Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Plant J ; 62(1): 52-63, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20070569

RESUMO

The self-incompatibility (SI) response occurs widely in flowering plants as a means of preventing self-fertilization. In these self/non-self discrimination systems, plant pistils reject self or genetically related pollen. In the Solanaceae, Plantaginaceae and Rosaceae, pistil-secreted S-RNases enter the pollen tube and function as cytotoxins to specifically arrest self-pollen tube growth. Recent studies have revealed that the S-locus F-box (SLF) protein controls the pollen expression of SI in these families. However, the precise role of SLF remains largely unknown. Here we report that PhSSK1 (Petunia hybrida SLF-interacting Skp1-like1), an equivalent of AhSSK1 of Antirrhinum hispanicum, is expressed specifically in pollen and acts as an adaptor in an SCF(Skp1-Cullin1-F-box)(SLF) complex, indicating that this pollen-specific SSK1-SLF interaction occurs in both Petunia and Antirrhinum, two species from the Solanaceae and Plantaginaceae, respectively. Substantial reduction of PhSSK1 in pollen reduced cross-pollen compatibility (CPC) in the S-RNase-based SI response, suggesting that the pollen S determinant contributes to inhibiting rather than protecting the S-RNase activity, at least in solanaceous plants. Furthermore, our results provide an example that a specific Skp1-like protein other than the known conserved ones can be recruited into a canonical SCF complex as an adaptor.


Assuntos
Endogamia , Petunia/genética , Proteínas de Plantas/metabolismo , Pólen/genética , Ribonucleases/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Sequência de Aminoácidos , Regulação da Expressão Gênica de Plantas , Técnicas de Silenciamento de Genes , Dados de Sequência Molecular , Proteínas de Plantas/genética , Ribonucleases/genética , Proteínas Ligases SKP Culina F-Box/genética , Alinhamento de Sequência
2.
Biochem Soc Trans ; 38(2): 622-6, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20298232

RESUMO

The recent identification of several proteins playing key roles in S-RNase-based gametophytic self-incompatibility has led both to a greater understanding of the molecular biology of this response, as well as to questions regarding the precise mechanism by which compatible pollen tubes are recognized and accepted. A proposed variant SCF(SLF) (where SCF is SSK1/cullin/F-box and SLF is S-locus F-box) ubiquitin ligase complex is thought to play a central role in recognizing and inhibiting non-self S-RNases, but the exact role of ubiquitination remains unclear. How the possible sequestration of non-self S-RNases in a pollen vacuolar compartment can be reconciled with the need for protein interaction between S-RNase and the SCF(SLF) complex needs to be determined. Current work to answer these questions focuses on more precisely defining quantitative protein interactions and subcellular localization of proteins involved in S-RNase-based gametophytic self-incompatibility.


Assuntos
Magnoliopsida/metabolismo , Magnoliopsida/fisiologia , Proteínas de Plantas/metabolismo , Ribonucleases/metabolismo , Cruzamentos Genéticos , Loci Gênicos , Células Germinativas Vegetais/metabolismo , Células Germinativas Vegetais/fisiologia , Endogamia , Magnoliopsida/genética , Modelos Biológicos , Proteínas de Plantas/genética , Proteínas de Plantas/fisiologia , Ligação Proteica/genética , Ligação Proteica/fisiologia , Ribonucleases/genética , Ribonucleases/fisiologia , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas Ligases SKP Culina F-Box/fisiologia , Distribuição Tecidual
3.
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
4.
Ann Surg Oncol ; 15(11): 3259-67, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18726131

RESUMO

BACKGROUND: We have shown that continuous systemic delivery of interferon beta (IFN-beta) remodels dysfunctional tumor vasculature, thereby improving tumor perfusion and enhancing delivery and efficacy of chemotherapeutic drugs. We hypothesized that because of their inherent tumor tropism, neural progenitor cells (NPCs) engineered to express IFN-beta could also effect maturation of tumor vasculature without generating high systemic levels of IFN-beta. METHODS: Mice with luciferase-expressing disseminated human neuroblastoma were divided into four groups of equal tumor burden by bioluminescence imaging: (1) untreated controls; (2) NPC-IFN-beta only; (3) cyclophosphamide (CTX) only; and (4) NPC-IFN-beta in combination with CTX. Two million NPC-IFN-beta cells were administered twice, 7 days apart, starting 21 days after tail vein administration of tumor cells. CTX was administered every 6 days for three doses. Mice were killed at 6 weeks, livers and kidneys weighed, and tumor removed for immunohistochemical staining for endothelial cells (CD34), pericytes (alpha-SMA), apoptosis (TUNEL [terminal deoxynucleotidyl transferase dUTP nick-end labeling]), and diI-labeled NPCs. RESULTS: Fluorescent-labeled NPCs confirmed localization of these cells to tumors. The alpha-SMA/CD34 ratio, a marker for vascular maturation, greatly increased in NPC-IFN-beta-treated tumors compared with controls. Bioluminescent signal from luciferase-expressing tumor cells, reflecting tumor burden, was lower with combination therapy than control or either monotherapy, and combination therapy resulted in far less tumor burden by weight in the kidneys and liver. CONCLUSIONS: Targeted delivery of IFN-beta with NPCs produced low circulating levels of IFN-beta, yet the maturing effect on the tumor vasculature and the enhanced efficacy of adjuvant therapy was maintained. Thus, combination therapy of NPC-IFN-beta with CTX warrants further investigation for the treatment of high-risk neuroblastoma patients.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Antivirais/administração & dosagem , Ciclofosfamida/uso terapêutico , Interferon beta/administração & dosagem , Neuroblastoma/terapia , Neurônios/fisiologia , Células-Tronco/fisiologia , Adenoviridae/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/métodos , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intravenosas , Masculino , Camundongos , Camundongos SCID , Neuroblastoma/genética , Neuroblastoma/patologia , Carga Tumoral
5.
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
6.
Surgery ; 142(2): 185-91, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17689684

RESUMO

BACKGROUND: Bortezomib is a proteasome inhibitor with pleiotropic antitumor activity. Here we investigate the antiangiogenic and antitumor efficacy of bortezomib against neuroblastoma both in vitro and in a murine model of localized and disseminated disease. METHODS: In vitro activity of bortezomib was assessed by evaluating its effect on cell proliferation and cell cycle status. Localized tumor burden was followed with caliper measurements and total-body bioluminescence in mice with disseminated disease. The antiangiogenic activity was evaluated with immunohistochemistry and human vascular endothelial growth factor (VEGF) enzyme-linked immunosorbent assay on tumor protein extracts. RESULTS: Bortezomib treatment resulted in dose and time-dependent decreases in cell proliferation and resulted in cell cycle arrest. In vivo, bortezomib restricted tumor growth in a model of localized disease and decreased bioluminescence in mice with disseminated disease. That decreased bioluminescence reflected decreased tumor burden was confirmed at necropsy by assessing disease in specific organs. In addition, treatment resulted in a decrease in intratumoral vessel counts and reduced tumor VEGF expression. CONCLUSION: Bortezomib shows significant activity against neuroblastoma in vitro, and it inhibits tumor growth and angiogenesis in vivo. These results suggest that clinical studies of bortezomib are warranted for the treatment of this difficult disease.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Pirazinas/farmacologia , Neoplasias de Tecidos Moles/tratamento farmacológico , Animais , Bortezomib , Linhagem Celular Tumoral , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos SCID , Neovascularização Patológica/patologia , Neuroblastoma/patologia , Neoplasias de Tecidos Moles/patologia , Tela Subcutânea , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Laparoendosc Adv Surg Tech A ; 25(9): 763-6, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26115333

RESUMO

BACKGROUND/PURPOSE: Intussusception is the most common cause of bowel obstruction in children from 3 months to 3 years of age. In the absence of peritonitis, initial treatment is either hydrostatic or pneumatic reduction. If these measures fail, operative intervention is required. In nonreducible cases, we propose the use of intraoperative hydrostatic enema to achieve or confirm reduction. In this study we describe a cohort of patients who have undergone laparoscopic-assisted hydrostatic reduction of intussusception (LAHRI). MATERIALS AND METHODS: This is a retrospective cohort study of all patients undergoing LAHRI from the years 2011 to 2013. We performed LAHRI in seven children 4 months to 2 years of age. All patients had ileocolic intussusception that failed initial reduction by radiographic enema. With the patient under general anesthesia, saline enema reduction was facilitated by direct laparoscopic visualization. RESULTS: In 2 of the 7 cases, intussusception reduction was visually confirmed in real time, and only a laparoscopic camera port was required. In 1 patient, the bowel was extensively dilated, requiring mini-laparotomy for visualization. The enema, however, reduced the intussusception without any need for manual reduction. In the remaining 4 cases, minimal laparoscopic manipulation was required after the enema failed to completely reduce the intussusceptum, but enema was used to confirm reduction. No child required bowel resection. CONCLUSIONS: In cases of failed reduction by contrast enema, we have demonstrated LAHRI to be a successful treatment modality. The technique has the advantage of little to no bowel manipulation and has evolved into one performed via a single umbilical port.


Assuntos
Doenças do Íleo/cirurgia , Intussuscepção/cirurgia , Pré-Escolar , Estudos de Coortes , Enema/métodos , Feminino , Humanos , Pressão Hidrostática , Doenças do Íleo/patologia , Lactente , Intussuscepção/patologia , Laparoscopia , Laparotomia/métodos , Masculino , Estudos Retrospectivos
8.
Obes Surg ; 13(1): 66-72, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12630616

RESUMO

BACKGROUND: Upper gastrointestinal (UGI) swallow radiographs following laparoscopic Roux-en-Y gastric bypass (LRYGBP) may detect an obstruction or an anastomotic leak. The aim of our study was to determine the efficacy of routine imaging following LRYGBP. METHODS: Radiograph reports were reviewed for 201 consecutive LRYGBP operations between April 1999 and June 2001. UGI swallow used Gastrografin, static films, fluoroscopic video, and a delayed image at 10 minutes. Mean values with one standard deviation were tested for significance (P < 0.05) using the Mann-Whitney U test statistic. RESULTS: Of 198 available reports, UGI detected jejunal efferent (Roux) limb narrowing (n = 17), partial obstruction (n = 12), anastomotic leak (n = 3), complete bowel obstruction (n = 3), diverticulum (n = 1), hiatal hernia (n = 1), and proximal Roux limb narrowing (n = 1). A normal study was reported in 160 cases (81%). Partial obstruction resolved without intervention. Complete obstruction required re-operation. Compared to 6 patients who developed delayed leaks, early identification of a leak by routine UGI swallow resulted in a shorter hospital stay (mean 7.7 +/- 1.5 days vs 40.2 +/- 12.3 days, P < 0.03). CONCLUSIONS: Early intervention after UGI swallow may lessen morbidity. Routine UGI swallow following LRYGBP does not obviate the importance of close clinical follow-up.


Assuntos
Meios de Contraste , Diatrizoato de Meglumina , Sistema Digestório/diagnóstico por imagem , Derivação Gástrica , Adulto , Idoso , Anastomose em-Y de Roux , Índice de Massa Corporal , Testes Diagnósticos de Rotina , Derivação Gástrica/efeitos adversos , Humanos , Laparoscopia , Tempo de Internação , Pessoa de Meia-Idade , Período Pós-Operatório , Radiografia , Estudos Retrospectivos
9.
Surgery ; 150(3): 497-504, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21878236

RESUMO

BACKGROUND: High-grade glioblastomas have immature, leaky tumor blood vessels that impede the efficacy of adjuvant therapy. We assessed the ability of human interferon (hIFN)-ß delivered locally via gene transfer to effect vascular stabilization in an orthotopic model of glioblastoma xenograft resection. METHODS: Xenografts were established by injecting 3 grade IV glioblastoma cell lines (GBM6-luc, MT330-luc, and SJG2-luc) into the cerebral cortex of nude rats. Tumors underwent subtotal resection, and then had gel foam containing an adeno-associated virus vector encoding either hIFN-ß or green fluorescence protein (control) placed in the resection cavity. The primary endpoint was stabilization of tumor vasculature, as evidenced by CD34, α-SMA, and CA IX staining. Overall survival was a secondary endpoint. RESULTS: hIFN-ß treatment altered the tumor vasculature of GBM6-luc and SJG2-luc xenografts, decreasing the density of endothelial cells, stabilizing vessels with pericytes, and decreasing tumor hypoxia. The mean survival for rats with these neoplasms was not improved, however. In rats with MT330-luc xenografts, hIFN-ß resulted in tumor regression with a 6-month survival of 55% (INF-ß group) and 9% (control group). CONCLUSION: The use of AAV hIFN-ß in our orthotopic model of glioblastoma resection stabilized tumor vasculature and improved survival in rats with MT330 xenografts.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/irrigação sanguínea , Glioblastoma/tratamento farmacológico , Interferon beta/administração & dosagem , Neovascularização Patológica/prevenção & controle , Animais , Biópsia por Agulha , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Circulação Cerebrovascular , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Transferência de Genes , Glioblastoma/mortalidade , Glioblastoma/patologia , Humanos , Imuno-Histoquímica , Infusões Intralesionais , Distribuição Aleatória , Ratos , Valores de Referência , Análise de Sobrevida , Transplante Heterólogo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Pediatr Surg ; 45(6): 1103-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20620303

RESUMO

PURPOSE: Resistance to angiogenesis inhibition can occur through the upregulation of alternative mediators of neovascularization. We used a combination of angiogenesis inhibitors with different mechanisms of action, interferon-beta (IFN-beta) and rapamycin, to target multiple angiogenic pathways to treat neuroblastoma xenografts. METHODS: Subcutaneous and retroperitoneal neuroblastoma xenografts (NB-1691 and SK-N-AS) were used. Continuous delivery of IFN-beta was achieved with adeno-associated virus vector-mediated, liver-targeted gene transfer. Rapamycin was delivered intraperitoneally (5 mg/kg per day). After 2 weeks of treatment, tumor size was measured, and tumor vasculature was evaluated with intravital microscopy and immunohistochemistry. RESULTS: Rapamycin and IFN-beta, alone and in combination, had little effect on tumor cell viability in vitro. In vivo, combination therapy led to fewer intratumoral vessels (69% of control), and the remaining vessels had an altered phenotype, being covered with significantly more pericytes (13x control). Final tumor size was significantly less than controls in all tumor models, with combination therapy having a greater antitumor effect than either monotherapy. CONCLUSION: The combination of IFN-beta and rapamycin altered the vasculature of neuroblastoma xenografts and resulted in significant tumor inhibition. The use of combinations of antiangiogenic agents should be further evaluated for the treatment of neuroblastoma and other solid tumors.


Assuntos
Interferon beta/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Neoplasias Retroperitoneais/tratamento farmacológico , Sirolimo/uso terapêutico , Actinas/metabolismo , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/uso terapêutico , Antígenos CD34/metabolismo , Análise por Conglomerados , Humanos , Imuno-Histoquímica , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/uso terapêutico , Injeções Intraperitoneais , Interferon beta/administração & dosagem , Masculino , Camundongos , Camundongos SCID , Neoplasias Experimentais/irrigação sanguínea , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neuroblastoma/irrigação sanguínea , Neuroblastoma/metabolismo , Neoplasias Retroperitoneais/irrigação sanguínea , Neoplasias Retroperitoneais/metabolismo , Sirolimo/administração & dosagem , Transplante Heterólogo , Células Tumorais Cultivadas
11.
Mol Cancer Ther ; 9(3): 761-71, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20197402

RESUMO

Ionizing radiation is an important component of multimodal therapy for alveolar rhabdomyosarcoma (ARMS). We sought to evaluate the ability of IFN-beta to enhance the activity of ionizing radiation. Rh-30 and Rh-41 ARMS cells were treated with IFN-beta and ionizing radiation to assess synergistic effects in vitro and as orthotopic xenografts in CB17 severe combined immunodeficient mice. In addition to effects on tumor cell proliferation and xenograft growth, changes in the tumor microenvironment including interstitial fluid pressure, perfusion, oxygenation, and cellular histology were assessed. A nonlinear regression model and isobologram analysis indicated that IFN-beta and ionizing radiation affected antitumor synergy in vitro in the Rh-30 cell line; the activity was additive in the Rh-41 cell line. In vivo continuous delivery of IFN-beta affected normalization of the dysfunctional tumor vasculature of both Rh-30 and Rh-41 ARMS xenografts, decreasing tumor interstitial fluid pressure, increasing tumor perfusion (as assessed by contrast-enhanced ultrasonography), and increasing oxygenation. Tumors treated with both IFN-beta and radiation were smaller than control tumors and those treated with radiation or IFN-beta alone. Additionally, treatment with high-dose IFN-beta followed by radiation significantly reduced tumor size compared with radiation treatment followed by IFN-beta. The combination of IFN-beta and ionizing radiation showed synergy against ARMS by sensitizing tumor cells to the cytotoxic effects of ionizing radiation and by altering tumor vasculature, thereby improving oxygenation. Therefore, IFN-beta and ionizing radiation may be an effective combination for treatment of ARMS.


Assuntos
Proliferação de Células/efeitos dos fármacos , Interferon beta/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Rabdomiossarcoma Alveolar/radioterapia , Carga Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos SCID , Neovascularização Patológica/radioterapia , Consumo de Oxigênio/efeitos dos fármacos , Radiação Ionizante , Radiossensibilizantes/farmacologia , Proteínas Recombinantes/farmacologia , Rabdomiossarcoma Alveolar/irrigação sanguínea , Rabdomiossarcoma Alveolar/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Int J Radiat Oncol Biol Phys ; 76(5): 1537-45, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20338480

RESUMO

PURPOSE: Ionizing radiation, an important component of glioma therapy, is critically dependent on tumor oxygenation. However, gliomas are notable for areas of necrosis and hypoxia, which foster radioresistance. We hypothesized that pharmacologic manipulation of the typically dysfunctional tumor vasculature would improve intratumoral oxygenation and, thus, the antiglioma efficacy of ionizing radiation. METHODS AND MATERIALS: Orthotopic U87 xenografts were treated with either continuous interferon-beta (IFN-beta) or bevacizumab, alone, or combined with cranial irradiation (RT). Tumor growth was assessed by quantitative bioluminescence imaging; the tumor vasculature using immunohistochemical staining, and tumor oxygenation using hypoxyprobe staining. RESULTS: Both IFN-beta and bevaziumab profoundly affected the tumor vasculature, albeit with different cellular phenotypes. IFN-beta caused a doubling in the percentage of area of perivascular cell staining, and bevacizumab caused a rapid decrease in the percentage of area of endothelial cell staining. However, both agents increased intratumoral oxygenation, although with bevacizumab, the effect was transient, being lost by 5 days. Administration of IFN-beta or bevacizumab before RT was significantly more effective than any of the three modalities as monotherapy or when RT was administered concomitantly with IFN-beta or bevacizumab or 5 days after bevacizumab. CONCLUSION: Bevacizumab and continuous delivery of IFN-beta each induced significant changes in glioma vascular physiology, improving intratumoral oxygenation and enhancing the antitumor activity of ionizing radiation. Additional investigation into the use and timing of these and other agents that modify the vascular phenotype, combined with RT, is warranted to optimize cytotoxic activity.


Assuntos
Inibidores da Angiogênese/farmacologia , Glioma/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Consumo de Oxigênio/efeitos dos fármacos , Tolerância a Radiação/efeitos dos fármacos , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Bevacizumab , Hipóxia Celular/efeitos dos fármacos , Glioma/metabolismo , Glioma/radioterapia , Interferon beta/farmacologia , Masculino , Camundongos , Camundongos SCID , Transplante Heterólogo
13.
J Pediatr Surg ; 44(1): 204-10; discussion 210-1, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19159744

RESUMO

PURPOSE: Osteoprotegerin (OPG) inhibits osteoclast activation and reduces osteolysis in bone tumors. We hypothesized that tumor-tropic neural progenitor cells (NPCs) engineered to express OPG would reduce neuroblastoma disease burden in the bone. METHODS: Stable expression of green fluorescent protein (NPC-GFP) and OPG (NPC-OPG) was established in human NPCs by lentivirus-mediated transduction. Bone disease was established by intrafemoral injection of luciferase-expressing human neuroblastoma (CHLA-255) cells into 20 SCID mice. Three weeks later, mice began receiving intravenous injection of 2 x 10(6) NPC-OPG or NPC-GFP (control) every 10 days x 3 doses. Disease was monitored with quantitative bioluminescence imaging and x-ray images, which were evaluated on a scale of 0 to 4. These studies were approved by the Institutional Animal Care and Use Committee. RESULTS: Osteoprotegerin treatment in vitro produced no direct toxicity to tumor cells. Coculture of tumor cells with bone marrow significantly increased activation of bone marrow-derived osteoclasts as assessed by tartrate-resistant acid phosphatase staining (156 +/- 10.8 osteoclasts per well) compared to bone marrow culture alone (91.67 +/- 4.7, P = .005). This increase was abrogated by adding OPG-containing media (68.3 +/- 2.8, P = .001). NPC-OPG slowed tumor progression (108-fold increase from pretreatment) compared to mice treated with NPC-GFP (538-fold), as judged by bioluminescence imaging. X-rays subjectively demonstrated less bone disease in NPC-OPG-treated mice (2.27 +/- 0.25) compared to NPC-GFP-treated mice (3.25 +/- 0.22, P = .04). CONCLUSIONS: Neural progenitor cell-mediated delivery of OPG slowed disease progression in a preclinical model of neuroblastoma bone metastasis. The decrease in bone disease was not from direct tumor cell toxicity but likely occurred indirectly through inhibition of osteoclast-directed bone resorption. Thus, targeted delivery of OPG by NPCs may be effective in the treatment of neuroblastoma bone metastasis.


Assuntos
Neoplasias Ósseas/patologia , Metástase Neoplásica/prevenção & controle , Neuroblastoma/patologia , Osteoprotegerina/farmacologia , Células-Tronco/metabolismo , Animais , Linhagem Celular Tumoral , Progressão da Doença , Proteínas de Fluorescência Verde/farmacologia , Humanos , Técnicas In Vitro , Camundongos , Camundongos SCID , Estatísticas não Paramétricas , Transdução Genética
14.
Surgery ; 144(2): 269-75, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18656635

RESUMO

BACKGROUND: We hypothesized that vascular endothelial growth factor (VEGF) contributes to autocrine stimulation of neuroblastoma and that inhibition of its signaling pathway contributes to the anticancer activity of bevacizumab, an anti-VEGF monoclonal antibody. METHODS: For in vitro studies, 2 neuroblastoma cell lines, CHLA-255 and NB1691, were treated with VEGF+/-bevacizumab. For in vivo studies, disseminated neuroblastoma was established by intravenous administration of luciferase-expressing tumor cells in SCID mice prior to bevacizumab treatment. RESULTS: Exogenous VEGF increased cell counts after 48 h (NB1691: 58,878 +/- 8279 vs 137,500 +/- 13,108 cells, P < .001; CHLA: 1.56 x 10(6) +/- 866 vs 1.81 x 10(6) +/- 2550 cells, P <.001); the addition of bevacizumab abrogated this stimulation. In vivo, mice with disseminated disease treated twice weekly with intraperitoneal bevacizumab had a decreased tumor burden at day 14 and prolonged survival (NB1691: 50 +/- 2 vs 43 +/- 2 days, P < .001; CHLA: 53 +/- 3 vs 42 +/- 1 days, P = .006). Interestingly, VEGF and basic fibroblast growth factor expression was increased in treated NB1691 tumors, which likely occurred in response to VEGF signaling inhibition. CONCLUSION: Our results suggest that VEGF has a role in neuroblastoma autocrine signaling. Maintenance therapy with bevacizumab may be useful for disease suppression after maximal cytoreductive therapy; however, upregulation of proangiogenic factors may provide resistance to this approach, which suggests that maximal antitumor efficacy may require combination therapy.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neuroblastoma/patologia , Inibidores da Angiogênese/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Antineoplásicos/farmacologia , Comunicação Autócrina , Bevacizumab , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos SCID , Neuroblastoma/tratamento farmacológico , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
15.
J Pediatr Surg ; 43(1): 177-82; discussion 182-3, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18206478

RESUMO

PURPOSE: Trichostatin A (TSA) is a potent histone deacetylase inhibitor and has demonstrated significant antitumor activity against a variety of cancer cell lines. Type I interferons have also shown significant antitumor as well as antiangiogenic activity. In this study, we examined the effectiveness of combination therapy of TSA and interferon beta (IFN-beta) on human neuroblastoma cells in vitro and in vivo using a murine model of retroperitoneal neuroblastoma. MATERIALS AND METHODS: For in vitro experiments, plated human neuroblastoma cells (NB-1643 and NB-1691) were treated with vehicle or with IFN-beta, TSA, or both for 24 hours. Cytotoxicity was assessed by counting cells and expressing the results as a percentage of controls. Expression of the tumor suppressor p21(Waf1) was assessed by Western blot. For in vivo experiments, retroperitoneal neuroblastomas were established in severe combined immune deficiency (SCID) mice. Interferon beta was given using a gene therapy approach, administering 1.5 x 10(10) particles of an adeno-associated virus vector encoding human IFN-beta (AAV hIFN-beta) via tail vein as a single dose per mouse. Trichostatin A was given at a dose of 5 mg/kg every 48 hours subcutaneously. Treatment groups included controls, AAV hIFN-beta alone, TSA alone, and AAV hIFN-beta together with TSA. Tumor volume was assessed 2 weeks after the treatment began. RESULTS: After 24 hours, treatment with IFN-beta, TSA, and a combination of both resulted in a 45.3%, 68.1%, and 75% reduction in cell count relative to controls in the NB-1691 cell line. In the NB-1643 line, cell counts were reduced by 23%, 58%, and 62.3% respectively. In addition, NB-1691 cells treated with TSA showed increased expression of p21(Waf1) on Western blot. For in vivo experiments, control-, AAV hIFN-beta-, TSA-, and combination-treated tumors had the following final volumes: 1577.7 +/- 264.2 mm(3) (n = 3); 128.5 +/- 74.4 mm(3) (n = 4; P = .0001); 1248.7 +/- 673.9 mm(3) (n = 4; P = .48); and 127.5 +/- 36.8 mm(3) (n = 4; P = .0007), respectively. CONCLUSION: Neuroblastoma, because of its unique biology, continues to be a challenging tumor to treat, and many times these tumors are refractory to standard chemotherapeutic regimens. These data show that both TSA and IFN-beta inhibit neuroblastoma growth and that the combination may potentially provide a unique way to treat this difficult disease.


Assuntos
Terapia Genética/métodos , Ácidos Hidroxâmicos/farmacologia , Interferon beta/farmacologia , Neuroblastoma/terapia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adenoviridae , Animais , Western Blotting , Linhagem Celular Tumoral/citologia , Linhagem Celular Tumoral/efeitos dos fármacos , Terapia Combinada , Modelos Animais de Doenças , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos SCID , Neuroblastoma/patologia , Probabilidade , Distribuição Aleatória , Sensibilidade e Especificidade
16.
J Pediatr Surg ; 43(6): 1159-63, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18558200

RESUMO

PURPOSE: Although occasionally difficult, distinguishing abdominal neuroblastoma (NBL) from Wilms tumor (WT) at presentation is important, as surgical management differs significantly. We reviewed our 20-year experience (1987-2006) treating patients with NBL, focusing on those with an initial diagnosis of WT, to determine presenting features that would have suggested the correct preoperative diagnosis. METHODS: Retrospective case cohort study reviewing charts and imaging of patients with NBL initially diagnosed clinically with WT. Preoperative symptoms, laboratory studies, and imaging were evaluated. Similar variables were assessed in the 20 patients with WT most recently treated at our institution. RESULTS: Nine patients with NBL were identified as those who had an exploratory laparotomy with a preoperative diagnosis of WT; 8 underwent nephrectomy at exploration. Children with NBL had symptoms such as fever and weight loss at presentation (67%) more often than patients with WT (20%). Preoperative computed tomography demonstrated intratumoral calcifications, vascular encasement, or both in 78% of patients with NBL but were never seen in WT patients. Of interest, preoperative urinary catecholamines were elevated in 5 patients ultimately diagnosed with NBL. CONCLUSION: Although NBL can be mistaken for WT at presentation, the presence of constitutional symptoms, or intratumoral calcification or vascular encasement on preoperative imaging should heighten suspicion for NBL. In addition, laboratory evaluation, including urinary catecholamines, should be completed before surgery when the etiology of an abdominal tumor is uncertain.


Assuntos
Erros de Diagnóstico/prevenção & controle , Neoplasias Renais/diagnóstico , Neuroblastoma/diagnóstico , Tumor de Wilms/diagnóstico , Biópsia por Agulha , Quimioterapia Adjuvante , Criança , Pré-Escolar , Estudos de Coortes , Diagnóstico Diferencial , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Imuno-Histoquímica , Neoplasias Renais/mortalidade , Neoplasias Renais/terapia , Laparotomia , Masculino , Estadiamento de Neoplasias , Nefrectomia/métodos , Neuroblastoma/mortalidade , Neuroblastoma/terapia , Cuidados Pré-Operatórios/métodos , Estudos Retrospectivos , Medição de Risco , Análise de Sobrevida , Resultado do Tratamento , Tumor de Wilms/mortalidade , Tumor de Wilms/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA