Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Eur J Nucl Med Mol Imaging ; 50(2): 287-301, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36271158

RESUMEN

BACKGROUND: ZED8 is a novel monovalent antibody labeled with zirconium-89 for the molecular imaging of CD8. This work describes nonclinical studies performed in part to provide rationale for and to inform expectations in the early clinical development of ZED8, such as in the studies outlined in clinical trial registry NCT04029181 [1]. METHODS: Surface plasmon resonance, X-ray crystallography, and flow cytometry were used to characterize the ZED8-CD8 binding interaction, its specificity, and its impact on T cell function. Immuno-PET with ZED8 was assessed in huCD8+ tumor-bearing mice and in non-human primates. Plasma antibody levels were measured by ELISA to determine pharmacokinetic parameters, and OLINDA 1.0 was used to estimate radiation dosimetry from image-derived biodistribution data. RESULTS: ZED8 selectively binds to human CD8α at a binding site approximately 9 Å from that of MHCI making mutual interference unlikely. The equilibrium dissociation constant (KD) is 5 nM. ZED8 binds to cynomolgus CD8 with reduced affinity (66 nM) but it has no measurable affinity for rat or mouse CD8. In a series of lymphoma xenografts, ZED8 imaging was able to identify different CD8 levels concordant with flow cytometry. In cynomolgus monkeys with tool compound 89Zr-aCD8v17, lymph nodes were conspicuous by imaging 24 h post-injection, and the pharmacokinetics suggested a flat-fixed first-in-human dose of 4 mg per subject. The whole-body effective dose for an adult human was estimated to be 0.48 mSv/MBq, comparable to existing 89Zr immuno-PET reagents. CONCLUSION: 89Zr immuno-PET with ZED8 appears to be a promising biomarker of tissue CD8 levels suitable for clinical evaluation in cancer patients eligible for immunotherapy.


Asunto(s)
Neoplasias , Tomografía de Emisión de Positrones , Adulto , Humanos , Ratones , Ratas , Animales , Tomografía de Emisión de Positrones/métodos , Indicadores y Reactivos/uso terapéutico , Distribución Tisular , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Inmunoterapia/métodos , Circonio/química , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral
3.
JCI Insight ; 5(7)2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32271166

RESUMEN

Systemic cytokine release and on-target/off-tumor toxicity to normal tissues are the main adverse effects limiting the clinical utility of T cell-redirecting therapies. This study was designed to determine how binding affinity for CD3 and tumor target HER2 impact the efficacy and nonclinical safety of anti-HER2/CD3 T cell-dependent antibodies (TDBs). Affinity was found to be a major determinant for the overall tolerability. Higher affinity for CD3 associated with rapidly elevated peripheral cytokine concentrations, weight loss in mice, and poor tolerability in cynomolgus monkeys. A TDB with lower CD3 affinity was better tolerated in cynomolgus monkeys compared with a higher CD3-affinity TDB. In contrast to tolerability, T cell binding affinity had only limited impact on in vitro and in vivo antitumor activity. High affinity for HER2 was critical for the tumor-killing activity of anti-HER2/CD3 TDBs, but higher HER2 affinity also associated with a more severe toxicity profile, including cytokine release and damage to HER2-expressing tissues. The tolerability of the anti-HER2/CD3 was improved by implementing a dose-fractionation strategy. Fine-tuning the affinities for both the tumor target and CD3 is likely a valuable strategy for achieving maximal therapeutic index of CD3 bispecific antibodies.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Afinidad de Anticuerpos , Antineoplásicos Inmunológicos/inmunología , Receptor ErbB-2/inmunología , Animales , Anticuerpos Biespecíficos/química , Antineoplásicos Inmunológicos/química , Complejo CD3/química , Células CHO , Cricetulus , Evaluación Preclínica de Medicamentos , Humanos , Macaca fascicularis , Receptor ErbB-2/química
4.
Cytokine X ; 2(4): 100042, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33458650

RESUMEN

Immunomodulatory therapeutics such as monoclonal antibodies (mAb) carry an inherent risk of undesired immune reactions. One such risk is cytokine release syndrome (CRS), a rapid systemic inflammatory response characterized by the secretion of pro-inflammatory cytokines from immune cells. It is crucial for patient safety to correctly identify potential risk of CRS prior to first-in-human dose administration. For this purpose, a variety of in vitro cytokine release assays (CRA) are routinely used as part of the preclinical safety assessment of novel therapeutic mAbs. One of the challenges for the development and comparison of CRA performance is the lack of availability of standard positive and negative control mAbs for use in assay qualification. To address this issue, the National Institute for Biological Standards and Control (NIBSC) developed a reference panel of lyophilised mAbs known to induce CRS in the clinic: human anti-CD52, mouse anti-CD3 and human superagonistic (SA) anti-CD28 mAb manufactured according to the respective published sequences of Campath-1H® (alemtuzumab, IgG1) , Orthoclone OKT-3® (muromonab, IgG2a) and TGN1412 (theralizumab, IgG4), as well as three isotype matched negative controls (human IgG1, mouse IgG2a and human IgG4, respectively). The relative capacity of these control mAbs to stimulate the release of IFN-γ, IL-2, TNF-α and IL-6 in vitro was evaluated in eleven laboratories in an international collaborative study mediated through the HESI Immuno-safety Technical Committee Cytokine Release Assay Working Group. Participants tested the NIBSC mAbs in a variety of CRA platforms established at each institution. This paper presents the results from the centralised cytokine quantification on all the plasma/supernatants corresponding to the stimulation of immune cells in the different CRA platforms by a single concentration of each mAb. Each positive control mAb induced significant cytokine release in most of the tested CRA platforms. There was a high inter-laboratory variability in the levels of cytokines produced, but similar patterns of response were observed across laboratories that replicated the cytokine release patterns previously published for the respective clinical therapeutic mAbs. Therefore, the positive and negative mAbs are suitable as a reference panel for the qualification and validation of CRAs, comparison of different CRA platforms (e.g. solid vs aqueous phase), and intra- and inter-laboratory comparison of CRA performance. Thus, the use of this panel of positive and negative control mAbs will increase the confidence in the robustness of a CRA platform to identify a potential CRS risk for novel immunomodulatory therapeutic candidates.

5.
Toxicol In Vitro ; 50: 109-123, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29427706

RESUMEN

Neurotoxicity is a major concern during drug development, and together with liver and cardio-toxicity, it is one of the main causes of clinical drug attrition. Current pre-clinical models may not sufficiently identify and predict the risk for central or peripheral nervous system toxicity. One such example is clinically dose-limiting neuropathic effects after the administration of chemotherapeutic agents. Thus, the need to establish novel in vitro tools to evaluate the risk of neurotoxicities, such as neuropathy, remains unmet in drug discovery. Though in vitro studies have been conducted using primary and immortalized cell lines, some limitations include the utility for higher throughput methodologies, method reproducibility, and species extrapolation. As a novel alternative, human induced-pluripotent stem cell (iPSC)-derived neurons appear promising for testing new drug candidates. These iPSC-derived neurons are readily available and can be manipulated as required. Here, we describe a novel approach to assess neurotoxicity caused by different classes of chemotherapeutics using kinetic monitoring of neurite dynamic changes and apoptosis in human iPSC-neurons. These studies show promising changes in neurite dynamics in response to clinical inducers of neuropathy, as well as the ability to rank-order and gather mechanistic insight into class-specific compound induced neurotoxicity. This platform can be utilized in early drug development, as part of a weight of evidence approach, to screen drug candidates, and potentially reduce clinical attrition due to neurotoxicity.


Asunto(s)
Antineoplásicos/toxicidad , Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes Inducidas/citología , Neuronas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Neuritas/efectos de los fármacos , Neuronas/metabolismo , Síndromes de Neurotoxicidad
6.
Invest Ophthalmol Vis Sci ; 58(10): 4274-4285, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28850638

RESUMEN

Purpose: Poly(lactic-co-glycolic) acid (PLGA) inserts have been successfully developed for the treatment of posterior eye disease as a means of reducing injection frequency of intravitreally administered therapeutics. PLGA microspheres are also of interest for the delivery of intravitreal drugs, since they offer the advantage of being easily injected without surgical procedures or large injectors. Methods: In the current study, the toxicity of PLGA microspheres and rods was investigated in nonhuman primates (NHPs) and rabbits. An in vitro assessment of cytokine responses to PLGA in peripheral blood mononuclear cells (PBMCs) and macrophages was also performed. Results: Intravitreal administration of 3, 10, or 12.5 mg/eye of PLGA microspheres in NHPs resulted in a severe immune response characterized by a foreign body response. Follow-up studies in the rabbit confirmed this finding for PLGA microspheres ranging in size from 20 to 100 µm. In contrast, administration of PLGA rod implants with a similar PLGA mass did not elicit a significant immune response. In vitro assays in PBMCs and macrophages confirmed proinflammatory cytokine release upon treatment with PLGA microspheres but not PLGA rods. Conclusions: These data demonstrate a lack of tolerability of PLGA microspheres upon intravitreal injection, and suggest that the size, shape, and/or surface area of PLGA depots are critical attributes in determining ocular toxicity.


Asunto(s)
Materiales Biocompatibles/toxicidad , Sistemas de Liberación de Medicamentos/efectos adversos , Ácido Láctico/toxicidad , Microesferas , Ácido Poliglicólico/toxicidad , Animales , Materiales Biocompatibles/administración & dosificación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Reacción a Cuerpo Extraño/metabolismo , Reacción a Cuerpo Extraño/patología , Inyecciones Intravítreas , Ácido Láctico/administración & dosificación , Macrófagos/metabolismo , Ácido Poliglicólico/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Conejos , Retina/efectos de los fármacos , Retina/patología , Cuerpo Vítreo/efectos de los fármacos , Cuerpo Vítreo/patología
7.
J Pharmacol Exp Ther ; 360(1): 226-238, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27821712

RESUMEN

Bruton's tyrosine kinase (BTK) is a member of the Tec family of cytoplasmic tyrosine kinases involved in B-cell and myeloid cell signaling. Small molecule inhibitors of BTK are being investigated for treatment of several hematologic cancers and autoimmune diseases. GDC-0853 ((S)-2-(3'-(hydroxymethyl)-1-methyl-5-((5-(2-methyl-4-(oxetan-3-yl)piperazin-1-yl)pyridin-2-yl)amino)-6-oxo-1,6-dihydro-[3,4'-bipyridin]-2'-yl)-7,7-dimethyl-3,4,7,8-tetrahydro-2H-cyclopenta[4,5]pyrrolo[1,2-a]pyrazin-1(6H)-one) is a selective and reversible oral small-molecule BTK inhibitor in development for the treatment of rheumatoid arthritis and systemic lupus erythematosus. In Sprague-Dawley (SD) rats, administration of GDC-0853 and other structurally diverse BTK inhibitors for 7 days or longer caused pancreatic lesions consisting of multifocal islet-centered hemorrhage, inflammation, fibrosis, and pigment-laden macrophages with adjacent lobular exocrine acinar cell atrophy, degeneration, and inflammation. Similar findings were not observed in mice or dogs at much higher exposures. Hemorrhage in the peri-islet vasculature emerged between four and seven daily doses of GDC-0853 and was histologically similar to spontaneously occurring changes in aging SD rats. This suggests that GDC-0853 could exacerbate a background finding in younger animals. Glucose homeostasis was dysregulated following a glucose challenge; however, this occurred only after 28 days of administration and was not directly associated with onset or severity of pancreatic lesions. There were no changes in other common serum biomarkers assessing endocrine and exocrine pancreatic function. Additionally, these lesions were not readily detectable via Doppler ultrasound, computed tomography, or magnetic resonance imaging. Our results indicate that pancreatic lesions in rats are likely a class effect of BTK inhibitors, which may exacerbate an islet-centered pathology that is unlikely to be relevant to humans.


Asunto(s)
Páncreas/efectos de los fármacos , Piperazinas/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Piridonas/toxicidad , Pirroles/toxicidad , Agammaglobulinemia Tirosina Quinasa , Animales , Perros , Relación Dosis-Respuesta a Droga , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Humanos , Masculino , Ratones , Páncreas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Ratas , Especificidad de la Especie
8.
Thromb Res ; 140 Suppl 1: S27-36, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27067975

RESUMEN

Metastasis is the main cause of death in cancer patients, and understanding mechanisms that control tumor cell dissemination may lead to improved therapy. Tumor cell adhesion receptors contribute to cancer spreading. We noted earlier that tumor cells can expressing the adhesion receptor integrin αvß3 in distinct states of activation, and found that cells which metastasize from the blood stream express it in a constitutively high affinity form. Here, we analyzed steps of the metastatic cascade in vivo and asked, when and how the affinity state of integrin αvß3 confers a critical advantage to cancer spreading. Following tumor cells by real time PCR, non-invasive bioluminescence imaging, intravital microscopy and histology allowed us to identify tumor cell extravasation from the blood stream as a rate-limiting step supported by high affinity αvß3. Successful transendothelial migration depended on cooperation between tumor cells and platelets involving the high affinity tumor cell integrin and release of platelet granules. Thus, this study identifies the high affinity conformer of integrin αvß3 and its interaction with platelets as critical for early steps during hematogenous metastasis and target for prevention of metastatic disease.


Asunto(s)
Plaquetas/patología , Integrina alfaVbeta3/metabolismo , Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes/patología , Animales , Plaquetas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Humanos , Integrina alfaVbeta3/análisis , Ratones SCID , Células Neoplásicas Circulantes/metabolismo
9.
Chem Biol ; 18(3): 299-303, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21439474

RESUMEN

We report a strategy for the generation of heterodimeric protein conjugates using an unnatural amino acid with orthogonal reactivity. This paper addresses the challenges of site-specificity and homogeneity with respect to the synthesis of bivalent proteins and antibody-drug conjugates. There are numerous antibody-drug conjugates in preclinical and clinical development, yet these are based either on nonspecific lysine coupling chemistry or on disulfide modification made difficult by the large number of cysteines in antibodies. Here, we describe a recombinant approach that can be used to rapidly generate a variety of constructs with defined conjugation sites. Moreover, this methodology results in homogeneous antibody conjugates whose biological, physical, and pharmacological properties can be quantitatively assessed and subsequently optimized. As proof of concept, we have generated anti-Her2 Fab-Saporin conjugates that demonstrate excellent potency in vitro.


Asunto(s)
Anticuerpos Monoclonales/química , Antineoplásicos Fitogénicos/química , Inmunotoxinas/química , Fenilalanina/análogos & derivados , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados , Antineoplásicos Fitogénicos/inmunología , Antineoplásicos Fitogénicos/toxicidad , Línea Celular Tumoral , Dimerización , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Inmunotoxinas/inmunología , Inmunotoxinas/toxicidad , Maleimidas/química , Fenilalanina/química , Receptor ErbB-2/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Saporinas , Trastuzumab
10.
J Mol Biol ; 406(4): 595-603, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21237172

RESUMEN

Immunoconjugates and multispecific antibodies are rapidly emerging as highly potent experimental therapeutics against cancer. We have developed a method to incorporate an unnatural amino acid, p-acetylphenylalanine (pAcPhe) into an antibody antigen binding fragment (Fab) targeting HER2 (human epidermal growth factor receptor 2), allowing site-specific labeling without disrupting antigen binding. Expression levels of the pAcPhe-containing proteins were comparable to that of wild-type protein in shake-flask and fermentation preparations. The pAcPhe-Fabs were labeled by reaction with hydroxylamine dye and biotin species to produce well-defined, singly conjugated Fabs. We then coupled a hydroxylamine biotin to the pAcPhe-Fab and demonstrated controlled assembly of Fabs in the presence of the tetrameric biotin-binding protein, NeutrAvidin. The position of Fab biotinylation dictates the geometry of multimer assembly, producing unique multimeric Fab structures. These assembled Fab multimers differentially attenuate Her2 phosphorylation in breast cancer cells that overexpress the Her2 receptor. Thus, an encoded unnatural amino acid produces a chemical "handle" by which immunoconjugates and multimers can be engineered.


Asunto(s)
Aminoácidos/metabolismo , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Fenilalanina/análogos & derivados , Multimerización de Proteína , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Cinética , Modelos Químicos , Modelos Moleculares , Fenilalanina/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Receptor ErbB-2/inmunología
11.
Clin Exp Metastasis ; 27(4): 217-31, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20225083

RESUMEN

Advanced metastatic disease is difficult to manage and specific therapeutic targets are rare. We showed earlier that metastatic breast cancer cells use the activated conformer of adhesion receptor integrin alphavbeta3 for dissemination. We now investigated if targeting this form of the receptor can impact advanced metastatic disease, and we analyzed the mechanisms involved. Treatment of advanced multi-organ metastasis in SCID mice with patient-derived scFv antibodies specific for activated integrin alphavbeta3 caused stagnation and regression of metastatic growth. The antibodies specifically localized to tumor lesions in vivo and inhibited alphavbeta3 ligand binding at nanomolar levels in vitro. At the cellular level, the scFs associated rapidly with high affinity alphavbeta3 and dissociated extremely slowly. Thus, the scFvs occupy the receptor on metastatic tumor cells for prolonged periods of time, allowing for inhibition of established cell interaction with natural alphavbeta3 ligands. Potential apoptosis inducing effects of the antibodies through interaction with caspase-3 were studied as potential additional mechanism of treatment response. However, in contrast to a previous concept, neither the RGD-containing ligand mimetic scFvs nor RGD peptides bound or activated caspase-3 at the cellular or molecular level. This indicates that the treatment effects seen in the animal model are primarily due to antibody interference with alphavbeta3 ligation. Inhibition of advanced metastatic disease by treatment with cancer patient derived single chain antibodies against the activated conformer of integrin alphavbeta3 identifies this form of the receptor as a suitable target for therapy.


Asunto(s)
Integrina alfaVbeta3/antagonistas & inhibidores , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/patología , Anticuerpos de Cadena Única/uso terapéutico , Animales , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Humanos , Integrina alfaVbeta3/inmunología , Integrina alfaVbeta3/metabolismo , Ratones , Ratones SCID , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/prevención & control , Anticuerpos de Cadena Única/inmunología
12.
Proc Natl Acad Sci U S A ; 106(26): 10666-71, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19541645

RESUMEN

The incidence of brain metastasis is rising and poses a severe clinical problem, as we lack effective therapies and knowledge of mechanisms that control metastatic growth in the brain. Here we demonstrate a crucial role for high-affinity tumor cell integrin alpha(v)beta(3) in brain metastatic growth and recruitment of blood vessels. Although alpha(v)beta(3) is frequently up-regulated in primary brain tumors and metastatic lesions of brain homing cancers, we show that it is the alpha(v)beta(3) activation state that is critical for brain lesion growth. Activated, but not non-activated, tumor cell alpha(v)beta(3) supports efficient brain metastatic growth through continuous up-regulation of vascular endothelial growth factor (VEGF) protein under normoxic conditions. In metastatic brain lesions carrying activated alpha(v)beta(3), VEGF expression is controlled at the post-transcriptional level and involves phosphorylation and inhibition of translational respressor 4E-binding protein (4E-BP1). In contrast, tumor cells with non-activated alpha(v)beta(3) depend on hypoxia for VEGF induction, resulting in reduced angiogenesis, tumor cell apoptosis, and inefficient intracranial growth. Importantly, the microenvironment critically influences the effects that activated tumor cell alpha(v)beta(3) exerts on tumor cell growth. Although it strongly promoted intracranial growth, the activation state of the receptor did not influence tumor growth in the mammary fat pad as a primary site. Thus, we identified a mechanism by which metastatic cells thrive in the brain microenvironment and use the high-affinity form of an adhesion receptor to grow and secure host support for proliferation. Targeting this molecular mechanism could prove valuable for the inhibition of brain metastasis.


Asunto(s)
Neoplasias Encefálicas/secundario , Integrina alfaVbeta3/metabolismo , Neoplasias Mamarias Experimentales/patología , Neovascularización Patológica/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hipoxia , Etiquetado Corte-Fin in Situ , Integrina alfaVbeta3/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones SCID , Mutación , Trasplante de Neoplasias , Fosfoproteínas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , Carga Tumoral , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
BMC Cancer ; 9: 206, 2009 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-19558675

RESUMEN

BACKGROUND: Glioblastoma are highly aggressive tumors with an average survival time of 12 months with currently available treatment. We have previously shown that specific embryonic neural progenitor cells (NPC) have the potential to target glioma growth in the CNS of rats. The neural progenitor cell treatment can cure approximately 40% of the animals with malignant gliomas with no trace of a tumor burden 6 months after finishing the experiment. Furthermore, the NPCs have been shown to respond to signals from the tumor environment resulting in specific migration towards the tumor. Based on these results we wanted to investigate what factors could influence the growth and progression of gliomas in our rodent model. METHODS: Using microarrays we screened for candidate genes involved in the functional mechanism of tumor inhibition by comparing glioma cell lines to neural progenitor cells with or without anti-tumor activity. The expression of candidate genes was confirmed at RNA level by quantitative RT-PCR and at the protein level by Western blots and immunocytochemistry. Moreover, we have developed in vitro assays to mimic the antitumor effect seen in vivo. RESULTS: We identified several targets involved in glioma growth and migration, specifically CXCL1, CD81, TPT1, Gas6 and AXL proteins. We further showed that follistatin secretion from the NPC has the potential to decrease tumor proliferation. In vitro co-cultures of NPC and tumor cells resulted in the inhibition of tumor growth. The addition of antibodies against proteins selected by gene and protein expression analysis either increased or decreased the proliferation rate of the glioma cell lines in vitro. CONCLUSION: These results suggest that these identified factors might be useful starting points for performing future experiments directed towards a potential therapy against malignant gliomas.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Glioma/metabolismo , Glioma/terapia , Neuronas/citología , Células Madre/citología , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inmunohistoquímica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Resultado del Tratamiento , Proteína Tumoral Controlada Traslacionalmente 1
14.
Exp Neurol ; 219(1): 363-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19427853

RESUMEN

Experimental stroke and excitotoxic brain lesion to the striatum increase the proliferation of cells residing within the ventricular wall and cause subsequent migration of newborn neuroblasts into the lesioned brain parenchyma. In this study, we clarify the different events of neurogenesis following striatal or cortical excitotoxic brain lesions in adult rats. Newborn cells were labeled by intraperitoneal injection of bromo-deoxy-uridine (BrdU), or by green fluorescent protein (GFP)-expressing lentiviral vectors injected into the subventricular zone (SVZ). We show that only neural progenitors born the first 5 days in the SVZ reside and expand within this neurogenic niche over time, and that these early labeled cells are more prone to migrate towards the striatum as neuroblasts. However, these neuroblasts could not mature into NeuN+ neurons in the striatum. Furthermore, we found that cortical lesions, close or distant from the SVZ, could not upregulate SVZ cell proliferation nor promote neurogenesis. Our study demonstrates that both the time window for labeling proliferating cells and the site of lesion are crucial when assessing neurogenesis following brain injury.


Asunto(s)
Lesión Encefálica Crónica/fisiopatología , Cuerpo Estriado/fisiopatología , Neurogénesis/fisiología , Neuronas/fisiología , Neurotoxinas/toxicidad , Células Madre/fisiología , Animales , Biomarcadores , Lesión Encefálica Crónica/inducido químicamente , Bromodesoxiuridina , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiopatología , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Neuronas/citología , Fenotipo , Ratas , Recuperación de la Función/fisiología , Regeneración/fisiología , Células Madre/citología , Factores de Tiempo , Regulación hacia Arriba/fisiología
15.
J Neurosci Res ; 85(10): 2147-59, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17526014

RESUMEN

Gliomas are the most common primary brain tumors and offer a poor prognosis in patients because of their infiltrative and treatment-resistant nature. The median survival time after diagnosis is approximately 11-12 months. There is a strong need for novel treatment modalities in targeting gliomas, and recent advances use neural progenitor cells as delivery systems for different therapeutic strategies. In this study, we show that rat embryonic neural progenitor cell (NPC) lines, transplanted at a distant site from a 3-day-preestablished glioma in the striatum, were able to migrate toward and colocalize with tumor isles without general spread into the brain parenchyma. Upon encounter with tumor, neural progenitor cells changed phenotype and became vimentin positive. These results demonstrate that transplanted neural progenitor cells respond to queues from a tumor and home to and exert an antitumor effect on the preestablished glioma, significantly decreasing the tumor volume with approximately 67% compared with control tumors after 1-2 weeks. Moreover, these early effects could be translated into increased survival times of animals treated with neural progenitor cell grafts 3 days after intrastriatal tumor inoculation. In contrast, there was no activation or migration of endogenous subventricular zone (SVZ) neuroblasts in response to an intrastriatal syngeneic tumor. In conclusion, NPC possess the ability to influence tumor growth as well as respond to queues from the tumor or tumor microenvironment, demonstrating a cross-talk between the cells.


Asunto(s)
Neoplasias Encefálicas/cirugía , Comunicación Celular , Glioma/cirugía , Neuronas/citología , Trasplante de Células Madre , Células Madre/citología , Células Madre/fisiología , Animales , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Cuerpo Estriado , Glioma/patología , Glioma/fisiopatología , Fenotipo , Ratas , Células Madre/metabolismo , Vimentina/metabolismo
16.
Exp Cell Res ; 312(8): 1265-76, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16434036

RESUMEN

We have earlier shown that the rat neural progenitor cell line HiB5 is capable of suppressing intracranial growth of glioma cells in Fisher rats. Unlike some neural progenitor cells, HiB5 cells have not shown homing capacity towards glioma cells growing intracranially. In this study, we have genetically modified HiB5 progenitor cells to over-express the chemokine receptor CXCR3. We show that the introduced receptor is functionally responding to ligand stimulation with increased phosphorylation levels of ERK and SAPK/JNK and a transcriptional response of an AP-1 reporter system introduced into HIB5 cells. These transfected progenitor cells migrate in vitro in response to IP-10 and I-TAC. Further, we show an enhanced in vivo migration of the CXCR3 transfected HiB5 cells over the corpus callosum towards an IP-10 and I-TAC expressing glioma, as compared to wild type HiB5 cells. Our data indicate that it is possible to take advantage of chemokines natural capacity to initiate migratory responses, and to use this ability to enhance tumor-inhibitory neural progenitor cells to target an intracranially growing glioma.


Asunto(s)
Neoplasias Encefálicas/inmunología , Movimiento Celular/inmunología , Quimiocinas/inmunología , Glioma/inmunología , Células Madre/inmunología , Animales , Neoplasias Encefálicas/fisiopatología , Neoplasias Encefálicas/terapia , Comunicación Celular/efectos de los fármacos , Comunicación Celular/inmunología , Línea Celular , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL10 , Quimiocina CXCL11 , Quimiocinas/metabolismo , Quimiocinas/farmacología , Quimiocinas CXC/inmunología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Marcación de Gen/métodos , Terapia Genética/métodos , Glioma/fisiopatología , Glioma/terapia , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Invasividad Neoplásica/fisiopatología , Invasividad Neoplásica/prevención & control , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores CXCR3 , Receptores de Quimiocina/agonistas , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/inmunología , Células Tumorales Cultivadas
17.
Cancer Res ; 64(15): 5347-54, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15289341

RESUMEN

Current therapies for gliomas often fail to address their infiltrative nature. Conventional treatments leave behind small clusters of neoplastic cells, resulting in eventual tumor recurrence. In the present study, we have evaluated the antitumor activity of neural progenitor cells against gliomas when stereotactically injected into nucleus Caudatus of Fisher rats. We show that the rat neural progenitor cell lines HiB5 and ST14A, from embryonic hippocampus and striatum primordium, respectively, are able to prolong animal survival and, in 25% of the cases, completely inhibit the outgrowth of N29 glioma compared with control animals. Delayed tumor outgrowth was also seen when HiB5 cells were inoculated at the site of tumor growth 1 week after tumor inoculation or when a mixture of tumor cells and HiB5 cells were injected s.c. into Fisher rats. HiB5 cells were additionally coinoculated together with two alternative rat gliomas, N32 and N25. N32 was growth inhibited, but rats inoculated with N25 cells did not show a prolonged survival. To evaluate the possibility of the involvement of the immune system in the tumor outgrowth inhibition, we show that HiB5 cells do not evoke an immune response when injected into Fisher rats. Furthermore, the rat neural progenitor cells produce all transforming growth factor beta isotypes, which could explain the observed immunosuppressive nature of these cells. Hence, some neural progenitor cells have the ability to inhibit tumor outgrowth when implanted into rats. These results indicate the usefulness of neural stem cells as therapeutically effective cells for the treatment of intracranial tumors.


Asunto(s)
Neoplasias Encefálicas/prevención & control , Neoplasias del Colon/prevención & control , Glioma/prevención & control , Neuronas/fisiología , Células Madre/fisiología , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Cuerpo Estriado/patología , Glioma/inmunología , Glioma/patología , Hipocampo/patología , Humanos , Inmunosupresores/metabolismo , Masculino , Neuronas/citología , Neuronas/trasplante , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Células Madre/citología , Tasa de Supervivencia , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...