Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Allergy Clin Immunol ; 133(4): 961-6, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24582311

RESUMEN

The present uncertainty of which live viral or bacterial vaccines can be given to immunodeficient patients and the growing neglect of societal adherence to routine immunizations has prompted the Medical Advisory Committee of the Immune Deficiency Foundation to issue recommendations based on published literature and the collective experience of the committee members. These recommendations address the concern for immunodeficient patients acquiring infections from healthy subjects who have not been immunized or who are shedding live vaccine-derived viral or bacterial organisms. Such transmission of infectious agents can occur within the hospital, clinic, or home or at any public gathering. Collectively, we define this type of transmission as close-contact spread of infectious disease that is particularly relevant in patients with impaired immunity who might have an infection when exposed to subjects carrying vaccine-preventable infectious diseases or who have recently received a live vaccine. Immunodeficient patients who have received therapeutic hematopoietic stem transplantation are also at risk during the time when immune reconstitution is incomplete or while they are receiving immunosuppressive agents to prevent or treat graft-versus-host disease. This review recommends the general education of what is known about vaccine-preventable or vaccine-derived diseases being spread to immunodeficient patients at risk for close-contact spread of infection and describes the relative risks for a child with severe immunodeficiency. The review also recommends a balance between the need to protect vulnerable subjects and their social needs to integrate into society, attend school, and benefit from peer education.


Asunto(s)
Infecciones Bacterianas/transmisión , Vacunas Bacterianas/efectos adversos , Huésped Inmunocomprometido , Vacunas Vivas no Atenuadas/efectos adversos , Vacunas Virales/efectos adversos , Virosis/transmisión , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/prevención & control , Vacunas Bacterianas/inmunología , Niño , Preescolar , Humanos , Síndromes de Inmunodeficiencia , Vacunas Vivas no Atenuadas/inmunología , Vacunas Virales/inmunología , Virosis/inmunología , Virosis/prevención & control
2.
Blood ; 120(18): 3635-46, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22968453

RESUMEN

We conducted a gene therapy trial in 10 patients with adenosine deaminase (ADA)-deficient severe combined immunodeficiency using 2 slightly different retroviral vectors for the transduction of patients' bone marrow CD34(+) cells. Four subjects were treated without pretransplantation cytoreduction and remained on ADA enzyme-replacement therapy (ERT) throughout the procedure. Only transient (months), low-level (< 0.01%) gene marking was observed in PBMCs of 2 older subjects (15 and 20 years of age), whereas some gene marking of PBMC has persisted for the past 9 years in 2 younger subjects (4 and 6 years). Six additional subjects were treated using the same gene transfer protocol, but after withdrawal of ERT and administration of low-dose busulfan (65-90 mg/m(2)). Three of these remain well, off ERT (5, 4, and 3 years postprocedure), with gene marking in PBMC of 1%-10%, and ADA enzyme expression in PBMC near or in the normal range. Two subjects were restarted on ERT because of poor gene marking and immune recovery, and one had a subsequent allogeneic hematopoietic stem cell transplantation. These studies directly demonstrate the importance of providing nonmyeloablative pretransplantation conditioning to achieve therapeutic benefits with gene therapy for ADA-deficient severe combined immunodeficiency.


Asunto(s)
Agammaglobulinemia/terapia , Trasplante de Médula Ósea/métodos , Terapia Genética/métodos , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas/métodos , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/deficiencia , Adolescente , Antígenos CD34/metabolismo , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Retroviridae/genética , Transducción Genética , Acondicionamiento Pretrasplante , Adulto Joven
3.
Clin Immunol ; 135(1): 72-83, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20123155

RESUMEN

The reasons underlying the occurrence of multiple revertant genotypes in Wiskott-Aldrich syndrome (WAS) patients remain unclear. We have identified more than 30 revertant genotypes in a C995T WAS patient having 10-15% revertant, WAS protein (WASp)-expressing circulating lymphocytes. Of 497 allospecific T-cell clones generated from the peripheral blood, 47.1% carried a revertant sequence. All revertant T-cell clones exhibited restoration of WASp expression. However, anti-CD3-induced proliferative responses varied greatly amongst revertants. Several revertant T-cell clones expressed an internally deleted WASp mutant lacking much of the proline-rich region. This potentially accounts for the reduced anti-CD3 proliferative responses of these T-cell clones. We found no evidence for an increased DNA mutation rate in this patient. We conclude that the diversity of revertant genotypes in our patient does not result from an extraordinary mutation rate and that the amino acid sequence space explored by WASp in revertant T-cells is significantly smaller than might have been predicted from the diversity of revertant genotypes.


Asunto(s)
Mosaicismo , Linfocitos T/inmunología , Proteína del Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/genética , Secuencia de Aminoácidos , Secuencia de Bases , Western Blotting , Células Clonales , Variación Genética , Genotipo , Humanos , ARN Mensajero/química , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Síndrome de Wiskott-Aldrich/inmunología , Proteína del Síndrome de Wiskott-Aldrich/inmunología
4.
Immunol Res ; 38(1-3): 274-84, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17917032

RESUMEN

The efforts to find satisfactory treatments for seriously ill patients with primary immunodeficiency have resulted in the development of important new therapeutic procedures with benefits reaching far beyond the relatively small number of patients affected with these rare disorders. Allogeneic bone marrow transplantation, immunoglobulin and enzyme replacement treatments and more recently gene therapy have all been introduced into clinical medicine as treatments for one or more of the primary immunodeficiency diseases. Beginning in 1990, gene-corrected T cells were first used to treat ADA deficiency SCID. With this demonstration that the gene-transfer procedure could be safely used to introduce functional transgenes into patient cells, clinical trials for a broad range of inherited disorders and cancer were started in the mid 90s. Of all these early clinical experiments, those addressing primary immunodeficiency have also been the most successful. Both ADA and X-SCID have now been cured using gene insertion into autologous bone marrow stem cells. In addition some patients with chronic granulomatous disease (CGD) have shown an unexpectedly high level of functionally corrected granulocytes in their blood following infusion of autologous gene-corrected bone marrow. There remain however a great many significant challenges to be overcome before gene therapy becomes the treatment of choice for these and other disorders. The use of genes as medicines is the most complex therapeutic system ever attempted and it may rake several more decades of work before its real potential as a treatment for both inherited and sporadic disorders if finally realized.


Asunto(s)
Terapia Genética/métodos , Síndromes de Inmunodeficiencia/terapia , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Ensayos Clínicos como Asunto , Genoma Humano , Enfermedad Granulomatosa Crónica/terapia , Humanos , Inmunodeficiencia Combinada Grave/terapia , Transgenes
5.
Hum Gene Ther ; 16(9): 1065-74, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16149905

RESUMEN

The present study examined the safety and relative in vivo survival of genetically engineered CD4+ T lymphocytes in human immunodeficiency virus (HIV)-infected individuals. Ten pairs of identical twins discordant for HIV infection were recruited, with the uninfected twin serving as the lymphocyte donor. Ten subjects were treated with a total of 19 separate infusions of retroviral vector-transduced CD4+ enriched T cells. Control (neo gene) or anti-HIV gene (antisense trans-activation response [TAR] element and/or trans-dominant Rev)-engineered lymphocytes were monitored in peripheral blood for 3 years, using a vector-specific PCR assay. Data from 9 of the 10 patients (15 of the 19 infusions) demonstrated preferential survival of CD4+ lymphocytes containing the anti-HIV gene(s) in the immediate weeks after infusion. In six of six patients studied long term (>100 weeks), only T cells containing the anti-HIV genes were consistently detected. In addition, a marked survival advantage of anti-HIV gene-containing T cells was observed in a patient treated during a period of high viral load. Thus, these data strongly support the hypothesis that anti-HIV genes afford a survival advantage to T cells and potential benefit to HIV-1+ individuals.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Enfermedades en Gemelos , Infecciones por VIH/inmunología , Infecciones por VIH/terapia , Transfusión de Linfocitos , Adulto , Terapia Antirretroviral Altamente Activa/métodos , Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular/genética , Vectores Genéticos , Humanos , Inmunoterapia Adoptiva , Transfusión de Linfocitos/métodos , Masculino , Persona de Mediana Edad , Retroviridae/genética , Trasplante Isogénico , Resultado del Tratamiento , Gemelos Monocigóticos
6.
Hum Gene Ther ; 13(1): 129-41, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11779417

RESUMEN

Preclinical and clinical studies with adenoviral vectors have clearly illustrated the potential advantages of this gene transfer system. However, many studies have also demonstrated potent immune responses directed at both vector and transduced cells. We examined in vitro responses of human peripheral blood mononuclear cells (PBMC) to virus exposure as a model for this host response. PBMC were isolated from normal donors and incubated with wild-type adenovirus (Ad5), Ad5 variants deleted for segments of E1 and/or E3, and empty viral capsids. Proinflammatory cytokine release was monitored for 96 hr. Induction of TNF-alpha by intact virions was low although stimulation by empty capsid gave a significant and sustained response. Induction of IL-6, GM-CSF, and a panel alpha- and beta-chemokines by intact virions was prominent, often approaching results obtained with 2.5 microg/ml of lipopolysaccharide (LPS). Responses were generally independent of virion genetic composition and were only partially blunted when UV-inactivated virus was used. Dose-response data showed 100-fold increases in virion concentration produced a maximum 3-fold increase in cytokine release, suggesting saturation. Surprisingly, prominent stimulation occurred after addition of empty capsid, which typically provoked responses equivalent to those seen with LPS stimulation. We present arguments that cellular signal transduction mechanisms activated by binding of virions/capsids stimulate transcription of proinflammatory cytokine genes.


Asunto(s)
Adenovirus Humanos/fisiología , Cápside/farmacología , Quimiocinas CXC , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Vectores Genéticos/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Interleucinas/metabolismo , Leucocitos Mononucleares/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células Cultivadas , Quimiocina CCL4 , Quimiocina CCL5/metabolismo , Quimiocina CXCL1 , Factores Quimiotácticos/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Sustancias de Crecimiento/metabolismo , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Lipopolisacáridos/farmacología , Proteínas Inflamatorias de Macrófagos/metabolismo
7.
Hum Gene Ther ; 13(13): 1605-10, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12228015

RESUMEN

The first approved clinical gene therapy trial for adenosine deaminase (ADA) deficiency employed autologous T cells grown in fetal calf serum (FCS)-supplemented medium and transduced with a retroviral vector (LASN) also produced in the presence of FCS. Ten years after their enrollment, both patients have circulating T cells containing vector DNA. However, whereas approximately 20% of the circulating T cells from patient 1 are still vector positive, less than 1% of patient 2's T cells have detectable vector. This difference appears to be not only a function of the original transduction efficiency and cell expansion capability in vitro, but also of the immune response that patient 2 developed to FCS components during the course of her treatment. In this study, serum samples from each patient were tested for antibodies to FCS by enzyme-linked immunosorbent assay and anti-FCS responses were demonstrated in both patients. Analysis of immunoglobulin classes revealed comparable levels of IgA and IgM anti-FCS titers. Patient 2, however, had significantly higher IgG responses to FCS than did patient 1. Investigation of the development of anti-FCS responses by IgG subclasses indicated that there was a different pattern in the development of IgG immunity to FCS between the two patients. In addition, significant antibody response to bovine lipoprotein was detected in patient 2, but not in patient 1 or in control samples. These findings suggest that the unique immune response mounted by patient 2 may have influenced the outcome of the gene transfer treatments in this patient.


Asunto(s)
Terapia Genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/terapia , Linfocitos T/inmunología , Adenosina Desaminasa/deficiencia , Animales , Proteínas Sanguíneas/inmunología , Transfusión de Sangre Autóloga , Bovinos , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos , Humanos , Retroviridae , Inmunodeficiencia Combinada Grave/genética , Linfocitos T/trasplante , Linfocitos T/virología , Transducción Genética
8.
J Neurosurg ; 99(4): 746-53, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14567611

RESUMEN

OBJECT: The aim of this study was to investigate further immunogene treatment of malignant brain tumor to improve its therapeutic efficacy. METHODS: Intratumoral dendritic cells pulsed with Semliki Forest virus (SFV)-interleukin-18 (IL-18) and/or systemic IL-12 were injected into mice bearing the B16 brain tumor. To study the immune mechanisms involved in tumor regression, we monitored the growth of implanted B16 brain tumor cells in T cell-depleted mice and IFNgamma-neutralized mice. To analyze the protective immunity created by tumor inoculation, B16 cells were injected into the left thighs of mice that had received an inoculation, and tumor growth was monitored. The local delivery of dendritic cells pulsed with IL-18 bound by SFV combined with the systemic administration of IL-12 enhanced the induction of the T helper type 1 response from tumor-specific CD4+ and CD8+ T cells and natural killer cells as well as antitumor immunity. Interferon-gamma is partly responsible for this IL-18-mediated antitumor immunity. Furthermore, the protective immunity is mediated mainly by CD8+ T cells. CONCLUSIONS: Immunogene therapy that combines the local administration of dendritic cells pulsed with IL-18 bound by SFV and the systemic administration of IL-12 may be an excellent candidate for the development of a new treatment protocol. A self-replicating SFV system may therefore open a novel approach for the treatment of malignant brain tumor.


Asunto(s)
Neoplasias Encefálicas , Células Dendríticas/inmunología , Células Dendríticas/virología , Ingeniería Genética/métodos , Terapia Genética/métodos , Glioma , Inmunoterapia Activa/métodos , Interleucina-12 , Interleucina-18 , Virus de los Bosques Semliki/inmunología , Animales , Formación de Anticuerpos , Antígenos CD/genética , Antígenos CD/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virología , Cricetinae , Cartilla de ADN/genética , Cartilla de ADN/inmunología , ADN Complementario/genética , ADN Complementario/inmunología , Glioma/inmunología , Glioma/terapia , Glioma/virología , Inmunogenética/métodos , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Interleucina-12/uso terapéutico , Interleucina-18/biosíntesis , Interleucina-18/inmunología , Interleucina-18/uso terapéutico , Células Asesinas Naturales/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Colaboradores-Inductores/inmunología , Transducción Genética , Células Tumorales Cultivadas/trasplante
11.
J Gene Med ; 6(3): 288-99, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15026990

RESUMEN

BACKGROUND: Hybrid adeno-retroviral vector systems utilize the high efficiency of adenovirus transduction to direct the in situ production of retroviral progeny. In this study, we show that a single-step transduction of glioma cells with trans-complementing hybrid adeno-retroviral vectors effectively turns these cells into retrovirus vector-producing cells, which in turn facilitates the transduction of adjacent cells. METHODS: We have adapted the adeno-retroviral hybrid viral vector system to enhance the ganciclovir (GCV) killing of glioma cells following transfer of the herpes simplex virus thymidine kinase (HSV-tk) gene. To assess the effect of the in situ production of retroviral vectors on the transduction efficiency of glioma cells, 9L cells were transduced with adeno-retroviral hybrid vectors that separately express a retroviral genome (AVC2.GCEGFP or AVC2.GCTK) and retroviral packaging proteins (AxTetGP and AxTetVSVG). The generation of an integrated HSV-tk provirus by trans-complementation of the adeno-retroviral vectors was verified by analysis of the flanking retroviral LTR sequences. Tumors established on nu/nu mice were injected with the viruses followed by intraperitoneal injections of either PBS or GCV. We also estimated the copy numbers of the HSV-tk transgene present in the tumors of the treated mice. To determine the expression pattern of the HSV-tk transcripts within a tumor, in situ hybridization analysis was performed using an RNA probe specific for HSV-tk. RESULTS: The co-transduction of rat 9L glioma cells with AVC2.GCEGFP together with vectors expressing packaging proteins of retroviruses increased the transduction efficiency. Transduction with AVC2.GCTK together with packaging vectors increased the in vitro sensitivity of cells to the pro-drug GCV by one log compared with control cells that were incapable of generating retrovirus. In vivo, the injection of established subcutaneous 9L tumors on athymic mice with a combination of AVC2.GCTK and packaging vectors followed by GCV treatment resulted in complete tumor regression in 50% of tumors at day 22, while no tumor regression was observed in control animals. Retroviral sequences diagnostic of 3' LTR reduplication in vivo were detected in genomic DNA extracted from the transduced tumors, indicating pro-viral integration of the retroviral genome derived from the adeno-retroviral hybrid vector. Furthermore, the relative copy number of the HSV-tk gene in tumors treated with the adeno-retroviral vectors was up to approximately 250-fold higher than in control tumors. In situ hybridization suggested dispersion of the HSV-tk product across a wider area of the tumor than in control tumors, which indicates the spread of the in situ generated retroviruses. CONCLUSIONS: Although the efficacy of this system has to be evaluated in orthotopic models, our observations suggest that this hybrid adeno-retroviral vector system could improve the suicide gene therapy of tumors.


Asunto(s)
Adenoviridae/genética , Genes Transgénicos Suicidas/genética , Terapia Genética , Glioma/terapia , Retroviridae/genética , Simplexvirus/genética , Timidina Quinasa/genética , Animales , Línea Celular Tumoral , Femenino , Citometría de Flujo , Ganciclovir/farmacología , Dosificación de Gen , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratas , Simplexvirus/enzimología , Transducción Genética , Integración Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Blood ; 101(7): 2563-9, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12456496

RESUMEN

The first human gene therapy experiment begun in September 1990 used a retroviral vector containing the human adenosine deaminase (ADA) cDNA to transduce mature peripheral blood lymphocytes from patients with ADA deficiency, an inherited disorder of immunity. Two patients who had been treated with intramuscular injections of pegylated bovine ADA (PEG-ADA) for 2 to 4 years were enrolled in this trial and each received a total of approximately 10(11) cells in 11 or 12 infusions over a period of about 2 years. No adverse events were observed. During and after treatment, the patients continued to receive PEG-ADA, although at a reduced dose. Ten years after the last cell infusion, approximately 20% of the first patient's lymphocytes still carry and express the retroviral gene, indicating that the effects of gene transfer can be remarkably long lasting. On the contrary, the persistence of gene-marked cells is very low (< 0.1%), and no expression of the transgene is detectable in lymphocytes from the second patient who developed persisting antibodies to components of the gene transfer system. Data collected from these original patients have provided novel information about the longevity of T lymphocytes in humans and persistence of gene expression in vivo from vectors driven by the Moloney murine leukemia virus long-terminal repeat (LTR) promoter. This long-term follow-up has also provided unique evidence supporting the safety of retroviral-mediated gene transfer and illustrates clear examples of both the potential and the pitfalls of gene therapy in humans.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Formación de Anticuerpos , Terapia Genética/métodos , Errores Innatos del Metabolismo de la Purina-Pirimidina/terapia , Adenosina Desaminasa/administración & dosificación , Adenosina Desaminasa/biosíntesis , Animales , Anticuerpos Heterófilos/sangre , Anticuerpos Antivirales/sangre , Bovinos , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/inmunología , Humanos , Estudios Longitudinales , Virus de la Leucemia Murina de Moloney/genética , Virus de la Leucemia Murina de Moloney/inmunología , Receptores de Antígenos de Linfocitos T/análisis , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA