Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
N Engl J Med ; 371(21): 1994-2004, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25409372

ABSTRACT

BACKGROUND: In patients with severe hemophilia B, gene therapy that is mediated by a novel self-complementary adeno-associated virus serotype 8 (AAV8) vector has been shown to raise factor IX levels for periods of up to 16 months. We wanted to determine the durability of transgene expression, the vector dose-response relationship, and the level of persistent or late toxicity. METHODS: We evaluated the stability of transgene expression and long-term safety in 10 patients with severe hemophilia B: 6 patients who had been enrolled in an initial phase 1 dose-escalation trial, with 2 patients each receiving a low, intermediate, or high dose, and 4 additional patients who received the high dose (2×10(12) vector genomes per kilogram of body weight). The patients subsequently underwent extensive clinical and laboratory monitoring. RESULTS: A single intravenous infusion of vector in all 10 patients with severe hemophilia B resulted in a dose-dependent increase in circulating factor IX to a level that was 1 to 6% of the normal value over a median period of 3.2 years, with observation ongoing. In the high-dose group, a consistent increase in the factor IX level to a mean (±SD) of 5.1±1.7% was observed in all 6 patients, which resulted in a reduction of more than 90% in both bleeding episodes and the use of prophylactic factor IX concentrate. A transient increase in the mean alanine aminotransferase level to 86 IU per liter (range, 36 to 202) occurred between week 7 and week 10 in 4 of the 6 patients in the high-dose group but resolved over a median of 5 days (range, 2 to 35) after prednisolone treatment. CONCLUSIONS: In 10 patients with severe hemophilia B, the infusion of a single dose of AAV8 vector resulted in long-term therapeutic factor IX expression associated with clinical improvement. With a follow-up period of up to 3 years, no late toxic effects from the therapy were reported. (Funded by the National Heart, Lung, and Blood Institute and others; ClinicalTrials.gov number, NCT00979238.).


Subject(s)
Factor IX/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Hemophilia B/therapy , Adult , Alanine Transaminase/blood , Dependovirus/genetics , Factor IX/metabolism , Follow-Up Studies , Gene Expression , Genetic Therapy/adverse effects , Hemophilia B/blood , Hemophilia B/genetics , Humans , Infusions, Intravenous , Male , Middle Aged , Transgenes , Young Adult
2.
Blood ; 121(17): 3335-44, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23426947

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors encoding human factor VIII (hFVIII) were systematically evaluated for hemophilia A (HA) gene therapy. A 5.7-kb rAAV-expression cassette (rAAV-HLP-codop-hFVIII-N6) containing a codon-optimized hFVIII cDNA in which a 226 amino acid (aa) B-domain spacer replaced the entire B domain and a hybrid liver-specific promoter (HLP) mediated 10-fold higher hFVIII levels in mice compared with non-codon-optimized variants. A further twofold improvement in potency was achieved by replacing the 226-aa N6 spacer with a novel 17-aa peptide (V3) in which 6 glycosylation triplets from the B domain were juxtaposed. The resulting 5.2-kb rAAV-HLP-codop-hFVIII-V3 cassette was more efficiently packaged within AAV virions and mediated supraphysiologic hFVIII expression (732 ± 162% of normal) in HA knock-out mice following administration of 2 × 10(12) vector genomes/kg, a vector dose shown to be safe in subjects with hemophilia B. Stable hFVIII expression at 15 ± 4% of normal was observed at this dose in a nonhuman primate. hFVIII expression above 100% was observed in 3 macaques that received a higher dose of either this vector or the N6 variant. These animals developed neutralizing anti-FVIII antibodies that were abrogated with transient immunosuppression. Therefore, rAAV-HLP-codop-hFVIII-V3 substantially improves the prospects of effective HA gene therapy.


Subject(s)
Dependovirus/genetics , Factor VIII/pharmacology , Genetic Therapy , Genetic Variation/genetics , Genetic Vectors/administration & dosage , Hemophilia A/therapy , Animals , Blotting, Western , Factor VIII/genetics , Factor VIII/immunology , Glycosylation , Hemophilia A/genetics , Humans , Immune Tolerance , Liver/metabolism , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/genetics , Peptide Fragments/metabolism , Promoter Regions, Genetic/genetics
3.
N Engl J Med ; 365(25): 2357-65, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22149959

ABSTRACT

BACKGROUND: Hemophilia B, an X-linked disorder, is ideally suited for gene therapy. We investigated the use of a new gene therapy in patients with the disorder. METHODS: We infused a single dose of a serotype-8-pseudotyped, self-complementary adenovirus-associated virus (AAV) vector expressing a codon-optimized human factor IX (FIX) transgene (scAAV2/8-LP1-hFIXco) in a peripheral vein in six patients with severe hemophilia B (FIX activity, <1% of normal values). Study participants were enrolled sequentially in one of three cohorts (given a high, intermediate, or low dose of vector), with two participants in each group. Vector was administered without immunosuppressive therapy, and participants were followed for 6 to 16 months. RESULTS: AAV-mediated expression of FIX at 2 to 11% of normal levels was observed in all participants. Four of the six discontinued FIX prophylaxis and remained free of spontaneous hemorrhage; in the other two, the interval between prophylactic injections was increased. Of the two participants who received the high dose of vector, one had a transient, asymptomatic elevation of serum aminotransferase levels, which was associated with the detection of AAV8-capsid-specific T cells in the peripheral blood; the other had a slight increase in liver-enzyme levels, the cause of which was less clear. Each of these two participants received a short course of glucocorticoid therapy, which rapidly normalized aminotransferase levels and maintained FIX levels in the range of 3 to 11% of normal values. CONCLUSIONS: Peripheral-vein infusion of scAAV2/8-LP1-hFIXco resulted in FIX transgene expression at levels sufficient to improve the bleeding phenotype, with few side effects. Although immune-mediated clearance of AAV-transduced hepatocytes remains a concern, this process may be controlled with a short course of glucocorticoids without loss of transgene expression. (Funded by the Medical Research Council and others; ClinicalTrials.gov number, NCT00979238.).


Subject(s)
Dependovirus , Factor IX/genetics , Genetic Therapy , Genetic Vectors , Hemophilia B/therapy , Adult , Dependovirus/genetics , Factor IX/therapeutic use , Genetic Therapy/adverse effects , Genetic Vectors/immunology , Humans , Infusions, Intravenous , Middle Aged , Transgenes/immunology
4.
Mol Ther ; 19(5): 876-85, 2011 May.
Article in English | MEDLINE | ID: mdl-21245849

ABSTRACT

Adeno-associated virus vectors (AAV) show promise for liver-targeted gene therapy. In this study, we examined the long-term consequences of a single intravenous administration of a self-complementary AAV vector (scAAV2/ 8-LP1-hFIXco) encoding a codon optimized human factor IX (hFIX) gene in 24 nonhuman primates (NHPs). A dose-response relationship between vector titer and transgene expression was observed. Peak hFIX expression following the highest dose of vector (2 × 10(12) pcr-vector genomes (vg)/kg) was 21 ± 3 µg/ml (~420% of normal). Fluorescent in-situ hybridization demonstrated scAAV provirus in almost 100% of hepatocytes at that dose. No perturbations of clinical or laboratory parameters were noted and vector genomes were cleared from bodily fluids by 10 days. Macaques transduced with 2 × 10(11) pcr-vg/kg were followed for the longest period (~5 years), during which time expression of hFIX remained >10% of normal level, despite a gradual decline in transgene copy number and the proportion of transduced hepatocytes. All macaques developed serotype-specific antibodies but no capsid-specific cytotoxic T lymphocytes were detected. The liver was preferentially transduced with 300-fold more proviral copies than extrahepatic tissues. Long-term biochemical, ultrasound imaging, and histologic follow-up of this large cohort of NHP revealed no toxicity. These data support further evaluation of this vector in hemophilia B patients.


Subject(s)
Capsid Proteins/metabolism , Dependovirus/genetics , Factor IX/metabolism , Genetic Therapy/methods , Hemophilia B/therapy , Animals , Capsid Proteins/genetics , Factor IX/genetics , Gene Expression , Genetic Vectors , HEK293 Cells , Hemophilia B/genetics , Humans , In Situ Hybridization, Fluorescence , Liver/metabolism , Macaca , Mice
5.
J Pediatr Surg ; 42(7): 1172-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17618876

ABSTRACT

BACKGROUND: We evaluated the potential of bioluminescence imaging (BLI) for early tumor detection, demonstrating occult sites of disseminated disease and assessing disease progression in a murine model of neuroblastoma. METHODS: Neuroblastoma cells engineered to express the enzyme firefly luciferase were used to establish localized tumors and disseminated disease in SCID mice. Bioluminescent signal intensity was measured at serial time points, and compared with traditional methods of evaluating tumor growth. RESULTS: Bioluminescence imaging detected subcutaneous and retroperitoneal tumors weeks before they were palpable or appreciable by ultrasound. Bioluminescent signal intensity at both sites then paralleled tumor growth. After intravenous administration of tumor cells, BLI revealed disseminated disease in the liver, lungs, and bone marrow, again weeks before any gross disease was present. The presence of tumor within these sites at early time points was confirmed by reverse transcriptase-polymerase chain reaction. Finally, BLI permitted a real-time, noninvasive, quantitative method for following response to therapy in a model of minimal residual disease. CONCLUSION: Bioluminescence imaging detects tumor much earlier than traditional methods. In addition, it can detect, quantify, and follow micrometastasis in real-time during disease progression. This methodology is extremely valuable for studying tumor tissue tropism, mechanisms of metastasis, and response to therapy in murine tumor models.


Subject(s)
Luminescent Measurements/methods , Neuroblastoma/pathology , Animals , Blotting, Southern , Disease Progression , Early Diagnosis , Immunoenzyme Techniques , Luciferases, Firefly , Mice , Mice, SCID , Regression Analysis , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
6.
Clin Cancer Res ; 13(13): 3942-50, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17606728

ABSTRACT

PURPOSE: Dysfunctional tumor vessels can be a significant barrier to effective cancer therapy. However, increasing evidence suggests that vascular endothelial growth factor (VEGF) inhibition can effect transient "normalization" of the tumor vasculature, thereby improving tumor perfusion and, consequently, delivery of systemic chemotherapy. We sought to examine temporal changes in tumor vascular function in response to the anti-VEGF antibody, bevacizumab. EXPERIMENTAL DESIGN: Established orthotopic neuroblastoma xenografts treated with bevacizumab were evaluated at serial time points for treatment-associated changes in intratumoral vascular physiology, penetration of systemically administered chemotherapy, and efficacy of combination therapy. RESULTS: After a single bevacizumab dose, a progressive decrease in tumor microvessel density to <30% of control was observed within 7 days. Assessment of the tumor microenvironment revealed a rapid, sustained decrease in both tumor vessel permeability and tumor interstitial fluid pressure, whereas intratumoral perfusion, as assessed by contrast-enhanced ultrasonography, was improved, although this latter change abated by 1 week. Intratumoral drug delivery mirrored these changes; penetration of chemotherapy was improved by as much as 81% when given 1 to 3 days after bevacizumab, compared with when both drugs were given concomitantly, or 7 days apart. Finally, administering topotecan to tumor-bearing mice 3 days after bevacizumab resulted in greater tumor growth inhibition (36% of control size) than with monotherapy (88% bevacizumab, 54% topotecan) or concomitant administration of the two drugs (44%). CONCLUSIONS: Bevacizumab-mediated VEGF blockade effects alterations in tumor vessel physiology that allow improved delivery and efficacy of chemotherapy, although careful consideration of drug scheduling is required to optimize antitumor activity.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Neuroblastoma/drug therapy , Animals , Antibodies, Monoclonal, Humanized , Bevacizumab , Cell Line, Tumor , Humans , Male , Mice , Microcirculation , Neoplasm Transplantation , Neovascularization, Pathologic , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phenotype , Pressure , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism
7.
Mol Cancer Res ; 5(6): 531-42, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17579115

ABSTRACT

IFNs have pleiotropic antitumor mechanisms of action. The purpose of this study was to further investigate the effects of IFN-beta on the vasculature of human xenografts in immunodeficient mice. We found that continuous, systemic IFN-beta delivery, established with liver-targeted adeno-associated virus vectors, led to sustained morphologic and functional changes of the tumor vasculature that were consistent with vessel maturation. These changes included increased smooth muscle cell coverage of tumor vessels, improved intratumoral blood flow, and decreased vessel permeability, tumor interstitial pressure, and intratumoral hypoxia. Although these changes in the tumor vasculature resulted in more efficient tumor perfusion, further tumor growth was restricted, as the mature vasculature seemed to be unable to expand to support further tumor growth. In addition, maturation of the intratumoral vasculature resulted in increased intratumoral penetration of systemically administered chemotherapy. Finally, molecular analysis revealed increased expression by treated tumors of angiopoietin-1, a cytokine known to promote vessel stabilization. Induction of angiopoietin-1 expression in response to IFN-beta was broadly observed in different tumor lines but not in those with defects in IFN signaling. In addition, IFN-beta-mediated vascular changes were prevented when angiopoietin signaling was blocked with a decoy receptor. Thus, we have identified an alternative approach for achieving sustained vascular remodeling-continuous delivery of IFN-beta. In addition to restricting tumor growth by inhibiting further angiogenesis, maturation of the tumor vasculature also improved the efficiency of delivery of adjuvant therapy. These results have significant implications for the planning of combination anticancer therapy.


Subject(s)
Interferon-beta/administration & dosage , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic , Angiopoietin-1/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytokines/metabolism , Dependovirus/metabolism , Humans , Hypoxia , Interferon-beta/therapeutic use , Male , Mice , Mice, SCID , Neoplasm Transplantation
8.
J Pediatr Surg ; 42(1): 48-53, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17208540

ABSTRACT

BACKGROUND: Interferon-beta (IFN-beta) has potent antitumor activity; however, systemic toxicity has limited its clinical use. We investigated the potential of targeted delivery using tumor-tropic neural progenitor cells (NPCs) transduced to express human IFN-beta (hIFN-beta). METHODS: Disseminated neuroblastoma was established in SCID mice by tail vein injection of tumor cells. Fourteen days after tumor cell inoculation, systemic disease was confirmed with bioluminescence imaging (BLI). Mice were then treated by intravenous injection of human F3.C1 NPCs that had been transduced with a replication deficient adenovirus to overexpress hIFN-beta (F3-IFN-beta). Two injections were given: the first at 14 days and the second at 28 days following tumor cell injection. Control mice received NPCs transduced with empty vector adenovirus at the same time points. Progression of disease was monitored using BLI. At sacrifice, organ weights and histology further evaluated tumor burden. RESULTS: After initiation of therapy, BLI demonstrated a significant decrease in the rate of disease progression in mice receiving F3-IFN-beta. At necropsy, control mice had bulky tumor replacing the liver and kidneys, as well as extensive retroperitoneal and mediastinal adenopathy. Impressively, these sites within mice receiving F3-IFN-beta therapy appeared grossly normal with the exception of small nodules within the kidneys of some of the F3-IFN-beta-treated mice. The accumulation of F3.C1 cells within sites of tumor growth was confirmed by fluorescence imaging. Importantly, systemic levels of hIFN-beta in the treated mice remained below detectable levels. CONCLUSIONS: These data indicate that in this model of disseminated neuroblastoma, the tumor-tropic property of F3.C1 NPCs was exploited to target delivery of IFN-beta to disseminated tissue foci, resulting in significant tumor growth delay. The described novel approach for effective IFN-beta therapy may circumvent limitations associated with the systemic toxicity of IFN-beta.


Subject(s)
Antineoplastic Agents/administration & dosage , Genetic Therapy/methods , Interferon-beta/administration & dosage , Stem Cells/physiology , Adenoviridae , Animals , Cell Line , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Disease Progression , Drug Delivery Systems/methods , Genetic Vectors , Humans , Injections, Intravenous , Mice , Mice, SCID , Neuroblastoma , Transduction, Genetic , Tumor Burden
9.
J Pediatr Surg ; 42(1): 160-5; discussion 165, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17208558

ABSTRACT

BACKGROUND: We have recently demonstrated that continuous delivery of interferon beta (IFN-beta) stabilizes solid tumor vasculature and improves tumor perfusion. In this study, we have further investigated the functional consequences of this effect by assessing delivery and efficacy of conventional chemotherapy against neuroblastoma xenografts when used in combination with IFN-beta. METHODS: Mice with established retroperitoneal tumors received adeno-associated virus vector encoding IFN-beta (AAV IFN-beta) or control vector. One week later, at 1 hour before sacrifice, a 1 mg/kg i.v. bolus of topotecan (TPT) was given. Intratumoral levels of TPT were measured by high-performance liquid chromatography and then standardized to plasma levels to determine tumor TPT penetration. Subsequent experiments evaluated the antitumor efficacy of topotecan alone or in combination with AAV IFN-beta. RESULTS: As observed in prior experiments, AAV IFN-beta resulted in a marked increase in tumor vessel association with stabilizing perivascular smooth muscle cells. These more "matured" vessels facilitated improved tumor TPT penetration (51.2% +/- 4.2%) compared with controls (30.8% +/- 4.7%, P = .004). In additional cohorts of mice, this resulted in an improved antitumor effect. Mice with established tumors (301.8 +/- 18.1 mm3) were treated with TPT (1 mg/kg daily for 5 days for 2 consecutive weeks) either alone or in combination with AAV IFN-beta (5 x 10(10) vector particles per mouse). Topotecan monotherapy resulted in a reduction in mean tumor volume of 12% (264.2 +/- 65.8 mm3, P = .66). However, when the same regimen was administered to mice receiving continuous IFN-beta therapy, a 61% (118.9 +/- 42.3 mm3, P = .004) reduction in mean tumor volume was achieved. CONCLUSION: Interferon beta-mediated vessel stabilization resulted in improved intratumoral delivery of systemically administered TPT, enhancing its antitumor efficacy. This approach of altering the tumor vasculature provides a strategy to help overcome solid tumor resistance to traditional cytotoxic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Vessels/drug effects , Interferon-beta/pharmacology , Neuroblastoma/metabolism , Topotecan/pharmacology , Adenoviridae , Animals , Disease Models, Animal , Genetic Vectors , Mice , Neoplasm Transplantation
10.
Blood ; 109(4): 1414-21, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17090654

ABSTRACT

The safety and efficacy of peripheral venous administration of a self-complementary adeno-associated viral vector encoding the human FIX gene (scAAV-LP1-hFIXco) was evaluated in nonhuman primates for gene therapy of hemophilia B. Peripheral vein infusion of 1x10(12) vg/kg scAAV-LP1-hFIXco pseudotyped with serotype 8 capsid, in 3 macaques, resulted in stable therapeutic expression (more than 9 months) of human FIX (hFIX) at levels (1.1+/-0.5 microg/mL, or 22% of normal) that were comparable to those achieved after direct delivery of the same vector dose into the portal circulation (1.3+/-0.3 microg/mL, or 26% of normal). Importantly, the pattern of vector biodistribution after systemic and portal vein administration of scAAV-LP1-hFIXco was almost identical. Additionally, comparable levels of gene transfer were achieved in macaques with preexisting immunity to AAV8 following peripheral vein administration of 1x10(12) vg/kg AAV5-pseudotyped scAAV-LP1-hFIXco. This confirms that alternative serotypes can circumvent preexisting naturally acquired immunity to AAV. Thus, peripheral venous administration of AAV5 and AAV8 vectors is safe and as effective at transducing the liver in nonhuman primates as direct vector administration into the portal circulation. These results should make vector administration to patients, especially those with a severe bleeding diathesis, significantly easier and safer.


Subject(s)
Factor IX/administration & dosage , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Hemophilia B/therapy , Liver/metabolism , Transduction, Genetic/methods , Animals , Antibody Formation , Factor IX/pharmacokinetics , Genetic Vectors/pharmacokinetics , Humans , Macaca , Tissue Distribution , Treatment Outcome
11.
Surgery ; 140(2): 227-35, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16904974

ABSTRACT

BACKGROUND: We investigated the antitumor and antiosteoclastic effects of zoledronate against human neuroblastoma in vitro and in a murine model of bone metastasis. METHODS: Antitumor activity of zoledronate against neuroblastoma cell lines was assessed by evaluating proliferation, apoptosis, and cell-cycle progression. A murine model of bone invasion was used to assess antiosteoclastic and antitumor activity in vivo. Mice were followed by radiographic and bioluminescence imaging. RESULTS: Treatment of human neuroblastoma cells resulted in a decrease in cell count, increase in apoptosis, and arrest of cell-cycle progression. In the model of bone invasion, mice were treated weekly with zoledronate or vehicle control 10 days after tumor cell inoculation. Five weeks later, radiographs revealed a large degree of osteolytic disease in control mice. Impressively, mice treated with zoledronate demonstrated minimal radiographic changes at this time. Bioluminescence imaging of these mice revealed a significant restriction of tumor growth during the course of therapy. CONCLUSIONS: Zoledronate exhibits significant antitumor activity against human neuroblastoma cells in vitro, prevents development of osteolytic lesions, and restricts tumor growth in a murine model of bone metastasis. These results suggest that clinical investigation of zoledronate or similar bisphosphonates as adjuvant therapy in neuroblastoma patients is warranted.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Neoplasms/drug therapy , Diphosphonates/therapeutic use , Imidazoles/therapeutic use , Neuroblastoma/drug therapy , Animals , Apoptosis/drug effects , Bone Density Conservation Agents/administration & dosage , Bone Neoplasms/pathology , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Diphosphonates/administration & dosage , Dose-Response Relationship, Drug , Humans , Imidazoles/administration & dosage , Mice , Mice, SCID , Neoplasm Transplantation , Neuroblastoma/pathology , Zoledronic Acid
12.
Blood ; 107(7): 2653-61, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16322469

ABSTRACT

Transduction with recombinant adeno-associated virus (AAV) vectors is limited by the need to convert its single-stranded (ss) genome to transcriptionally active double-stranded (ds) forms. For AAV-mediated hemophilia B (HB) gene therapy, we have overcome this obstacle by constructing a liver-restricted mini-human factor IX (hFIX) expression cassette that can be packaged as complementary dimers within individual AAV particles. Molecular analysis of murine liver transduced with these self-complementary (sc) vectors demonstrated rapid formation of active ds-linear genomes that persisted stably as concatamers or monomeric circles. This unique property resulted in a 20-fold improvement in hFIX expression in mice over comparable ssAAV vectors. Administration of only 1 x 10(10) scAAV particles led to expression of hFIX at supraphysiologic levels (8I U/mL) and correction of the bleeding diathesis in FIX knock-out mice. Of importance, therapeutic levels of hFIX (3%-30% of normal) were achieved in nonhuman primates using a significantly lower dose of scAAV than required with ssAAV. Furthermore, AAV5-pseudotyped scAAV vectors mediated successful transduction in macaques with pre-existing immunity to AAV8. Hence, this novel vector represents an important advance for hemophilia B gene therapy.


Subject(s)
Dependovirus/physiology , Factor IX/genetics , Hemophilia B/therapy , Liver/physiology , Liver/virology , Animals , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Genome, Viral , Humans , Macaca mulatta , Male , Mice , Primates , Transduction, Genetic/methods
13.
Clin Cancer Res ; 11(16): 6020-9, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16115947

ABSTRACT

PURPOSE: Type I IFNs (IFN-alpha/beta) have shown significant antitumor activity in preclinical models but limited efficacy and significant toxicity in clinical trials. We hypothesized that the antitumor activity of type I IFNs could be enhanced by chronic, low-dose systemic delivery and sought to test this in murine neuroblastoma models. EXPERIMENTAL DESIGN: Continuous liver-generated expression of human IFN-beta (hINF-beta) was achieved through a gene therapy-mediated approach using adeno-associated virus vectors encoding hIFN-beta (AAV hINF-beta). Orthotopic localized retroperitoneal and disseminated models of neuroblastoma were established using three different xenografts. Immunohistochemical analysis and ELISA were used to evaluate the antiangiogenic effect of therapy. RESULTS: The development of both localized orthotopic (retroperitoneal) and disseminated neuroblastoma was prevented in all mice expressing hINF-beta. Continued growth of established retroperitoneal tumors, treated with AAV hINF-beta as monotherapy, was significantly restricted, and survival for mice with established, disseminated disease was significantly prolonged following administration of AAV hINF-beta. Analysis of treated tumors revealed a significant antiangiogenic effect. Mean intratumoral vessel density was diminished and expression of the angiogenic factors vascular endothelial growth factor and basic fibroblast growth factor were both decreased. Finally, combination therapy in which AAV hIFN-beta was used together with low-dose cyclophosphamide resulted in regression of both established retroperitoneal and disseminated disease. CONCLUSIONS: AAV-mediated delivery of hIFN-beta when used as monotherapy was able to restrict neuroblastoma growth due in part to inhibition of angiogenesis. When used in combination with conventional chemotherapy, AAV hIFN-beta was able to effect complete tumor regression.


Subject(s)
Cyclophosphamide/therapeutic use , Interferon Type I/genetics , Neuroblastoma/therapy , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Cyclophosphamide/pharmacology , Dependovirus/genetics , Dose-Response Relationship, Drug , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Interferon Type I/metabolism , Interferon Type I/pharmacology , Mice , Mice, SCID , Neuroblastoma/genetics , Neuroblastoma/pathology , Recombinant Proteins , Remission Induction , Survival Analysis , Treatment Outcome
14.
Mol Ther ; 11(6): 875-88, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15922958

ABSTRACT

A detailed comparison of recombinant adeno-associated viral (rAAV) vectors of serotypes 2, 5, and 8 was performed in mice and nonhuman primates. Differences within the capsid proteins and viral terminal repeats of rAAV-2 and -5 did not significantly influence their ability to transduce murine liver. However, vectors pseudotyped with AAV-8 capsid (rAAV-2/8) mediated transgene expression more rapidly and from lower doses than possible with rAAV-2 and -5, although expression declined from peak values in a distinct dose-dependent manner prior to reaching steady-state levels. Nevertheless, at all time points and vector doses, rAAV-2/8 transgene levels were 17- to 84-fold higher than with rAAV-2 or -5 due to a more rapid conversion of the single-stranded genome to transcriptionally active stable duplex DNA. In nonhuman primates, liver-targeted administration of rAAV-5 and rAAV-2/8 vectors established therapeutic levels of transgene expression. The importance of preexisting serotype immunity was highlighted by the inability to achieve successful transduction in the presence of serotype-specific antibodies, although this impediment was successfully avoided through the use of alternative serotypes. In summary, serotype-specific differences in transduction biology and the appreciation of preexisting immunity will likely influence the selection of the rAAV serotype for future clinical trials.


Subject(s)
Capsid Proteins/immunology , Dependovirus/genetics , Dependovirus/immunology , Genetic Vectors , Liver/metabolism , Transduction, Genetic , Animals , Antibody Formation , DNA, Viral/analysis , Dependovirus/classification , Genetic Therapy , Genetic Vectors/immunology , Hemophilia B/therapy , Hepatocytes/chemistry , Liver/cytology , Macaca mulatta , Male , Mice , Serotyping , Transgenes
15.
Cancer Lett ; 228(1-2): 163-70, 2005 Oct 18.
Article in English | MEDLINE | ID: mdl-15927362

ABSTRACT

Angiogenesis appears to be a fundamental requirement for tumor growth, invasion and metastasis. Evidence also exists to suggest that inhibition of tumor-associated angiogenesis can retard tumor growth and prevent tumor spread. Several naturally occurring angiogenesis inhibitors have been identified, including type I interferons (alpha/beta). These proteins are potent inhibitors of angiogenesis and may also have direct anti-tumor and immunomodulatory effects. Because anti-angiogenic therapy is likely cytostatic, long-term delivery of angiogenesis inhibitors may be required for the successful treatment of cancer. We have, therefore, explored the utility of a gene therapy-mediated approach for the delivery of interferon-beta and tested this approach, both alone and in combination with conventional chemotherapy, in murine models of neuroblastoma.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Interferons/therapeutic use , Neovascularization, Pathologic/drug therapy , Neuroblastoma/drug therapy , Genetic Therapy , Humans , Neuroblastoma/blood supply , Neuroblastoma/therapy
16.
Mol Ther ; 11(2): 300-10, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15668142

ABSTRACT

To inhibit tumor-induced angiogenesis, the VEGF signaling pathway was targeted using AAV vectors encoding a VEGF decoy receptor, a truncated, soluble form of the murine VEGF receptor-2 (tsFlk-1). This approach initially had significant anti-neuroblastoma efficacy in murine xenograft models of local and metastatic disease, but when higher circulating levels of tsFlk-1 were established, tumor growth was more aggressive than even in control mice. Part of the mechanism for this apparent tumor resistance was increased human VEGF expression by the tumor cells. However, further investigation revealed that although a greater amount of VEGF could be bound by higher levels of tsFlk-1, more VEGF also existed in an unbound state and was, therefore, available to support angiogenesis. This novel, tumor-independent mechanism for resistance to antiangiogenic strategies suggests that careful titering of angiogenesis inhibitors may be required to achieve maximal antitumor efficacy and avoid therapy resistance mediated, in part, by ligand bioavailability. This has important implications for therapeutic strategies that use decoy receptors and other agents, such as antibodies, to bind angiogenic factors, in an attempt to inhibit tumor neovascularization.


Subject(s)
Neovascularization, Pathologic , Neuroblastoma/blood supply , Neuroblastoma/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Biological Availability , Cell Line, Tumor , Gene Expression , Humans , Ligands , Mice , Neuroblastoma/genetics , Neuroblastoma/pathology , Solubility , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/genetics , Xenograft Model Antitumor Assays
17.
J Virol Methods ; 121(2): 209-15, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15381358

ABSTRACT

Recombinant vectors based on the recently isolated AAV serotype 8 (rAAV-8) shows great promise for gene therapy, particularly for disorders affecting the liver. Transition of this vector system to the clinic, however, is limited by the lack of an efficient scaleable purification method. In this report, we describe a simple method for purification of rAAV-8 vector particles based on ion exchange chromatography that generates vector stocks with greater than 90% purity. The average yield of purified rAAV-8 from five different vector preparation was 41%. Electron microscopy of these purified stocks revealed typical icosohedral virions with less than 10% empty particles. Liver targeted delivery of ion-exchange purified rAAV-8 vector encoding the human factor IX (hFIX) gene, resulted in plasma hFIX levels approaching 30% of normal in immunocompetent mice, which is 20-fold higher than observed with an equivalent number of rAAV-5 ion exchange purified vector particles. The method takes less then 5 h to process and purify rAAV-8 vector from producer cells and represents a significant advance on the CsCl density centrifugation technique in current use for purification of rAAV-8 vector systems and will likely facilitate the transition of the rAAV-8 vector system to the clinic.


Subject(s)
Dependovirus/isolation & purification , Genetic Vectors/isolation & purification , Animals , Chromatography, Ion Exchange , Dependovirus/genetics , Factor IX/analysis , Factor IX/genetics , Genetic Vectors/administration & dosage , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Recombination, Genetic
18.
Surgery ; 136(2): 183-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15300178

ABSTRACT

PURPOSE: We tested the hypothesis that the antiangiogenic activity of the type I interferons (IFNs), could affect tumor engraftment and growth in murine xenograft models of neuroblastoma. METHODS: Subcutaneous and retroperitoneal human neuroblastoma xenografts were established in SCID mice. Five days after tumor cell inoculation, daily subcutaneous injections of human IFN-alpha at several different doses were initiated and continued for 30 days. The effectiveness of continuous delivery of low-dose interferon was then tested using a gene therapy approach in which an adeno-associated virus vector encoding IFN-beta (rAAV-IFN-beta) was used to mediate expression prior to retroperitoneal tumor implantation. RESULTS: Subcutaneous and retroperitoneal tumors were significantly smaller in IFN-alpha-treated mice, as compared with control mice. Intratumoral basic fibroblast growth factor and vascular endothelial growth factor expression were also decreased, as was mean intratumoral endothelial cell density. Interestingly, the lower doses of IFN-alpha were more effective than the higher dose. No tumors developed in any of the mice given rAAV-IFN-beta, whereas each of the mice that received control vector developed large tumors. CONCLUSIONS: Treatment with IFN had a significant impact on neuroblastoma engraftment and growth in mice, particularly when delivered continuously using a gene therapy approach. This activity appears to be mediated in part by inhibition of tumor-induced angiogenesis through the downregulation of tumor-elaborated factors, including basic fibroblast growth factor and vascular endothelial growth factor.


Subject(s)
Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Neovascularization, Pathologic/prevention & control , Neuroblastoma/blood supply , Animals , Cell Division/drug effects , Cell Line, Tumor , Dependovirus/genetics , Genetic Therapy , Humans , Interferon-beta/genetics , Male , Mice , Mice, SCID , Neoplasm Transplantation , Neuroblastoma/pathology , Neuroblastoma/therapy , Transplantation, Heterologous
19.
J Pediatr Surg ; 39(3): 405-11; discussion 405-11, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15017561

ABSTRACT

PURPOSE: Concomitant resistance, the phenomenon whereby a primary malignancy inhibits the growth of metastatic lesions, is likely caused by the production of endogenous anti-angiogenic factors. The purpose of this study was to evaluate the influence of the angiogenesis inhibitor, endostatin, expressed by primary sites of neuroblastoma, on synchronous disease. METHODS: Two neuroblastoma models were used. In one, the growth of a second primary tumor in mice with an already established primary tumor was compared with tumor growth in nave mice. In the other, the growth of liver metastases arising spontaneously from a subcutaneous tumor was compared in mice in which the primary tumor was either excised or left in place. Systemic endostatin levels and endothelial cell density, vascularity, and degree of apoptosis in the secondary tumors and liver metastasis were evaluated. RESULTS: Subcutaneous tumors in mice with preexisting neuroblastoma were significantly smaller than in mice without an established primary tumor; systemic endostatin levels at the time of tumor implantation in mice with preexisting tumors were nearly 3 times that of nave mice. Decreased angiogenesis and increased apoptosis were seen in the secondary tumors of mice with preexisting tumors. Similarly, the weight of liver metastases was significantly less in mice in which the primary tumor was left in place as compared with those in which the primary tumor had been excised. Systemic endostatin levels in this model paralleled the status of the primary tumor; levels decreased with primary tumor excision but increased when the primary tumor was retained and allowed to grow. Although no difference in microvessel density was seen between groups in the liver metastasis, more tumor cell apoptosis was seen in liver metastasis when the primary tumor was retained. Thus, the presence of an established primary neuroblastoma had a significant inhibitory effect on the growth of secondary disease in both models and was associated with elevated systemic endostatin levels. CONCLUSIONS: Concomitant antitumoral resistance occurred in these experimental murine models of neuroblastoma and appears to be caused, at least in part, by angiogenesis inhibition mediated by endostatin elaborated from primary tumors.


Subject(s)
Angiogenesis Inhibitors/physiology , Endostatins/physiology , Liver Neoplasms/blood supply , Liver Neoplasms/secondary , Neoplasms, Second Primary/blood supply , Neuroblastoma/metabolism , Skin Neoplasms/metabolism , Analysis of Variance , Angiogenesis Inhibitors/biosynthesis , Angiography , Animals , Apoptosis , Cell Line, Tumor , Disease Models, Animal , Endostatins/biosynthesis , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, SCID , Neoplasm Transplantation , Neovascularization, Pathologic , Neuroblastoma/pathology , Neuroblastoma/physiopathology , Skin Neoplasms/pathology , Skin Neoplasms/physiopathology
20.
Blood ; 102(2): 480-8, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12637328

ABSTRACT

A systematic evaluation of the influence of sex on transduction by recombinant adeno-associated viral vector (rAAV) indicated that transgene expression after liver-targeted delivery of vector particles was between 5- to 13-fold higher in male mice compared with female mice, irrespective of the proviral promoter or cDNA and mouse strain. Molecular analysis revealed that the rAAV genome was stably retained in male liver at levels that were 7-fold higher than those observed in females. Further, the sex difference in transduction was observed with AAV-2- and AAV-5-based vectors, which use distinct receptor complexes for infection. In concordance with the differences in AAV transduction, gel shift analysis with nuclear extracts derived from the liver of mice and humans revealed substantially higher binding of host nuclear protein to the rep-binding site (RBS) of AAV inverted terminal repeat (ITR) in males compared with females. Transduction efficiency and binding of nuclear protein to RBS was dramatically reduced in male mice by castration. In contrast, although oophorectomy did not significantly influence rAAV transduction, administration of 5alpha dihydrotestosterone, prior to gene transfer, increased stable hepatocyte gene transfer in females to levels observed in male mice, implying that androgens significantly influence hepatocyte gene transfer. Interestingly, sex did not have a significant effect on AAV gene transfer into nonhepatic tissue, indicating that there are distinct tissue- and sex-specific differences in the mechanisms responsible for efficient transduction with this vector. These results have significant implications for gene therapy of autosomal and acquired disorders affecting the liver.


Subject(s)
Androgens/physiology , Dependovirus/genetics , Genetic Vectors/genetics , Liver/metabolism , Sex Characteristics , Transduction, Genetic , Animals , CHO Cells , Carcinoma, Hepatocellular/pathology , Cricetinae , Cricetulus , Dihydrotestosterone/administration & dosage , Dihydrotestosterone/pharmacology , Drug Implants , Female , Gene Expression Regulation, Viral/drug effects , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Liver/virology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, SCID , Orchiectomy , Organ Specificity , Ovariectomy , Receptors, Virus/physiology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/genetics , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...