Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
MAbs ; 16(1): 2329321, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38494955

RESUMEN

Antibody-mediated delivery of immunogenic viral CD8+ T-cell epitopes to redirect virus-specific T cells toward cancer cells is a promising new therapeutic avenue to increase the immunogenicity of tumors. Multiple strategies for viral epitope delivery have been shown to be effective. So far, most of these have relied on a free C-terminus of the immunogenic epitope for extracellular delivery. Here, we demonstrate that antibody-epitope conjugates (AECs) with genetically fused epitopes to the N-terminus of the antibody can also sensitize tumors for attack by virus-specific CD8+ T cells. AECs carrying epitopes genetically fused at the N-terminus of the light chains of cetuximab and trastuzumab demonstrate an even more efficient delivery of the T-cell epitopes compared to AECs with the epitope fused to the C-terminus of the heavy chain. We demonstrate that this increased efficiency is not caused by the shift in location of the cleavage site from the N- to the C-terminus, but by its increased proximity to the cell surface. We hypothesize that this facilitates more efficient epitope delivery. These findings not only provide additional insights into the mechanism of action of AECs but also broaden the possibilities for genetically fused AECs as an avenue for the redirection of multiple virus-specific T cells toward tumors.


Asunto(s)
Inmunoconjugados , Neoplasias , Humanos , Epítopos de Linfocito T , Linfocitos T CD8-positivos , Anticuerpos , Neoplasias/terapia
2.
Exp Neurol ; 355: 114120, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35605669

RESUMEN

Gene therapy is a powerful approach to promote spinal cord regeneration. For a clinical application it is important to restrict therapeutic gene expression to the appropriate time window to limit unwanted side effects. The doxycycline (dox)-inducible system is a widely used regulatable gene expression platform, however, this system depends on a bacterial-derived immunogenic transactivator. The foreign origin of this transactivator prevents reliable regulation of therapeutic gene expression and currently limits clinical translation. The glycine-alanine repeat (GAR) of Epstein-Barr virus nuclear antigen-1 protein inhibits its presentation to cytotoxic T cells, allowing virus-infected cells to evade the host immune system. We developed a chimeric transactivator (GARrtTA) and show that GARrtTA has an immune-evading advantage over "classical" rtTA in vivo. Direct comparison of lentiviral vectors expressing rtTA and GARrtTA in the rat spinal cord shows that the GARrtTA system is inducible for 6 doxycycline-cycles over a 47 week period, whereas with the rtTA-based system luciferase reporter expression declines during the 3rd cycle and is no longer re-inducible, indicating that GARrtTA provides an immune-advantage over rtTA. Immunohistochemistry revealed that GARrtTA expressing cells in the spinal cord appear healthier and survive better than rtTA expressing cells. Characterization of the immune response shows that expression of GARrtTA, in contrast to rtTA, does not recruit cytotoxic T-cells to the transduced spinal cord. This study demonstrates that fusion of the GAR domain to rtTA results in a functional doxycycline-inducible transactivator with a clear immune-advantage over the classical rtTA in vivo.


Asunto(s)
Doxiciclina , Infecciones por Virus de Epstein-Barr , Animales , Doxiciclina/farmacología , Regulación de la Expresión Génica , Terapia Genética/métodos , Herpesvirus Humano 4/genética , Ratas , Médula Espinal , Transactivadores/genética
3.
Gene Ther ; 24(7): 385-391, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28485720

RESUMEN

Transplantation of allogeneic cells as well as of genetically corrected autologous cells are potent approaches to restore cellular functions in patients suffering from genetic diseases. The recipient's immune responses against non-self-antigens may compromise the survival of the grafted cells. Recipients of the graft may therefore require lifelong treatment with immunosuppressive drugs. An alternative approach to reduce graft rejection could involve the use of immune-evasion molecules. Expression of such molecules in cells of the graft may subvert recognition by the host's immune system. Viruses in particular are masters of exploitation and modulation of their hosts immune response. The Herpesviridae family provides a proof of concept for this as these viruses are capable to establish latency and a lifelong persistence in the infected hosts. While several viral proteins involved in immune evasion have been characterized, the Herpesviridae also encode a multitude of viral microRNA (miRNAs). Several of these miRNAs have been demonstrated to reduce the sensitivity of the infected cells to the destructive action of the host's immune cells. In this review, the miRNAs of some common herpesviruses that are associated with immune modulation will be discussed with a focus on their potential use in strategies aiming at generating non-immunogenic cells for transplantation.


Asunto(s)
Terapia Genética/métodos , Rechazo de Injerto/terapia , Evasión Inmune , MicroARNs/genética , ARN Viral/genética , Animales , Herpesviridae/genética , Humanos
4.
Gene Ther ; 22(3): 267-79, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25588743

RESUMEN

The capacity to modify the reovirus genome facilitates generation of new therapeutic reoviruses. We describe a method for generating replication-competent reoviruses carrying a heterologous transgene. The strategy is based on the expanded-tropism reovirus mutant jin-3, which can infect cells independent of the reovirus receptor junction-adhesion molecule A (JAM-A). Jin-3 harbors a mutation in the S1 segment, resulting in a G196R substitution in the tail of the spike protein σ1. The use of the jin-3 tail-encoding S1 segment allows replacing the codons for the JAM-A-binding head domain by up to 522 nucleotides of foreign sequences, without exceeding the size of the wild-type S1 segment. We inserted the codons for the porcine teschovirus-1 2A element fused with those encoding the fluorescent protein iLOV. Replicating rS1His-2A-iLOV reoviruses were generated by co-transfection of expression plasmids for all reovirus segments. These reoviruses contain the S1His-2A-iLOV segment in the absence of the wild-type S1 segment. Density-gradient centrifugation confirmed the association of the σ1-tail fragment with the capsid. Both JAM-A-positive and -negative cells exposed to the rS1His-2A-iLOV reoviruses exhibited iLOV fluorescence, confirming the jin-3-derived expanded-tropism phenotype. These data demonstrated the feasibility of generating decapitated replication-competent T3D reoviruses carrying a heterologous transgene.


Asunto(s)
Proteínas de la Cápside/genética , Codón/genética , Reoviridae/fisiología , Transgenes , Replicación Viral , Animales , Secuencia de Bases , Proteínas de la Cápside/metabolismo , Línea Celular , Cricetinae , Humanos , Datos de Secuencia Molecular , Viroterapia Oncolítica
5.
Ann Rheum Dis ; 74(10): 1915-23, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24818634

RESUMEN

OBJECTIVE: Mast cells may play a role in rheumatoid arthritis (RA), but activation of human mast cells in autoimmune settings has been little studied. Toll-like receptors (TLR) and Fcγ receptors (FcγR) are important receptors for cellular activation in the joint, but expression and stimulation of these receptors in human mast cells or the functional interplay between these pathways is poorly understood. Here, we analysed triggering of human mast cells via these receptors in the context of anti-citrullinated protein antibody-positive (ACPA+) RA. METHODS: RNA and protein expression of TLRs and FcγR was quantified using PCR and flow cytometry, respectively. Mast cells were stimulated with TLR ligands (including HSP70) combined with IgG immune complexes and IgG-ACPA. RESULTS: Human mast cells expressed TLRs and produced cytokines in response to TLR ligands. Both cultured and synovial mast cells expressed FcγRIIA, and triggering of this receptor by IgG immune complexes synergised with activation by TLR ligands, leading to two- to fivefold increased cytokine levels. Mast cells produced cytokines in response to ACPA immune complexes in a citrulline-specific manner, which synergised in the presence of HSP70. CONCLUSIONS: Our data show that synovial mast cells express FcγRIIA and that mast cells can be activated by IgG-ACPA and TLR ligands. Importantly, combined stimulation via TLRs and immune complexes leads to synergy in cytokine production. These findings suggest mast cells are important targets for TLR ligands and immune complexes, and that combined activation of mast cells via these pathways greatly enhances inflammation in synovial tissue of RA patients.


Asunto(s)
Mastocitos/inmunología , Péptidos Cíclicos/inmunología , Receptores Toll-Like/biosíntesis , Complejo Antígeno-Anticuerpo/inmunología , Artritis Reumatoide/inmunología , Células Cultivadas , Citocinas/biosíntesis , Regulación de la Expresión Génica/inmunología , Humanos , Inmunoglobulina G/inmunología , Ligandos , Osteoartritis/inmunología , ARN Mensajero/genética , Receptores de IgG/inmunología , Membrana Sinovial/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
6.
Gene Ther ; 21(6): 549-57, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24694534

RESUMEN

Viral vector-mediated gene transfer of neurotrophic factors is an emerging and promising strategy to promote the regeneration of injured peripheral nerves. Unfortunately, the chronic exposure to neurotrophic factors results in local trapping of regenerating axons or other unwanted side effects. Therefore, tight control of therapeutic gene expression is required. The tetracycline/doxycycline-inducible system is considered to be one of the most promising systems for regulating heterologous gene expression. However, an immune response directed against the transactivator protein rtTA hampers further translational studies. Immunogenic proteins fused with the Gly-Ala repeat of the Epstein-Barr virus Nuclear Antigen-1 protein have been shown to successfully evade the immune system. In this article, we used this strategy to demonstrate that a chimeric transactivator, created by fusing the Gly-Ala repeat with rtTA and embedded in a lentiviral vector (i) retained its transactivator function in vitro, in muscle explants, and in vivo following injection into the rat peripheral nerve, (ii) exhibited a reduced leaky expression, and (iii) had an immune-evasive advantage over rtTA as shown in a novel bioassay for human antigen presentation. The current findings are an important step toward creating a clinically applicable potentially immune-evasive tetracycline-regulatable viral vector system.


Asunto(s)
Vectores Genéticos/farmacología , Nervios Periféricos/efectos de los fármacos , Tetraciclina/farmacología , Animales , Secuencia de Bases , Femenino , Regulación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Células HEK293 , Humanos , Técnicas In Vitro , Lentivirus/genética , Datos de Secuencia Molecular , Músculo Esquelético/fisiología , Ratas Wistar , Linfocitos T Citotóxicos/inmunología , Transactivadores/genética , Transactivadores/metabolismo
7.
Gene Ther ; 21(6): 609-17, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24739522

RESUMEN

In the canonical pathway, infection of cells by the wild-type mammalian orthoreovirus Type 3 Dearing (T3D) is dependent on the interaction of the viral spike protein σ1 with the high-affinity cellular receptor junction adhesion molecule-A (JAM-A). We previously demonstrated that the human glioblastoma cell line U-118 MG does not express JAM-A and resists reovirus T3D infection in standard cell culture conditions (SCCC). Heterologous JAM-A expression sensitises U-118 MG cells to reovirus T3D. Here we studied reovirus infection in U-118 MG cells grown in spheroid cultures with the premise that cells in such cultures resemble cells in tumours more than those grown under standard adherent cell culture conditions on a plastic surface. Although the U-118 MG cells in spheroids do not express JAM-A, they are susceptible to reovirus T3D infection. We show that this can be attributed to factors secreted by cells in the spheroids. The concentration of active extracellular proteases cathepsin B and L in the medium of spheroid cultures was increased 19- and 24-fold, respectively, as compared with SCCC. These enzymes can convert the reovirus particles into a form that can infect the U-118 MG cells independent of JAM-A. Taken together, these data demonstrate that infection of tumour cells by wild-type reovirus T3D is not strictly dependent on the expression of JAM-A on the cell surface.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Glioblastoma/patología , Glioblastoma/virología , Orthoreovirus Mamífero 3/patogenicidad , Receptores de Superficie Celular/metabolismo , Esferoides Celulares/virología , Catepsina B/metabolismo , Catepsina L/metabolismo , Humanos
8.
Cancer Gene Ther ; 20(9): 507-13, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23907517

RESUMEN

Glioblastoma (GB) is a devastating disease for which new treatment modalities are needed. Efficacious therapy requires the removal of stem-cell like cells, these cells drive tumor progression because of their ability to self-renew and differentiate. In glioblastoma, the GB stem-like cells (GSC) form a small population of tumor cells and possess high resistance to chemo and radiation therapies. To assess the sensitivity of GSC to reovirus-mediated cytolysis, a panel of GSC cultures was exposed to wild-type reovirus Type 3 Dearing (T3D) and its junction adhesion molecule-A (JAM-A)-independent mutant, jin-1. Several parameters were evaluated, including the fraction of cells expressing the JAM-A reovirus receptor, the fraction of cells synthesizing reovirus proteins, the number of infectious reovirus particles required to reduce cell viability, the amount of infectious progeny reovirus produced and the capacity of the reoviruses to infect the GSC in 3-dimensional (3D) tumor cell spheroids. Our data demonstrate a marked heterogeneity in the susceptibility of the cultures to reovirus-induced cytolysis. While in monolayer cultures the jin-1 reovirus was generally more cytolytic than the wild-type reovirus T3D, in the 3D GSC spheroids, these viruses were equally effective. Despite the variation in reovirus sensitivity between the different GSC cultures, our data support the use of reovirus as an oncolytic agent. It remains to be established whether the variation in the reovirus sensitivity correlates with a patient's response to reovirus therapy. Moreover, our data show that the expression of the JAM-A receptor is not a major determinant of reovirus sensitivity in 3D GSC cultures.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/virología , Reoviridae/fisiología , Tropismo Viral , Adulto , Anciano , Neoplasias Encefálicas/virología , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Efecto Citopatogénico Viral , Femenino , Glioblastoma/virología , Humanos , Masculino , Persona de Mediana Edad , Virus Oncolíticos/fisiología , Receptores de Superficie Celular/metabolismo , Esferoides Celulares , Células Tumorales Cultivadas
9.
Differentiation ; 85(3): 101-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23665895

RESUMEN

The generation of human induced pluripotent stem cells (hiPSCs) requires the collection of donor tissue, but clinical circumstances in which the interests of patients have highest priority may compromise the quality and availability of cells that are eventually used for reprogramming. Here we compared (i) skin biopsies stored in standard physiological salt solution for up to two weeks (ii) blood outgrowth endothelial cells (BOECs) isolated from fresh peripheral blood and (iii) children's milk teeth lost during normal replacement for their ability to form somatic cell cultures suitable for reprogramming to hiPSCs. We derived all hiPSC lines using the same reprogramming method (a conditional (FLPe) polycistronic lentivirus) and under similar conditions (same batch of virus, fetal calf serum and feeder cells). Skin fibroblasts could be reprogrammed robustly even after long-term biopsy storage. Generation of hiPSCs from juvenile dental pulp cells gave similar high efficiencies, but that of BOECs was lower. In terms of invasiveness of biopsy sampling, biopsy storage and reprogramming efficiencies skin fibroblasts appeared best for the generation of hiPSCs, but where non-invasive procedures are required (e.g., for children and minors) dental pulp cells from milk teeth represent a valuable alternative.


Asunto(s)
Células Endoteliales/citología , Células Madre Pluripotentes Inducidas/citología , Piel/citología , Diente Primario/citología , Biopsia , Células Sanguíneas/citología , Diferenciación Celular/genética , Genes/genética , Humanos , Lentivirus , Cloruro de Sodio
10.
Hum Gene Ther ; 23(1): 70-82, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21875358

RESUMEN

The efficacy of adenovirus (Ad)-based gene therapy of solid tumors, such as prostate cancer, is limited. One of the many problems is that the virus infects many different cell types in the body, resulting in high toxicity, whereas the target cancer cells are often less prone to wild-type Ad infection. Our aim was to develop genetically de- and retargeted Ad vectors to reduce off-target effects and increase target infection for prostate cancer. We have previously reported an Ad5 vector specific for the cancer-associated receptor Her2/neu, created by inserting Her2/neu-reactive Affibody(®) molecules (ZH) into the HI loop of a coxsackievirus and adenovirus receptor binding-ablated fiber (Ad[ZH/1]). In addition to virus retargeting to Her2/neu, this virus was further modified from wild-type Ad by changing the RGD motif in the penton base to EGD and by substitution of the KKTK motif in the third shaft repeat to RKSK, resulting in the vector Ad[ZH/3]. The ZH-containing vectors could be produced to high titers and were specific for their target, resulting in efficient infection and killing of Her2/neu-positive androgen-dependent PC346C prostate cancer cells in vitro. Here we show that the oncolytic Ad[ZH/3] vector significantly prolonged survival time and reduced serum prostate-specific antigen levels in an orthotopic prostate tumor model in nude mice to the same extent as wild-type Ad5. Our results show that Her2/neu targeting using Ad-based vectors for prostate cancer is feasible and may serve as a basis for the development of gene therapy of human prostate cancer as well as other Her2/neu-expressing cancers.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/uso terapéutico , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/terapia , Receptor ErbB-2/metabolismo , Adenoviridae/metabolismo , Animales , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Necrosis , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Receptor ErbB-2/genética , Factores de Tiempo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Gene Ther ; 19(1): 58-68, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22015640

RESUMEN

Oncolytic adenoviruses are promising anticancer agents. To study and optimize their tumor-killing potency, genuine tumor models are required. Here we describe the use of the chicken chorioallantoic membrane (CAM) tumor model in studies on oncolytic adenoviral vectors. Suspensions of human melanoma, colorectal carcinoma and glioblastoma multiforme cell lines were grafted on the CAM of embryonated chicken eggs. All cell lines tested formed 5-10 mm size tumors, which recapitulated hallmarks of corresponding human specimens. Furthermore, melanoma tumors were injected with adenoviral vector-carrying gene encoding the fusion protein of parainfluenza virus type 5. This led to the induction of cell fusion and syncytia formation in the infected cells. At 6 days post-injection, histological and immunohistochemical analyses of tumor sections confirmed adenovirus replication and syncytia formation. These results demonstrate that the CAM model allows rapid assessment of oncolytic viruses in three-dimensional tumors. Hence, this model constitutes an easy and affordable system for preclinical characterization of viral oncolytic agents that may precede the mandatory process of animal testing. Application of this model will help reducing the use of human xenografts in mice for preclinical evaluation of oncolytic viruses and other anticancer agents.


Asunto(s)
Membrana Corioalantoides/virología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Adenoviridae/genética , Animales , Línea Celular Tumoral , Pollos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Neoplasias Colorrectales/virología , Vectores Genéticos/genética , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/terapia , Glioblastoma/virología , Humanos , Inmunohistoquímica , Melanoma/genética , Melanoma/patología , Melanoma/terapia , Melanoma/virología
12.
Gene Ther ; 19(9): 899-906, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22011643

RESUMEN

Human adenoviruses have a great potential as anticancer agents. One strategy to improve their tumor-cell specificity and anti-tumor efficacy is to include tumor-specific targeting ligands in the viral capsid. This can be achieved by fusion of polypeptide-targeting ligands with the minor capsid protein IX. Previous research suggested that protein IX-mediated targeting is limited by inefficient release of protein IX-fused ligands from their cognate receptors in the endosome. This thwarts endosomal escape of the virus particles. Here we describe that the targeted transduction of tumor cells is augmented by a cathepsin-cleavage site between the protein IX anchor and the HER2/neu-binding ZH Affibody molecule as ligand. The cathepsin-cleavage site did not interfere with virus production and incorporation of the Affibody molecules in the virus capsid. Virus particles harboring the cleavable protein IX-ligand fusion in their capsid transduced the HER2/neu-positive SKOV-3 ovarian carcinoma cells with increased efficiency in monolayer cultures, three-dimensional spheroid cultures and in SKOV-3 tumors grown on the chorioallantoic membrane of embryonated chicken eggs. These data show that inclusion of a cathepsin-cleavage sequence between protein IX and a high-affinity targeting ligand enhances targeted transduction. This modification further augments the applicability of protein IX as an anchor for coupling tumor-targeting ligands.


Asunto(s)
Adenovirus Humanos/genética , Proteínas de la Cápside/metabolismo , Catepsinas/química , Vectores Genéticos , Ligandos , Transducción Genética , Línea Celular Tumoral , Marcación de Gen , Humanos , Neoplasias/terapia , Proteínas Recombinantes de Fusión/química
13.
Leukemia ; 25(9): 1471-83, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21617701

RESUMEN

Severe combined immunodeficiency (SCID) patients with an inactivating mutation in recombination activation gene 1 (RAG1) lack B and T cells due to the inability to rearrange immunoglobulin (Ig) and T-cell receptor (TCR) genes. Gene therapy is a valid treatment option for RAG-SCID patients, especially for patients lacking a suitable bone marrow donor, but developing such therapy has proven challenging. As a preclinical model for RAG-SCID, we used Rag1-/- mice and lentiviral self-inactivating (SIN) vectors harboring different internal elements to deliver native or codon-optimized human RAG1 sequences. Treatment resulted in the appearance of B and T cells in peripheral blood and developing B and T cells were detected in central lymphoid organs. Serum Ig levels and Ig and TCR Vß gene segment usage was comparable to wild-type (WT) controls, indicating that RAG-mediated rearrangement took place. Remarkably, relatively low frequencies of B cells produced WT levels of serum immunoglobulins. Upon stimulation of the TCR, corrected spleen cells proliferated and produced cytokines. In vivo challenge resulted in production of antigen-specific antibodies. No leukemia development as consequence of insertional mutagenesis was observed. The functional reconstitution of the B- as well as the T-cell compartment provides proof-of-principle for therapeutic RAG1 gene transfer in Rag1-/- mice using lentiviral SIN vectors.


Asunto(s)
Terapia Genética , Vectores Genéticos/administración & dosificación , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Lentivirus/genética , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/terapia , Animales , Linfocitos B/fisiología , Western Blotting , Médula Ósea/metabolismo , Médula Ósea/patología , Trasplante de Médula Ósea , Proliferación Celular , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Reordenamiento Génico , Técnicas de Transferencia de Gen , Humanos , Inmunoglobulina G/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Receptores de Antígenos de Linfocitos T/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Linfocitos T/fisiología , Transgenes/fisiología
14.
Gene Ther ; 15(24): 1567-78, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18650851

RESUMEN

Human Orthoreovirus Type 3 Dearing is not pathogenic to humans and has been evaluated clinically as an oncolytic agent. Its transduction efficiency and the tumor cell selectivity may be enhanced by incorporating ligands for alternative receptors. However, the genetic modification of reoviruses has been difficult, and genetic targeting of reoviruses has not been reported so far. Here we describe a technique for generating genetically targeted reoviruses. The propagation of wild-type reoviruses on cells expressing a modified sigma 1-encoding segment embedded in a conventional RNA polymerase II transcript leads to substitution of the wild-type genome segment by the modified version. This technique was used for generating reoviruses that are genetically targeted to an artificial receptor expressed on U118MG cells. These cells lack the junction adhesion molecule-1 and therefore resist infection by wild-type reoviruses. The targeted reoviruses were engineered to carry the ligand for this receptor at the C terminus of the sigma 1 spike protein. This demonstrates that the C terminus of the sigma 1 protein is a suitable locale for the insertion of oligopeptide ligands and that targeting of reoviruses is feasible. The genetically targeted viruses can be propagated using the modified U118MG cells as helper cells. This technique may be applicable for the improvement of human reoviruses as oncolytic agents.


Asunto(s)
Moléculas de Adhesión Celular/genética , Terapia Genética/métodos , Orthoreovirus Mamífero 3/genética , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Secuencia de Aminoácidos , Efecto Espectador , Línea Celular Tumoral , Supervivencia Celular , Clonación Molecular , Marcación de Gen , Ingeniería Genética , Humanos , Fragmentos de Inmunoglobulinas , Moléculas de Adhesión de Unión , Ligandos , Datos de Secuencia Molecular , Neoplasias/patología , Neoplasias/virología , Alineación de Secuencia , Transducción Genética/métodos
15.
Gene Ther ; 15(13): 978-89, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18323790

RESUMEN

Adenovirus vectors have great potential in cancer gene therapy. Targeting of cancer-testis (CT) antigens, which are specifically presented at the surface of tumor cells by human leukocyte antigen (HLA) class I molecules, is an attractive option. In this study, a single-chain T-cell receptor (scTCR) directed against the CT antigen melanoma-associated antigen (MAGE)-A1 in complex with the HLA class I molecule of haplotype HLA-A1 is fused with the C terminus of the adenovirus minor capsid protein IX. Propagation of a protein-IX (pIX)-gene-deleted human adenovirus 5 (HAdV-5) vector on cells that constitutively express the pIXscTCR fusion protein yielded viral particles with the pIXscTCR fusion protein incorporated in their capsid. Generated particles specifically transduced melanoma cell lines expressing the HLA-A1/MAGE-A1 target complex with at least 10-fold higher efficiency than control viruses. Whereas loading of HLA-A1-positive cells with MAGE-A1 peptides leads to enhanced transduction of the cells, the efficiency of virus transduction is strongly reduced if the HLA-A1 molecules are not accessible at the target cell. Taken together, these data provide proof of principle that pIXscTCR fusions can be used to target HAdV-5 vectors to tumor cells expressing intracellular CT antigens.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Melanoma/terapia , Transducción Genética/métodos , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Proteínas de la Cápside/genética , Línea Celular Tumoral , Citotoxicidad Inmunológica , Citometría de Flujo , Marcación de Gen , Ingeniería Genética , Vectores Genéticos/genética , Antígeno HLA-A1/inmunología , Humanos , Masculino , Melanoma/inmunología , Melanoma/metabolismo , Antígenos Específicos del Melanoma , Proteínas de Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/genética
16.
Cancer Gene Ther ; 15(5): 284-92, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18259212

RESUMEN

Reovirus T3D preferentially kills tumor cells expressing Ras oncogenes and has shown great promise as an anticancer agent in various preclinical tumor models. Here, we investigated whether reovirus can infect and kill tumor cell cultures and tissue fragments isolated from resected human colorectal tumors, and whether this was affected by the presence of endogenous oncogenic KRAS. Tissue fragments and single-cell populations isolated from human colorectal tumor biopsies were infected with reovirus virions or with intermediate subviral particles (ISVPs). Reovirus virions were capable of infecting neither single-cell tumor cell populations nor small fragments of intact viable tumor tissue. However, infection of tumor cells with ISVPs resulted in transient viral protein synthesis, irrespective of the presence of oncogenic KRAS, but this did not lead to the production of infectious virus particles, and tumor cell viability was largely unaffected. ISVPs failed to infect intact tissue fragments. Thermolysin treatment of tumor tissue liberated single cells from the tissue and allowed infection with ISVPs, but this did not result in the production of infectious virus particles. Immunohistochemistry on tissue microarrays showed that junction adhesion molecule 1, the major cellular reovirus receptor, was improperly localized in the cytoplasm of colorectal tumor cells and was expressed at very low levels in liver metastases. This may contribute to the observed resistance of tumor cells to reovirus T3D virions. We conclude that infection of human colorectal tumor cells by reovirus T3D requires processing of virions to ISVPs, but that oncolysis is prevented by a tumor cell response that aborts viral protein synthesis and the generation of infectious viral particles, irrespective of KRAS mutation status.


Asunto(s)
Neoplasias Colorrectales/virología , Orthoreovirus Mamífero 3 , Infecciones por Reoviridae/complicaciones , Biopsia , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias del Colon/cirugía , Neoplasias del Colon/virología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/cirugía , ADN de Neoplasias/genética , ADN de Neoplasias/aislamiento & purificación , ADN Viral/genética , ADN Viral/aislamiento & purificación , Citometría de Flujo , Genes ras , Humanos , Orthoreovirus Mamífero 3/fisiología , Metástasis de la Neoplasia , Infecciones por Reoviridae/genética , Ensayo de Placa Viral , Replicación Viral
17.
Gene Ther ; 15(4): 239-46, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18046427

RESUMEN

The development of efficient and safe vectors for gene delivery paved the way for evolution of gene therapy as a new modality for treatment of various inherited disorders and for cancer. The current vectors, viral and non-viral, have their limitations. Innate and adaptive immune responses to vector particles and components may restrict the efficiency of gene transfer and the persistence of expression of the transgene. Results from preclinical studies in animals and more recently data from clinical studies have demonstrated the potential impact of the cellular and the humoral immune response on the therapeutic efficacy. Not only the vector components, but also the transgene products may induce an immune response that negatively affects the therapeutic efficacy. The induction of a cytotoxic T-cell response to transgene-encoded peptides, as well as the production of antibodies directed against secreted proteins have been reported in preclinical and clinical studies, and these may thwart those applications that require long-term expression. Here we will review some of the options to blunt the acquired immune responses to transgene-encoded polypeptides.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/inmunología , Inmunidad Innata , Transgenes/inmunología , Humanos , Linfocitos T Citotóxicos/inmunología
18.
Cancer Gene Ther ; 14(5): 468-79, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17273181

RESUMEN

In order to use adenovirus (Ad) type 5 (Ad5) for cancer gene therapy, Ad needs to be de-targeted from its native receptors and re-targeted to a tumor antigen. A limiting factor for this has been to find a ligand that (i) binds a relevant target, (ii) is able to fold correctly in the reducing environment of the cytoplasm and (iii) when incorporated at an optimal position on the virion results in a virus with a low physical particle to plaque-forming units ratio to diminish the viral load to be administered to a future patient. Here, we present a solution to these problems by producing a genetically re-targeted Ad with a tandem repeat of the HER2/neu reactive Affibody molecule (ZH) in the HI-loop of a Coxsackie B virus and Ad receptor (CAR) binding ablated fiber genetically modified to contain sequences for flexible linkers between the ZH and the knob sequences. ZH is an Affibody molecule specific for the extracellular domain of human epidermal growth factor receptor 2 (HER2/neu) that is overexpressed in inter alia breast and ovarian carcinomas. The virus presented here exhibits near wild-type growth characteristics, infects cells via HER2/neu instead of CAR and represents an important step toward the development of genetically re-targeted adenoviruses with clinical relevance.


Asunto(s)
Adenoviridae/genética , Antígenos de Neoplasias/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Proteínas Recombinantes de Fusión/genética , Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/terapia , Femenino , Humanos , Ligandos , Neoplasias Ováricas/terapia , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/inmunología , Células Tumorales Cultivadas
19.
Gene Ther ; 14(8): 664-70, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17268536

RESUMEN

Recombinant adenoviruses are frequently used as gene transfer vehicles for therapeutic gene delivery. Strategies to amend their tropism include the incorporation of polypeptides with high affinity for cellular receptors. Single-chain antibodies have a great potential to achieve such cell type specificity. In this study, we evaluated the efficiency of incorporation of a single-chain antibody fused with the adenovirus minor capsid protein IX in the capsid of adenovirus type 5 vectors. To this end, the codons for the single-chain antibody fragments (scFv) 13R4 were fused with those encoding of pIX via a 75-Angstrom spacer sequence. The 13R4 is a hyper-stable single-chain antibody directed against beta-galactosidase, which was selected for its capacity to fold correctly in a reducing environment such as the cytoplasm. A lentiviral vector was used to stably express the pIX.flag.75.13R4.MYC.HIS fusion gene in 911 helper cells. Upon propagation of pIX-gene deleted human adenovirus-5 vectors on these cells, the pIX-fusion protein was efficiently incorporated in the capsid. Here, the 13R4 scFv was functional as was evident from its capacity to bind its ligand beta-galactosidase. These data demonstrate that the minor capsid protein IX can be used as an anchor for incorporation of single-chain antibodies in the capsids of adenovirus vectors.


Asunto(s)
Adenoviridae/genética , Proteínas de la Cápside/genética , Ingeniería Genética , Vectores Genéticos/genética , Fragmentos de Inmunoglobulinas/genética , Adenoviridae/ultraestructura , Western Blotting/métodos , Línea Celular Transformada , Expresión Génica , Terapia Genética , Vectores Genéticos/análisis , Humanos , Inmunohistoquímica , Lentivirus/genética , Microscopía Inmunoelectrónica , Proteínas Recombinantes de Fusión/genética , Transducción Genética/métodos , beta-Galactosidasa/genética
20.
Cancer Gene Ther ; 14(2): 211-9, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17082793

RESUMEN

Clinical trials in malignant glioma have demonstrated excellent safety of recombinant adenovirus type 5 (Ad5) but lack of convincing efficacy. The overall low expression levels of the Coxsackie and Adenovirus receptor and the presence of high anti-Ad5-neutralizing antibody (NAb) titers in the human population are considered detrimental for consistency of clinical results. To identify an adenoviral vector better suited to infect primary glioma cells, we tested a library of fiber-chimeric Ad5-based adenoviral vectors on 12 fresh human glioma cell suspensions. Significantly improved marker gene expression was obtained with several Ad5-chimeric vectors, predominantly vectors carrying fiber molecules derived from B-group viruses (Ad11, Ad16, Ad35 and Ad50). We next tested Ad35 sero prevalence in sera derived from 90 Dutch cancer patients including 30 glioma patients and investigated the transduction efficiency of this vector in glioma cell suspensions. Our results demonstrate that the sero prevalence and the titers of NAb against Ad35 are significantly lower than against Ad5. Also, recombinant Ad35 has significantly increased ability to transfer a gene to primary glioma cells compared to Ad5. We thus conclude that Ad35 represents an interesting candidate vector for gene therapy of malignant glioma.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Terapia Genética , Vectores Genéticos , Glioma/terapia , Secuencia de Bases , Neoplasias Encefálicas/inmunología , Cartilla de ADN , Glioma/inmunología , Humanos , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...