Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cytotherapy ; 22(11): 642-652, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32747299

RESUMEN

Background aims: E3L is an immediate-early protein of vaccinia virus (VV) that is detected within 0.5 h of infection, potentially before the many immune evasion genes of vaccinia can exert their protective effects. E3L is highly conserved among orthopoxviruses and hence could provide important protective T-cell epitopes that should be retained in any subunit or attenuated vaccine. We have therefore evaluated the immunogenicity of E3L in healthy VV-vaccinated donors. Methods: Peripheral blood mononuclear cells from healthy volunteers (n = 13) who had previously received a smallpox vaccine (Dryvax) were activated and expanded using overlapping E3L peptides and their function, specificity and antiviral activity was analyzed. E3L-specific T cells were expanded from 7 of 12 (58.3%) vaccinated healthy donors. Twenty-five percent of these produced CD8+ T-cell responses and 87.5% produced CD4+ T cells. We identified epitopes restricted by HLA-B35 and HLA-DR15. Results: E3L-specific T cells killed peptide-loaded target cells as well as vaccinia-infected cells, but only CD8+ T cells could prevent the spread of infectious virus in virus inhibition assays. The epitopes recognized by E3L-specific T cells were shared with monkeypox, and although there was a single amino acid change in the variola epitope homolog, it was recognized by vaccinia-specific T-cells. Conclusions: It might be important to include E3L in any deletion mutant or subunit vaccine and E3L could provide a useful antigen to monitor protective immunity in humans.


Asunto(s)
Antígenos Virales/inmunología , Vacuna contra Viruela/inmunología , Viruela/inmunología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/genética , Humanos , Interferón gamma/metabolismo , Leucocitos Mononucleares/inmunología , Péptidos/química , Péptidos/inmunología , Viruela/prevención & control , Donantes de Tejidos , Vacunación , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Virión/inmunología , Replicación Viral/fisiología
2.
Clin Cancer Res ; 23(14): 3499-3509, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28183713

RESUMEN

Purpose: The multiple mechanisms used by solid tumors to suppress tumor-specific immune responses are a major barrier to the success of adoptively transferred tumor-specific T cells. As viruses induce potent innate and adaptive immune responses, we hypothesized that the immunogenicity of viruses could be harnessed for the treatment of solid tumors if virus-specific T cells (VST) were modified with tumor-specific chimeric antigen receptors (CAR). We tested this hypothesis using VZV-specific T cells (VZVST) expressing a CAR for GD2, a disialoganglioside expressed on neuroblastoma and certain other tumors, so that the live-attenuated VZV vaccine could be used for in vivo stimulation.Experimental Design: We generated GMP-compliant, GD2.CAR-modified VZVSTs from healthy donors and cancer patients by stimulation of peripheral blood mononuclear cells with overlapping peptide libraries spanning selected VZV antigens, then tested their ability to recognize and kill GD2- and VZV antigen-expressing target cells.Results: Our choice of VZV antigens was validated by the observation that T cells specific for these antigens expanded in vivo after VZV vaccination. VZVSTs secreted cytokines in response to VZV antigens, killed VZV-infected target cells and limited infectious virus spread in autologous fibroblasts. However, while GD2.CAR-modified VZVSTs killed neuroblastoma cell lines on their first encounter, they failed to control tumor cells in subsequent cocultures. Despite this CAR-specific dysfunction, CAR-VZVSTs retained functional specificity for VZV antigens via their TCRs and GD2.CAR function was partially rescued by stimulation through the TCR or exposure to dendritic cell supernatants.Conclusions: Vaccination via the TCR may provide a means to reactivate CAR-T cells rendered dysfunctional by the tumor microenvironment (NCT01953900). Clin Cancer Res; 23(14); 3499-509. ©2017 AACR.


Asunto(s)
Neuroblastoma/tratamiento farmacológico , Proteínas de Fusión Oncogénica/administración & dosificación , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Adolescente , Adulto , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Niño , Preescolar , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/inmunología , Femenino , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Ratones , Persona de Mediana Edad , Neuroblastoma/inmunología , Neuroblastoma/patología , Proteínas de Fusión Oncogénica/inmunología , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Microambiente Tumoral/inmunología , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Mol Ther ; 23(2): 387-95, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25366030

RESUMEN

Antiretroviral therapy (ART) is unable to eradicate human immunodeficiency virus type 1 (HIV-1) infection. Therefore, there is an urgent need to develop novel therapies for this disease to augment anti-HIV immunity. T cell therapy is appealing in this regard as T cells have the ability to proliferate, migrate, and their antigen specificity reduces the possibility of off-target effects. However, past human studies in HIV-1 infection that administered T cells with limited specificity failed to provide ART-independent, long-term viral control. In this study, we sought to expand functional, broadly-specific cytotoxic T cells (HXTCs) from HIV-infected patients on suppressive ART as a first step toward developing cellular therapies for implementation in future HIV eradication protocols. Blood samples from seven HIV+ patients on suppressive ART were used to derive HXTCs. Multiantigen specificity was achieved by coculturing T cells with antigen-presenting cells pulsed with peptides representing Gag, Pol, and Nef. All but two lines were multispecific for all three antigens. HXTCs demonstrated efficacy as shown by release of proinflammatory cytokines, specific lysis of antigen-pulsed targets, and the ability to suppress HIV replication in vitro. In conclusion, we are able to generate broadly-specific cytotoxic T cell lines that simultaneously target multiple HIV antigens and show robust antiviral function.


Asunto(s)
Epítopos de Linfocito T/inmunología , Antígenos VIH/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Terapia Antirretroviral Altamente Activa , Técnicas de Cultivo Celular por Lotes , Citocinas/metabolismo , Infecciones por VIH/terapia , Infecciones por VIH/virología , Humanos , Memoria Inmunológica , Inmunofenotipificación , Inmunoterapia , Fenotipo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/metabolismo
4.
Mol Ther ; 23(3): 602-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25531693

RESUMEN

Pexa-Vec (pexastimogene devacirepvec, JX-594) is an oncolytic and immunotherapeutic vaccinia virus designed to destroy cancer cells through viral lysis and induction of granulocyte-macrophage colony-stimulating factor (GM-CSF)-driven tumor-specific immunity. Pexa-Vec has undergone phase 1 and 2 testing alone and in combination with other therapies in adult patients, via both intratumoral and intravenous administration routes. We sought to determine the safety of intratumoral administration in pediatric patients. In a dose-escalation study using either 10(6) or 10(7) plaque-forming units per kilogram, we performed one-time injections in up to three tumor sites in five pediatric patients and two injections in one patient. Ages at study entry ranged from 4 to 21 years, and their cancer diagnoses included neuroblastoma, hepatocellular carcinoma, and Ewing sarcoma. All toxicities were ≤ grade 3. The most common side effects were sinus fever and sinus tachycardia. All three patients at the higher dose developed asymptomatic grade 1 treatment-related skin pustules that resolved within 3-4 weeks. One patient showed imaging evidence suggestive of antitumor biological activity. The two patients tested for cellular immunoreactivity to vaccinia antigens showed strong responses. Overall, our study suggests Pexa-Vec is safe to administer to pediatric patients by intratumoral administration and could be studied further in this patient population.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Vacunas contra el Cáncer/inmunología , Rayos gamma , Inmunoterapia/métodos , Viroterapia Oncolítica/métodos , Virus Vaccinia/inmunología , Adolescente , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/administración & dosificación , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Niño , Preescolar , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Humanos , Inyecciones Intralesiones , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Masculino , Estadificación de Neoplasias , Neuroblastoma/inmunología , Neuroblastoma/patología , Neuroblastoma/terapia , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/patología , Sarcoma de Ewing/terapia , Vacunación , Virus Vaccinia/genética , Adulto Joven
5.
J Immunother ; 37(4): 193-203, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24714353

RESUMEN

Antigen-specific T cells provide a therapy for cancer that is highly specific, self-replicating, and potentially devoid of toxicity. Ideally, tumor-specific T cells should recognize multiple epitopes on multiple antigens to prevent tumor immune escape. However the large-scale expansion of such broad-spectrum T cells has been limited by the availability of potent autologous antigen-presenting cells that can present antigens on the polymorphic array of each patient's HLA allotype. We evaluated a novel antigen-presenting complex (KATpx) in which antigens in the form of peptide libraries can be presented by autologous activated T cells, whereas costimulation is complemented in trans by an HLA-negative K562 cell line genetically modified to express CD80, CD83, CD86, and 4-1BBL (K562cs). The additional costimulation provided by K562cs significantly enhanced T-cell expansion in culture over autologous activated T cells alone while maintaining antigen specificity. We validated this antigen-presenting system by generating Epstein-Barr virus (EBV) antigen-specific T cells from healthy donors and from patients with EBV-positive malignancies including nasopharyngeal carcinoma and multiply relapsed EBV-positive lymphoma. These T cells were specific for EBNA1, LMP1, and LMP2, the viral antigens expressed in these type 2 latency EBV-associated malignancies. The KATpx system consistently activated and expanded antigen-specific T cells both from healthy donors and from 5 of 6 patients with lymphoma and 6 of 6 with nasopharyngeal carcinoma, while simplifying the process for generating APCs by eliminating the need for live virus (EBV) or viral vectors to force expression of transgenic EBV antigens. Hence, KATpx provides a robust, reliable, and scalable process to expand tumor-directed T cells for the treatment of virus-associated cancers.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Subgrupos de Linfocitos T/inmunología , Adenoviridae/genética , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Antígenos Virales/química , Antígenos Virales/inmunología , Carcinoma , Línea Celular Transformada , Citotoxicidad Inmunológica , Epítopos de Linfocito T/inmunología , Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Vectores Genéticos/genética , Humanos , Activación de Linfocitos/inmunología , Linfoma/inmunología , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/inmunología , Péptidos/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Transducción Genética , Proteínas de la Matriz Viral/inmunología
6.
Nat Med ; 19(3): 329-36, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23396206

RESUMEN

Oncolytic viruses and active immunotherapeutics have complementary mechanisms of action (MOA) that are both self amplifying in tumors, yet the impact of dose on subject outcome is unclear. JX-594 (Pexa-Vec) is an oncolytic and immunotherapeutic vaccinia virus. To determine the optimal JX-594 dose in subjects with advanced hepatocellular carcinoma (HCC), we conducted a randomized phase 2 dose-finding trial (n=30). Radiologists infused low- or high-dose JX-594 into liver tumors (days 1, 15 and 29); infusions resulted in acute detectable intravascular JX-594 genomes. Objective intrahepatic Modified Response Evaluation Criteria in Solid Tumors (mRECIST) (15%) and Choi (62%) response rates and intrahepatic disease control (50%) were equivalent in injected and distant noninjected tumors at both doses. JX-594 replication and granulocyte-macrophage colony-stimulating factor (GM-CSF) expression preceded the induction of anticancer immunity. In contrast to tumor response rate and immune endpoints, subject survival duration was significantly related to dose (median survival of 14.1 months compared to 6.7 months on the high and low dose, respectively; hazard ratio 0.39; P=0.020). JX-594 demonstrated oncolytic and immunotherapy MOA, tumor responses and dose-related survival in individuals with HCC.


Asunto(s)
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Viroterapia Oncolítica , Virus Vaccinia/genética , Anciano , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/mortalidad , Relación Dosis-Respuesta Inmunológica , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inmunoterapia , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/mortalidad , Masculino , Persona de Mediana Edad , Virus Oncolíticos/metabolismo , Tasa de Supervivencia , Virus Vaccinia/fisiología , Replicación Viral
7.
Cytotherapy ; 14(9): 1131-43, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22900959

RESUMEN

BACKGROUND AIMS: Interest in natural killer (NK) cell-based immunotherapy has resurged since new protocols for the purification and expansion of large numbers of clinical-grade cells have become available. METHODS: We have successfully adapted a previously described NK expansion method that uses K562 cells expressing interleukin (IL)-15 and 4-1 BB Ligand (BBL) (K562-mb15-41BBL) to grow NK cells in novel gas-permeable static cell culture flasks (G-Rex). RESULTS: Using this system we produced up to 19 × 10(9) functional NK cells from unseparated apheresis products, starting with 15 × 10(7) CD3(-) CD56 (+) NK cells, within 8-10 days of culture. The G-Rex yielded a higher fold expansion of NK cells than conventional gas-permeable bags and required no cell manipulation or feeding during the culture period. We also showed that K562-mb15-41BBL cells up-regulated surface HLA class I antigen expression upon stimulation with the supernatants from NK cultures and stimulated alloreactive CD8 (+) T cells within the NK cultures. However, these CD3 (+) T cells could be removed successfully using the CliniMACS system. We describe our optimized NK cell cryopreservation method and show that the NK cells are viable and functional even after 12 months of cryopreservation. CONCLUSIONS: We have successfully developed a static culture protocol for large-scale expansion of NK cells in the gas permeable G-Rex system under good manufacturing practice (GMP) conditions. This strategy is currently being used to produce NK cells for cancer immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva , Células K562/citología , Células Asesinas Naturales/citología , Linfocitos T , Ligando 4-1BB/metabolismo , Eliminación de Componentes Sanguíneos , Técnicas de Cultivo de Célula , Supervivencia Celular , Criopreservación , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
8.
Hum Mol Genet ; 20(R1): R93-9, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21415041

RESUMEN

Adoptive immunotherapy is an appealing approach to cancer treatment, with the potential for more precise targeting and reduced toxicity. While early clinical trial data using adoptive T cells against post-transplant virus-associated hematologic malignancies, lymphoma and melanoma have been promising, treating other solid tumors has proven to be more challenging. Adoptive lymphocytes have been genetically modified in many ways to improve activity and circumvent tumor evasion, including transfer of transgenic T-cell receptors and chimeric antigen receptors to redirect T cell and natural killer cell antigen specificity. Gene transfer may also allow expression of homeostatic cytokines or their receptors to overcome the lack of stimulatory signals or expression of dominant-negative receptors for inhibitory cytokines to compensate for an immunosuppressive tumor milieu. In addition, suicide genes can install a 'safety switch' on adoptively transferred cells to allow ablation if necessary. Although further refinement and validation are necessary, these genetic modification strategies offer hope for significant improvements in cancer immunotherapy.


Asunto(s)
Técnicas de Transferencia de Gen , Inmunoterapia Adoptiva/métodos , Neoplasias/inmunología , Neoplasias/terapia , Animales , Humanos , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/terapia , Neoplasias/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
9.
J Immunother ; 33(3): 305-15, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20445351

RESUMEN

The clinical manufacture of antigen-specific cytotoxic T lymphocytes (CTLs) for adoptive immunotherapy is limited by the complexity and time required to produce large numbers with the desired function and specificity. The culture conditions required are rigorous, and in some cases only achieved in 2-cm wells in which cell growth is limited by gas exchange, nutrients, and waste accumulation. Bioreactors developed to overcome these issues tend to be complex, expensive, and not always conducive to CTL growth. We observed that antigen-specific CTLs undergo 7 to 10 divisions poststimulation. However, the expected CTL numbers were achieved only in the first week of culture. By recreating the culture conditions present during this first week-low frequency of antigen-specific T cells and high frequency of feeder cells-we were able to increase CTL expansion to expected levels that could be sustained for several weeks without affecting phenotype or function. However, the number of 24-well plates needed was excessive and cultures required frequent media changes, increasing complexity and manufacturing costs. Therefore, we evaluated novel gas-permeable culture devices (G-Rex) with a silicone membrane at the base allowing gas exchange to occur uninhibited by the depth of the medium above. This system effectively supports the expansion of CTL and actually increases output by up to 20-fold while decreasing the required technician time. Importantly, this amplified cell expansion is not because of more cell divisions but because of reduced cell death. This bioprocess optimization increased T-cell output while decreasing the complexity and cost of CTL manufacture, making cell therapy more accessible.


Asunto(s)
Antígenos/inmunología , Proliferación Celular , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Humanos , Inmunoterapia Adoptiva/métodos , Células K562 , Recuento de Linfocitos , Reproducibilidad de los Resultados , Linfocitos T Citotóxicos/trasplante , Factores de Tiempo
10.
Development ; 134(11): 2115-24, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17507412

RESUMEN

In the developing ventral midbrain, the signaling molecule sonic hedgehog (SHH) is sufficient to specify a striped pattern of cell fates (midbrain arcs). Here, we asked whether and precisely how hedgehog (HH) signaling might be necessary for ventral midbrain patterning. By blocking HH signaling by in ovo misexpression of Ptc1(Delta)(loop2), we show that HH signaling is necessary and can act directly at a distance to specify midbrain cell fates. Ventral midbrain progenitors extinguish their dependence upon HH in a spatiotemporally complex manner, completing cell-fate specification at the periphery by Hamburger and Hamilton stage 13. Thus, patterning at the lateral periphery of the ventral midbrain is accomplished early, when the midbrain is small and the HH signal needs to travel relatively short distances (approximately 30 cell diameters). Interestingly, single-cell injections demonstrate that patterning in the midbrain occurs within the context of cortex-like radial columns of cells that can share HH blockade and are cytoplasmically connected by gap junctions. HH blockade results in increased cell scatter, disrupting the spatial coherence of the midbrain arc pattern. Finally, HH signaling is required for the integrity and the signaling properties of the boundaries of the midbrain (e.g. the midbrain-hindbrain boundary, the dorsoventral boundary), its perturbations resulting in abnormal cell mixing across 'leaky' borders.


Asunto(s)
Proteínas Hedgehog/metabolismo , Mesencéfalo/embriología , Morfogénesis/fisiología , Transducción de Señal/fisiología , Animales , Bromodesoxiuridina , Diferenciación Celular/genética , Proliferación Celular , Embrión de Pollo , Electroporación , Hibridación in Situ , Microscopía Fluorescente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...