Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Stem Cells Transl Med ; 10(12): 1581-1587, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34515419

RESUMEN

Debilitating diseases of the eye represent a large unmet medical need potentially addressable with stem cell-based approaches. Over the past decade, the California Institute for Regenerative Medicine (CIRM) has funded and supported the translation, from early research concepts to human trials, of therapeutic stem cell approaches for dry age-related macular degeneration, retinitis pigmentosa, and limbal stem cell deficiency. This article chronicles CIRM's journey in the ophthalmology field and discusses some key challenges and questions that were addressed along the way as well as questions that remain.


Asunto(s)
Oftalmología , Humanos , Medicina Regenerativa , Trasplante de Células Madre , Células Madre
2.
J Exp Med ; 198(6): 925-35, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12963695

RESUMEN

Human macrophages found in juxtaposition to fragmented elastin in vivo express the elastolytic matrix metalloproteinases (MMPs) progelatinase B, prometalloelastase, and promatrilysin. Though MMPs can degrade a range of extracellular matrix components, increasing evidence suggests that preferred targets in vivo include nonmatrix substrates such as chemokines and growth factors. Hence, the means by which MMPs participate in elastin turnover remain undefined as does the identity of the elastolysins. Herein, human macrophage cultures have been established that express a complement of elastolytic proteinases similar, if not identical, to that found in vivo. Under plasminogen-free conditions, macrophages preferentially use metalloelastase to mediate elastolysis via a process that deposits active enzyme on elastin surfaces. By contrast, in the presence of plasminogen, human macrophages up-regulate proteolysis 10-fold by processing promatrilysin to an active elastolysin via a urokinase-type plasminogen activator-dependent pathway. Matrilysin-deficient human macrophages fail to mediate an elastolytic response despite the continued expression of gelatinase B and metalloelastase. Thus, acting in concert with cosecreted cysteine proteinases whose activities are constrained to sites of macrophage-elastin contact (Punturieri, A., S. Filippov, E. Allen, I. Caras, R. Murray, V. Reddy, and S.J. Weiss. 2000. J. Exp. Med. 192:789-799), matrilysin confers macrophages with their most potent MMP-dependent elastolytic system.


Asunto(s)
Elastina/metabolismo , Macrófagos/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Animales , Aorta/citología , Aorta/metabolismo , Catepsinas/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados , Medio de Cultivo Libre de Suero , Inhibidores Enzimáticos/metabolismo , Humanos , Macrófagos/citología , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Elastasa Pancreática/genética , Elastasa Pancreática/metabolismo , Plasminógeno/metabolismo , Activadores Plasminogénicos/metabolismo
3.
Blood ; 112(4): 1329-37, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17906076

RESUMEN

Currently, no approved monoclonal antibody (mAb) therapies exist for human multiple myeloma (MM). Here we characterized cell surface CS1 as a novel MM antigen and further investigated the potential therapeutic utility of HuLuc63, a humanized anti-CS1 mAb, for treating human MM. CS1 mRNA and protein was highly expressed in CD138-purified primary tumor cells from the majority of MM patients (more than 97%) with low levels of circulating CS1 detectable in MM patient sera, but not in healthy donors. CS1 was expressed at adhesion-promoting uropod membranes of polarized MM cells, and short interfering RNA (siRNA) targeted to CS1 inhibited MM cell adhesion to bone marrow stromal cells (BMSCs). HuLuc63 inhibited MM cell binding to BMSCs and induced antibody-dependent cellular cytotoxicity (ADCC) against MM cells in dose-dependent and CS1-specific manners. HuLuc63 triggered autologous ADCC against primary MM cells resistant to conventional or novel therapies, including bortezomib and HSP90 inhibitor; and pretreatment with conventional or novel anti-MM drugs markedly enhanced HuLuc63-induced MM cell lysis. Administration of HuLuc63 significantly induces tumor regression in multiple xenograft models of human MM. These results thus define the functional significance of CS1 in MM and provide the preclinical rationale for testing HuLuc63 in clinical trials, either alone or in combination.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Receptores Inmunológicos/inmunología , Animales , Antígenos de Neoplasias , Médula Ósea , Humanos , Ratones , Mieloma Múltiple/patología , ARN Mensajero/análisis , Receptores Inmunológicos/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Células del Estroma , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Stem Cells Transl Med ; 9(8): 821-826, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32281289

RESUMEN

A key implication of the cancer stem cell model is that for a cancer therapy to be curative, it is imperative to eliminate the cancer stem cells (CSCs) that drive tumor progression. The California Institute for Regenerative Medicine is supporting two novel approaches that target CSCs, one an antibody-mediated immunotherapy targeting CD47 and the other an antibody targeting ROR1. This article summarizes the evidence that CSCs are targeted and discusses the results of early clinical trials within the context of the CSC model.


Asunto(s)
Ensayos Clínicos como Asunto/normas , Inmunoterapia/métodos , Neoplasias/terapia , Células Madre Neoplásicas/patología , Humanos
5.
J Immunol Methods ; 315(1-2): 80-7, 2006 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-16935294

RESUMEN

Confirmatory assays for immunogenicity testing typically involve testing a sample in the presence or absence of excess drug. A decrease in assay signal in the presence of drug is taken to indicate the presence of anti-drug antibodies (ADAb) and the sample is confirmed positive. While there is widespread acceptance of the principle, there are currently no published guidelines for determining how much the signal should be reduced for a sample to be confirmed positive. In this report we address this issue using a novel approach employing a Student's t-test. The basic premise is to assess if an observed decrease in signal in the presence of drug is greater than what might be expected to occur as a result of the normal variation in the system. A key component of the method involves being able to capture and measure all of the normal variation. This requires a modification of commonly employed methods of sample preparation. We validated the method and tested samples from a clinical study. In addition, we reanalyzed the data to see what would have been the outcome had we used two other common approaches for confirmatory assays, one based on a minimum percent decrease in signal to confirm positivity (arbitrarily set at 25%), and one requiring a minimum drop in signal, set by a low quality control (QC) sample. The t-test approach proved superior over a wide range of assay signals and appeared to result in fewer false negatives.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Interpretación Estadística de Datos , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Humanos , Reproducibilidad de los Resultados
6.
J Exp Ther Oncol ; 5(4): 273-86, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17024968

RESUMEN

Integrin alpha5beta1, the principal fibronectin receptor, is an important survival factor, playing a key role in angiogenesis. Angiogenesis is critical for tumor growth, and anti-angiogenic therapies have met clinical success. To validate the therapeutic potential of an anti-alpha5beta1 strategy, we generated volociximab (M200) a chimeric human IgG4 version of the alpha5beta1 function-blocking murine antibody IIA1; and F200, the Fab derivative. Volociximab, F200 and IIA1 showed similar activity by ELISA (EC50= 0.2nM), Biacore (Kd= 0.1-0.4nM) and inhibition of fibronectin binding (IC50= 2-3nM). The inhibitory potential of alpha5beta1 antibodies was compared to HuMV833, an anti-VEGF antibody. Both volociximab and HuMV833 inhibited HUVEC proliferation (IC50 of volociximab = 0.2-0.5nM; IC50 of HuMV833 = 45nM). However, IIA1, volociximab and F200 were also potent inhibitors of an in vitro model of angiogenesis (HUVEC tube formation assay), unlike HuMV833. Additionally, volociximab inhibited in vitro tube formation induced by VEGF and/or bFGF, suggesting a mechanism of action independent of growth factor stimulus. In fact, inhibition of alpha5beta1 function by volociximab induced apoptosis of actively proliferating, but not resting, endothelial cells. Volociximab does not cross-react with rodent alpha5beta1, therefore in vivo validation of an anti-alpha5beta1 approach was conducted in a cynomolgus model of choroidal revascularization. Volociximab and F200 were potent inhibitors of neovessel formation in this model. These data demonstrate that volociximab has therapeutic potential in diseases in which new vessel formation is a component of the pathology.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos/uso terapéutico , Evaluación Preclínica de Medicamentos , Integrina alfa5beta1/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino , Células COS , Chlorocebus aethiops , Matriz Extracelular/metabolismo , Humanos , Concentración 50 Inhibidora , Integrina alfa5beta1/química , Cinética , Macaca fascicularis , Degeneración Macular/tratamiento farmacológico , Neovascularización Patológica , Rituximab
7.
Oncogene ; 22(38): 6005-12, 2003 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-12955079

RESUMEN

We have utilized oligonucleotide microarrays to identify novel genes of potential clinical and biological importance in prostate cancer. RNA from 74 prostate cancers and 164 normal body samples representing 40 different tissues were analysed using a customized Affymetrix GeneChip oligonucleotide microarray representative of over 90% of the expressed human genome. The gene for the zinc transporter ZnT4 was one of several genes that displayed significantly higher expression in prostate cancer compared to normal tissues from other organs. A polyclonal antipeptide antibody was used to demonstrate ZnT4 expression in the epithelium of all 165 elements of benign and 326 elements of localized prostate cancers examined and in nine of 10 advanced prostate cancer specimens by immunohistochemistry. Interestingly, decreased intensity of ZnT4 immunoreactivity occurred in the progression from benign to invasive localized prostate cancer and to metastatic disease. Immunofluorescence analysis and surface biotinylation studies of cells expressing ZnT4 localised the protein to intracellular vesicles and to the plasma membrane. These findings are consistent with a role for ZnT4 in vesicular transport of zinc to the cell membrane and potentially in efflux of zinc in the prostate.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Adolescente , Adulto , Secuencia de Aminoácidos , Proteínas de Transporte de Catión , Membrana Celular/metabolismo , Progresión de la Enfermedad , Humanos , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Próstata/fisiología , Hiperplasia Prostática/genética , Neoplasias de la Próstata/genética , Valores de Referencia , Vesículas Transportadoras/metabolismo
8.
Mol Cancer Ther ; 3(8): 921-32, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15299075

RESUMEN

Current treatments for advanced stage, hormone-resistant prostate cancer are largely ineffective, leading to high patient mortality and morbidity. To fulfill this unmet medical need, we used global gene expression profiling to identify new potential antibody-drug conjugate (ADC) targets that showed maximal prostate cancer-specific expression. TMEFF2, a gene encoding a plasma membrane protein with two follistatin-like domains and one epidermal growth factor-like domain, had limited normal tissue distribution and was highly overexpressed in prostate cancer. Immunohistochemistry analysis using a specific monoclonal antibody (mAb) to human TMEFF2 showed significant protein expression in 74% of primary prostate cancers and 42% of metastatic lesions from lymph nodes and bone that represented both hormone-naïve and hormone-resistant disease. To evaluate anti-TMEFF2 mAbs as potential ADCs, one mAb was conjugated to the cytotoxic agent auristatin E via a cathepsin B-sensitive valine-citrulline linker. This ADC, Pr1-vcMMAE, was used to treat male severe combined immunodeficient mice bearing xenografted LNCaP and CWR22 prostate cancers expressing TMEFF2. Doses of 3 to 10 mg/kg of this specific ADC resulted in significant and sustained tumor growth inhibition, whereas an isotype control ADC had no significant effect. Similar efficacy and specificity was shown with huPr1-vcMMAE, a humanized anti-TMEFF2 ADC. No overt in vivo toxicity was observed with either murine or human ADC, despite significant cross-reactivity of anti-TMEFF2 mAb with the murine TMEFF2 protein, implying minimal toxicity to other body tissues. These data support the further evaluation and clinical testing of huPr1-vcMMAE as a novel therapeutic for the treatment of metastatic and hormone-resistant prostate cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Proteínas de la Membrana/química , Proteínas de la Membrana/inmunología , Proteínas de Neoplasias/química , Proteínas de Neoplasias/inmunología , Oligopéptidos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Antineoplásicos/química , Encéfalo/metabolismo , Membrana Celular/metabolismo , Proliferación Celular , Clonación Molecular , ADN Complementario/metabolismo , Citometría de Flujo , Folistatina/química , Humanos , Hibridomas/química , Inmunohistoquímica , Cinética , Metástasis Linfática , Masculino , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligopéptidos/química , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie , Factores de Tiempo , Transfección
9.
Stem Cells Transl Med ; 4(8): 863-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26136501

RESUMEN

Heart disease due to myocardial infarction and the ensuing heart failure represent a major unmet medical need. Approved treatments do not prevent loss of cardiac muscle or reduce scar formation, both of which weaken heart function. Cell-based therapies currently being investigated both preclinically and clinically have the potential to address these underlying problems either by actually replacing lost tissue or by supplying paracrine growth factors that may have multiple beneficial effects such as reduction of inflammation, increase of blood supply, improvement in cell survival, and reduction of scar size. The best cell types, stage of disease to target, and delivery method to improve heart function are currently unclear. The California Institute for Regenerative Medicine supports multiple different cell-therapy strategies for heart disease, offering hope that improved treatments will be available for patients in the future.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Infarto del Miocardio/terapia , Medicina Regenerativa , Trasplante de Células Madre/tendencias , Insuficiencia Cardíaca/terapia , Humanos , Infarto del Miocardio/fisiopatología , Miocardio/patología
10.
Stem Cells Transl Med ; 3(12): 1393-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25378652

RESUMEN

Debilitating eye diseases such as age-related macular degeneration and retinitis pigmentosa currently represent a large unmet medical need that could potentially be addressed by stem cell therapy. A number of novel stem cell-based cellular therapies are now under development to treat a variety of eye diseases. The approaches being taken by the California Institute for Regenerative Medicine, together with its grantees, are discussed.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Retinitis Pigmentosa/terapia , Trasplante de Células Madre , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA