Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Intervalo de año de publicación
1.
Nat Biotechnol ; 2023 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-37932420

RESUMEN

The development of liver-based adeno-associated virus (AAV) gene therapies is facing concerns about limited efficiency and durability of transgene expression. We evaluated nonhuman primates following intravenous dosing of AAV8 and AAVrh10 vectors for over 2 years to better define the mechanism(s) of transduction that affect performance. High transduction of non-immunogenic transgenes was achieved, although expression declined over the first 90 days to reach a lower but stable steady state. More than 10% of hepatocytes contained single nuclear domains of vector DNA that persisted despite the loss of transgene expression. Greater reductions in vector DNA and RNA were observed with immunogenic transgenes. Genomic integration of vector sequences, including complex concatemeric structures, were detected in 1 out of 100 cells at broadly distributed loci that were not in proximity to genes associated with hepatocellular carcinoma. Our studies suggest that AAV-mediated transgene expression in primate hepatocytes occurs in two phases: high but short-lived expression from episomal genomes, followed by much lower but stable expression, likely from integrated vectors.

2.
Hum Gene Ther ; 34(21-22): 1081-1094, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37930949

RESUMEN

Integration of naturally occurring adeno-associated viruses (AAV; wild-type AAV [wtAAV]) and those used in gene therapy (recombinant AAV [rAAV]) into host genomic DNA has been documented for over two decades. Results from mouse and dog studies have raised concerns of insertional mutagenesis and clonal expansion following AAV exposure, particularly in the context of gene therapy. This study aimed to characterize the genomic location, abundance, and expansion of wtAAV and rAAV integrations in macaque and human genomes. Using an unbiased, next-generation sequencing-based approach, we identified the genome-wide integration loci in tissue samples (primarily liver) in 168 nonhuman primates (NHPs) and 85 humans naïve to rAAV exposure and 86 NHPs treated with rAAV in preclinical studies. Our results suggest that rAAV and wtAAV integrations exhibit similar, broad distribution patterns across species, with a higher frequency in genomic regions highly vulnerable to DNA damage or close to highly transcribed genes. rAAV exhibited a higher abundance of unique integration loci, whereas wtAAV integration loci were associated with greater clonal expansion. This expansive and detailed characterization of AAV integration in NHPs and humans provides key translational insights, with important implications for the safety of rAAV as a gene therapy vector.


Asunto(s)
Dependovirus , Macaca , Animales , Humanos , Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Hígado , Macaca/genética , Integración Viral/genética
3.
Hum Gene Ther ; 33(21-22): 1174-1186, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36375122

RESUMEN

Transthyretin amyloidosis (ATTR) is a progressive and fatal disease caused by transthyretin (TTR) amyloid fibril accumulation in tissues, which disrupts organ function. As the TTR protein is primarily synthesized by the liver, liver transplantation can cure familial ATTR but is not an option for the predominant age-related wild-type ATTR. Approved treatment approaches include TTR stabilizers and an RNA-interference therapeutic, but these require regular re-administration. Gene editing could represent an effective one-time treatment. We evaluated adeno-associated virus (AAV) vector-delivered, gene-editing meganucleases to reduce TTR levels. We used engineered meganucleases targeting two different sites within the TTR gene. AAV vectors expressing TTR meganuclease transgenes were first tested in immunodeficient mice expressing the human TTR sequence delivered using an AAV vector and then against the endogenous TTR gene in rhesus macaques. Following a dose of 3 × 1013 genome copies per kilogram, we detected on-target editing efficiency of up to 45% insertions and deletions (indels) in the TTR genomic DNA locus and >80% indels in TTR RNA, with a concomitant decrease in serum TTR levels of >95% in macaques. The significant reduction in serum TTR levels following TTR gene editing indicates that this approach could be an effective treatment for ATTR.


Asunto(s)
Neuropatías Amiloides Familiares , Dependovirus , Humanos , Ratones , Animales , Dependovirus/genética , Dependovirus/metabolismo , Macaca mulatta/genética , Macaca mulatta/metabolismo , Neuropatías Amiloides Familiares/terapia , Neuropatías Amiloides Familiares/tratamiento farmacológico , Prealbúmina/genética , Prealbúmina/metabolismo , Prealbúmina/uso terapéutico , ARN/uso terapéutico
4.
Mol Ther ; 29(6): 2019-2029, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-33609733

RESUMEN

Gene disruption via programmable, sequence-specific nucleases represents a promising gene therapy strategy in which the reduction of specific protein levels provides a therapeutic benefit. Proprotein convertase subtilisin/kexin type 9 (PCSK9), an antagonist of the low-density lipoprotein (LDL) receptor, is a suitable target for nuclease-mediated gene disruption as an approach to treat hypercholesterolemia. We sought to determine the long-term durability and safety of PCSK9 knockdown in non-human primate (NHP) liver by adeno-associated virus (AAV)-delivered meganuclease following our initial report on the feasibility of this strategy. Six previously treated NHPs and additional NHPs administered AAV-meganuclease in combination with corticosteroid treatment or an alternative AAV serotype were monitored for a period of up to 3 years. The treated NHPs exhibited a sustained reduction in circulating PCSK9 and LDL cholesterol (LDL-c) through the course of the study concomitant with stable gene editing of the PCSK9 locus. Low-frequency off-target editing remained stable, and no obvious adverse changes in histopathology of the liver were detected. We demonstrate similar on-target nuclease activity in primary human hepatocytes using a chimeric liver-humanized mouse model. These studies demonstrate that targeted in vivo gene disruption exerts a lasting therapeutic effect and provide pivotal data for safety considerations, which support clinical translation.


Asunto(s)
Edición Génica , Lipoproteínas LDL/metabolismo , Proproteína Convertasa 9/genética , Animales , Sistemas CRISPR-Cas , Dependovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Lipoproteínas LDL/genética , Hígado/metabolismo , Ratones , Ratones Noqueados , Primates , Proproteína Convertasa 9/metabolismo
5.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33411694

RESUMEN

Gene editing holds the potential to correct mutations and cure devastating genetic disorders. The technology has not yet proven efficacious for therapeutic use in CNS diseases with ubiquitous neuronal defects. Angelman syndrome (AS), a severe neurodevelopmental disorder, is caused by a lack of maternal expression of the UBE3A gene. Because of genomic imprinting, only neurons are affected. One therapeutic approach focuses on the intact paternal UBE3A copy in patients with AS that is silenced by an antisense transcript (UBE3A-ATS). We show here that gene editing of Ube3a-ATS in the mouse brain resulted in the formation of base pair insertions/deletions (indels) in neurons and the subsequent unsilencing of the paternal Ube3a allele in neurons, which partially corrected the behavioral phenotype of a murine AS model. This study provides compelling evidence to further investigate editing of the homologous region of the human UBE3A-ATS because this may provide a lasting therapeutic effect for patients with AS.


Asunto(s)
Síndrome de Angelman/metabolismo , Síndrome de Angelman/terapia , Encéfalo/metabolismo , Sistemas CRISPR-Cas , Edición Génica , ARN sin Sentido/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Síndrome de Angelman/genética , Animales , Humanos , Ratones , ARN sin Sentido/genética , Ubiquitina-Proteína Ligasas/genética
6.
Mol Ther ; 29(3): 1047-1056, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33359790

RESUMEN

Our group previously used adeno-associated viral vectors (AAVs) to express an engineered meganuclease specific for a sequence in the PCSK9 gene (M2PCSK9), a clinical target for treating coronary heart disease. Upon testing this nuclease in non-human primates, we observed specific editing characterized by several insertions and deletions (indels) in the target sequence as well as indels in similar genomic sequences. We hypothesized that high nuclease expression increases off-target editing. Here, we reduced nuclease expression using two strategies. The first was a self-targeting strategy that involved inserting the M2PCSK9 target sequence into the AAV genome that expresses the nuclease and/or fusing the nuclease to a specific peptide to promote its degradation. The second strategy used a shortened version of the parental promoter to reduce nuclease expression. Mice administered with these second-generation AAV vectors showed reduced PCSK9 expression due to the nuclease on-target activity and reduced off-target activity. All vectors induced a stable reduction of PCSK9 in primates treated with self-targeting and short-promoter AAVs. Compared to the meganuclease-expressing parental AAV vector, we observed a significant reduction in off-target activity. In conclusion, we increased the in vivo nuclease specificity using a clinically relevant strategy that can be applied to other genome-editing nucleases.


Asunto(s)
Dependovirus/genética , Endonucleasas/genética , Edición Génica , Vectores Genéticos/genética , Lipoproteínas LDL/sangre , Inhibidores de PCSK9 , Regiones Promotoras Genéticas , Animales , Humanos , Ratones , Primates , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo
7.
BMC Genomics ; 21(1): 810, 2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33218308

RESUMEN

An amendment to this paper has been published and can be accessed via the original article.

8.
Mol Ther Oncolytics ; 18: 546-555, 2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32839735

RESUMEN

Recombinant vesicular stomatitis virus (VSV)-fusion and hemagglutinin (FH) was developed by substituting the promiscuous VSV-G glycoprotein (G) gene in the backbone of VSV with genes encoding for the measles virus envelope proteins F and H. Hybrid VSV-FH exhibited a multifaceted mechanism of cancer-cell killing and improved neurotolerability over parental VSV in preclinical studies. In this study, we evaluated VSV-FH in vitro and in vivo in models of hepatobiliary and pancreatic cancers. Our results indicate that high intrahepatic doses of VSV-FH did not result in any significant toxicity and were well tolerated by transgenic mice expressing the measles virus receptor CD46. Furthermore, a single intratumoral treatment with VSV-FH yielded improved survival and complete tumor regressions in a proportion of mice in the Hep3B hepatocellular carcinoma model but not in mice xenografted with BxPC-3 pancreatic cancer cells. Our preliminary findings indicate that VSV-FH can induce potent oncolysis in hepatocellular and pancreatic cancer cell lines with concordant results in vivo in hepatocellular cancer and discordant in pancreatic cancer without the VSV-mediated toxic effects previously observed in laboratory animals. Further study of VSV-FH as an oncolytic virotherapy is warranted in hepatocellular carcinoma and pancreatic cancer to understand broader applicability and mechanisms of sensitivity and resistance.

9.
BMC Genomics ; 21(1): 239, 2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32183699

RESUMEN

BACKGROUND: Identifying nuclease-induced double-stranded breaks in DNA on a genome-wide scale is critical for assessing the safety and efficacy of genome editing therapies. We previously demonstrated that after administering adeno-associated viral (AAV) vector-mediated genome-editing strategies in vivo, vector sequences integrated into the host organism's genomic DNA at double-stranded breaks. Thus, identifying the genomic location of inserted AAV sequences would enable us to identify DSB events, mainly derived from the nuclease on- and off-target activity. RESULTS: Here, we developed a next-generation sequencing assay that detects insertions of specific AAV vector sequences called inverted terminal repeats (ITRs). This assay, ITR-Seq, enables us to identify off-target nuclease activity in vivo. Using ITR-Seq, we analyzed liver DNA samples of rhesus macaques treated with AAV vectors expressing a meganuclease. We found dose-dependent off-target activity and reductions in off-target events induced by further meganuclease development. In mice, we identified the genomic locations of ITR integration after treatment with Cas9 nucleases and their corresponding single-guide RNAs. CONCLUSIONS: In sum, ITR-Seq is a powerful method for identifying off-target sequences induced by AAV vector-delivered genome-editing nucleases. ITR-Seq will help us understand the specificity and efficacy of different genome-editing nucleases in animal models and clinical studies. This information can help enhance the safety profile of gene-editing therapies.


Asunto(s)
Roturas del ADN de Doble Cadena , Edición Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Animales , Proteína 9 Asociada a CRISPR , Dependovirus , Vectores Genéticos/genética , Macaca mulatta , Ratones , ARN Guía de Kinetoplastida , Secuencias Repetidas Terminales/genética
10.
Sci Adv ; 6(7): eaax5701, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32095520

RESUMEN

Ornithine transcarbamylase (OTC) deficiency is an X-linked urea cycle disorder associated with high mortality. Although a promising treatment for late-onset OTC deficiency, adeno-associated virus (AAV) neonatal gene therapy would only provide short-term therapeutic effects as the non-integrated genome gets lost during hepatocyte proliferation. CRISPR-Cas9-mediated homology-directed repair can correct a G-to-A mutation in 10% of OTC alleles in the livers of newborn OTC spfash mice. However, an editing vector able to correct one mutation would not be applicable for patients carrying different OTC mutations, plus expression would not be fast enough to treat a hyperammonemia crisis. Here, we describe a dual-AAV vector system that accomplishes rapid short-term expression from a non-integrated minigene and long-term expression from the site-specific integration of this minigene without any selective growth advantage for OTC-positive cells in newborns. This CRISPR-Cas9 gene-targeting approach may be applicable to all patients with OTC deficiency, irrespective of mutation and/or clinical state.


Asunto(s)
Sistemas CRISPR-Cas/genética , Marcación de Gen , Terapia Genética , Mutación/genética , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/terapia , Animales , Reparación del ADN/genética , Dependovirus/genética , Proteínas en la Dieta , Modelos Animales de Enfermedad , Sitios Genéticos , Vectores Genéticos/metabolismo , Mutación INDEL/genética , Hígado/enzimología , Hígado/patología , Masculino , Ratones , Ornitina Carbamoiltransferasa/genética , Ornitina Carbamoiltransferasa/metabolismo , Factores de Tiempo
11.
Blood ; 133(26): 2745-2752, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-30975639

RESUMEN

Many genetic diseases, including hemophilia, require long-term therapeutic effects. Despite the initial success of liver-directed adeno-associated virus (AAV) gene therapy for hemophilia in clinical trials, long-term sustained therapeutic effects have yet to be seen. One explanation for the gradual decline of efficacy over time is that the nonintegrating AAV vector genome could be lost during cell division during hepatocyte turnover, albeit at a slow pace in adults. Readministering the same vector is challenging as a result of the AAV-neutralizing antibodies elicited by the initial treatment. Here, we investigated the use of clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated homology-directed gene targeting for sustained treatment of hemophilia B. We developed a donor vector containing a promoterless partial human factor IX (FIX) complementary DNA carrying the hyperactive FIX Padua mutation. A single injection of dual AAV vectors in newborn and adult FIX-knockout (FIX-KO) mice led to stable expression of FIX at or above the normal levels for 8 months. Eight weeks after the vector treatment, we subjected a subgroup of newborn and adult treated FIX-KO mice to a two-thirds partial hepatectomy; all of these animals survived the procedure without any complications or interventions. FIX levels persisted at similar levels for 24 weeks after partial hepatectomy, indicating stable genomic targeting. Our results lend support for the use of a CRISPR/Cas9 approach to achieve lifelong expression of therapeutic proteins.


Asunto(s)
Sistemas CRISPR-Cas , Factor IX/genética , Marcación de Gen/métodos , Hemofilia B/genética , Hemostasis/genética , Animales , Animales Recién Nacidos , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos , Humanos , Ratones , Ratones Noqueados
12.
Nat Biotechnol ; 36(8): 717-725, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29985478

RESUMEN

Clinical translation of in vivo genome editing to treat human genetic diseases requires thorough preclinical studies in relevant animal models to assess safety and efficacy. A promising approach to treat hypercholesterolemia is inactivating the secreted protein PCSK9, an antagonist of the LDL receptor. Here we show that single infusions in six non-human primates of adeno-associated virus vector expressing an engineered meganuclease targeting PCSK9 results in dose-dependent disruption of PCSK9 in liver, as well as a stable reduction in circulating PCSK9 and serum cholesterol. Animals experienced transient, asymptomatic elevations of serum transaminases owing to the formation of T cells against the transgene product. Vector DNA and meganuclease expression declined rapidly, leaving stable populations of genome-edited hepatocytes. A second-generation PCSK9-specific meganuclease showed reduced off-target cleavage. These studies demonstrate efficient, physiologically relevant in vivo editing in non-human primates, and highlight safety considerations for clinical translation.


Asunto(s)
Colesterol/sangre , Desoxirribonucleasas/metabolismo , Hígado/enzimología , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo , Animales , Dependovirus/genética , Edición Génica , Vectores Genéticos , Células HEK293 , Hepatocitos/metabolismo , Humanos , Hipercolesterolemia/enzimología , Hipercolesterolemia/terapia , Células Madre Pluripotentes Inducidas/metabolismo , Macaca mulatta , Masculino , Ratones , Ratones Noqueados , Receptores de LDL/antagonistas & inhibidores
13.
Front Oncol ; 7: 127, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28695108

RESUMEN

Therapy resistance and tumor recurrence are often linked to a small refractory and highly tumorigenic subpopulation of neoplastic cells, known as cancer stem cells (CSCs). A putative marker of CSCs is CD133 (prominin-1). We have previously described a CD133-targeted oncolytic measles virus (MV-CD133) as a promising approach to specifically eliminate CD133-positive tumor cells. Selectivity was introduced at the level of cell entry by an engineered MV hemagglutinin (H). The H protein was blinded for its native receptors and displayed a CD133-specific single-chain antibody fragment (scFv) as targeting domain. Interestingly, MV-CD133 was more active in killing CD133-positive tumors than the unmodified MV-NSe despite being highly selective for its target cells. To further enhance the antitumoral activity of MV-CD133, we here pursued arming technologies, receptor extension, and chimeras between MV-CD133 and vesicular stomatitis virus (VSV). All newly generated viruses including VSV-CD133 were highly selective in eliminating CD133-positive cells. MV-CD46/CD133 killed in addition CD133-negative cells being positive for the MV receptors. In an orthotopic glioma model, MV-CD46/CD133 and MVSCD-CD133, which encodes the super cytosine deaminase, were most effective. Notably, VSV-CD133 caused fatal neurotoxicity in this tumor model. Use of CD133 as receptor could be excluded as being causative. In a subcutaneous tumor model of hepatocellular cancer, VSV-CD133 revealed the most potent oncolytic activity and also significantly prolonged survival of the mice when injected intravenously. Compared to MV-CD133, VSV-CD133 infected a more than 104-fold larger area of the tumor within the same time period. Our data not only suggest new concepts and approaches toward enhancing the oncolytic activity of CD133-targeted oncolytic viruses but also raise awareness about careful toxicity testing of novel virus types.

14.
Mol Ther Oncolytics ; 2: 15012, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27119107

RESUMEN

The hybrid oncolytic vesicular stomatitis virus (VSV-FH) deleted for its G glycoprotein and displaying the measles virus (MV) envelope glycoproteins (hemagglutinin H and fusion F) is fusogenic, infects cells via any of the three MV receptors and has potent oncolytic activity against subcutaneous and disseminated myeloma tumors. To tailor VSV-FH as an oncolytic virus for ovarian cancer, we ablated its natural tropism and retargeted the virus by display of a single-chain antibody (scFv) with specificity to the HER-2/neu receptor. A panel of six VSVFH-αHER2 viruses displaying anti-HER2 scFv that bind to the same HER2 epitope but with different K d (10(-6) to 10(-11) M, VSVFH-αHER2#6 to #11, respectively) were rescued and characterized. A K d of at least 10(-8) M is required for infection of HER-2 positive SKOV3ip.1 cells. The higher affinity viruses (>10(-8) M) were able to infect and fuse SKOV3ip.1 cells more efficiently, inducing more extensive cytopathic effects. We next compared the antitumor potency of the viruses against SKOV3ip.1 tumor xenografts. In contrast to the saline-treated animals, one intratumoral injection of VSVFH-αHER2#9, #10, or #11 resulted in efficient tumor control. There was no significant difference between viruses with an affinity higher than 10(-9) M in terms of oncolytic potency. VSVFH-αHER2 virus may be a promising agent for the treatment of HER-2 positive malignancies.

15.
J Virol ; 88(15): 8332-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24829351

RESUMEN

UNLABELLED: VSV-FH is a hybrid vesicular stomatitis virus (VSV) with a deletion of its G glycoprotein and encoding the measles virus (MV) fusion (F) and hemagglutinin (H) envelope glycoproteins. VSV-FH infects cells expressing MV receptors and is fusogenic and effective against myeloma xenografts in mice. We evaluated the fusogenic activities of MV and VSV-FH in relationship to the density of receptor on the target cell surface and the kinetics of F and H expression in infected cells. Using a panel of cells expressing increasing numbers of the MV receptor CD46, we evaluated syncytium size in MV- or VSV-FH-infected cells. VSV-FH is not fusogenic at low CD46 density but requires less CD46 for syncytium formation than MV. The size of each syncytium is larger in VSV-FH-infected cells at a specific CD46 density. While syncytium size reached a plateau and did not increase further in MV-infected CHO cells expressing ≥4,620 CD46 copies/cell, there was a corresponding increase in syncytium size with increases in CD46 levels in VSV-FH-infected CD46-expressing CHO (CHO-CD46) cells. Further analysis in VSV-FH-infected cell lines shows earlier and higher expression of F and H mRNAs and protein. However, VSV-FH cytotoxic activity was reduced by pretreatment of the cells with type I interferon. In contrast, the cytopathic effects are not affected in MV-infected cells. In summary, VSV-FH has significant advantages over MV as an oncolytic virus due to its higher viral yield, faster replication kinetics, and larger fusogenic capabilities but should be used in cancer types with defective interferon signaling pathways. IMPORTANCE: We studied the cytotoxic activity of a vesicular stomatitis/measles hybrid virus (VSV-FH), which is superior to that of measles virus (MV), in different cancer cell lines. We determined that viral RNA and protein were produced faster and in higher quantities in VSV-FH-infected cells. This resulted in the formation of larger syncytia, higher production of infectious particles, and a more potent cytopathic effect in permissive cells. Importantly, VSV-FH, similar to MV, can discriminate between low- and high-expressing CD46 cells, a phenotype important for cancer therapy as the virus will be able to preferentially infect cancer cells that overexpress CD46 over low-CD46-expressing normal cells.


Asunto(s)
Glicoproteínas/metabolismo , Hemaglutininas/metabolismo , Virus del Sarampión/fisiología , Recombinación Genética , Vesiculovirus/fisiología , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Animales , Células CHO , Cricetinae , Cricetulus , Células Gigantes/virología , Glicoproteínas/genética , Hemaglutininas/genética , Virus del Sarampión/genética , Virus del Sarampión/crecimiento & desarrollo , Proteína Cofactora de Membrana/metabolismo , Receptores Virales/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vesiculovirus/genética , Vesiculovirus/crecimiento & desarrollo , Proteínas Virales de Fusión/genética
16.
J Virol ; 88(4): 2195-204, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24335299

RESUMEN

Measles virus (MV) immunosuppression is due to infection of SLAM-positive immune cells, whereas respiratory shedding and virus transmission are due to infection of nectin4-positive airway epithelial cells. The vaccine lineage MV strain Edmonston (MV-Edm) acquired an additional tropism for CD46 which is the basis of its oncolytic specificity. VSVFH is a vesicular stomatitis virus (VSV) encoding the MV-Edm F and H entry proteins in place of G. The virus spreads faster than MV-Edm and is highly fusogenic and a potent oncolytic. To determine whether ablating nectin4 tropism from VSVFH might prevent shedding, increasing its safety profile as an oncolytic, or might have any effect on CD46 binding, we generated VSVFH viruses with H mutations that disrupt attachment to SLAM and/or nectin4. Disruption of nectin4 binding reduced release of VSVFH from the basolateral side of differentiated airway epithelia composed of Calu-3 cells. However, because nectin4 and CD46 have substantially overlapping receptor binding surfaces on H, disruption of nectin4 binding compromised CD46 binding and greatly diminished the oncolytic potency of these viruses on human cancer cells. Thus, our results support continued preclinical development of VSVFH without ablation of nectin4 binding.


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular/deficiencia , Virus del Sarampión/inmunología , Receptores de Superficie Celular/metabolismo , Vesiculovirus/inmunología , Esparcimiento de Virus/genética , Animales , Células CHO , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Cricetinae , Cricetulus , Cartilla de ADN/genética , Humanos , Immunoblotting , Proteína Cofactora de Membrana/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Células Vero , Esparcimiento de Virus/fisiología
17.
Mol Ther ; 21(10): 1930-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23842448

RESUMEN

Oncolytic viruses are structurally and biologically diverse, spreading through tumors and killing them by various mechanisms and with different kinetics. Here, we created a hybrid vesicular stomatitis/measles virus (VSV/MV) that harnesses the safety of oncolytic MV, the speed of VSV, and the tumor killing mechanisms of both viruses. Oncolytic MV targets CD46 and kills by forcing infected cells to fuse with uninfected neighbors, but propagates slowly. VSV spreads rapidly, directly lysing tumor cells, but is neurotoxic and loses oncolytic potency when neuroattenuated by conventional approaches. The hybrid VSV/MV lacks neurotoxicity, replicates rapidly with VSV kinetics, and selectively targets CD46 on tumor cells. Its in vivo performance in a myeloma xenograft model was substantially superior to either MV or widely used recombinant oncolytic VSV-M51.


Asunto(s)
Ingeniería Genética , Virus del Sarampión/fisiología , Proteína Cofactora de Membrana/efectos de los fármacos , Mieloma Múltiple/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Virus de la Estomatitis Vesicular Indiana/fisiología , Animales , Encéfalo/patología , Encéfalo/virología , Células CHO , Línea Celular Tumoral , Chlorocebus aethiops , Cricetulus , Modelos Animales de Enfermedad , Humanos , Virus del Sarampión/genética , Ratones , Ratones SCID , Ratones Transgénicos , Mieloma Múltiple/patología , Neuronas/patología , Neuronas/virología , Virus Oncolíticos/genética , Células Vero , Virus de la Estomatitis Vesicular Indiana/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Hum Gene Ther ; 23(5): 484-91, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22171635

RESUMEN

Oncolytic vesicular stomatitis virus (VSV) has potent antitumor activity, but infects a broad range of cell types. Here, we used the measles virus (MV) hemagglutinin (H) and fusion (F) envelope glycoproteins to redirect VSV entry and infection specifically to tumor-associated receptors. Replication-defective VSV, deleted of its glycoprotein gene (VSVΔG), was pseudotyped with MV-F and MV-H displaying single-chain antibodies (scFv) specific for epidermal growth factor receptor (EGFR), folate receptor (FR), or prostate membrane-specific antigen (PSMA). Viral titers were ∼10(5) PFU/ml, but could be concentrated to 10(7) PFU/ml. Immunoblotting confirmed incorporation of the MV-H-scFv and MV-F into functional VSV virions. Although VSV-G was able to infect all tumor cell lines tested, the retargeted VSV infected only cells that expressed the targeted receptor. In vivo specificities of the EGFR-, FR-, and PSMA-retargeted VSV were assessed by intratumoral injection into human tumor xenografts. Analysis of green fluorescent protein reporter gene expression indicated that VSV infection was restricted to receptor-positive tumors. In summary, we have demonstrated for the first time that VSV can be efficiently retargeted to different cellular receptors using the measles display technology, yielding retargeted VSV vectors that are highly specific for tumors that express the relevant receptor.


Asunto(s)
Hemaglutininas Virales/genética , Virus del Sarampión/genética , Neoplasias/terapia , Viroterapia Oncolítica , Vesiculovirus/genética , Proteínas Virales de Fusión/genética , Animales , Antígenos de Superficie/inmunología , Antígenos de Superficie/metabolismo , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Femenino , Receptor 1 de Folato/inmunología , Receptor 1 de Folato/metabolismo , Vectores Genéticos/administración & dosificación , Glutamato Carboxipeptidasa II/inmunología , Glutamato Carboxipeptidasa II/metabolismo , Proteínas Fluorescentes Verdes/análisis , Células HEK293 , Humanos , Inyecciones Intralesiones , Masculino , Ratones , Ratones SCID , Proteínas Recombinantes de Fusión , Anticuerpos de Cadena Única/genética , Vesiculovirus/metabolismo
19.
MedUNAB ; 15(2): 77-77, 2012.
Artículo en Español | LILACS | ID: biblio-998619

RESUMEN

Para conmemorar los quince años de MedUNAB el comité estudiantil quiso escribir sobre el significado que tiene para cada uno de sus miembros pertenecer a ella. Es así como en este editorial recogemos las distintas experiencias que se han tenido, algunas de años, otras de meses.


To commemorate the fifteenth anniversary of MedUNAB, the student committee wanted to write about the meaning of belonging to the journals. That is how in this editorial we collect the different experiences that have been had, some of years, others of month


Asunto(s)
Ciencias de la Salud , Editorial , Educación , Publicación Periódica , Humanismo
20.
J Virol ; 83(17): 8819-31, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19553303

RESUMEN

Rotaviruses have a genome composed of 11 segments of double-stranded RNA (dsRNA) surrounded by three protein layers. The virus contains an RNA-dependent RNA polymerase that synthesizes RNA transcripts corresponding to all segments of the viral genome. These transcripts direct the synthesis of the viral proteins and also serve as templates for the synthesis of the complementary strand to form the dsRNA genome. In this work, we analyzed the kinetics of transcription and replication of the viral genome throughout the replication cycle of the virus using quantitative reverse transcription-PCR. The role of the proteins that form double-layered particles ([DLPs] VP1, VP2, VP3, and VP6) in replication and transcription of the viral genome was analyzed by silencing their expression in rotavirus-infected cells. All of them were shown to be essential for the replication of the dsRNA genome since in their absence there was little synthesis of viral mRNA and dsRNA. The characterization of the kinetics of RNA transcription and replication of the viral genome under conditions where these proteins were silenced provided direct evidence for a second round of transcription during the replication of the virus. Interestingly, despite the decrease in mRNA accumulation when any of the four proteins was silenced, the synthesis of viral proteins decreased when VP2 and VP6 were knocked down, whereas the absence of VP1 and VP3 did not have a severe impact on viral protein synthesis. Characterization of viral particle assembly in the absence of VP1 and VP3 showed that while the formation of triple-layered particles and DLPs was decreased, the amount of assembled lower-density particles, often referred to as empty particles, was not different from the amount in control-infected cells, suggesting that viral particles can assemble in the absence of either VP1 or VP3.


Asunto(s)
ARN Viral/biosíntesis , Rotavirus/fisiología , Transcripción Genética , Replicación Viral , Técnicas de Silenciamiento del Gen/métodos , Cinética , Interferencia de ARN , ARN Bicatenario/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Proteínas Estructurales Virales/antagonistas & inhibidores , Proteínas Estructurales Virales/metabolismo , Ensamble de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...