Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cells ; 33(10): 2985-94, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26260958

RESUMEN

The treatment of human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer has been revolutionized by trastuzumab. However, longer survival of these patients now predisposes them to forming HER2 positive brain metastases, as the therapeutic antibodies cannot cross the blood brain barrier. The current oncologic repertoire does not offer a rational, nontoxic targeted therapy for brain metastases. In this study, we used an established human neural stem cell line, HB1.F3 NSCs and generated a stable pool of cells secreting a high amount of functional full-length anti-HER2 antibody, equivalent to trastuzumab. Anti-HER2Ab secreted by the NSCs (HER2Ab-NSCs) specifically binds to HER2 overexpressing human breast cancer cells and inhibits PI3K-Akt signaling. This translates to HER2Ab-NSC inhibition of breast cancer cell growth in vitro. Preclinical in vivo experiments using HER2Ab overexpressing NSCs in a breast cancer brain metastases (BCBM) mouse model demonstrate that intracranial injection of HER2Ab-NSCs significantly improves survival. In effect, these NSCs provide tumor localized production of HER2Ab, minimizing any potential off-target side effects. Our results establish HER2Ab-NSCs as a novel, nontoxic, and rational therapeutic approach for the successful treatment of HER2 overexpressing BCBM, which now warrants further preclinical and clinical investigation.


Asunto(s)
Anticuerpos Antiidiotipos/biosíntesis , Neoplasias Encefálicas/terapia , Neoplasias de la Mama/tratamiento farmacológico , Células-Madre Neurales/metabolismo , Receptor ErbB-2/biosíntesis , Animales , Anticuerpos Antiidiotipos/inmunología , Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Células-Madre Neurales/inmunología , Células-Madre Neurales/trasplante , Receptor ErbB-2/inmunología , Trastuzumab/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Sci Transl Med ; 5(184): 184ra59, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23658244

RESUMEN

High-grade gliomas are extremely difficult to treat because they are invasive and therefore not curable by surgical resection; the toxicity of current chemo- and radiation therapies limits the doses that can be used. Neural stem cells (NSCs) have inherent tumor-tropic properties that enable their use as delivery vehicles to target enzyme/prodrug therapy selectively to tumors. We used a cytosine deaminase (CD)-expressing clonal human NSC line, HB1.F3.CD, to home to gliomas in mice and locally convert the prodrug 5-fluorocytosine to the active chemotherapeutic 5-fluorouracil. In vitro studies confirmed that the NSCs have normal karyotype, tumor tropism, and CD expression, and are genetically and functionally stable. In vivo biodistribution studies demonstrated NSC retention of tumor tropism, even in mice pretreated with radiation or dexamethasone to mimic clinically relevant adjuvant therapies. We evaluated safety and toxicity after intracerebral administration of the NSCs in non-tumor-bearing and orthotopic glioma-bearing immunocompetent and immunodeficient mice. We detected no difference in toxicity associated with conversion of 5-fluorocytosine to 5-fluorouracil, no NSCs outside the brain, and no histological evidence of pathology or tumorigenesis attributable to the NSCs. The average tumor volume in mice that received HB1.F3.CD NSCs and 5-fluorocytosine was about one-third that of the average volume in control mice. On the basis of these results, we conclude that combination therapy with HB1.F3.CD NSCs and 5-fluorocytosine is safe, nontoxic, and effective in mice. These data have led to approval of a first-in-human study of an allogeneic NSC-mediated enzyme/prodrug-targeted cancer therapy in patients with recurrent high-grade glioma.


Asunto(s)
Glioma/tratamiento farmacológico , Glioma/terapia , Células-Madre Neurales/citología , Profármacos/uso terapéutico , Animales , Línea Celular , Citosina Desaminasa/metabolismo , Femenino , Citometría de Flujo , Flucitosina/metabolismo , Flucitosina/uso terapéutico , Fluorouracilo/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Células-Madre Neurales/metabolismo , Profármacos/metabolismo
3.
PLoS One ; 7(4): e35150, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22539956

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is the most aggressive type of malignant primary brain tumors in adults. Molecular and genetic analysis has advanced our understanding of glioma biology, however mapping the cellular composition of the tumor microenvironment is crucial for understanding the pathology of this dreaded brain cancer. In this study we identified major cell populations attracted by glioma using orthotopic rodent models of human glioma xenografts. Marker-specific, anatomical and morphological analyses revealed a robust influx of host cells into the main tumor bed and tumor satellites. METHODOLOGY/PRINCIPAL FINDINGS: Human glioma cell lines and glioma spheroid orthotopic implants were used in rodents. In both models, the xenografts recruited large numbers of host nestin-expressing cells, which formed a 'network' with glioma. The host nestin-expressing cells appeared to originate in the subventricular zone ipsilateral to the tumor, and were clearly distinguishable from pericytes that expressed smooth muscle actin. These distinct cell populations established close physical contact in a 'pair-wise' manner and migrated together to the deeper layers of tumor satellites and gave rise to tumor vasculature. The GBM biopsy xenografts displayed two different phenotypes: (a) low-generation tumors (first in vivo passage in rats) were highly invasive and non-angiogenic, and host nestin-positive cells that infiltrated into these tumors displayed astrocytic or elongated bipolar morphology; (b) high-generation xenografts (fifth passage) had pronounced cellularity, were angiogenic with 'glomerulus-like' microvascular proliferations that contained host nestin-positive cells. Stromal cell-derived factor-1 and its receptor CXCR4 were highly expressed in and around glioma xenografts, suggesting their role in glioma progression and invasion. CONCLUSIONS/SIGNIFICANCE: Our data demonstrate a robust migration of nestin-expressing host cells to glioma, which together with pericytes give rise to tumor vasculature. Mapping the cellular composition of glioma microenvironment and deciphering the complex 'crosstalk' between tumor and host may ultimately aid the development of novel anti-glioma therapies.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Animales , Neoplasias Encefálicas/patología , Comunicación Celular , Línea Celular Tumoral , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Femenino , Glioma/patología , Humanos , Inmunohistoquímica , Proteínas de Filamentos Intermediarios/metabolismo , Ratones , Invasividad Neoplásica , Neovascularización Patológica , Proteínas del Tejido Nervioso/metabolismo , Nestina , Fenotipo , Ratas , Receptores CXCR4/metabolismo , Trasplante Heterólogo , Microambiente Tumoral
4.
Biochim Biophys Acta ; 1816(2): 191-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21767610

RESUMEN

Antibodies and antibody conjugates have emerged as important tools for cancer therapy. However, a major therapeutic challenge for the use of antibodies is their inability to cross the blood-brain barrier (BBB) to reach tumors localized in the central nervous system (CNS). Multiple methods have been developed to enhance antibody delivery to the CNS, including direct injection, mechanical or biochemical disruption of the BBB, conjugation to a 'molecular Trojan horse', cationization, encapsulation in nanoparticles and liposomes, and more recently, stem cell-mediated antibody delivery. In this review, we discuss each of these approaches, highlighting their successes and the obstacles that remain to be overcome.


Asunto(s)
Anticuerpos/administración & dosificación , Encéfalo/metabolismo , Animales , Anticuerpos/metabolismo , Barrera Hematoencefálica , Difusión , Humanos , Liposomas , Nanopartículas , Células Madre/fisiología , Transcitosis
5.
Stem Cells ; 28(11): 2084-7, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21089119

RESUMEN

Monoclonal antibodies are important tools for cancer therapy, however, three factors limit their effectiveness: toxicity, poor tumor penetration, and inability to cross the blood-brain barrier. This review discusses the emerging field of stem cell-mediated antibody delivery and how this approach may improve antibody therapy of cancer by overcoming these obstacles.


Asunto(s)
Anticuerpos/inmunología , Neoplasias/terapia , Células Madre/metabolismo , Animales , Humanos , Modelos Biológicos , Neoplasias/inmunología , Células Madre/citología
6.
PLoS One ; 4(12): e8314, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20016813

RESUMEN

BACKGROUND: Recombinant monoclonal antibodies have emerged as important tools for cancer therapy. Despite the promise shown by antibody-based therapies, the large molecular size of antibodies limits their ability to efficiently penetrate solid tumors and precludes efficient crossing of the blood-brain-barrier into the central nervous system (CNS). Consequently, poorly vascularized solid tumors and CNS metastases cannot be effectively treated by intravenously-injected antibodies. The inherent tumor-tropic properties of human neural stem cells (NSCs) can potentially be harnessed to overcome these obstacles and significantly improve cancer immunotherapy. Intravenously-delivered NSCs preferentially migrate to primary and metastatic tumor sites within and outside the CNS. Therefore, we hypothesized that NSCs could serve as an ideal cellular delivery platform for targeting antibodies to malignant tumors. METHODS AND FINDINGS: As proof-of-concept, we selected Herceptin (trastuzumab), a monoclonal antibody widely used to treat HER2-overexpressing breast cancer. HER2 overexpression in breast cancer is highly correlated with CNS metastases, which are inaccessible to trastuzumab therapy. Therefore, NSC-mediated delivery of trastuzumab may improve its therapeutic efficacy. Here we report, for the first time, that human NSCs can be genetically modified to secrete anti-HER2 immunoglobulin molecules. These NSC-secreted antibodies assemble properly, possess tumor cell-binding affinity and specificity, and can effectively inhibit the proliferation of HER2-overexpressing breast cancer cells in vitro. We also demonstrate that immunoglobulin-secreting NSCs exhibit preferential tropism to tumor cells in vivo, and can deliver antibodies to human breast cancer xenografts in mice. CONCLUSIONS: Taken together, these results suggest that NSCs modified to secrete HER2-targeting antibodies constitute a promising novel platform for targeted cancer immunotherapy. Specifically, this NSC-mediated antibody delivery system has the potential to significantly improve clinical outcome for patients with HER2-overexpressing breast cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Neuronas/citología , Células Madre/metabolismo , Animales , Anticuerpos Monoclonales Humanizados , Especificidad de Anticuerpos/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Inmunoglobulina G/inmunología , Ratones , Ratones Desnudos , Neuronas/efectos de los fármacos , Especificidad de Órganos/efectos de los fármacos , Receptor ErbB-2/inmunología , Células Madre/efectos de los fármacos , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto
7.
PLoS One ; 4(9): e7218, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19787043

RESUMEN

BACKGROUND: Treatment strategies for the highly invasive brain tumor, glioblastoma multiforme, require that cells which have invaded into the surrounding brain be specifically targeted. The inherent tumor-tropism of neural stem cells (NSCs) to primary and invasive tumor foci can be exploited to deliver therapeutics to invasive brain tumor cells in humans. Use of the strategy of converting prodrug to drug via therapeutic transgenes delivered by immortalized therapeutic NSC lines have shown efficacy in animal models. Thus therapeutic NSCs are being proposed for use in human brain tumor clinical trials. In the context of NSC-based therapies, MRI can be used both to non-invasively follow dynamic spatio-temporal patterns of the NSC tumor targeting allowing for the optimization of treatment strategies and to assess efficacy of the therapy. Iron-labeling of cells allows their presence to be visualized and tracked by MRI. Thus we aimed to iron-label therapeutic NSCs without affecting their cellular physiology using a method likely to gain United States Federal Drug Administration (FDA) approval. METHODOLOGY: For human use, the characteristics of therapeutic Neural Stem Cells must be clearly defined with any pertubation to the cell including iron labeling requiring reanalysis of cellular physiology. Here, we studied the effect of iron-loading of the therapeutic NSCs, with ferumoxide-protamine sulfate complex (FE-Pro) on viability, proliferation, migratory properties and transgene expression, when compared to non-labeled cells. FE-Pro labeled NSCs were imaged by MRI at tumor sites, after intracranial administration into the hemisphere contralateral to the tumor, in an orthotopic human glioma xenograft mouse model. CONCLUSION: FE-Pro labeled NSCs retain their proliferative status, tumor tropism, and maintain stem cell character, while allowing in vivo cellular MRI tracking at 7 Tesla, to monitor their real-time migration and distribution at brain tumor sites. Of significance, this work directly supports the use of FE-Pro-labeled NSCs for real-time tracking in the clinical trial under development: "A Pilot Feasibility Study of Oral 5-Fluorocytosine and Genetically modified Neural Stem Cells Expressing Escherichia coli Cytosine Deaminase for Treatment of Recurrent High-Grade Gliomas".


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Hierro/metabolismo , Imagen por Resonancia Magnética/métodos , Neuronas/citología , Células Madre/citología , Animales , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Dextranos , Modelos Animales de Enfermedad , Óxido Ferrosoférrico/química , Glioma/metabolismo , Humanos , Nanopartículas de Magnetita , Ratones , Trasplante de Neoplasias , Protaminas/química
8.
Stem Cells ; 26(6): 1406-13, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18403751

RESUMEN

Human neural and mesenchymal stem cells have been identified for cell-based therapies in regenerative medicine and as vehicles for delivering therapeutic agents to areas of injury and tumors. However, the signals required for homing and recruitment of stem cells to these sites are not well understood. Urokinase plasminogen activator (uPA) and urokinase plasminogen activator receptor (uPAR) are involved in chemotaxis and cell guidance during normal development and are upregulated in invasive tumors. Here we provided evidence that activation of uPA and uPAR in malignant solid tumors (brain, lung, prostate, and breast) augments neural and mesenchymal stem cell tropism. Expression levels of uPAR on human solid tumor cell lines correlated with levels of uPA and soluble uPAR in tumor cell-conditioned media. Cytokine expression profiles of these tumor-conditioned media were determined by protein arrays. Among 79 cytokines investigated, interleukin (IL)-6, IL-8, and monocyte chemoattractant protein-1 were the most highly expressed cytokines in uPAR-positive tumors. We provided evidence that human recombinant uPA induced stem cell migration, whereas depletion of uPA from PC-3 prostate cancer cell-conditioned medium blocked stem cell migration. Furthermore, retrovirus-mediated overexpression of uPA and uPAR in neuroblastoma (NB1691) cells induced robust migration of stem cells toward NB1691 cell-conditioned media, compared with media derived from wild-type NB1691 cells. We conclude that expression of uPA and uPAR in cancer cells underlies a novel mechanism of stem cell tropism to malignant solid tumors, which may be important for development of optimal stem cell-based therapies. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Neoplasias/fisiopatología , Receptores de Superficie Celular/fisiología , Células Madre/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Neoplasias Encefálicas/fisiopatología , Neoplasias de la Mama/fisiopatología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/fisiopatología , Masculino , Mesencéfalo/embriología , Mesencéfalo/fisiopatología , Células Madre Mesenquimatosas/citología , Neuroblastoma , Reacción en Cadena de la Polimerasa , Neoplasias de la Próstata/fisiopatología , Receptores de Superficie Celular/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Células Madre/citología , Activador de Plasminógeno de Tipo Uroquinasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...