Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(7): e0301664, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38985719

RESUMEN

Influenza viruses constitute a major threat to human health globally. The viral surface glycoprotein hemagglutinin (HA) is the immunodominant antigen, contains the site for binding to the cellular receptor (RBS), and it is the major target of neutralizing antibody responses post-infection. We developed llama-derived single chain antibody fragments (VHHs) specific for type A influenza virus. Four VHHs were identified and further characterized. VHH D81 bound residues in the proximity of the C-terminal region of HA1 of H1 and H5 subtypes, and showed weak neutralizing activity, whereas VHH B33 bound residues in the proximity of the N-terminal region of the HA's stem domain (HA2) of H1, H5, and H9 subtypes, and showed no neutralizing activity. Of most relevance, VHHs E13 and G41 recognized highly conserved conformational epitopes on the H1 HA's globular domain (HA1) and showed high virus neutralizing activity (ranging between 0.94 to 0.01µM), when tested against several human H1N1 isolates. Additionally, E13 displayed abrogated virus replication of a panel of H1N1 strains spanning over 80 years of antigenic drift and isolated from human, avian, and swine origin. Interestingly, E13 conferred protection in vivo at a dose as low as 0.05 mg/kg. Mice treated with E13 intranasally resulted in undetectable virus challenge loads in the lungs at day 4 post-challenge. The transfer of sterilizing pan-H1 immunity, by a dose in the range of micrograms given intranasally, is of major significance for a monomeric VHH and supports the further development of E13 as an immunotherapeutic agent for the mitigation of influenza infections.


Asunto(s)
Anticuerpos Neutralizantes , Camélidos del Nuevo Mundo , Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Anticuerpos de Dominio Único , Animales , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Anticuerpos de Dominio Único/inmunología , Anticuerpos Neutralizantes/inmunología , Ratones , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Camélidos del Nuevo Mundo/inmunología , Anticuerpos Antivirales/inmunología , Femenino , Gripe Humana/inmunología , Gripe Humana/prevención & control , Gripe Humana/virología , Epítopos/inmunología , Perros , Ratones Endogámicos BALB C
2.
Biotechnol Bioeng ; 118(9): 3348-3358, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33624832

RESUMEN

Single-domain antibodies (sdAbs) offer the affinity and therapeutic value of conventional antibodies, with increased stability and solubility. Unlike conventional antibodies, however, sdAbs do not benefit from a platform manufacturing process. While successful production of a variety of sdAbs has been shown in numerous hosts, purification methods are often molecule specific or require affinity tags, which generally cannot be used in clinical manufacturing due to regulatory concerns. Here, we have developed a broadly applicable production and purification process for sdAbs in Komagataella phaffii (Pichia pastoris) and demonstrated the production of eight different sdAbs at a quality appropriate for nonclinical studies. We developed a two-step, integrated purification process without the use of affinity resins and showed that modification of a single process parameter, pH of the bridging buffer, was required for the successful purification of a variety of sdAbs. Further, we determined that this parameter can be predicted based only on the biophysical characteristics of the target molecule. Using these methods, we produced nonclinical quality sdAbs as few as 5 weeks after identifying the product sequence. Nonclinical studies of three different sdAbs showed that molecules produced using our platform process conferred protection against viral shedding of rotavirus or H1N1 influenza and were equivalent to similar molecules produced in Escherichia coli and purified using affinity tags.


Asunto(s)
Anticuerpos Antivirales , Subtipo H1N1 del Virus de la Influenza A/inmunología , Rotavirus/inmunología , Saccharomycetales/crecimiento & desarrollo , Anticuerpos de Cadena Única , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/aislamiento & purificación
3.
Mol Neurobiol ; 55(11): 8637-8650, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29582397

RESUMEN

The specific roles of Notch in progressive adulthood neurodegenerative disorders have begun to be unraveled in recent years. A number of independent studies have shown significant increases of Notch expression in brains from patients at later stages of sporadic Alzheimer's disease (AD). However, the impact of Notch canonical signaling activation in the pathophysiology of AD is still elusive. To further investigate this issue, 2-month-old wild-type (WT) and hemizygous McGill-R-Thy1-APP rats (Tg(+/-)) were injected in CA1 with lentiviral particles (LVP) expressing the transcriptionally active fragment of Notch, known as Notch Intracellular Domain (NICD), (LVP-NICD), or control lentivirus particles (LVP-C). The Tg(+/-) rat model captures presymptomatic aspects of the AD pathology, including intraneuronal amyloid beta (Aß) accumulation and early cognitive deficits. Seven months after LVP administration, Morris water maze test was performed, and brains isolated for biochemical and histological analysis. Our results showed a learning impairment and a worsening of spatial memory in LVP-NICD- as compared to LVP-C-injected Tg(+/-) rats. In addition, immuno histochemistry, ELISA multiplex, Western blot, RT-qPCR, and 1H-NMR spectrometry of cerebrospinal fluid (CSF) indicated that chronic expression of NICD promoted hippocampal vessel thickening with accumulation of Aß in brain microvasculature, alteration of blood-brain barrier (BBB) permeability, and a decrease of CSF glucose levels. These findings suggest that, in the presence of early Aß pathology, expression of NICD may contribute to the development of microvascular abnormalities, altering glucose transport at the BBB with impact on early decline of spatial learning and memory.


Asunto(s)
Enfermedad de Alzheimer/patología , Vasos Sanguíneos/patología , Glucosa/metabolismo , Hipocampo/metabolismo , Trastornos de la Memoria/patología , Receptores Notch/química , Receptores Notch/metabolismo , Memoria Espacial , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/fisiopatología , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Modelos Animales de Enfermedad , Vectores Genéticos/metabolismo , Células HEK293 , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Inflamación/patología , Lentivirus/genética , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/fisiopatología , Microvasos/patología , Dominios Proteicos , Espectroscopía de Protones por Resonancia Magnética , Ratas Transgénicas , Ratas Wistar
4.
Mol Neurobiol ; 55(2): 1068-1081, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28092084

RESUMEN

When disrupted, iron homeostasis negatively impacts oligodendrocyte (OLG) differentiation and impairs myelination. To better understand myelin formation and OLG maturation, in vivo and in vitro studies were conducted to evaluate the effect of iron deficiency (ID) not only on OLG maturation but also on astrocytes (AST) and microglial cells (MG). In vivo experiments in an ID model were carried out to describe maturational events during OLG and AST development and the reactive profile of MG during myelination when iron availability is lower than normal. In turn, in vitro assays were conducted to explore proliferating and maturational states of each glial cell type derived from control or ID conditions. Studies targeted NG2, PDGFRα, CNPAse, CC1, and MBP expression in OLG, GFAP and S100 expression in AST, and CD11b, ED1, and cytokine expression in MG, as well as BrDU incorporation in the three cell types. Our results show that ID affected OLG development at early stages, not only reducing their maturation capacity but also increasing their proliferation and affecting their morphological complexity. AST ID proliferated more than control ones and were more immature, much like OLG. Cytokine expression in ID animals reflected an anti-inflammatory state which probably influenced OLG maturation. These results show that ID conditions alter all glial cells and may impact myelin formation, which could be regulated by a mechanism involving a cross talk between AST, MG, and oligodendrocyte progenitors (OPC).


Asunto(s)
Anemia Ferropénica/metabolismo , Astrocitos/metabolismo , Hierro/metabolismo , Microglía/metabolismo , Oligodendroglía/metabolismo , Animales , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Homeostasis/fisiología , Vaina de Mielina/metabolismo , Ratas , Ratas Wistar
5.
Mol Cell Neurosci ; 67: 75-83, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26051800

RESUMEN

Transforming growth factor beta 1 (TGF-beta1), an anti-inflammatory cytokine, has been shown to have pro-neurogenic effects on adult Neural Stem Cells (aNSC) from the dentate gyrus and in vivo models. Here, we expanded the observation of the pro-neurogenic effect of TGF-beta1 on aNSC from the subventricular zone (SVZ) of adult rats and performed a functional genomic analysis to identify candidate genes to mediate its effect. 10 candidate genes were identified by microarray analysis and further validated by qRT-PCR. Of these, Fibulin-2 was increased 477-fold and its inhibition by siRNA blocks TGF-beta1 pro-neurogenic effect. Curiously, Fibulin-2 was not expressed by aNSC but by a GFAP-positive population in the culture, suggesting an indirect mechanism of action. TGF-beta1 also induced Fibulin-2 in the SVZ in vivo. Interestingly, 5 out of the 10 candidate genes identified are known to interact with integrins, paving the way for exploring their functional role in adult neurogenesis. In conclusion, we have identified 10 genes with putative pro-neurogenic effects, 5 of them related to integrins and provided proof that Fibulin-2 is a major mediator of the pro-neurogenic effects of TGF-beta1. These data should contribute to further exploring the molecular mechanism of adult neurogenesis of the genes identified and the involvement of the integrin pathway on adult neurogenesis.


Asunto(s)
Células Madre Adultas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Factor de Crecimiento Transformador beta1/farmacología , Células Madre Adultas/citología , Células Madre Adultas/efectos de los fármacos , Animales , Astrocitos/metabolismo , Proteínas de Unión al Calcio/genética , Células Cultivadas , Proteínas de la Matriz Extracelular/genética , Integrinas/metabolismo , Ventrículos Laterales/citología , Ventrículos Laterales/crecimiento & desarrollo , Ventrículos Laterales/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Ratas , Ratas Wistar , Factor de Crecimiento Transformador beta1/metabolismo
6.
Clin Cancer Res ; 20(6): 1555-1565, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24501391

RESUMEN

PURPOSE: Glioblastoma multiforme is the most common primary brain cancer in adults. Chemotherapy with temozolomide (TMZ) significantly prolongs the survival of patients with glioblastoma multiforme. However, the three-year survival is still approximately 5%. Herein, we combined intratumoral administration of an adenoviral vector expressing Flt3L (Ad-Flt3L) with systemic temozolomide to assess its impact on therapeutic efficacy. EXPERIMENTAL DESIGN: Wild-type or immunodeficient mice bearing intracranial glioblastoma multiforme or metastatic melanoma were treated with an intratumoral injection of Ad-Flt3L alone or in combination with the conditionally cytotoxic enzyme thymidine kinase (Ad-TK), followed by systemic administration of ganciclovir and temozolomide. We monitored survival and measured the tumor-infiltrating immune cells. RESULTS: Although treatment with temozolomide alone led to a small improvement in median survival, when used in combination with gene therapy-mediated immunotherapy, it significantly increased the survival of tumor-bearing mice. The antitumor effect was further enhanced by concomitant intratumoral administration of Ad-TK, leading to 50% to 70% long-term survival in all tumor models. Although temozolomide reduced the content of T cells in the tumor, this did not affect the therapeutic efficacy. The antitumor effect of Ad-Flt3L+Ad-TK+TMZ required an intact immune system because the treatment failed when administered to knock out mice that lacked lymphocytes or dendritic cells. CONCLUSIONS: Our results challenge the notion that chemotherapy leads to a state of immune-suppression which impairs the ability of the immune system to mount an effective antitumor response. Our work indicates that temozolomide does not inhibit antitumor immunity and supports its clinical implementation in combination with immune-mediated therapies.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/patología , Dacarbazina/análogos & derivados , Glioblastoma/patología , Inmunoterapia/métodos , Adenoviridae , Animales , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Terapia Genética/métodos , Vectores Genéticos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Temozolomida , Timidina Quinasa/genética , Timidina Quinasa/inmunología
7.
BMC Vet Res ; 10: 8, 2014 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24401205

RESUMEN

BACKGROUND: Bovine herpesvirus type 1 (BoHV-1) is the causative agent of respiratory and genital tract infections; causing a high economic loss in all continents. Use of marker vaccines in IBR eradication programs is widely accepted since it allows for protection of the animals against the disease while adding the possibility of differentiating vaccinated from infected animals.The aim of the present study was the development and evaluation of safety and efficacy of a glycoprotein E-deleted (gE-) BoHV-1 marker vaccine strain (BoHV-1ΔgEßgal) generated by homologous recombination, replacing the viral gE gene with the ß-galactosidase (ßgal) gene. RESULTS: In vitro growth kinetics of the BoHV-1ΔgEßgal virus was similar to BoHV-1 LA. The immune response triggered by the new recombinant strain in cattle was characterized both as live attenuated vaccine (LAV) and as an inactivated vaccine. BoHV-1ΔgEßgal was highly immunogenic in both formulations, inducing specific humoral and cellular immune responses. Antibody titers found in animals vaccinated with the inactivated vaccine based on BoHV-1ΔgEßgal was similar to the titers found for the control vaccine (BoHV-1 LA). In the same way, titers of inactivated vaccine groups were significantly higher than any of the LAV immunized groups, independently of the inoculation route (p < 0.001). Levels of IFN-γ were significantly higher (p < 0.001) in those animals that received the LAV compared to those that received the inactivated vaccine. BoHV-1ΔgEßgal exhibited an evident attenuation when administered as a LAV; no virus was detected in nasal secretions of vaccinated or sentinel animals during the post-vaccination period. BoHV-1ΔgEßgal, when used in either formulation, elicited an efficient immune response that protected animals against challenge with virulent wild-type BoHV-1. Also, the deletion of the gE gene served as an immunological marker to differentiate vaccinated animals from infected animals. All animals vaccinated with the BoHV-1ΔgE ßgal strain were protected against disease after challenge and shed significantly less virus than control calves, regardless of the route and formulation they were inoculated. CONCLUSIONS: Based on its attenuation, immunogenicity and protective effect after challenge, BoHV-1ΔgEßgal virus is an efficient and safe vaccine candidate when used either as inactivated or as live attenuated forms.


Asunto(s)
Infecciones por Herpesviridae/veterinaria , Herpesvirus Bovino 1/metabolismo , Proteínas Virales/metabolismo , Vacunas Virales/inmunología , Animales , Bovinos , Línea Celular , Perros , Femenino , Eliminación de Gen , Regulación Viral de la Expresión Génica/fisiología , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/virología , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/inmunología , Embarazo , Complicaciones Infecciosas del Embarazo/prevención & control , Complicaciones Infecciosas del Embarazo/veterinaria , Complicaciones Infecciosas del Embarazo/virología , Vacunas Atenuadas , Vacunas de Productos Inactivados , Proteínas Virales/genética , Vacunas Virales/efectos adversos
8.
Front Cell Neurosci ; 7: 53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23641196

RESUMEN

Neuroinflammation has received increased attention as a target for putative neuroprotective therapies in Parkinson's Disease (PD). Two prototypic pro-inflammatory cytokines interleukin-1ß (IL-1) and tumor necrosis factor-α (TNF) have been implicated as main effectors of the functional consequences of neuroinflammation on neurodegeneration in PD models. In this review, we describe that the functional interaction between these cytokines in the brain differs from the periphery (e.g., their expression is not induced by each other) and present data showing predominantly a toxic effect of these cytokines when expressed at high doses and for a sustained period of time in the substantia nigra pars compacta (SN). In addition, we highlight opposite evidence showing protective effects of these two main cytokines when conditions of duration, amount of expression or state of activation of the target or neighboring cells are changed. Furthermore, we discuss these results in the frame of previous disappointing results from anti-TNF-α clinical trials against Multiple Sclerosis, another neurodegenerative disease with a clear neuroinflammatory component. In conclusion, we hypothesize that the available evidence suggests that the duration and dose of IL-1ß or TNF-α expression is crucial to predict their functional effect on the SN. Since these parameters are not amenable for measurement in the SN of PD patients, we call for an in-depth analysis to identify downstream mediators that could be common to the toxic (and not the protective) effects of these cytokines in the SN. This strategy could spare the possible neuroprotective effect of these cytokines operative in the patient at the time of treatment, increasing the probability of efficacy in a clinical setting. Alternatively, receptor-specific agonists or antagonists could also provide a way to circumvent undesired effects of general anti-inflammatory or specific anti-IL-1ß or TNF-α therapies against PD.

9.
Toxicol Appl Pharmacol ; 268(3): 318-30, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23403069

RESUMEN

Adenoviral vectors (Ads) are promising gene delivery vehicles due to their high transduction efficiency; however, their clinical usefulness has been hampered by their immunogenicity and the presence of anti-Ad immunity in humans. We reported the efficacy of a gene therapy approach for glioma consisting of intratumoral injection of Ads encoding conditionally cytotoxic herpes simplex type 1 thymidine kinase (Ad-TK) and the immunostimulatory cytokine fms-like tyrosine kinase ligand 3 (Ad-Flt3L). Herein, we report the biodistribution, efficacy, and neurological and systemic effects of a bicistronic high-capacity Ad, i.e., HC-Ad-TK/TetOn-Flt3L. HC-Ads elicit sustained transgene expression, even in the presence of anti-Ad immunity, and can encode large therapeutic cassettes, including regulatory elements to enable turning gene expression "on" or "off" according to clinical need. The inclusion of two therapeutic transgenes within a single vector enables a reduction of the total vector load without adversely impacting efficacy. Because clinically the vectors will be delivered into the surgical cavity, normal regions of the brain parenchyma are likely to be transduced. Thus, we assessed any potential toxicities elicited by escalating doses of HC-Ad-TK/TetOn-Flt3L (1×10(8), 1×10(9), or 1×10(10) viral particles [vp]) delivered into the rat brain parenchyma. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points. The results indicate that doses up to 1×10(9) vp of HC-Ad-TK/TetOn-Flt3L can be safely delivered into the normal rat brain and underpin further developments for its implementation in a phase I clinical trial for glioma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Ensayos Clínicos Fase I como Asunto/métodos , Citotoxinas/administración & dosificación , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Glioblastoma/tratamiento farmacológico , Inmunización/métodos , Adenoviridae/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Citotoxinas/efectos adversos , Citotoxinas/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Quimioterapia Combinada , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Vectores Genéticos/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Ratas , Ratas Endogámicas Lew , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Resultado del Tratamiento
10.
Hum Gene Ther Methods ; 23(4): 271-84, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22950971

RESUMEN

Adenoviral vectors (Ads) have been evaluated in clinical trials for glioma. However, systemic immunity against the vectors can hamper therapeutic efficacy. We demonstrated that combined immunostimulation and cytotoxic gene therapy provides long-term survival in preclinical glioma models. Because helper-dependent high-capacity Ads (HC-Ads) elicit sustained transgene expression, in the presence of antiadenoviral immunity, we engineered HC-Ads encoding conditional cytotoxic herpes simplex type 1 thymidine kinase and immunostimulatory cytokine Fms-like tyrosine kinase ligand-3 under the control of the TetOn system. Escalating doses of combined HC-Ads (1×10(8), 1×10(9), and 1×10(10) viral particles [VP]) were delivered into the rat brain. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points after vector delivery. Histopathological analysis did not reveal any evidence of toxicity or long-term inflammation at the lower doses tested. Vector genomes were restricted to the injection site. Serum chemistry did not uncover adverse systemic side effects at any of the doses tested. Taken together, our data indicate that doses of up to 1×10(9) VP of each HC-Ad can be safely administered into the normal brain. This comprehensive toxicity and biodistribution study will lay the foundations for implementation of a phase 1 clinical trial for GBM using HC-Ads.


Asunto(s)
Adenoviridae/genética , Encéfalo/metabolismo , Vectores Genéticos/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Análisis Químico de la Sangre , Encéfalo/efectos de los fármacos , Encéfalo/patología , Ensayos Clínicos Fase I como Asunto , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Vectores Genéticos/toxicidad , Glioma/terapia , Herpesvirus Humano 1/enzimología , Humanos , Masculino , Ratas , Ratas Endogámicas Lew , Timidina Quinasa/genética , Distribución Tisular , Transducción Genética , Tirosina Quinasa 3 Similar a fms/genética
11.
Neurotherapeutics ; 9(4): 827-43, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22996231

RESUMEN

Immune-mediated gene therapy using adenovirus expressing Flt3 ligand and thymidine kinase followed by ganciclovir administration (Flt3/TK) effectively elicits tumor regression in preclinical glioma models. Herein, we assessed new strategies to optimize Flt3L/TK therapeutic efficacy in a refractory RG2 orthotopic glioblastoma model. Specifically, we aimed to optimize the therapeutic efficacy of Flt3L/TK treatment in the RG2 model by overexpressing the following genes within the brain tumor microenvironment: 1) a TK mutant with enhanced cytotoxicity (SR39 mutant TK), 2) Flt3L-IgG fusion protein that has a longer half-life, 3) CD40L to stimulate DC maturation, 4) T helper cell type 1 polarizing dendritic cell cytokines interleukin-12 or C-X-C motif ligand 10 chemokine (CXCL)-10, 5) C-C motif ligand 2 chemokine (CCL2) or C-C motif ligand 3 chemokine (CCL3) to enhance dendritic cell recruitment into the tumor microenvironment, 6) T helper cell type 1 cytokines interferon-γ or interleukin-2 to enhance effector T-cell functions, and 7) IκBα or p65RHD (nuclear factor kappa-B [NF-κB] inhibitors) to suppress the function of Foxp3+ Tregs and enhanced effector T-cell functions. Anti-tumor immunity and tumor specific effector T-cell functions were assessed by cytotoxic T lymphocyte assay and intracellular IFN-γ staining. Our data showed that overexpression of interferon-γ or interleukin-2, or inhibition of the nuclear factor kappa-B within the tumor microenvironment, enhanced cytotoxic T lymphocyte-mediated immune responses and successfully extended the median survival of rats bearing intracranial RG2 when combined with Flt3L/TK. These findings indicate that enhancement of T-cell functions constitutes a critical therapeutic target to overcome immune evasion and enhance therapeutic efficacy for brain cancer. In addition, our study provides novel targets to be used in combination with immune-therapeutic strategies for glioblastoma, which are currently being tested in the clinic.


Asunto(s)
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Inmunoterapia/métodos , Transducción de Señal , Linfocitos T/inmunología , Adenoviridae/genética , Animales , Antivirales/uso terapéutico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Ganciclovir/uso terapéutico , Vectores Genéticos , Glioblastoma/genética , Glioblastoma/inmunología , Humanos , Interleucina-2/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Proteínas de la Membrana/uso terapéutico , FN-kappa B/inmunología , Ratas , Proteínas Recombinantes/uso terapéutico , Timidina Quinasa/uso terapéutico , Microambiente Tumoral/inmunología
12.
Proc Natl Acad Sci U S A ; 109(20): 7835-40, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22547816

RESUMEN

Following antigen recognition on target cells, effector T cells establish immunological synapses and secrete cytokines. It is thought that T cells secrete cytokines in one of two modes: either synaptically (i.e., toward antigenic target cells) or multidirectionally, affecting a wider population of cells. This paradigm predicts that synaptically secreted cytokines such as IFN-γ will preferentially signal to antigenic target cells contacted by the T cell through an immunological synapse. Despite its physiological significance, this prediction has never been tested. We developed a live-cell imaging system to compare the responses of target cells and nonantigenic bystanders to IFN-γ secreted by CD8+, antigen-specific, cytotoxic T cells. Both target cells and surrounding nontarget cells respond robustly. This pattern of response was detected even at minimal antigenic T-cell stimulation using low doses of antigenic peptide, or altered peptide ligands. Although cytotoxic immunological synapses restrict killing to antigenic target cells, the effects of IFN-γ are more widespread.


Asunto(s)
Sinapsis Inmunológicas/inmunología , Interferón gamma/metabolismo , Linfocitos T Citotóxicos/inmunología , Adenoviridae , Análisis de Varianza , Astrocitos/inmunología , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Procesamiento de Imagen Asistido por Computador , Interferón gamma/inmunología , Microscopía/métodos
13.
Mol Ther ; 20(4): 808-19, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22233583

RESUMEN

The adaptive immune response to viral vectors reduces vector-mediated transgene expression from the brain. It is unknown, however, whether this loss is caused by functional downregulation of transgene expression or death of transduced cells. Herein, we demonstrate that during the elimination of transgene expression, the brain becomes infiltrated with CD4(+) and CD8(+) T cells and that these T cells are necessary for transgene elimination. Further, the loss of transgene-expressing brain cells fails to occur in the absence of IFNγ, perforin, and TNFα receptor. Two methods to induce severe immune suppression in immunized animals also fail to restitute transgene expression, demonstrating the irreversibility of this process. The need for cytotoxic molecules and the irreversibility of the reduction in transgene expression suggested to us that elimination of transduced cells is responsible for the loss of transgene expression. A new experimental paradigm that discriminates between downregulation of transgene expression and the elimination of transduced cells demonstrates that transduced cells are lost from the brain upon the induction of a specific antiviral immune response. We conclude that the anti-adenoviral immune response reduces transgene expression in the brain through loss of transduced cells.


Asunto(s)
Encéfalo/citología , Interferón gamma/metabolismo , Perforina/metabolismo , Transducción Genética/métodos , Transgenes/genética , Factor de Necrosis Tumoral alfa/metabolismo , Adenoviridae/genética , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Transgénicos
14.
Neoplasia ; 13(10): 947-60, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22028620

RESUMEN

We have demonstrated that modifying the tumor microenvironment through intratumoral administration of adenoviral vectors (Ad) encoding the conditional cytotoxic molecule, i.e., HSV1-TK and the immune-stimulatory cytokine, i.e., fms-like tyrosine kinase 3 ligand (Flt3L) leads to T-cell-dependent tumor regression in rodent models of glioblastoma. We investigated the role of B cells during immune-mediated glioblastoma multiforme regression. Although treatment with Ad-TK+Ad-Flt3L induced tumor regression in 60% of wild-type (WT) mice, it completely failed in B-cell-deficient Igh6(-/-) mice. Tumor-specific T-cell precursors were detected in Ad-TK+Ad-Flt3L-treated WT mice but not in Igh6(-/-) mice. The treatment also failed in WT mice depleted of total B cells or marginal zone B cells. Because we could not detect circulating antibodies against tumor cells and the treatment was equally efficient in WT mice and in mice with B-cell-specific deletion of Prdm 1 (encoding Blimp-1), in which B cells are present but unable to fully differentiate into antibody-secreting plasma cells, tumor regression in this model is not dependent on B cells' production of tumor antigen-specific immunoglobulins. Instead, B cells seem to play a role as antigen-presenting cells (APCs). Treatment with Ad-TK+Ad-Flt3L led to an increase in the number of B cells in the cervical lymph nodes, which stimulated the proliferation of syngeneic T cells and induced clonal expansion of antitumor T cells. Our data show that B cells act as APCs, playing a critical role in clonal expansion of tumor antigen-specific T cells and brain tumor regression.


Asunto(s)
Linfocitos B/inmunología , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Linfocitos B/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Citotoxicidad Inmunológica/inmunología , Femenino , Glioblastoma/genética , Glioblastoma/patología , Herpesvirus Humano 1/enzimología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Linfocitos T/metabolismo , Timidina Quinasa/genética , Timidina Quinasa/inmunología , Timidina Quinasa/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
15.
PLoS One ; 6(8): e23523, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21897844

RESUMEN

CD8(+) T cells infiltrate the brain during an anti-viral immune response. Within the brain CD8(+) T cells recognize cells expressing target antigens, become activated, and secrete IFNγ. However, there are no methods to recognize individual cells that respond to IFNγ. Using a model that studies the effects of the systemic anti-adenoviral immune response upon brain cells infected with an adenoviral vector in mice, we describe a method that identifies individual cells that respond to IFNγ. To identify individual mouse brain cells that respond to IFNγ we constructed a series of adenoviral vectors that contain a transcriptional response element that is selectively activated by IFNγ signaling, the gamma-activated site (GAS) promoter element; the GAS element drives expression of a transgene, Cre recombinase (Ad-GAS-Cre). Upon binding of IFNγ to its receptor, the intracellular signaling cascade activates the GAS promoter, which drives expression of the transgene Cre recombinase. We demonstrate that upon activation of a systemic immune response against adenovirus, CD8(+) T cells infiltrate the brain, interact with target cells, and cause an increase in the number of cells expressing Cre recombinase. This method can be used to identify, study, and eventually determine the long term fate of infected brain cells that are specifically targeted by IFNγ. The significance of this method is that it will allow to characterize the networks in the brain that respond to the specific secretion of IFNγ by anti-viral CD8(+) T cells that infiltrate the brain. This will allow novel insights into the cellular and molecular responses underlying brain immune responses.


Asunto(s)
Antivirales/metabolismo , Encéfalo/citología , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón gamma/metabolismo , Imagen Molecular/métodos , Transducción de Señal/inmunología , Adenoviridae/genética , Animales , Encéfalo/metabolismo , Encéfalo/virología , Femenino , Genes Reporteros/genética , Vectores Genéticos/genética , Herpesvirus Humano 1/genética , Integrasas/metabolismo , Interferón gamma/genética , Masculino , Ratones , Regiones Promotoras Genéticas/genética , Especificidad de la Especie , Timidina Quinasa/genética
16.
Anticancer Agents Med Chem ; 11(8): 729-38, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21707497

RESUMEN

Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. GBM is very aggressive due to its poor cellular differentiation and invasiveness, which makes complete surgical resection virtually impossible. Therefore, GBM's invasive nature as well as its intrinsic resistance to current treatment modalities makes it a unique therapeutic challenge. Extensive examination of human GBM specimens has uncovered that these tumors overexpress a variety of receptors that are virtually absent in the surrounding non-neoplastic brain. Human GBMs overexpress receptors for cytokines, growth factors, ephrins, urokinase-type plasminogen activator (uPA), and transferrin, which can be targeted with high specificity by linking their ligands with highly cytotoxic molecules, such as Diptheria toxin and Pseudomonas exotoxin A. We review the preclinical development and clinical translation of targeted toxins for GBM. In view of the clinical experience, we conclude that although these are very promising therapeutic modalities for GBM patients, efforts should be focused on improving the delivery systems utilized in order to achieve better distribution of the immuno-toxins in the tumor/resection cavity. Delivery of targeted toxins using viral vectors would also benefit enormously from improved strategies for local delivery.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Inmunotoxinas/administración & dosificación , Inmunotoxinas/metabolismo , Animales , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos/métodos , Humanos
17.
Curr Gene Ther ; 11(3): 155-80, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21453286

RESUMEN

The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of 15-18 months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted; this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/terapia , Toxinas Biológicas/uso terapéutico , Animales , Neoplasias Encefálicas/genética , Terapia Combinada , Marcación de Gen , Glioma/genética , Humanos , Inmunoterapia , Interferones/uso terapéutico , Interleucinas/uso terapéutico , Modelos Biológicos
18.
Proc Natl Acad Sci U S A ; 107(46): 20021-6, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21030678

RESUMEN

Restricting the cytotoxicity of anticancer agents by targeting receptors exclusively expressed on tumor cells is critical when treating infiltrative brain tumors such as glioblastoma multiforme (GBM). GBMs express an IL-13 receptor (IL13Rα2) that differs from the physiological IL4R/IL13R receptor. We developed a regulatable adenoviral vector (Ad.mhIL-4.TRE.mhIL-13-PE) encoding a mutated human IL-13 fused to Pseudomonas exotoxin (mhIL-13-PE) that specifically binds to IL13Rα2 to provide sustained expression, effective anti-GBM cytotoxicity, and minimal neurotoxicity. The therapeutic Ad also encodes mutated human IL-4 that binds to the physiological IL4R/IL13R without interacting with IL13Rα2, thus inhibiting potential binding of mhIL-13-PE to normal brain cells. Using intracranial GBM xenografts and syngeneic mouse models, we tested the Ad.mhIL-4.TRE.mhIL-13-PE and two protein formulations, hIL-13-PE used in clinical trials (Cintredekin Besudotox) and a second-generation mhIL-13-PE. Cintredekin Besudotox doubled median survival without eliciting long-term survival and caused severe neurotoxicity; mhIL-13-PE led to ∼40% long-term survival, eliciting severe neurological toxicity at the high dose tested. In contrast, Ad-mediated delivery of mhIL-13-PE led to tumor regression and long-term survival in over 70% of the animals, without causing apparent neurotoxicity. Although Cintredekin Besudotox was originally developed to target GBM, when tested in a phase III trial it failed to achieve clinical endpoints and revealed neurotoxicity. Limitations of Cintredekin Besudotox include its short half-life, which demanded frequent or continued administration, and binding to IL4R/IL13R, present in normal brain cells. These shortcomings were overcome by our therapeutic Ad, thus representing a significant advance in the development of targeted therapeutics for GBM.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Citotoxinas/genética , Citotoxinas/uso terapéutico , Técnicas de Transferencia de Gen , Terapia Genética , Glioma/tratamiento farmacológico , Adenoviridae/genética , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Exotoxinas/genética , Exotoxinas/uso terapéutico , Vectores Genéticos/genética , Glioma/patología , Humanos , Inmunocompetencia/inmunología , Interleucina-13/genética , Interleucina-13/uso terapéutico , Ratones , Ratones Desnudos , Mutación/genética , Neurotoxinas/toxicidad , Pseudomonas/metabolismo , Transgenes/genética , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
19.
PLoS One ; 5(6): e11074, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20552015

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults and carries a dismal prognosis. We have developed a conditional cytotoxic/immunotherapeutic approach using adenoviral vectors (Ads) encoding the immunostimulatory cytokine, human soluble fms-like tyrosine kinase 3 ligand (hsFlt3L) and the conditional cytotoxic molecule, i.e., Herpes Simplex Type 1- thymide kinase (TK). This therapy triggers an anti-tumor immune response that leads to tumor regression and anti-tumor immunological memory in intracranial rodent cancer models. We aim to test the efficacy of this immunotherapy in dogs bearing spontaneous GBM. In view of the controversy regarding the effect of human cytokines on dog immune cells, and considering that the efficacy of this treatment depends on hsFlt3L-stimulated dendritic cells (DCs), in the present work we tested the ability of Ad-encoded hsFlt3L to generate DCs from dog peripheral blood and compared its effects with canine IL-4 and GM-CSF. METHODOLOGY/PRINCIPAL FINDINGS: Our results demonstrate that hsFlT3L expressed form an Ad vector, generated DCs from peripheral blood cultures with very similar morphological and phenotypic characteristics to canine IL-4 and GM-CSF-cultured DCs. These include phagocytic activity and expression of CD11c, MHCII, CD80 and CD14. Maturation of DCs cultured under both conditions resulted in increased secretion of IL-6, TNF-alpha and IFN-gamma. Importantly, hsFlt3L-derived antigen presenting cells showed allostimulatory potential highlighting their ability to present antigen to T cells and elicit their proliferation. CONCLUSIONS/SIGNIFICANCE: These results demonstrate that hsFlt3L induces the proliferation of canine DCs and support its use in upcoming clinical trials for canine GBM. Our data further support the translation of hsFlt3L to be used for dendritic cells' vaccination and gene therapeutic approaches from rodent models to canine patients and its future implementation in human clinical trials.


Asunto(s)
Neoplasias Encefálicas/veterinaria , Células Dendríticas/inmunología , Enfermedades de los Perros/terapia , Glioma/veterinaria , Inmunoterapia , Proteínas de la Membrana/inmunología , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Proliferación Celular , Células Cultivadas , Enfermedades de los Perros/inmunología , Perros , Vectores Genéticos , Glioma/inmunología , Glioma/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Herpesvirus Humano 1/genética , Interleucina-4/administración & dosificación , Activación de Linfocitos , Proteínas de la Membrana/genética , Fagocitosis
20.
J Virol ; 84(12): 6007-17, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20375153

RESUMEN

Glioblastoma multiforme (GBM) is a deadly primary brain tumor. Conditional cytotoxic/immune-stimulatory gene therapy (Ad-TK and Ad-Flt3L) elicits tumor regression and immunological memory in rodent GBM models. Since the majority of patients enrolled in clinical trials would exhibit adenovirus immunity, which could curtail transgene expression and therapeutic efficacy, we used high-capacity adenovirus vectors (HC-Ads) as a gene delivery platform. Herein, we describe for the first time a novel bicistronic HC-Ad driving constitutive expression of herpes simplex virus type 1 thymidine kinase (HSV1-TK) and inducible Tet-mediated expression of Flt3L within a single-vector platform. We achieved anti-GBM therapeutic efficacy with no overt toxicities using this bicistronic HC-Ad even in the presence of systemic Ad immunity. The bicistronic HC-Ad-TK/TetOn-Flt3L was delivered into intracranial gliomas in rats. Survival, vector biodistribution, neuropathology, systemic toxicity, and neurobehavioral deficits were assessed for up to 1 year posttreatment. Therapeutic efficacy was also assessed in animals preimmunized against Ads. We demonstrate therapeutic efficacy, with vector genomes being restricted to the brain injection site and an absence of overt toxicities. Importantly, antiadenoviral immunity did not inhibit therapeutic efficacy. These data represent the first report of a bicistronic vector platform driving the expression of two therapeutic transgenes, i.e., constitutive HSV1-TK and inducible Flt3L genes. Further, our data demonstrate no promoter interference and optimum gene delivery and expression from within this single-vector platform. Analysis of the efficacy, safety, and toxicity of this bicistronic HC-Ad vector in an animal model of GBM strongly supports further preclinical testing and downstream process development of HC-Ad-TK/TetOn-Flt3L for a future phase I clinical trial for GBM.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Glioma/genética , Glioma/terapia , Herpesvirus Humano 1/enzimología , Timidina Quinasa/uso terapéutico , Proteínas Virales/uso terapéutico , Tirosina Quinasa 3 Similar a fms/uso terapéutico , Adenoviridae/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Terapia Genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glioma/metabolismo , Herpesvirus Humano 1/genética , Humanos , Ratas , Ratas Endogámicas Lew , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA