Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
N Engl J Med ; 371(21): 1994-2004, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25409372

RESUMO

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.).


Assuntos
Fator IX/genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Hemofilia B/terapia , Adulto , Alanina Transaminase/sangue , Dependovirus/genética , Fator IX/metabolismo , Seguimentos , Expressão Gênica , Terapia Genética/efeitos adversos , Hemofilia B/sangue , Hemofilia B/genética , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Transgenes , Adulto Jovem
2.
Blood ; 121(17): 3335-44, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23426947

RESUMO

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.


Assuntos
Dependovirus/genética , Fator VIII/farmacologia , Terapia Genética , Variação Genética/genética , Vetores Genéticos/administração & dosagem , Hemofilia A/terapia , Animais , Western Blotting , Fator VIII/genética , Fator VIII/imunologia , Glicosilação , Hemofilia A/genética , Humanos , Tolerância Imunológica , Fígado/metabolismo , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Regiões Promotoras Genéticas/genética
3.
N Engl J Med ; 365(25): 2357-65, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-22149959

RESUMO

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.).


Assuntos
Dependovirus , Fator IX/genética , Terapia Genética , Vetores Genéticos , Hemofilia B/terapia , Adulto , Dependovirus/genética , Fator IX/uso terapêutico , Terapia Genética/efeitos adversos , Vetores Genéticos/imunologia , Humanos , Infusões Intravenosas , Pessoa de Meia-Idade , Transgenes/imunologia
4.
Mol Ther ; 19(5): 876-85, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21245849

RESUMO

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.


Assuntos
Proteínas do Capsídeo/metabolismo , Dependovirus/genética , Fator IX/metabolismo , Terapia Genética/métodos , Hemofilia B/terapia , Animais , Proteínas do Capsídeo/genética , Fator IX/genética , Expressão Gênica , Vetores Genéticos , Células HEK293 , Hemofilia B/genética , Humanos , Hibridização in Situ Fluorescente , Fígado/metabolismo , Macaca , Camundongos
5.
Mol Cancer Ther ; 7(12): 3852-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19056675

RESUMO

Although temozolomide has shown clinical activity against neuroblastoma, this activity is likely limited by the DNA repair enzyme O6-methylguanine DNA methyltransferase (MGMT). We hypothesized that IFN-beta could sensitize neuroblastoma cells to the cytotoxic effects of temozolomide through its ability to down-regulate MGMT expression. In vitro proliferation of three neuroblastoma cell lines treated with IFN-beta and temozolomide alone or in combination was examined. Antitumor activity was assessed in both localized and disseminated neuroblastoma xenografts using single-agent and combination therapy, with continuous delivery of IFN-beta being established by a liver-targeted adeno-associated virus-mediated approach. Two neuroblastoma cell lines (NB-1691 and SK-N-AS) were found to have high baseline levels of MGMT expression, whereas a third cell line (CHLA-255) had low levels. Temozolomide had little effect on in vitro proliferation of the neuroblastoma cell lines with high MGMT expression, but pretreatment with IFN-beta significantly decreased MGMT expression and cell counts (NB-1691: 36 +/- 3% of control, P = 0.0008; SK-N-AS: 54 +/- 7% control, P = 0.003). In vivo, NB-1691 tumors in CB17-SCID mice treated with the combination of IFN-beta and temozolomide had lower MGMT expression and a significantly reduced tumor burden, both localized [percent initial tumor volume: 2,516 +/- 680% (control) versus 1,272 +/- 330% (temozolomide), P = 0.01; 1,348 +/- 220%, P = 0.03 (IFN-beta); 352 +/- 110%, P = 0.0001 (combo)] and disseminated [bioluminescent signal: control (1.32e10 +/- 6.5e9) versus IFN-beta (2.78e8 +/- 3.09e8), P = 0.025, versus temozolomide (2.06e9 +/- 1.55e9), P = 0.1, versus combination (2.13e7 +/- 7.67e6), P = 0.009]. IFN-beta appears to sensitize neuroblastoma cells to the cytotoxic effects of temozolomide through attenuation of MGMT expression. Thus, IFN-beta and temozolomide may be a useful combination for treating children with this difficult disease.


Assuntos
Antineoplásicos/farmacologia , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Interferon beta/metabolismo , Interleucinas/fisiologia , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Animais , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Humanos , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Temozolomida
6.
Mol Cancer Res ; 5(6): 531-42, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17579115

RESUMO

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.


Assuntos
Interferon beta/administração & dosagem , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neovascularização Patológica , Angiopoietina-1/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Dependovirus/metabolismo , Humanos , Hipóxia , Interferon beta/uso terapêutico , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias
7.
Clin Cancer Res ; 13(13): 3942-50, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17606728

RESUMO

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.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacologia , Anticorpos Monoclonais/farmacologia , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Neuroblastoma/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados , Bevacizumab , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Microcirculação , Transplante de Neoplasias , Neovascularização Patológica , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Fenótipo , Pressão , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Clin Cancer Res ; 23(21): 6441-6449, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28939747

RESUMO

Purpose: Anti-GD2 mAbs, acting via antibody-dependent cell-mediated cytotoxicity, may enhance the effects of chemotherapy. This pilot trial investigated a fixed dose of a unique anti-GD2 mAb, hu14.18K322A, combined with chemotherapy, cytokines, and haploidentical natural killer (NK) cells.Experimental Design: Children with recurrent/refractory neuroblastoma received up to six courses of hu14.18K322A (40 mg/m2/dose, days 2-5), GM-CSF, and IL2 with chemotherapy: cyclophosphamide/topotecan (courses 1,2), irinotecan/temozolomide (courses 3,4), and ifosfamide/carboplatin/etoposide (courses 5,6). Parentally derived NK cells were administered with courses 2, 4, and 6. Serum for pharmacokinetic studies of hu14.18K322A, soluble IL2 receptor alpha (sIL2Rα) levels, and human antihuman antibodies (HAHA) were obtained.Results: Thirteen heavily pretreated patients (9 with prior anti-GD2 therapy) completed 65 courses. One patient developed an unacceptable toxicity (grade 4 thrombocytopenia >35 days). Four patients discontinued treatment for adverse events (hu14.18K322A allergic reaction, viral infection, surgical death, second malignancy). Common toxicities included grade 3/4 myelosuppression (13/13 patients) and grade 1/2 pain (13/13 patients). Eleven patients received 29 NK-cell infusions. The response rate was 61.5% (4 complete responses, 1 very good partial response, 3 partial responses) and five had stable disease. The median time to progression was 274 days (range, 239-568 days); 10 of 13 patients (77%) survived 1 year. Hu14.18K322A pharmacokinetics was not affected by chemotherapy or HAHA. All patients had increased sIL2Rα levels, indicating immune activation.Conclusions: Chemotherapy plus hu14.18K322A, cytokines, and NK cells is feasible and resulted in clinically meaningful responses in patients with refractory/recurrent neuroblastoma. Further studies of this approach are warranted in patients with relapsed and newly diagnosed neuroblastoma. Clin Cancer Res; 23(21); 6441-9. ©2017 AACR.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Terapia Baseada em Transplante de Células e Tecidos , Gangliosídeos/antagonistas & inibidores , Recidiva Local de Neoplasia/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Adolescente , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Carboplatina/administração & dosagem , Criança , Pré-Escolar , Terapia Combinada , Ciclofosfamida/administração & dosagem , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Intervalo Livre de Doença , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/sangue , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/classificação , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Etoposídeo/administração & dosagem , Feminino , Gangliosídeos/imunologia , Humanos , Ifosfamida/administração & dosagem , Lactente , Interleucina-2/sangue , Subunidade alfa de Receptor de Interleucina-2/sangue , Irinotecano , Células Matadoras Naturais/imunologia , Masculino , Recidiva Local de Neoplasia/sangue , Recidiva Local de Neoplasia/patologia , Neuroblastoma/sangue , Neuroblastoma/patologia , Temozolomida , Topotecan/administração & dosagem , Resultado do Tratamento
9.
Surgery ; 140(2): 227-35, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16904974

RESUMO

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.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Difosfonatos/uso terapêutico , Imidazóis/uso terapêutico , Neuroblastoma/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Conservadores da Densidade Óssea/administração & dosagem , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Difosfonatos/administração & dosagem , Relação Dose-Resposta a Droga , Humanos , Imidazóis/administração & dosagem , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neuroblastoma/patologia , Ácido Zoledrônico
10.
Clin Cancer Res ; 11(16): 6020-9, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16115947

RESUMO

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.


Assuntos
Ciclofosfamida/uso terapêutico , Interferon Tipo I/genética , Neuroblastoma/terapia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Ciclofosfamida/farmacologia , Dependovirus/genética , Relação Dose-Resposta a Droga , Terapia Genética/métodos , Vetores Genéticos/genética , Humanos , Interferon Tipo I/metabolismo , Interferon Tipo I/farmacologia , Camundongos , Camundongos SCID , Neuroblastoma/genética , Neuroblastoma/patologia , Proteínas Recombinantes , Indução de Remissão , Análise de Sobrevida , Resultado do Tratamento
11.
Cancer Res ; 62(11): 3077-83, 2002 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12036917

RESUMO

It is now well established that tumor growth is angiogenesis dependent. Inhibition of angiogenesis, therefore, is likely to be an effective anticancer approach. A gene therapy-mediated approach to the delivery of antiangiogenic agents using adeno-associated virus (AAV) vectors has a number of advantages, including the potential for sustained expression. We have constructed a rAAV vector in which the expression of a soluble, truncated form of the vascular endothelial growth factor receptor-2 (Flk-1), a known inhibitor of endothelial cell activation, is driven by a composite beta-actin-based promoter. After intraportal injection of this vector, high-level, stable transgene expression was generated in mice. This established a systemic state of angiogenesis inhibition; sera from these mice inhibited endothelial cell activation in vitro and Matrigel plug neovascularization in vivo. Significant antitumor efficacy was observed in two murine models of pediatric kidney tumors. Tumor development was prevented in 10 of 15 (67%) mice, with significant growth restriction of tumors in the remaining mice. For the first time, long-term, in vivo expression of a functional angiogenesis inhibitor has been established using rAAV, with resultant anticancer efficacy in a relevant, orthotopic tumor model. These findings establish the feasibility of using rAAV vectors in antiangiogenic gene therapy.


Assuntos
Neoplasias Renais/terapia , Fígado/metabolismo , Neovascularização Patológica/terapia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Tumor de Wilms/terapia , Animais , Divisão Celular/genética , Divisão Celular/fisiologia , Dependovirus/genética , Fatores de Crescimento Endotelial/antagonistas & inibidores , Fatores de Crescimento Endotelial/biossíntese , Fatores de Crescimento Endotelial/genética , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Terapia Genética/métodos , Humanos , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/genética , Neoplasias Renais/patologia , Fígado/fisiologia , Linfocinas/antagonistas & inibidores , Linfocinas/biossíntese , Linfocinas/genética , Camundongos , Camundongos SCID , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/biossíntese , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/antagonistas & inibidores , Receptores de Fatores de Crescimento/biossíntese , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento do Endotélio Vascular , Transdução Genética , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Tumor de Wilms/irrigação sanguínea , Tumor de Wilms/genética , Tumor de Wilms/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cancer Lett ; 228(1-2): 163-70, 2005 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-15927362

RESUMO

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.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Interferons/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Terapia Genética , Humanos , Neuroblastoma/irrigação sanguínea , Neuroblastoma/terapia
13.
Cancer Gene Ther ; 10(3): 161-7, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12637936

RESUMO

Destruction and remodeling of the extracellular matrix occurs during the formation of new blood vessels that are required for tumor growth. We sought to determine whether gene-therapy mediated in vivo delivery of tissue inhibitor of matrix metalloproteinase-3 (TIMP-3), using retroviral vector-producer cells, could suppress angiogenesis and subsequent tumor growth in a murine neuroblastoma model. Tumor volume 28 days after coinjection of tumor cells with producer cells generating TIMP-3-encoding retroviral vectors was 21% that of controls, as was the mean tumor vascular index, a measure of blood vessel maturity. When tumors were allowed to reach a mean volume of 0.05 cm(3) before treatment, their size 2 weeks later was 47% relative to controls; larger tumors were not significantly affected. When producer cells were injected at surgical sites following excision of subcutaneous tumors, local recurrence 14 days later was only 22% in TIMP-3 producer cell treated mice as compared to 71% in controls. Unsuccessful transduction of melanoma cells in situ, another tumor of neural crest origin, resulted in unimpaired tumor growth, despite the fact that these tumors are susceptible to TIMP-3 overexpression, demonstrating the importance of tumor cell transduction in this approach. Thus, retroviral vector-producer cell-mediated in vivo gene transfer of TIMP-3 to tumor cells can significantly restrict tumor-induced angiogenesis and tumor growth. This approach may be an effective adjuvant in the treatment of neuroblastoma and other solid tumors refractory to traditional therapy, although it appears to be most effective in smaller tumors or in the setting of minimal residual disease, and the tumor cells must be susceptible to retroviral vector-mediated transduction.


Assuntos
Inibidores da Angiogênese/genética , Vetores Genéticos/administração & dosagem , Neuroblastoma/terapia , Retroviridae/genética , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Animais , Linhagem Celular Tumoral , Matriz Extracelular , Camundongos , Camundongos SCID , Neoplasia Residual , Neovascularização Patológica , Neuroblastoma/irrigação sanguínea , Neuroblastoma/patologia
14.
Surgery ; 136(2): 183-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15300178

RESUMO

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.


Assuntos
Interferon-alfa/farmacologia , Interferon beta/farmacologia , Neovascularização Patológica/prevenção & controle , Neuroblastoma/irrigação sanguínea , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dependovirus/genética , Terapia Genética , Humanos , Interferon beta/genética , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neuroblastoma/patologia , Neuroblastoma/terapia , Transplante Heterólogo
15.
J Virol Methods ; 121(2): 209-15, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15381358

RESUMO

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.


Assuntos
Dependovirus/isolamento & purificação , Vetores Genéticos/isolamento & purificação , Animais , Cromatografia por Troca Iônica , Dependovirus/genética , Fator IX/análise , Fator IX/genética , Vetores Genéticos/administração & dosagem , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Recombinação Genética
16.
J Am Coll Surg ; 199(1): 78-86, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15217634

RESUMO

BACKGROUND: A gene therapy-mediated approach to the delivery of antiangiogenic agents using adeno-associated virus (AAV) vectors has a number of advantages including the potential for sustained expression. The purpose of this study was to attempt to restrict the growth of disseminated neuroblastoma through delivery of a truncated, soluble form of the vascular endothelial growth factor receptor-2 (VEGFR-2 fetal liver kinase [Flk]-1), a decoy receptor for VEGF. STUDY DESIGN: Mice underwent portal vein injection of vector, either AAV-CAGG-tsFlk-1 or control vector. Subsequent truncated soluble fetal liver kinase-1 (tsFlk-1) expression was confirmed and quantified by ELISA. After 8 weeks, mice were given human neuroblastoma cells through the tail vein to establish disseminated disease and were sacrificed after 40 days. The weight of liver metastasis was measured. Intrahepatic tumor microvessel density and levels of apoptosis were determined. Survival was assessed in a second cohort of mice. RESULTS: After intraportal injection of AAV-CAGG-tsFlk-1, high-level, stable transgene expression was generated. Sera from these mice inhibited endothelial cell activation in vitro and Matrigel plug (BD Biosciences) neovascularization in vivo, suggesting that a systemic state of angiogenesis inhibition had been established. After neuroblastoma injection, mice expressing tsFlk-1 had a smaller tumor burden in the liver when sacrificed, as compared with control mice. The decrease in tumor weight in the liver correlated with lower intratumoral microvessel density and higher levels of tumor cell apoptosis in the tsFlk-1-treated tumors, supporting the hypothesis that decreased angiogenesis had occurred. In a second cohort of mice, survival of tsFlk-1-expressing mice after tumor cell challenge was longer than in control mice. CONCLUSIONS: Longterm in vivo expression of a functional VEGF inhibitor was established using an AAV-mediated gene therapy approach, and it demonstrated antiangiogenic and anticancer efficacy in a murine model of disseminated neuroblastoma.


Assuntos
Inibidores da Angiogênese/biossíntese , Técnicas de Transferência de Genes , Neoplasias Hepáticas/terapia , Neuroblastoma/terapia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Linhagem Celular , Dependovirus/genética , Vetores Genéticos/genética , Humanos , Camundongos , Modelos Animais , Metástase Neoplásica , Transplante de Neoplasias , Análise de Sobrevida , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico
17.
J Pediatr Surg ; 48(1): 39-46, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23331791

RESUMO

INTRODUCTION: The anti-tumor activity of angiogenesis inhibitors is often limited by the development of resistance to these drugs. Here we establish HIF-1α as a major factor in the development of this resistance in neuroblastoma xenografts. METHODS: Neuroblastoma xenografts were established by injecting unmodified SKNAS or NB-1691 cells (2 × 10(6) cells), or cells in which HIF-1α expression had been knocked down with shRNA, into the retroperitoneal space of SCID mice. Treatment of established tumors included bevacizumab (5mg/kg q2wk), sunitinib (40 mg/kg qd), or topotecan (0.5mg/kg qd) alone or in combination for a total of two weeks. RESULTS: NB-1691 xenografts showed no difference in relative growth in HIF-1α knockdowns compared to control tumors (73.33 ± 7.90 vs 79.94 ± 6.15, p=0.528). However, HIF-1α knockdowns demonstrated relative final volumes that were significantly lower than unmodified tumors when both were treated with bevacizumab (35.88 ± 4.24 vs 53.57 ± 6.61, p=0.0544) or sunitinib (12.46 ± 2.59 vs 36.36 ± 4.82, p=0.0024). Monotherapy of unmodified xenografts with bevacizumab, sunitinib, or topotecan was largely ineffective. Relative final volumes of NB-1691 xenografts were significantly less in cohorts treated with sunitinib+topotecan (4.78 ± 0.77 vs 39.17 ± 2.44 [sunitinib alone], p=0.011) and bevacizumab+topotecan (13.63 ± 1.55 vs 48.16 ± 9.94 [bevacizumab alone], p=0.014). CONCLUSION: Upregulation of HIF-1α appears to be a significant mechanism of resistance to antiangiogenic therapies in neuroblastoma. Suppressing HIF-1α with low-dose topotecan potentiates the effects of the antiangiogenic drugs in a mouse model.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neuroblastoma/tratamento farmacológico , Inibidores da Angiogênese/administração & dosagem , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Bevacizumab , Linhagem Celular Tumoral , Esquema de Medicação , Humanos , Indóis/administração & dosagem , Injeções Intraperitoneais , Camundongos , Camundongos SCID , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Pirróis/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sunitinibe , Inibidores da Topoisomerase I/administração & dosagem , Topotecan/administração & dosagem , Resultado do Tratamento , Carga Tumoral
18.
J Pediatr Surg ; 48(1): 47-55, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23331792

RESUMO

PURPOSE: Osteoprotegerin (OPG) is a decoy receptor for the Receptor of NF-κB (RANK) ligand that can inhibit osteoclastogenesis. Previous studies have suggested that Mammalian Target of Rapamycin (mTOR) inhibition upregulates OPG production. We tested the hypothesis that the mTOR inhibitor rapamycin could inhibit neuroblastoma bone metastases through its action on OPG. EXPERIMENTAL DESIGN: An orthotopic model of bone metastasis was established. Mice with established disease were subsequently treated with rapamycin (5mg/kg IP daily) or vehicle control (DMSO 1:1000). X-rays were obtained twice a week to detect pathologic fractures. Serum OPG levels were measured by ELISA after two weeks of treatment. RESULTS: Mice with bone disease receiving rapamycin had increased serum levels of OPG in the CHLA-20 mice compared to controls (36.89 pg/mL ± 3.90 vs 18.4 pg/mL ± 1.67, p=0.004) and NB1691 tumor-bearing groups (46.03 ± 2.67 pg/mL vs 17.96 ± 1.84pg/mL, p=0.001), and a significantly longer median time to pathologic fractures with CHLA-20 (103 days vs 74.5 days, p=0.014) and NB1691 xenografts. CONCLUSION: In a xenograft model, increased OPG expression correlated with a delay to pathologic fracture suggesting a potential role for mTOR inhibitors in the treatment of neuroblastoma bone metastases.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Neuroblastoma/tratamento farmacológico , Neuroblastoma/secundário , Osteoprotegerina/sangue , Sirolimo/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores/sangue , Neoplasias Ósseas/sangue , Neoplasias Ósseas/complicações , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Esquema de Medicação , Ensaio de Imunoadsorção Enzimática , Fraturas Espontâneas/etiologia , Fraturas Espontâneas/prevenção & controle , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos SCID , Neuroblastoma/sangue , Neuroblastoma/complicações , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Resultado do Tratamento
19.
PLoS One ; 8(2): e51309, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23408929

RESUMO

Ionizing radiation (IR) is an essential component of therapy for alveolar rhabdomyosarcoma. Nuclear factor-kappaB (NF-κΒ) transcription factors are upregulated by IR and have been implicated in radioresistance. We evaluated the ability of curcumin, a putative NF-κΒ inhibitor, and cells expressing genetic NF- κΒ inhibitors (IκBα and p100 super-repressor constructs) to function as a radiosensitizer. Ionizing radiation induced NF-κΒ activity in the ARMS cells in vitro in a dose- and time-dependent manner, and upregulated expression of NF-κΒ target proteins. Pretreatment of the cells with curcumin inhibited radiation-induced NF-κΒ activity and target protein expression. In vivo, the combination of curcumin and IR had synergistic antitumor activity against Rh30 and Rh41 ARMS xenografts. The greatest effect occurred when tumor-bearing mice were treated with curcumin prior to IR. Immunohistochemistry revealed that combination therapy significantly decreased tumor cell proliferation and endothelial cell count, and increased tumor cell apoptosis. Stable expression of the super-repressor, SR-IκBα, that blocks the classical NF-κB pathway, increased sensitivity to IR, while expression of SR-p100, that blocks the alternative pathway, did not. Our results demonstrate that curcumin can potentiate the antitumor activity of IR in ARMS xenografts by suppressing a classical NF-κΒ activation pathway induced by ionizing radiation. These data support testing of curcumin as a radiosensitizer for the clinical treatment of alveolar rhabdomyosarcoma. IMPACT OF WORK: The NF-κΒ protein complex has been linked to radioresistance in several cancers. In this study, we have demonstrated that inhibiting radiation-induced NF-κΒ activity by either pharmacologic (curcumin) or genetic (SR-IκBα) means significantly enhanced the efficacy of radiation therapy in the treatment of alveolar rhabdomyosarcoma cells and xenografts. These data suggest that preventing the radiation-induced activation of the NF-κΒ pathway is a promising way to improve the antitumor efficacy of ionizing radiation and warrants clinical trials.


Assuntos
Curcumina/farmacologia , NF-kappa B/metabolismo , Tolerância a Radiação , Radiação Ionizante , Rabdomiossarcoma Alveolar/patologia , Animais , Apoptose , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Imuno-Histoquímica , Camundongos , Rabdomiossarcoma Alveolar/irrigação sanguínea , Rabdomiossarcoma Alveolar/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Pediatr Surg ; 47(1): 183-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22244414

RESUMO

PURPOSE: Rapamycin inhibits vascular endothelial growth factor expression. Vascular endothelial growth factor is a tumor-elaborated protein that stimulates neovascularization. This inhibition can cause transient "normalization" of the generally dysfunctional tumor vasculature, resulting in improved tumor perfusion and oxygenation. We hypothesized that this may potentiate the antitumor effects of adjuvant ionizing radiation. METHODS: Mice bearing orthotopic Rh30 alveolar rhabdomyosarcomas were treated with rapamycin (5 mg/kg intraperitoneally daily ×5). Tumors were then evaluated for changes in intratumoral oxygenation, perfusion, vessel permeability, and microvessel density. Additional tumor-bearing mice were treated with 5 doses of rapamycin, irradiation (4 Gy), or 5 doses of rapamycin with irradiation administered on the first or sixth day of rapamycin treatment. RESULTS: Although tumor vessel permeability changed only minimally, microvessel density decreased (3153 ± 932 vs 20,477 ± 3717.9 pixels per high-power field), whereas intratumoral oxygenation increased significantly (0.0385 ± 0.0141 vs 0.0043 ± 0.0023 mm Hg/mm(3)) after 5 doses of rapamycin. Contrast-enhanced ultrasound demonstrated a significantly increased rate of change of signal intensity after 5 days of rapamycin, suggesting improved intratumoral perfusion. Tumor volume 14 days after treatment was smallest in mice treated with the combination of rapamycin given before irradiation. CONCLUSION: Combination therapy with rapamycin given before irradiation to normalize the tumor vasculature, thereby improving tumor oxygenation, increased the sensitivity of alveolar rhabdomyosarcoma xenografts to adjuvant irradiation.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Rabdomiossarcoma Alveolar/tratamento farmacológico , Rabdomiossarcoma Alveolar/radioterapia , Sirolimo/uso terapêutico , Animais , Terapia Combinada , Camundongos , Camundongos SCID , Transplante de Neoplasias , Radiação Ionizante , Rabdomiossarcoma Alveolar/irrigação sanguínea , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA