Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oncotarget ; 8(32): 52045-52060, 2017 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-28881712

RESUMEN

Cancers employ a number of mechanisms to evade host immune responses. Here we report the effects of tumor-shed antigen CA125/MUC16 on suppressing IgG1-mediated antibody-dependent cellular cytotoxicity (ADCC). This evidence stems from prespecified subgroup analysis of a Phase 3 clinical trial testing farletuzumab, a monoclonal antibody to folate receptor alpha, plus standard-of-care carboplatin-taxane chemotherapy in patients with recurrent platinum-sensitive ovarian cancer. Patients with low serum CA125 levels treated with farletuzumab demonstrated improvements in progression free survival (HR 0.49, p = 0.0028) and overall survival (HR 0.44, p = 0.0108) as compared to placebo. Farletuzumab's pharmacologic activity is mediated in part through ADCC. Here we show that CA125 inhibits ADCC by directly binding to farletuzumab that in turn perturbs Fc-γ receptor engagement on effector cells.

2.
Oncotarget ; 6(28): 25429-40, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26327620

RESUMEN

Over-expression of endosialin/CD248 (herein referred to as CD248) has been associated with increased tumor microvasculature in various tissue origins which makes it an attractive anti-angiogenic target. In an effort to target CD248, we have generated a human CD248 knock-in mouse line and MORAb-004, the humanized version of the mouse anti-human CD248 antibody Fb5. Here, we report that MORAb-004 treatment significantly impacted syngeneic tumor growth and tumor metastasis in the human CD248 knock-in mice. In comparison with untreated tumors, MORAb-004 treated tumors displayed overall shortened and distorted blood vessels. Immunofluorescent staining of tumor sections revealed drastically more small and dysfunctional vessels in the treated tumors. The CD248 levels on cell surfaces of neovasculature pericytes were significantly reduced due to its internalization. This reduction of CD248 was also accompanied by reduced α-SMA expression, depolarization of pericytes and endothelium, and ultimately dysfunctional microvessels. These results suggest that MORAb-004 reduced CD248 on pericytes, impaired tumor microvasculature maturation and ultimately suppressed tumor development.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Microvasos/efectos de los fármacos , Neovascularización Patológica , Pericitos/efectos de los fármacos , Actinas/metabolismo , Inhibidores de la Angiogénesis/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Transporte Biológico , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Microvasos/inmunología , Microvasos/metabolismo , Microvasos/patología , Metástasis de la Neoplasia , Pericitos/inmunología , Pericitos/metabolismo , Pericitos/patología , Interferencia de ARN , Factores de Tiempo , Transfección , Carga Tumoral/efectos de los fármacos
3.
Cancer Biol Ther ; 14(11): 1032-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24025360

RESUMEN

Because of its high mortality rate, ovarian cancer is a leading cause of death among women and a highly unmet medical need. New therapeutic agents that are effective and well tolerated are needed and cancer antigen-specific monoclonal antibodies that have direct pharmacologic effects or can stimulate immunological responses represent a promising class of agents for the treatment of this disease. The human folate receptor α (FOLR1), which is overexpressed in ovarian cancer but largely absent in normal tissues, appears to play a role in the transformed phenotype in ovarian cancer, cisplatin sensitivity, and growth in depleted folate conditions and therefore has potential as a target for passive immunotherapy. The anti-FOLR1 monoclonal antibody MORAb-003 (farletuzumab) was previously shown to elicit antibody dependent cellular cytotoxicity (ADCC) and inhibit tumor growth of human tumor xenografts in nude mice. Because of its promising preclinical profile, farletuzumab has been evaluated in clinical trials as a potential therapeutic agent for ovarian cancer. In this report, we demonstrated that farletuzumab's antitumor effect against an experimental model of ovarian cancer is mediated by its ADCC activity.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Antineoplásicos/farmacología , Receptor 1 de Folato/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Femenino , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Receptores de IgG/metabolismo
4.
J Immune Based Ther Vaccines ; 8: 9, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21176153

RESUMEN

BACKGROUND: Staphylococcal enterotoxins are considered potential biowarfare agents that can be spread through ingestion or inhalation. Staphylococcal enterotoxin B (SEB) is a widely studied superantigen that can directly stimulate T-cells to release a massive amount of proinflammatory cytokines by bridging the MHC II molecules on an antigen presenting cell (APC) and the Vß chains of the T-cell receptor (TCR). This potentially can lead to toxic, debilitating and lethal effects. Currently, there are no preventative measures for SEB exposure, only supportive therapies. METHODS: To develop a potential therapeutic candidate to combat SEB exposure, we have generated three human B-cell hybridomas that produce human monoclonal antibodies (HuMAbs) to SEB. These HuMAbs were screened for specificity, affinity and the ability to block SEB activity in vitro as well as its lethal effect in vivo. RESULTS: The high-affinity HuMAbs, as determined by BiaCore analysis, were specific to SEB with minimal crossreactivity to related toxins by ELISA. In an immunoblotting experiment, our HuMAbs bound SEB mixed in a cell lysate and did not bind any of the lysate proteins. In an in vitro cell-based assay, these HuMAbs could inhibit SEB-induced secretion of the proinflammatory cytokines (INF-γ and TNF-α) by primary human lymphocytes with high potency. In an in vivo LPS-potentiated mouse model, our lead antibody, HuMAb-154, was capable of neutralizing up to 100 µg of SEB challenge equivalent to 500 times over the reported LD50 (0.2 µg) , protecting mice from death. Extended survival was also observed when HuMAb-154 was administered after SEB challenge. CONCLUSION: We have generated high-affinity SEB-specific antibodies capable of neutralizing SEB in vitro as well as in vivo in a mouse model. Taken together, these results suggest that our antibodies hold the potential as passive immunotherapies for both prophylactic and therapeutic countermeasures of SEB exposure.

5.
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
6.
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
7.
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.

8.
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
9.
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
10.
Plant Biotechnol J ; 3(4): 399-407, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17173628

RESUMEN

The reversible inhibition of DNA repair is a novel approach to maximize genetic diversity within a plant's genome in order to generate offspring exhibiting important de novo output traits. This process is based on the inhibition of the evolutionarily conserved mismatch repair (MMR) system. In this process, a human dominant negative MMR gene allele is introduced into the germline of a target plant, yielding progeny that can be screened to identify variants with commercially important agronomic output traits. Using this novel strategy, we generated MMR-deficient Arabidopsis thaliana plants that showed genome-wide instability of nucleotide repeats associated with chromosomal microsatellites, in addition to base substitution mutations. Functional screenings of the MMR-deficient Arabidopsis offspring identified variants expressing selectable traits (ethylene insensitivity and salt tolerance), as well as plants exhibiting altered morphologic traits (albinos and dwarfs). We determined by segregation analyses of variant plants that the de novo phenotypes were due to both recessive and dominant genetic mutations. Mutations caused by MMR deficiency showed a different spectrum compared with those derived using ethylmethane sulphonate (EMS) mutagenesis. Our finding demonstrates the feasibility of using reversible MMR deficiency via transient expression of a single human gene product to enhance genetic diversity in plants.

11.
Science ; 302(5646): 842-6, 2003 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-14593172

RESUMEN

Functional analysis of a genome requires accurate gene structure information and a complete gene inventory. A dual experimental strategy was used to verify and correct the initial genome sequence annotation of the reference plant Arabidopsis. Sequencing full-length cDNAs and hybridizations using RNA populations from various tissues to a set of high-density oligonucleotide arrays spanning the entire genome allowed the accurate annotation of thousands of gene structures. We identified 5817 novel transcription units, including a substantial amount of antisense gene transcription, and 40 genes within the genetically defined centromeres. This approach resulted in completion of approximately 30% of the Arabidopsis ORFeome as a resource for global functional experimentation of the plant proteome.


Asunto(s)
Arabidopsis/genética , Genoma de Planta , ARN Mensajero/genética , ARN de Planta/genética , Transcripción Genética , Mapeo Cromosómico , Cromosomas de las Plantas/genética , Clonación Molecular , Biología Computacional , ADN Complementario/genética , ADN Intergénico , Etiquetas de Secuencia Expresada , Perfilación de la Expresión Génica , Genes de Plantas , Genómica , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Sistemas de Lectura Abierta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA