valuable information on the content of the human genome, including the structure and chromosomal location of many disease-associated loci. This wealth of information has resulted in the identification of novel targets that are amenable to biopharmaceutical product development, leading to an overall enhanced pace of therapeutic development for a broad array of disease indications. Historically, small chemical entities have been designed as therapies to gene products that are encoded by intracellular proteins, enzymes and channels. With the advent and development of biologically based (protein- and cell-based) entities (BBEs) therapies, it is now possible to create molecules that have exquisite specificity for disease targets and spare unwanted pharmacologic activity against normal tissues. In addition to the specificity, BBEs generally have lower toxicity profiles when compared with small chemical entities, making biologically-based therapeutic approaches even more attractive. One of the difficulties that has been encountered with BBEs is the ability to produce compounds with maximal pharmacologic activity as well as establishing systems that can manufacture these complex biological molecules in sufficient quantities to meet clinical demand. This review discusses a broad enabling platform technology called morphogenics that can rapidly yield robust systems that can overcome present manufacturing shortfalls as well as accelerate the development of highly efficacious BBEs from those with insufficient pharmacological activity.

6.
Proc Natl Acad Sci U S A ; 103(10): 3557-62, 2006 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-16505368

RESUMEN

Current strategies for the production of therapeutic mAbs include the use of mammalian cell systems to recombinantly produce Abs derived from mice bearing human Ig transgenes, humanization of rodent Abs, or phage libraries. Generation of hybridomas secreting human mAbs has been previously reported; however, this approach has not been fully exploited for immunotherapy development. We previously reported the use of transient regulation of cellular DNA mismatch repair processes to enhance traits (e.g., affinity and titers) of mAb-producing cell lines, including hybridomas. We reasoned that this process, named morphogenics, could be used to improve suboptimal hybridoma cells generated by means of ex vivo immunization and immortalization of antigen-specific human B cells for therapeutic Ab development. Here we present a platform process that combines hybridoma and morphogenics technologies for the generation of fully human mAbs specific for disease-associated human antigens. We were able to generate hybridoma lines secreting mAbs with high binding specificity and biological activity. One mAb with strong neutralizing activity against human granulocyte-macrophage colony-stimulating factor was identified that is now considered for preclinical development for autoimmune disease indications. Moreover, these hybridoma cells have proven suitable for genetic optimization using the morphogenics process and have shown potential for large-scale manufacturing.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/uso terapéutico , Linfocitos B/inmunología , Hibridomas/inmunología , Inmunoterapia/métodos , Secuencia de Aminoácidos , Animales , Antígenos/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Disparidad de Par Base , Células Cultivadas , Reparación del ADN , Epítopos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Cambio de Clase de Inmunoglobulina , Técnicas In Vitro , Ratones , Datos de Secuencia Molecular , Pruebas de Neutralización
7.
Ann N Y Acad Sci ; 1059: 86-96, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16382047

RESUMEN

Mutations in DNA mismatch repair (MMR) genes lead to genetically hypermutable cells. Germline mutations in MMR genes in man have been linked to the genetic predisposition to hereditary nonpolyposis colon cancer and a number of other inherited and sporadic malignancies. The ability to modulate the MMR process (referred to as morphogenics) in model systems offers a powerful tool for generating functional diversity in cells and multicellular organisms via the perpetual genomewide accumulation of randomized point and slippage mutation(s). Morphogenics is a platform process that employs a dominant negative MMR gene to create genetic diversity within defined cellular systems and results in a wide range of phenotypes, thus enabling the development and improvement of pharmaceutical products and the discovery of new pharmaceutical targets. Libraries of morphogenics-derived siblings are generated through random mutagenesis from naturally occurring DNA polymerase-induced mutations that occur during DNA replication. Morphogenic cells are screened in high-throughput assays to identify subclones with desired phenotypes for pathway discovery and/or product development. Morphogenics has been successfully applied to a wide range of hosts, including mammalian cells, transgenic mice, plants, yeast, and bacteria. Manipulation of these systems via morphogenics has led to the discovery of novel disease-associated phenotypes in targeted model systems. Moreover, morphogenics has been successfully applied to antibody-producing cell lines to yield subclones producing antibodies with enhanced binding affinities for therapeutic use, as well as to derive subclones with enhanced titers that are suitable for scaleable manufacturing. The selective manipulation of the MMR process via morphogenics is a platform technology that offers many advantages for the discovery of druggable targets, as well as for the development of novel pharmaceutical products.


Asunto(s)
Diseño de Fármacos , Mutación , Disparidad de Par Base , Línea Celular , Reparación del ADN , Enzimas Reparadoras del ADN , Replicación del ADN , Mutación de Línea Germinal , Humanos , Modelos Genéticos , Proteínas MutL , Mutagénesis , Proteínas de Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...