Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Lancet ; 383(9923): 1138-46, 2014 Mar 29.
Article in English | MEDLINE | ID: mdl-24412048

ABSTRACT

BACKGROUND: Parkinson's disease is typically treated with oral dopamine replacement therapies; however, long-term treatment leads to motor complications and, occasionally, impulse control disorders caused by intermittent stimulation of dopamine receptors and off-target effects, respectively. We aimed to assess the safety, tolerability, and efficacy of bilateral, intrastriatal delivery of ProSavin, a lentiviral vector-based gene therapy aimed at restoring local and continuous dopamine production in patients with advanced Parkinson's disease. METHODS: We undertook a phase 1/2 open-label trial with 12-month follow-up at two study sites (France and UK) to assess the safety and efficacy of ProSavin after bilateral injection into the putamen of patients with Parkinson's disease. All patients were then enrolled in a separate open-label follow-up study of long-term safety. Three doses were assessed in separate cohorts: low dose (1·9×10(7) transducing units [TU]); mid dose (4·0×10(7) TU); and high dose (1×10(8) TU). Inclusion criteria were age 48-65 years, disease duration 5 years or longer, motor fluctuations, and 50% or higher motor response to oral dopaminergic therapy. The primary endpoints of the phase 1/2 study were the number and severity of adverse events associated with ProSavin and motor responses as assessed with Unified Parkinson's Disease Rating Scale (UPDRS) part III (off medication) scores, at 6 months after vector administration. Both trials are registered at ClinicalTrials.gov, NCT00627588 and NCT01856439. FINDINGS: 15 patients received ProSavin and were followed up (three at low dose, six mid dose, six high dose). During the first 12 months of follow-up, 54 drug-related adverse events were reported (51 mild, three moderate). Most common were increased on-medication dyskinesias (20 events, 11 patients) and on-off phenomena (12 events, nine patients). No serious adverse events related to the study drug or surgical procedure were reported. A significant improvement in mean UPDRS part III motor scores off medication was recorded in all patients at 6 months (mean score 38 [SD 9] vs 26 [8], n=15, p=0·0001) and 12 months (38 vs 27 [8]; n=15, p=0·0001) compared with baseline. INTERPRETATION: ProSavin was safe and well tolerated in patients with advanced Parkinson's disease. Improvement in motor behaviour was observed in all patients. FUNDING: Oxford BioMedica.


Subject(s)
Antiparkinson Agents/administration & dosage , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Infectious Anemia Virus, Equine/genetics , Parkinson Disease/therapy , Transfection/methods , Aged , Antiparkinson Agents/adverse effects , Dopa Decarboxylase/genetics , Dopamine/biosynthesis , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/virology , Follow-Up Studies , GTP Cyclohydrolase/administration & dosage , GTP Cyclohydrolase/adverse effects , GTP Cyclohydrolase/genetics , Genetic Therapy/adverse effects , Genetic Vectors/adverse effects , Humans , Injections, Intralesional , Male , Middle Aged , Putamen , Transgenes/genetics , Tyrosine 3-Monooxygenase/administration & dosage , Tyrosine 3-Monooxygenase/adverse effects , Tyrosine 3-Monooxygenase/genetics
2.
Sci Transl Med ; 1(2): 2ra4, 2009 Oct 14.
Article in English | MEDLINE | ID: mdl-20368163

ABSTRACT

In Parkinson's disease, degeneration of specific neurons in the midbrain can cause severe motor deficits, including tremors and the inability to initiate movement. The standard treatment is administration of pharmacological agents that transiently increase concentrations of brain dopamine and thereby discontinuously modulate neuronal activity in the striatum, the primary target of dopaminergic neurons. The resulting intermittent dopamine alleviates parkinsonian symptoms but is also thought to cause abnormal involuntary movements, called dyskinesias. To investigate gene therapy for Parkinson's disease, we simulated the disease in macaque monkeys by treating them with the complex I mitochondrial inhibitor 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, which induces selective degeneration of dopamine-producing neurons. In this model, we demonstrated that injection of a tricistronic lentiviral vector encoding the critical genes for dopamine synthesis (tyrosine hydroxylase, aromatic L-amino acid decarboxylase, and guanosine 5'-triphosphate cyclohydrolase 1) into the striatum safely restored extracellular concentrations of dopamine and corrected the motor deficits for 12 months without associated dyskinesias. Gene therapy-mediated dopamine replacement may be able to correct Parkinsonism in patients without the complications of dyskinesias.


Subject(s)
Disease Models, Animal , Dopamine/genetics , Genetic Therapy , Parkinson Disease/therapy , Animals , Dopamine/deficiency , Dyskinesias/complications , Genetic Vectors , Lentivirus/genetics , Macaca mulatta , Motor Activity/genetics , Parkinson Disease/complications
3.
Int Immunol ; 20(8): 1057-66, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18567615

ABSTRACT

UNLABELLED: The cancer vaccine TroVax, modified vaccinia Ankara encoding the tumor-associated antigen 5T4, has been tested in phase I and II studies in colorectal cancer patients. Monitoring of 5T4-specific immune responses in patients receiving TroVax is critical since it could inform future refinements to the therapeutic or provide a surrogate marker of clinical efficacy. Tumor-specific cytotoxic T lymphocyte (CTL) are considered to be a key component of an effective anti-cancer immune response. Though numerous techniques have been employed to identify CTL epitopes, many are labor intensive, of variable reliability or biased toward common alleles such as human leukocyte antigen (HLA)-A2. A new high-throughput technique, iTopia, enables peptides to be evaluated on the basis of their physical binding properties for HLA alleles. This technique has been utilized to rapidly screen a panel of overlapping peptides, spanning the length of 5T4. Initially, peptides which bound to four class I alleles (A*0101, A*0201, A*0301 and B*0702) were identified and their physical binding characteristics assessed further by analysis of relative affinity and complex stability. 46 putative CTL epitopes have been identified which bind to at least one of the four HLA alleles. Using PBMCs from patients vaccinated with TroVax, we have used the interferon gamma (IFN gamma) ELISpot assay to validate one predicted A1 and two A2 epitopes. CONCLUSION: iTopia represents a rapid and high-throughput technique to identify CTL epitopes.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Colorectal Neoplasms/immunology , Epitopes, T-Lymphocyte/immunology , Membrane Glycoproteins/immunology , Neoplasms, Glandular and Epithelial/immunology , T-Lymphocytes, Cytotoxic/metabolism , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Epitope Mapping , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/metabolism , HLA-A Antigens/chemistry , HLA-A Antigens/metabolism , HLA-B Antigens/chemistry , HLA-B Antigens/metabolism , Humans , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Glandular and Epithelial/therapy , Peptides/chemistry , Peptides/immunology , Peptides/therapeutic use , Protein Binding , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Vaccines, DNA
4.
Cancer Immunol Immunother ; 57(7): 977-86, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18060404

ABSTRACT

Modified vaccinia Ankara (MVA) encoding the tumor antigen 5T4 (TroVax) has been evaluated in an open label phase II study in metastatic colorectal cancer patients. The primary objective was to assess the safety and immunogenicity of TroVax injected before, during and after treatment with 5-fluorouracil, leukovorin and irinotecan. TroVax was administered to 19 patients with metastatic colorectal cancer. Twelve patients had blood samples taken following each of the six injections and were considered to be evaluable for assessment of immunological responses. Both antibody and cellular responses specific for the tumor antigen 5T4 and the viral vector MVA were monitored throughout the study. Administration of TroVax alongside chemotherapy was safe and well tolerated with no SAEs attributed to the vaccine and no enhancement of chemo-related toxicity. Of the 12 patients who were evaluable for assessment of immune responses, ten mounted 5T4-specific antibody responses with titers ranging from 10 to > 5,000. IFNgamma ELISPOT responses specific for 5T4 were detected in 11 patients with frequencies exceeding one in 1,000 PBMCs in five patients. Eight patients presented with elevated circulating CEA concentrations, six of whom showed decreases in excess of 50% during chemotherapy and four had CEA levels which remained stable for > 1 month following completion of chemotherapy. Of the 19 intention to treat (ITT) patients, one had a CR, six had PRs and five had SD. Potent 5T4-specific cellular and/or humoral immune responses were induced in all 12 evaluable patients and were detectable in most patients during the period in which chemotherapy was administered. These data demonstrate that TroVax can be layered on top of chemotherapy regimens without any evidence of enhanced toxicity or reduced immunological or therapeutic efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer Vaccines/therapeutic use , Colorectal Neoplasms/drug therapy , Aged , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Colorectal Neoplasms/immunology , Combined Modality Therapy , Female , Fluorouracil/administration & dosage , Humans , Immunotherapy , Irinotecan , Leucovorin/administration & dosage , Male , Middle Aged , Vaccines, DNA
5.
Clin Cancer Res ; 13(15 Pt 1): 4487-94, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671134

ABSTRACT

PURPOSE: The attenuated strain of vaccinia virus, modified vaccinia Ankara (MVA) encoding the tumor antigen 5T4 (TroVax), has been evaluated in an open-label phase II study in metastatic colorectal cancer patients. The primary objective was to assess the safety and immunogenicity of TroVax injected before, during, and after treatment with cycles of 5-fluorouracil, folinic acid, and oxaliplatin. EXPERIMENTAL DESIGN: TroVax was administered to 17 patients with metastatic colorectal cancer. In total, 11 patients were considered to be evaluable for assessment of immunologic responses having received a total of six injections of TroVax, administered before, during, and following completion of chemotherapy. Antibody and cellular responses specific for 5T4 and MVA were monitored throughout the study. RESULTS: Administration of TroVax alongside 5-fluorouracil, folinic acid, and oxaliplatin was safe and well tolerated with no serious adverse events attributed to TroVax. Ten of the 11 evaluable patients mounted 5T4-specific antibody responses with titers ranging from 10 to >1,000. IFNgamma enzyme-linked immunospot responses specific for 5T4 were detected in 10 patients with precursor frequencies exceeding 1 in 1,000 peripheral blood mononuclear cells in 4 patients. Of the 11 evaluable patients, 6 had complete or partial responses. 5T4-specific immune responses, but not MVA-specific immune responses, correlated with clinical benefit. CONCLUSIONS: Potent 5T4-specific cellular and/or antibody responses were induced in all evaluable patients and were still detectable during the period in which chemotherapy was administered. These results suggest that TroVax can be added to chemotherapy regimens without any evidence of enhanced toxicity or reduced immunologic efficacy and may provide additional clinical benefit.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer Vaccines/therapeutic use , Colorectal Neoplasms/therapy , Vaccinia virus/genetics , Adult , Aged , Cancer Vaccines/immunology , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Female , Fluorouracil/administration & dosage , Humans , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Lymphatic Metastasis , Male , Membrane Glycoproteins/immunology , Middle Aged , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Tomography, X-Ray Computed , Vaccination , Vaccines, DNA
6.
J Gene Med ; 9(5): 345-56, 2007 May.
Article in English | MEDLINE | ID: mdl-17366519

ABSTRACT

Pseudotyping viral vectors with vesicular stomatitis virus glycoprotein (VSV-G) enables the transduction of an extensive range of cell types from different species. We have discovered two important parameters of the VSV-G-pseudotyping phenomenon that relate directly to the transduction potential of lentiviral vectors: (1) the glycosylation status of VSV-G, and (2) the quantity of glycoprotein associated with virions. We measured production-cell and virion-associated quantities of two isoform variants of VSV-G, which differ in their glycosylation status, VSV-G1 and VSV-G2, and assessed the impact of this difference on the efficiency of mammalian cell transduction by lentiviral vectors. The glycosylation of VSV-G at N336 allowed greater maximal expression of VSV-G in HEK293T cells, thus facilitating vector pseudotyping. The transduction of primate cell lines was substantially affected (up to 50-fold) by the degree of VSV-G1 or VSV-G2 incorporation, whereas other cell lines, such as D17 (canine), were less sensitive to virion-associated VSV-G1/2 quantities. These data indicate that the minimum required concentration of virion-associated VSV-G differs substantially between cell species/types. The implications of these data with regard to VSV-G-pseudotyped vector production, titration, and use in host-cell restriction studies, are discussed.


Subject(s)
Genetic Vectors , Lentivirus/genetics , Membrane Glycoproteins/genetics , Transduction, Genetic , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/genetics , Animals , Cell Line , Glycosylation , Humans , Membrane Glycoproteins/analysis , Membrane Glycoproteins/chemistry , Protein Isoforms/analysis , Protein Isoforms/chemistry , Species Specificity , Viral Envelope Proteins/analysis , Viral Envelope Proteins/chemistry , Viral Proteins/analysis , Viral Proteins/chemistry , Viral Proteins/genetics , Virion/chemistry
7.
Mol Ther ; 14(4): 536-45, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16950499

ABSTRACT

Lentiviral vectors based on equine infectious anemia virus (EIAV) stably integrate into dividing and nondividing cells such as neurons, conferring long-term expression of their transgene. The integration profile of an EIAV vector was analyzed in dividing HEK293T cells, alongside an HIV-1 vector as a control, and compared to a random dataset generated in silico. A multivariate regression model was generated and the influence of the following parameters on integration site selection determined: (a) within/not within a gene, (b) GC content within 20 kb, (c) within 10 kb of a CpG island, (d) gene density within a 2-Mb window, and (e) chromosome number. The majority of the EIAV integration sites (68%; n = 458) and HIV-1 integration sites (72%; n = 162) were within a gene, and both vectors favored AT-rich regions. Sites within genes were examined using a second model to determine the influence of the gene-specific parameters, gene region, and transcriptional activity. Both EIAV and HIV-1 vectors preferentially integrated within active genes. Unlike the gammaretrovirus MLV, EIAV and HIV-1 vectors do not integrate preferentially into the promoter region or the 5' end of the transcription unit.


Subject(s)
Genetic Vectors/genetics , Infectious Anemia Virus, Equine/genetics , Base Composition/genetics , Base Sequence , Cell Line , Chromosomes, Human/genetics , Gene Expression , Genome, Human/genetics , Humans , Transcription, Genetic/genetics
8.
Clin Cancer Res ; 12(11 Pt 1): 3416-24, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740766

ABSTRACT

PURPOSE: The highly attenuated strain of vaccinia virus, modified vaccinia Ankara (MVA), encoding the tumor antigen 5T4 (termed TroVax), has been evaluated in an open-label phase I/II study in colorectal cancer patients. The primary objectives were to assess the safety and immunogenicity of ascending doses of TroVax and to determine the biodistribution of the vector. EXPERIMENTAL DESIGN: TroVax was given to 22 patients with metastatic colorectal cancer. Seventeen patients received doses of TroVax ranging from 5 x 10(7) up to 5 x 10(8) plaque-forming units at 0, 4, and 8 weeks and were considered to be evaluable for assessment of immunologic responses. Both antibody and cellular responses specific for the tumor antigen 5T4 and the viral vector were monitored throughout the study. RESULTS: TroVax was well tolerated in all patients with no serious adverse events attributed to vaccination. Of 17 evaluable patients, 16 showed 5T4-specific cellular responses whereas 14 had detectable antibody levels following vaccination. TroVax was able to boost 5T4-specific immune responses in the presence of MVA neutralizing antibodies. Periods of disease stabilization ranging from 3 to 18 months were observed in five patients, all of whom mounted 5T4-specific immune responses. Furthermore, statistical analysis showed a positive association between the development of a 5T4 (but not MVA) antibody response and patient survival or time to disease progression. CONCLUSION: These data indicate that vaccination with TroVax is safe and well tolerated and that immune responses to 5T4 can be induced without any evidence of autoimmune toxicity. Furthermore, 5T4-specific antibody responses correlate with evidence of disease control.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Colorectal Neoplasms/therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adult , Aged , Antigen-Antibody Reactions , Antigens, Neoplasm/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Cell Proliferation/drug effects , Colorectal Neoplasms/immunology , Colorectal Neoplasms/secondary , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions , Female , Follow-Up Studies , Humans , Male , Middle Aged , Survival Rate , Time Factors , Treatment Outcome , Vaccination , Vaccines, DNA
9.
Hum Gene Ther ; 17(1): 1-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16409120

ABSTRACT

The management of disorders of the nervous system remains a medical challenge. The key goals are to understand disease mechanisms, to validate therapeutic targets, and to develop new therapeutic strategies. Viral vector-mediated gene transfer can meet these goals and vectors based on lentiviruses have particularly useful features. Lentiviral vectors can deliver 8 kb of sequence, they mediate gene transfer into any neuronal cell type, expression and therapy are sustained, and normal cellular functions in vitro and in vivo are not compromised. After delivery into the nervous system they induce no significant immune responses, there are no unwanted side effects of the vectors per se to date, and manufacturing and safety testing for clinical applications are well advanced. There are now numerous examples of effective long-term treatment of animal models of neurological disorders, such as Parkinson's disease, Alzheimer's disease, Huntington's disease, motor neuron diseases, lysosomal storage diseases, and spinal injury, using a range of therapeutic genes expressed in lentiviral vectors. Significant issues remain in some areas of neural gene therapy including defining the optimum therapeutic gene(s), increasing the specificity of delivery, regulating expression of potentially toxic genes, and designing clinically relevant strategies. We discuss the applications of lentiviral vectors in therapy and research and highlight the essential features that will ensure their translation to the clinic in the near future.


Subject(s)
Central Nervous System Diseases/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Lentivirus/genetics , Alzheimer Disease/therapy , Animals , Central Nervous System/physiology , Central Nervous System/virology , Humans , Huntington Disease/therapy , Lysosomal Storage Diseases/therapy , Motor Neuron Disease/therapy , Neurobiology/methods , Neurons/physiology , Neurons/virology , Parkinson Disease/therapy , Spinal Cord Injuries/therapy
10.
Nat Neurosci ; 9(2): 243-50, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16388307

ABSTRACT

The embryonic CNS readily undergoes regeneration, unlike the adult CNS, which has limited axonal repair after injury. Here we tested the hypothesis that retinoic acid receptor beta2 (RARbeta2), critical in development for neuronal growth, may enable adult neurons to grow in an inhibitory environment. Overexpression of RARbeta2 in adult rat dorsal root ganglion cultures increased intracellular levels of cyclic AMP and stimulated neurite outgrowth. Stable RARbeta2 expression in DRG neurons in vitro and in vivo enabled their axons to regenerate across the inhibitory dorsal root entry zone and project into the gray matter of the spinal cord. The regenerated neurons enhanced second-order neuronal activity in the spinal cord, and RARbeta2-treated rats showed highly significant improvement in sensorimotor tasks. These findings show that RARbeta2 induces axonal regeneration programs within injured neurons and may thus offer new therapeutic opportunities for CNS regeneration.


Subject(s)
Nerve Regeneration/physiology , Neurons, Afferent/metabolism , Receptors, Retinoic Acid/metabolism , Spinal Cord/metabolism , Animals , Cells, Cultured , Ganglia, Spinal/injuries , Ganglia, Spinal/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Rats , Rats, Wistar , Recovery of Function/physiology
11.
Cancer Immunol Immunother ; 55(9): 1081-90, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16311730

ABSTRACT

5T4 is a tumor associated antigen that is expressed on the surface of a wide spectrum of human adenocarcinomas. The highly attenuated virus, modified vaccinia Ankara, has been engineered to express human 5T4 (h5T4). In a pre-clinical murine model, the recombinant virus (TroVax) induces protection against challenge with CT26-h5T4 (a syngeneic tumor line expressing h5T4). Anti-tumor activity is long lived, with protection still evident 6 months after the final vaccination. In a therapeutic setting, injection of mice with TroVax results in a reduction in tumor burden of >90%. Depletion of CD8+ T cells has no effect upon therapy in the active treatment model, whereas depletion of CD4+ T cells completely abrogates anti-tumor activity. In a prophylactic setting, depletion of CD4+ and CD8+ T cells after the induction of a h5T4 immune response has no deleterious effect on protection following challenge with CT26-h5T4. In light of these studies, the role of antibodies in protection against tumor challenge was investigated. 5T4 specific polyclonal serum decreased tumor burden by approximately 70%. Thus, we conclude that CD4+ T cells are essential for the induction of a protective immune response and that antibodies are the likely effector moiety in this xenogeneic murine tumor model.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Carcinoma/therapy , Colonic Neoplasms/therapy , Vaccinia virus/genetics , Animals , Antibodies/metabolism , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , Antigens, Surface/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/pharmacology , Carcinoma/immunology , Colonic Neoplasms/immunology , Disease Models, Animal , Female , Humans , Immunotherapy, Active , Infusions, Parenteral , Membrane Glycoproteins , Mice , Mice, Inbred BALB C , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Vaccines, DNA , Vaccinia virus/immunology
12.
Nat Med ; 11(4): 429-33, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15768029

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease resulting in the selective death of motor neurons in the brain and spinal cord. Some familial cases of ALS are caused by dominant mutations in the gene encoding superoxide dismutase (SOD1). The emergence of interfering RNA (RNAi) for specific gene silencing could be therapeutically beneficial for the treatment of such dominantly inherited diseases. We generated a lentiviral vector to mediate expression of RNAi molecules specifically targeting the human SOD1 gene (SOD1). Injection of this vector into various muscle groups of mice engineered to overexpress a mutated form of human SOD1 (SOD1(G93A)) resulted in an efficient and specific reduction of SOD1 expression and improved survival of vulnerable motor neurons in the brainstem and spinal cord. Furthermore, SOD1 silencing mediated an improved motor performance in these animals, resulting in a considerable delay in the onset of ALS symptoms by more than 100% and an extension in survival by nearly 80% of their normal life span. These data are the first to show a substantial extension of survival in an animal model of a fatal, dominantly inherited neurodegenerative condition using RNAi and provide the highest therapeutic efficacy observed in this field to date.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Nerve Degeneration , RNA Interference , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/therapy , Animals , Disease Models, Animal , Genetic Therapy , Genetic Vectors , Humans , Lentivirus/genetics , Mice , Mice, Transgenic , Mutation , RNA, Small Interfering , Survival Rate , Transfection
13.
Mol Ther ; 11(1): 89-95, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15585409

ABSTRACT

Nutrient deprivation during ischemia leads to severe insult to neurons causing widespread excitotoxic damage in specific brain regions such as the hippocampus. One possible strategy for preventing neurodegeneration is to express therapeutic proteins in the brain to protect against excitotoxicity. We investigated the utility of equine infectious anemia virus (EIAV)-based vectors as genetic tools for delivery of therapeutic proteins in an in vivo excitotoxicity model. The efficacy of these vectors at preventing cellular loss in target brain areas following excitotoxic insult was also assessed. EIAV vectors generated to overexpress the human antiapoptotic Bcl-2 or growth factor glial-derived neurotrophic factor (GDNF) genes protected against glutamate-induced toxicity in cultured hippocampal neurons. In an in vivo excitotoxicity model, adult Wistar rats received a unilateral dose of the glutamate receptor agonist N-methyl-D-aspartate to the hippocampus that induced a large lesion in the CA1 region. Neuronal loss could not be protected by prior transduction of a control vector expressing beta-galactosidase. In contrast, EIAV-mediated expression of Bcl-2 and GDNF significantly reduced lesion size thus protecting the hippocampus from excitotoxic damage. These results demonstrate that EIAV vectors can be effectively used to deliver putative neuroprotective genes to target brain areas and prevent cellular loss in the event of a neurological insult. Therefore these lentiviral vectors provide potential therapeutic tools for use in cases of acute neurotrauma such as cerebral ischemia.


Subject(s)
Hippocampus/drug effects , Hippocampus/physiopathology , Lentivirus/genetics , Nerve Growth Factors/metabolism , Neuroprotective Agents/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Cell Line , Genetic Vectors/genetics , Glial Cell Line-Derived Neurotrophic Factor , Hippocampus/metabolism , Hippocampus/pathology , Humans , N-Methylaspartate/toxicity , Nerve Growth Factors/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Tissue Culture Techniques
14.
J Clin Invest ; 114(12): 1726-31, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15599397

ABSTRACT

Spinal muscular atrophy (SMA) is a frequent recessive autosomal disorder. It is caused by mutations or deletion of the telomeric copy of the survival motor neuron (SMN) gene, leading to depletion in SMN protein levels. The treatment rationale for SMA is to halt or delay the degeneration of motor neurons, but to date there are no effective drug treatments for this disease. We have previously demonstrated that pseudotyping of the nonprimate equine infectious anemia virus (using the lentivector gene transfer system) with the glycoprotein of the Evelyn-Rokitnicki-Abelseth strain of the rabies virus confers retrograde axonal transport on these vectors. Here, we report that lentivector expressing human SMN was successfully used to restore SMN protein levels in SMA type 1 fibroblasts. Multiple single injections of a lentiviral vector expressing SMN in various muscles of SMA mice restored SMN to motor neurons, reduced motor neuron death, and increased the life expectancy by an average of 3 and 5 days (20% and 38%) compared with LacZ and untreated animals, respectively. Further extension of survival by SMN expression constructs will likely require a knowledge of when and/or where high levels of SMN are needed.


Subject(s)
Lentivirus/genetics , Nerve Tissue Proteins/genetics , Animals , Cell Line , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein , Disease Models, Animal , Fibroblasts/metabolism , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Humans , Immunohistochemistry , Lac Operon , Mice , Microscopy, Fluorescence , Motor Neurons/metabolism , RNA-Binding Proteins , SMN Complex Proteins , Time Factors
15.
J Gene Med ; 6(9): 951-62, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15352068

ABSTRACT

Vectors based on lentiviruses efficiently deliver genes into many different types of primary neurons from a broad range of species including man and the resulting gene expression is long term. These vectors are opening up new approaches for the treatment of neurological diseases such as Parkinson's disease (PD), Huntington's disease (HD), and motor neuron diseases (MNDs). Numerous animal studies have now been undertaken with these vectors and correction of disease models has been obtained. Lentiviral vectors also provide a new strategy for in vivo modeling of human diseases; for example, the lentiviral-mediated overexpression of mutated human alpha-synuclein or huntingtin genes in basal ganglia induces neuronal pathology in animals resembling PD and HD in man. These vectors have been refined to a very high level and can be produced safely for the clinic. This review will describe the general features of lentiviral vectors with particular emphasis on vectors derived from the non-primate lentivirus, equine infectious anemia virus (EIAV). It will then describe some key examples of genetic correction and generation of genetic animal models of neurological diseases. The prospects for clinical application of lentiviral vectors for the treatment of PD and MNDs will also be outlined.


Subject(s)
Central Nervous System Diseases/therapy , Genetic Therapy/methods , Genetic Vectors , Lentivirus/genetics , Animals , Disease Models, Animal , Gene Expression , Humans , Huntington Disease/therapy , Motor Neuron Disease/therapy , Parkinson Disease/therapy
16.
Nature ; 429(6990): 413-7, 2004 May 27.
Article in English | MEDLINE | ID: mdl-15164063

ABSTRACT

Amyotrophic lateral sclerosis (ALS) causes adult-onset, progressive motor neuron degeneration in the brain and spinal cord, resulting in paralysis and death three to five years after onset in most patients. ALS is still incurable, in part because its complex aetiology remains insufficiently understood. Recent reports have indicated that reduced levels of vascular endothelial growth factor (VEGF), which is essential in angiogenesis and has also been implicated in neuroprotection, predispose mice and humans to ALS. However, the therapeutic potential of VEGF for the treatment of ALS has not previously been assessed. Here we report that a single injection of a VEGF-expressing lentiviral vector into various muscles delayed onset and slowed progression of ALS in mice engineered to overexpress the gene coding for the mutated G93A form of the superoxide dismutase-1 (SOD1(G93A)) (refs 7-10), even when treatment was only initiated at the onset of paralysis. VEGF treatment increased the life expectancy of ALS mice by 30 per cent without causing toxic side effects, thereby achieving one of the most effective therapies reported in the field so far.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Axonal Transport , Disease Models, Animal , Infectious Anemia Virus, Equine/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/therapeutic use , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain Stem/pathology , Disease Progression , Gene Expression , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Injections, Intramuscular , Mice , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Point Mutation/genetics , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Survival Rate , Time Factors , Transgenes/genetics , Vascular Endothelial Growth Factor A/adverse effects , Vascular Endothelial Growth Factor A/metabolism
17.
Neuroreport ; 15(6): 985-90, 2004 Apr 29.
Article in English | MEDLINE | ID: mdl-15076720

ABSTRACT

Vectors based on lentiviruses are opening up new approaches for the treatment of neurodegenerative diseases. Currently, the equine infectious anaemia virus (EIAV) vector is one of the most attractive gene delivery systems with respect to neuronal tropism. The aim was to validate EIAV-lentiviral vectors as a gene delivery system for neurotrophic factor genes in an animal model of Parkinson's disease. EIAV carrying the glial cell line-derived neurotrophic factor (GDNF) gene was unilaterally injected into rat striatum and above the substantia nigra (SN). One week later, the rats received a 6-OHDA lesion into the ipsilateral striatum. GDNF delivery led to extensive expression of GDNF protein within the striatum. In addition, near complete protection against dopaminergic cell death was observed in the GDNF-treated group.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/therapeutic use , Nerve Growth Factors/biosynthesis , Neuroprotective Agents/therapeutic use , Parkinson Disease/prevention & control , Animals , Female , Genetic Vectors/genetics , Glial Cell Line-Derived Neurotrophic Factor , Infectious Anemia Virus, Equine/genetics , Nerve Growth Factors/genetics , Parkinson Disease/genetics , Rats , Rats, Sprague-Dawley
18.
Mol Ther ; 9(1): 101-11, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14741783

ABSTRACT

We have developed a non-primate-based lentiviral vector based on the equine infectious anemia virus (EIAV) for efficient gene transfer to the central and peripheral nervous systems. Previously we have demonstrated that pseudotyping lentiviral vectors with the rabies virus glycoprotein confers retrograde axonal transport to these vectors. In the present study we have successfully produced high-titer EIAV vectors pseudotyped with envelope glycoproteins from Rhabdovirus vesicular stomatitis virus (VSV) serotypes (Indiana and Chandipura strains); rabies virus [various Evelyn-Rokitnicki-Abelseth ERA strains and challenge virus standard (CVS)]; Lyssavirus Mokola virus, a rabies-related virus; and Arenavirus lymphocytic choriomeningitis virus (LCMV). These vectors were delivered to the striatum or spinal cord of adult rats or muscle of neonatal mice by direct injection. We report that the lentiviral vectors pseudotyped with envelopes from the VSV Indiana strain, wild-type ERA, and CVS strains resulted in strong transduction in the striatum, while Mokola- and LCMV-pseudotyped vectors exhibited moderate and weak transduction, respectively. Furthermore ERA- and CVS-pseudotyped lentiviral vectors demonstrated retrograde transport and expression in distal neurons after injection in brain, spinal cord, and muscle. The differences in transduction efficiencies and retrograde transport conferred by these envelope glycoproteins present novel opportunities in designing therapeutic strategies for different neurological diseases.


Subject(s)
Genetic Vectors , Infectious Anemia Virus, Equine/genetics , Nervous System/metabolism , Transduction, Genetic , Viral Envelope Proteins/genetics , Animals , Biological Transport , Corpus Striatum/cytology , Gene Expression/physiology , Genetic Therapy , Glycoproteins/metabolism , Injections , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/metabolism , Lyssavirus/genetics , Male , Mice , Muscle, Skeletal , Nervous System Diseases/therapy , Rabies virus/genetics , Rabies virus/metabolism , Rats , Rats, Wistar , Spinal Cord , Viral Envelope Proteins/metabolism
19.
Mol Ther ; 7(5 Pt 1): 588-96, 2003 May.
Article in English | MEDLINE | ID: mdl-12718901

ABSTRACT

Due to the complexity of brain function and the difficulty in monitoring alterations in neuronal gene expression, the potential of lentiviral gene therapy vectors to treat disorders of the CNS has been difficult to fully assess. In this study, we have assessed the utility of a third-generation equine infectious anemia virus (EIAV) in the Brattleboro rat model of diabetes insipidus, in which a mutation in the arginine vasopressin (AVP) gene results in the production of nonfunctional mutant AVP precursor protein. Importantly, by using this model it is possible to monitor the success of the gene therapy treatment by noninvasive assays. Injection of an EIAV-CMV-AVP vector into the supraoptic nuclei of the hypothalamus resulted in expression of functional AVP peptide in magnocellular neurons. This was accompanied by a 100% recovery in water homeostasis as assessed by daily water intake, urine production, and urine osmolality lasting for a 1-year measurement period. These data show that a single gene defect leading to a neurological disorder can be corrected with a lentiviral-based strategy. This study highlights the potential of using viral gene therapy for the long-term treatment of disorders of the CNS.


Subject(s)
Arginine Vasopressin/genetics , Diabetes Insipidus, Neurogenic/therapy , Genetic Therapy , Genetic Vectors , Infectious Anemia Virus, Equine/genetics , Supraoptic Nucleus/metabolism , Animals , Arginine Vasopressin/metabolism , Diabetes Insipidus, Neurogenic/metabolism , Diabetes Insipidus, Neurogenic/pathology , Homeostasis , Humans , In Situ Hybridization , Male , Rats , Rats, Brattleboro , Rats, Inbred WKY , Supraoptic Nucleus/pathology , Time Factors , Vasoconstrictor Agents/metabolism , Water/metabolism
20.
Mol Cancer Ther ; 1(12): 1129-37, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12481437

ABSTRACT

The human oncofetal antigen 5T4 (h5T4) is a transmembrane glycoprotein overexpressed by a wide spectrum of cancers, including colorectal, ovarian, and gastric, but with a limited normal tissue expression. Such properties make 5T4 an excellent putative target for cancer immunotherapy. The murine homologue of 5T4 (m5T4) has been cloned and characterized, which allows for the evaluation of immune intervention strategies in "self-antigen" in vivo tumor models. We have constructed recombinant vaccinia viruses based on the highly attenuated and modified vaccinia virus ankara (MVA strain), expressing h5T4 (MVA-h5T4), m5T4 (MVA-m5T4), and Escherichia coli LacZ (MVA-LacZ). Immunization of BALB/c and C57BL/6 mice with MVA-h5T4 and MVA-m5T4 constructs induced antibody responses to human and mouse 5T4, respectively. C57BL/6 and BALB/c mice vaccinated with MVA-h5T4 were challenged with syngeneic tumor line transfectants, B16 melanoma, and CT26 colorectal cells that express h5T4. MVA-h5T4-vaccinated mice showed significant tumor retardation compared with mice vaccinated with MVA-LacZ or PBS. In active treatment studies, inoculation with MVA-h5T4 was able to treat established CT26-h5T4 lung tumor and to a lesser extent B16.h5T4 s.c. tumors. Additionally, when C57BL/6 mice vaccinated with MVA-m5T4 were challenged with B16 cells expressing m5T4, resulting growth of the tumors was significantly retarded compared with control animals. Furthermore, mice vaccinated with MVA-m5T4 showed no signs of autoimmune toxicity. These data support the use of MVA-5T4 for tumor immunotherapy.


Subject(s)
Cancer Vaccines , Immunotherapy/methods , Membrane Glycoproteins/metabolism , Neoplasms/drug therapy , Vaccinia virus/metabolism , Animals , Antigens, Neoplasm/chemistry , Biomarkers, Tumor/chemistry , Blotting, Western , Cell Separation , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Melanoma, Experimental , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/cytology , Time Factors , Transfection , Tumor Cells, Cultured , Vaccinia virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...