Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 9(3): e93321, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24681846

RESUMEN

The clinical success of adoptive immunotherapy of cancer relies on the selection of target antigens that are highly expressed in tumor cells but absent in essential normal tissues. A group of genes that encode the cancer/testis or cancer germline antigens have been proposed as ideal targets for immunotherapy due to their high expression in multiple cancer types and their restricted expression in immunoprivileged normal tissues. In the present work we report the isolation and characterization of human T cell receptors (TCRs) with specificity for synovial sarcoma X breakpoint 2 (SSX2), a cancer/testis antigen expressed in melanoma, prostate cancer, lymphoma, multiple myeloma and pancreatic cancer, among other tumors. We isolated seven HLA-A2 restricted T cell receptors from natural T cell clones derived from tumor-infiltrated lymph nodes of two SSX2-seropositive melanoma patients, and selected four TCRs for cloning into retroviral vectors. Peripheral blood lymphocytes (PBL) transduced with three of four SSX2 TCRs showed SSX241-49 (KASEKIFYV) peptide specific reactivity, tumor cell recognition and tetramer binding. One of these, TCR-5, exhibited tetramer binding in both CD4 and CD8 cells and was selected for further studies. Antigen-specific and HLA-A*0201-restricted interferon-γ release, cell lysis and lymphocyte proliferation was observed following culture of TCR engineered human PBL with relevant tumor cell lines. Codon optimization was found to increase TCR-5 expression in transduced T cells, and this construct has been selected for development of clinical grade viral vector producing cells. The tumor-specific pattern of expression of SSX2, along with the potent and selective activity of TCR-5, makes this TCR an attractive candidate for potential TCR gene therapy to treat multiple cancer histologies.


Asunto(s)
Antígenos de Neoplasias/inmunología , Epítopos/inmunología , Antígeno HLA-A2/inmunología , Proteínas de Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Represoras/inmunología , Testículo/inmunología , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Epítopos/genética , Humanos , Inmunoterapia Adoptiva/métodos , Interferón gamma/inmunología , Masculino , Proteínas de Neoplasias/genética , Proteínas Represoras/genética
2.
J Immunother ; 36(2): 133-51, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23377668

RESUMEN

Nine cancer patients were treated with adoptive cell therapy using autologous anti-MAGE-A3 T-cell receptors (TCR)-engineered T cells. Five patients experienced clinical regression of their cancers including 2 on-going responders. Beginning 1-2 days postinfusion, 3 patients (#'s 5, 7, and 8) experienced mental status changes, and 2 patients (5 and 8) lapsed into comas and subsequently died. Magnetic resonance imagining analysis of patients 5 and 8 demonstrated periventricular leukomalacia, and examination of their brains at autopsy revealed necrotizing leukoencephalopathy with extensive white matter defects associated with infiltration of CD3(+)/CD8(+) T cells. Patient 7, developed Parkinson-like symptoms, which resolved over 4 weeks and fully recovered. Immunohistochemical staining of patient and normal brain samples demonstrated rare positively staining neurons with an antibody that recognizes multiple MAGE-A family members. The TCR used in this study recognized epitopes in MAGE-A3/A9/A12. Molecular assays of human brain samples using real-time quantitative-polymerase chain reaction, Nanostring quantitation, and deep-sequencing indicated that MAGE-A12 was expressed in human brain (and possibly MAGE-A1, MAGE-A8, and MAGE-A9). This previously unrecognized expression of MAGE-A12 in human brain was possibly the initiating event of a TCR-mediated inflammatory response that resulted in neuronal cell destruction and raises caution for clinical applications targeting MAGE-A family members with highly active immunotherapies.


Asunto(s)
Antígenos de Neoplasias , Terapia Genética/métodos , Inmunoterapia Adoptiva , Melanoma/terapia , Proteínas de Neoplasias , Receptores de Antígenos de Linfocitos T , Adulto , Anciano , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/uso terapéutico , Encéfalo/metabolismo , Células Dendríticas/inmunología , Femenino , Técnicas de Transferencia de Gen , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Inmunoterapia Adoptiva/mortalidad , Masculino , Melanoma/inmunología , Antígenos Específicos del Melanoma , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/uso terapéutico , ARN Mensajero/análisis , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/uso terapéutico , Vacunas de Subunidad/inmunología , Adulto Joven
3.
Hum Gene Ther ; 23(10): 1043-53, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22780919

RESUMEN

No curative treatment exists for glioblastoma, with median survival times of less than 2 years from diagnosis. As an approach to develop immune-based therapies for glioblastoma, we sought to target antigens expressed in glioma stem cells (GSCs). GSCs have multiple properties that make them significantly more representative of glioma tumors than established glioma cell lines. Epidermal growth factor receptor variant III (EGFRvIII) is the result of a novel tumor-specific gene rearrangement that produces a unique protein expressed in approximately 30% of gliomas, and is an ideal target for immunotherapy. Using PCR primers spanning the EGFRvIII-specific deletion, we found that this tumor-specific gene is expressed in three of three GCS lines. Based on the sequence information of seven EGFRvIII-specific monoclonal antibodies (mAbs), we assembled chimeric antigen receptors (CARs) and evaluated the ability of CAR-engineered T cells to recognize EGFRvIII. Three of these anti-EGFRvIII CAR-engineered T cells produced the effector cytokine, interferon-γ, and lysed antigen-expressing target cells. We concentrated development on a CAR produced from human mAb 139, which specifically recognized GSC lines and glioma cell lines expressing mutant EGFRvIII, but not wild-type EGFR and did not recognize any normal human cell tested. Using the 139-based CAR, T cells from glioblastoma patients could be genetically engineered to recognize EGFRvIII-expressing tumors and could be expanded ex vivo to large numbers, and maintained their antitumor activity. Based on these observations, a γ-retroviral vector expressing this EGFRvIII CAR was produced for clinical application.


Asunto(s)
Neoplasias Encefálicas/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Receptores ErbB/inmunología , Glioma/inmunología , Glioma/terapia , Inmunoterapia Adoptiva , Células Madre Neoplásicas/patología , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Ingeniería Genética , Vectores Genéticos/genética , Glioma/patología , Humanos , Ratones , Células 3T3 NIH , Receptores de Antígenos/inmunología , Transducción Genética
4.
J Immunother ; 35(5): 430-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22576348

RESUMEN

The clinical application of interleukin-12 (IL-12) has been hindered by the toxicity associated with its systemic administration. To potentially overcome this problem, we developed a promoter designed to direct IL-12 expression within the tumor environment using an inducible composite promoter containing binding motifs for the nuclear factor of activated T cells (NFAT) linked to a minimal IL-2 promoter. In this study, the NFAT promoter was coupled to a single-chain human IL-12 gene and inserted into 2 γ-retroviral self-inactivating vectors (SERS.NFAT.hIL12 and SERS.NFAT.hIL12.PA2) and 1 γ-retroviral vector (MSGV1.NFAT.hIL.12 PA2). Peripheral blood lymphocytes (PBLs) were double transduced with an antigen-specific T-cell receptor and the 3 NFAT.hIL12 vectors. Evaluation of inducible IL-12 expression, transduction efficiency, and vector production considerations led to the choice of the MSGV1.NFAT.hIL12.PA2 vector for clinical application. MSGV1.NFAT.hIL12.PA2 PG13 retroviral vector producer cell clones were screened by transduction of tumor antigen-specific PBLs. On the basis of expression studies in PBL, clone D3 was chosen to produce clinical-grade viral vector supernatant and was demonstrated to efficiently transduce young tumor-infiltrating lymphocytes (TIL). The vector-transduced young TIL with known tumor recognition demonstrated specific inducible IL-12 production after coculture with HLA-matched tumor targets and had augmented effector function as demonstrated by increased IFN-γ secretion. These results support the clinical application of adoptive transfer of young TIL engineered with the NFAT.hIL12 vector as a new approach for cancer immunotherapy.


Asunto(s)
Gammaretrovirus/inmunología , Interleucina-12/biosíntesis , Neoplasias/terapia , Traslado Adoptivo , Técnicas de Cocultivo , Gammaretrovirus/genética , Expresión Génica , Vectores Genéticos , Antígenos HLA/inmunología , Humanos , Inmunoterapia , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/inmunología , Regiones Promotoras Genéticas , Receptores de Antígenos de Linfocitos T/biosíntesis , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Transducción Genética
5.
J Transl Med ; 10: 29, 2012 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-22330761

RESUMEN

BACKGROUND: There has been significant progress in the last two decades on the design of chimeric antigen receptors (CAR) for adoptive immunotherapy targeting tumor-associated antigens. Structurally CARs consist of a single chain antibody fragment directed against a tumor-associated antigen fused to an extracellular spacer and transmembrane domain followed by T cell cytoplasmic signaling moieties. Currently several clinical trials are underway using gene modified peripheral blood lymphocytes (PBL) with CARs directed against a variety of tumor associated antigens. Despite the improvements in the design of CARs and expansion of the number of target antigens, there is no universal flow cytometric method available to detect the expression of CARs on the surface of transduced lymphocytes. METHODS: Currently anti-fragment antigen binding (Fab) conjugates are most widely used to determine the expression of CARs on gene-modified lymphocytes by flow cytometry. The limitations of these reagents are that many of them are not commercially available, generally they are polyclonal antibodies and often the results are inconsistent. In an effort to develop a simple universal flow cytometric method to detect the expression of CARs, we employed protein L to determine the expression of CARs on transduced lymphocytes. Protein L is an immunoglobulin (Ig)-binding protein that binds to the variable light chains (kappa chain) of Ig without interfering with antigen binding site. Protein L binds to most classes of Ig and also binds to single-chain antibody fragments (scFv) and Fab fragments. RESULTS: We used CARs derived from both human and murine antibodies to validate this novel protein L based flow cytometric method and the results correlated well with other established methods. Activated human PBLs were transduced with retroviral vectors expressing two human antibody based CARs (anti-EGFRvIII, and anti-VEGFR2), two murine antibody derived CARs (anti-CSPG4, and anti-CD19), and two humanized mouse antibody based CARs (anti-ERBB2, and anti-PSCA). Transduced cells were stained first with biotin labeled protein L followed by phycoerythrin (PE)-conjugated streptavidin (SA) and analyzed by flow cytometry. For comparison, cells were stained in parallel with biotin conjugated goat-anti-mouse Fab or CAR specific fusion proteins. Using protein L, all CAR transduced lymphocytes exhibited specific staining pattern ranging from 40 to 80% of positive cells (compared to untransduced cells) and staining was comparable to the pattern observed with anti-Fab antibodies. CONCLUSION: Our data demonstrate the feasibility of employing Protein L as a general reagent for the detection of CAR expression on transduced lymphocytes by flow cytometry.


Asunto(s)
Proteínas Bacterianas/metabolismo , Citometría de Flujo/métodos , Receptores de Antígenos/metabolismo , Proteínas Recombinantes/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Humanos , Indicadores y Reactivos , Linfocitos/metabolismo , Ratones , Proteína Estafilocócica A/metabolismo , Volumetría
6.
Cancer Res ; 71(12): 4192-204, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21546573

RESUMEN

Cancer-testis antigens (CTA), such as NY-ESO-1, MAGE-A1, and MAGE-A3, are immunogenic proteins encoded by genes, which are normally expressed only in male germ cells but are activated by ill-defined epigenetic mechanisms in human tumors, including lung cancers. Previously, we reported induction of these CTAs in cancer cells, but not normal cells, by DNA-demethylating agents and histone deacetylase inhibitors using clinically achievable exposure conditions. In the present study, we evaluated chromatin alterations associated with repression/activation of cancer-testis genes in lung cancer cells to further develop gene-induction regimens for cancer immunotherapy. Repression of NY-ESO-1, MAGE-A1, and MAGE-A3 coincided with DNA hypermethylation, recruitment, and binding of polycomb-group proteins, and histone heterochromatin modifications within the promoters of these genes. Derepression coincided with DNA demethylation, dissociation of polycomb proteins, and presence of euchromatin marks within the respective promoters. Short hairpin RNAs were used to inhibit several histone methyltransferases (KMT) and histone demethylases (KDM) that mediate histone methylation and repress gene expression. Knockdown of KMT6, KDM1, or KDM5B markedly enhanced deoxyazacytidine (DAC)-mediated activation of these cancer-testis genes in lung cancer cells. DZNep, a pharmacologic inhibitor of KMT6 expression, recapitulated the effects of KMT6 knockdown. Following DAC-DZNep exposure, lung cancer cells were specifically recognized and lysed by allogeneic lymphocytes expressing recombinant T-cell receptors recognizing NY-ESO-1 and MAGE-A3. Combining DNA-demethylating agents with compounds, such as DZNep, that modulate histone lysine methylation may provide a novel epigenetic strategy to augment cancer-testis gene expression as an adjunct to adoptive cancer immunotherapy.


Asunto(s)
Antígenos de Neoplasias/genética , Inhibidores Enzimáticos/farmacología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Inmunoterapia Adoptiva , Neoplasias Pulmonares/terapia , Antígenos Específicos del Melanoma/genética , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Línea Celular Tumoral , Cromatina/química , Metilación de ADN , Perfilación de la Expresión Génica , Histona Metiltransferasas , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/inmunología , Lisina/metabolismo , Metilación
7.
J Immunol ; 186(2): 685-96, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149604

RESUMEN

Adoptive immunotherapy using TCR-engineered PBLs against melanocyte differentiation Ags mediates objective tumor regression but is associated with on-target toxicity. To avoid toxicity to normal tissues, we targeted cancer testis Ag (CTA) MAGE-A3, which is widely expressed in a range of epithelial malignancies but is not expressed in most normal tissues. To generate high-avidity TCRs against MAGE-A3, we employed a transgenic mouse model that expresses the human HLA-A*0201 molecule. Mice were immunized with two HLA-A*0201-restricted peptides of MAGE-A3: 112-120 (KVAELVHFL) or MAGE-A3: 271-279 (FLWGPRALV), and T cell clones were generated. MAGE-A3-specific TCR α- and ß-chains were isolated and cloned into a retroviral vector. Expression of both TCRs in human PBLs demonstrated Ag-specific reactivity against a range of melanoma and nonmelanoma tumor cells. The TCR against MAGE-A3: 112-120 was selected for further development based on superior reactivity against tumor target cells. Interestingly, peptide epitopes from MAGE-A3 and MAGE-A12 (and to a lesser extent, peptides from MAGE-A2 and MAGE-A6) were recognized by PBLs engineered to express this TCR. To further improve TCR function, single amino acid variants of the CDR3 α-chain were generated. Substitution of alanine to threonine at position 118 of the α-chain in the CDR3 region of the TCR improved its functional avidity in CD4 and CD8 cells. On the basis of these results, a clinical trial is planned in which patients bearing a variety of tumor histologies will receive autologous PBLs that have been transduced with this optimized anti-MAGE-A3 TCR.


Asunto(s)
Antígenos de Neoplasias/inmunología , Pruebas Inmunológicas de Citotoxicidad , Epítopos de Linfocito T/metabolismo , Antígenos HLA-A/metabolismo , Proteínas de Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/biosíntesis , Secuencia de Bases , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/terapia , Línea Celular Transformada , Línea Celular Tumoral , Técnicas de Cocultivo , Pruebas Inmunológicas de Citotoxicidad/métodos , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/inmunología , Femenino , Antígenos HLA-A/biosíntesis , Antígenos HLA-A/inmunología , Antígeno HLA-A2 , Humanos , Inmunoterapia Adoptiva/métodos , Interferón gamma/metabolismo , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/terapia , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Familia de Multigenes/inmunología , Proteínas de Neoplasias/administración & dosificación , Proteínas de Neoplasias/biosíntesis , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante
8.
Methods Mol Biol ; 515: 137-50, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19378113

RESUMEN

In the last decade lentiviral gene transfer vectors have gained significant place both in basic science and gene therapy applications. A number of gene transfer applications would benefit from vectors capable of expressing multiple genes. This chapter focuses on production of bicistronic and tricistronic lentiviral vectors based on the internal ribosomal entry site (IRES) sequence of encephalomyocarditis virus (EMCV) and/or foot-and-mouth disease virus (FMDV) cleavage factor 2A. Multigene vectors produced high titer viral particles and were able to simultaneously express two or three transgenes in transduced cells. The level of expression of individual transgenes varied depending on the transgene itself, its position within the construct, the total number of transgenes expressed, the strategy used for multigene expression, and the number of copies of proviral insertions.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/genética , VIH-1/genética , Lentivirus/genética , Línea Celular , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Genes Reporteros/genética , Humanos , Técnicas de Cultivo de Tejidos
9.
Stem Cells ; 25(2): 490-9, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17284653

RESUMEN

Progress toward clinical application of ESC-derived hematopoietic cellular transplantation will require rigorous evaluation in a large animal allogeneic model. However, in contrast to human ESCs (hESCs), efforts to induce conclusive hematopoietic differentiation from rhesus macaque ESCs (rESCs) have been unsuccessful. Characterizing these poorly understood functional differences will facilitate progress in this area and likely clarify the critical steps involved in the hematopoietic differentiation of ESCs. To accomplish this goal, we compared the hematopoietic differentiation of hESCs with that of rESCs in both EB culture and stroma coculture. Initially, undifferentiated rESCs and hESCs were adapted to growth on Matrigel without a change in their phenotype or karyotype. Subsequent differentiation of rESCs in OP9 stroma led to the development of CD34(+)CD45(-) cells that gave rise to endothelial cell networks in methylcellulose culture. In the same conditions, hESCs exhibited convincing hematopoietic differentiation. In cytokine-supplemented EB culture, rESCs demonstrated improved hematopoietic differentiation with higher levels of CD34(+) and detectable levels of CD45(+) cells. However, these levels remained dramatically lower than those for hESCs in identical culture conditions. Subsequent plating of cytokine-supplemented rhesus EBs in methylcellulose culture led to the formation of mixed colonies of erythroid, myeloid, and endothelial cells, confirming the existence of bipotential hematoendothelial progenitors in the cytokine-supplemented EB cultures. Evaluation of four different rESC lines confirmed the validity of these disparities. Although rESCs have the potential for hematopoietic differentiation, they exhibit a pause at the hemangioblast stage of hematopoietic development in culture conditions developed for hESCs.


Asunto(s)
Células Madre Embrionarias/citología , Sistema Hematopoyético/citología , Macaca mulatta , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Colágeno/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Citocinas/farmacología , Combinación de Medicamentos , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Fibroblastos , Perfilación de la Expresión Génica , Sistema Hematopoyético/efectos de los fármacos , Humanos , Cariotipificación , Laminina/efectos de los fármacos , Ratones , Fenotipo , Proteoglicanos/efectos de los fármacos , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
10.
Cancer Immunol Immunother ; 56(6): 885-95, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17102977

RESUMEN

Prostatic acid phosphatase (PAP) is a prostate cancer tumor antigen and a prostate-specific protein shared by rats and humans. Previous studies indicated that Copenhagen rats immunized with a recombinant vaccinia virus expressing human PAP (hPAP) developed PAP-specific cytotoxic T cells (CTL) with cross reactivity to rat PAP (rPAP) and evidence of prostate inflammation. Viral delivery of vaccine antigens is an active area of clinical investigation. However, a potential difficulty with viral-based immunizations is that immune responses elicited to the viral vector might limit the possibility of multiple immunizations. In this paper, we investigate the ability of another genetic immunization method, a DNA vaccine encoding PAP, to elicit antigen-specific CD8+ T cell immune responses. Specifically, Lewis rats were immunized with either a plasmid DNA-based (pTVG-HP) or vaccinia-based (VV-HP) vaccine each encoding hPAP. We determined that rats immunized with a DNA vaccine encoding hPAP developed a Th1-biased immune response as indicated by proliferating PAP-specific CD4+ and CD8+ cells and IFNgamma production. Rats immunized with vaccinia virus encoding PAP did not develop a PAP-specific response unless boosted with a heterologous vaccination scheme. Most importantly, multiple immunizations with a DNA vaccine encoding the rat PAP homologue (pTVG-RP) could overcome peripheral self-tolerance against rPAP and generate a Th1-biased antigen-specific CD4+ and CD8+ T cell response. Overall, DNA vaccines provide a safe and effective method of generating prostate antigen-specific T cell responses. These findings support the investigation of PAP-specific DNA vaccines in human clinical trials.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Neoplasias de la Próstata/terapia , Proteínas Tirosina Fosfatasas/inmunología , Vacunas de ADN/inmunología , Fosfatasa Ácida , Animales , Autoantígenos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Vectores Genéticos/inmunología , Humanos , Masculino , Plásmidos , Neoplasias de la Próstata/inmunología , Ratas , Ratas Endogámicas Lew , Transducción Genética , Vacunas de ADN/administración & dosificación , Virus Vaccinia/genética
11.
Cancer Immunol Immunother ; 55(12): 1504-14, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16612600

RESUMEN

Fusion proteins consisting of the ligand-binding domain of CTLA4 covalently attached to an antigen (Ag) are potent immunogens. This fusion strategy effectively induces Ag-specific immunity both when introduced as a DNA-based vaccine and as a recombinant protein. CTLA4 is a ligand for B7 molecules expressed on the surface of antigen-presenting cells (APCs), and this interaction is critical for the fusion protein to stimulate Ag-specific immunity. We show that interaction of the fusion protein with either B7-1 or B7-2 is sufficient to stimulate immune activity, and that T cells are essential for the development of IgG responses. In addition, we demonstrate that human dendritic cells (DCs) pulsed with CTLA4-Ag fusion proteins can efficiently present Ag to T cells and induce an Ag-specific immune response in vitro. These studies provide further mechanistic understanding of the process by which CTLA4-Ag fusion proteins stimulate the immune system, and represent an efficient means of generating Ag-specific T cells for immunotherapy.


Asunto(s)
Antígenos CD/genética , Antígenos de Diferenciación/genética , Antígenos/inmunología , Activación de Linfocitos , Proteínas Recombinantes de Fusión/inmunología , Animales , Formación de Anticuerpos , Antígenos/genética , Antígenos CD/química , Antígenos de Diferenciación/química , Antígeno B7-1/inmunología , Antígeno B7-2/inmunología , Antígeno CTLA-4 , Células Dendríticas/efectos de los fármacos , Humanos , Inmunoglobulina G/inmunología , Ligandos , Ratones , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología
12.
Virol J ; 3: 14, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16539700

RESUMEN

BACKGROUND: A number of gene therapy applications would benefit from vectors capable of expressing multiple genes. In this study we explored the feasibility and efficiency of expressing two or three transgenes in HIV-1 based lentiviral vector. Bicistronic and tricistronic self-inactivating lentiviral vectors were constructed employing the internal ribosomal entry site (IRES) sequence of encephalomyocarditis virus (EMCV) and/or foot-and-mouth disease virus (FMDV) cleavage factor 2A. We employed enhanced green fluorescent protein (eGFP), O6-methylguanine-DNA-methyltransferase (MGMT), and homeobox transcription factor HOXB4 as model genes and their expression was detected by appropriate methods including fluorescence microscopy, flow cytometry, immunocytochemistry, biochemical assay, and western blotting. RESULTS: All the multigene vectors produced high titer virus and were able to simultaneously express two or three transgenes in transduced cells. However, the level of expression of individual transgenes varied depending on: the transgene itself; its position within the construct; the total number of transgenes expressed; the strategy used for multigene expression and the average copy number of pro-viral insertions. Notably, at limiting MOI, the expression of eGFP in a bicistronic vector based on 2A was approximately 4 times greater than that of an IRES based vector. CONCLUSION: The small and efficient 2A sequence can be used alone or in combination with an IRES for the construction of multicistronic lentiviral vectors which can express encoded transgenes at functionally relevant levels in cells containing an average of one pro-viral insert.


Asunto(s)
Virus de la Fiebre Aftosa/genética , Vectores Genéticos/genética , Lentivirus/genética , Virus de la Encefalomiocarditis/genética , Terapia Genética , Proteínas Fluorescentes Verdes/genética , Células HeLa , Proteínas de Homeodominio/genética , Humanos , Células K562 , O(6)-Metilguanina-ADN Metiltransferasa/genética , Ribosomas/metabolismo , Factores de Transcripción/genética , Transgenes
13.
Hum Gene Ther ; 15(8): 758-69, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15319033

RESUMEN

Lentiviral vectors are capable of efficiently transducing nondividing and slowly dividing cells, including hematopoietic stem cells, resulting in stable integration and sustained transgene expression. We constructed human immunodeficiency virus type 1-based self-inactivating lentiviral vectors to express either wild-type or an O6-benzylguanine (O6-beG)-resistant mutant form of the human O6-alkylguanine-DNA methyltransferase (MGMT; DNA-O6-methylguanine:[protein]-L-cysteine S-methyltransferase, EC 2.1.1.63) and transduced K562 and granulocyte colony-stimulating factor-mobilized human peripheral blood CD34+ cells. After transduction, K562 cells expressed high levels of MGMT as determined by Western blot, immunocytochemistry, and biochemical assay. A colony-forming survival assay showed significant protection against O6-beG plus 1,3-bis(2-chloroethyl)-nitrosourea (BCNU) or temozolomide (TMZ) toxicity. Similarly, a single transduction of CD34+ cells resulted in a 13- to 14-fold increase in the level of MGMT expression. In comparison with non-transduced cells, mutant MGMTP140K-transduced CD34+ cells showed significant resistance against the combined toxicity of O6-beG with either TMZ or BCNU: there was an approximately 9-fold increase in the survival of colony-forming cells as indicated by the IC50 values after O6-beG plus TMZ treatment and an approximately 5-fold increase in the case of O6-beG plus BCNU treatment. These results show that lentivirus-mediated expression of MGMTP140K can efficiently protect the hematopoietic compartment against the combined toxicity of O6-beG plus TMZ or BCNU.


Asunto(s)
Dacarbazina/análogos & derivados , Regulación Enzimológica de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/genética , Células Madre Hematopoyéticas/enzimología , Lentivirus/genética , O(6)-Metilguanina-ADN Metiltransferasa/genética , Antígenos CD34/metabolismo , Western Blotting , Carmustina/toxicidad , Línea Celular , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN , Dacarbazina/toxicidad , Citometría de Flujo , Vectores Genéticos/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Inmunohistoquímica , Concentración 50 Inhibidora , Reacción en Cadena de la Polimerasa , Temozolomida , Transducción Genética , Transgenes/genética , Células Tumorales Cultivadas
14.
Hum Gene Ther ; 14(6): 497-507, 2003 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-12718761

RESUMEN

The use of lentiviral vectors for basic research and potential future clinical applications requires methodologies that can accurately determine lentiviral titers and monitor viral transgene expression within target cells, beyond the context of reporter genes typically used for this purpose. Here we describe a quantitative RT-PCR (qRT-PCR) method that achieves both goals using primer sequences that are specific for the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE), an enhancer contained in many retroviral vectors and that is incorporated in the 3' UTR of nascent transgene transcripts. Quantitation of titers of three recombinant lentiviruses, genetically identical except for the transgene, demonstrated consistent differences in titer that were likely due to transgene-associated toxicity in producer cells and target cells. Viruses encoding the tumor-associated antigens tyrosinase and neo-poly(A) polymerase yielded reproducibly lower titers than a virus encoding enhanced green fluorescent protein (GFP) at the viral RNA, integrated DNA, and transgene mRNA levels, as measured by WPRE qPCR. Furthermore, the magnitude of differences in expression of the three transgenes in transduced target cells could not have been predicted by measuring vector DNA integration events. Since transgene expression in target cells is the most common goal of lentiviral transduction, and since methods to quantify transgene expression on the protein level are not always readily available, qRT-PCR based on a nucleotide sequence included in the transcript provides a useful tool for titering novel recombinant lentiviruses.


Asunto(s)
Vectores Genéticos , Lentivirus/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transgenes , Regiones no Traducidas 3'/genética , Línea Celular , Línea Celular Transformada , Elementos de Facilitación Genéticos/genética , Citometría de Flujo , Expresión Génica , Genes Reguladores/genética , Proteínas Fluorescentes Verdes , Células HeLa , Virus de Hepatitis/genética , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , ARN Mensajero/biosíntesis , Transducción Genética , Virión/genética , Integración Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...