Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Angiogenesis ; 25(1): 71-86, 2022 02.
Article in English | MEDLINE | ID: mdl-34170441

ABSTRACT

Aberrant variations in angiogenesis have been observed in tumor tissues with abnormal stiffness of extracellular matrix (ECM). However, it remains largely unclear how ECM stiffness influences tumor angiogenesis. Numerous studies have reported that vascular endothelial growth factor-A (VEGF-A) released from tumor cells plays crucial roles in angiogenesis. Hence, we demonstrated the role of ECM stiffness in VEGF-A release from neuroblastoma (NB) cells and the underlying mechanisms. Based on 17 NB clinical samples, a negative correlation was observed between the length of blood vessels and stiffness of NB tissues. In vitro, an ECM stiffness of 30 kPa repressed the secretion of VEGF165 from NB cells which subsequently inhibited the tube formation of human umbilical vein endothelial cells (HUVECs). Knocked down VEGF165 in NB cells or blocked VEGF165 with neutralizing antibodies both repressed the tube formation of HUVECs. Specifically, 30 kPa ECM stiffness repressed the expression and nuclear accumulation of Yes-associated protein (YAP) to regulate the expression of Serine/Arginine Splicing Factor 1 (SRSF1) via Runt-related transcription factor 2 (RUNX2), which may then subsequently induce the expression and secretion of VEGF165 in NB tumor cells. Through implantation of 3D col-Tgels with different stiffness into nude mice, the inhibitory effect of 30 kPa on NB angiogenesis was confirmed in vivo. Furthermore, we found that the inhibitory effect of 30 kPa stiffness on NB angiogenesis was reversed by YAP overexpression, suggesting the important role of YAP in NB angiogenesis regulated by ECM stiffness. Overall, our work not only showed a regulatory effect of ECM stiffness on NB angiogenesis, but also revealed a new signaling axis, YAP-RUNX2-SRSF1, that mediates angiogenesis by regulating the expression and secretion of VEGF165 from NB cells. ECM stiffness and the potential molecules revealed in the present study may be new therapeutic targets for NB angiogenesis.


Subject(s)
Cell Cycle Proteins/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Neovascularization, Pathologic/metabolism , Neuroblastoma , Serine-Arginine Splicing Factors/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Core Binding Factor Alpha 1 Subunit/genetics , Extracellular Matrix , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Neuroblastoma/blood supply , Vascular Endothelial Growth Factor A/genetics
2.
Cell Tissue Res ; 372(2): 269-275, 2018 05.
Article in English | MEDLINE | ID: mdl-29032465

ABSTRACT

Hypoxia (i.e., low oxygen levels) is a known feature of aggressive tumors. Cells, including tumor cells, respond to conditions of insufficient oxygen by activating a transcriptional program mainly driven by hypoxia-inducible factors (HIF)-1 and HIF-2. Both HIF-1α and HIF-2α expression levels have been shown to correlate to patient outcome in various tumor forms and in neuroblastoma, a solid childhood tumor of the sympathetic nervous system, in particular, HIF-2α marks a subpopulation of immature neural crest-like perivascularly located cells and associates with aggressive disease and distant metastasis. It has for long been recognized that the HIF-α subunits are oxygen-dependently regulated at the post-translational level, via ubiquitination and proteasomal degradation. Evidence of oxygen-independent mechanisms of regulation, transcriptional control of EPAS1/HIF2A and possible cytoplasmic activities of HIF-2α has also emerged during recent years. In this review, we discuss these non-conventional actions of HIF-2α, its putative role as a therapeutic target and the constraints it carries, as well as the importance of HIF-2 activity in a vascularized setting, the so-called pseudo-hypoxic state.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Animals , Cell Hypoxia , Humans , Neovascularization, Pathologic/metabolism , Neuroblastoma/blood supply , Neuroblastoma/genetics , Oxygen/metabolism , Treatment Outcome
3.
Bull Math Biol ; 80(8): 2026-2048, 2018 08.
Article in English | MEDLINE | ID: mdl-29948886

ABSTRACT

Neuroblastoma is the leading cause of cancer death in young children. Although treatment for neuroblastoma has improved, the 5-year survival rate of patients still remains less than half. Recent studies have indicated that bevacizumab, an anti-VEGF drug used in treatment of several other cancer types, may be effective for treating neuroblastoma as well. However, its effect on neuroblastoma has not been well characterized. While traditional experiments are costly and time-consuming, mathematical models are capable of simulating complex systems quickly and inexpensively. In this study, we present a model of vascular tumor growth of neuroblastoma IMR-32 that is complex enough to replicate experimental data across a range of tumor cell properties measured in a suite of in vitro and in vivo experiments. The model provides quantitative insight into tumor vasculature, predicting a linear relationship between vasculature and tumor volume. The tumor growth model was coupled with known pharmacokinetics and pharmacodynamics of the VEGF blocker bevacizumab to study its effect on neuroblastoma growth dynamics. The results of our model suggest that total administered bevacizumab concentration per week, as opposed to dosage regimen, is the major determining factor in tumor suppression. Our model also establishes an exponentially decreasing relationship between administered bevacizumab concentration and tumor growth rate.


Subject(s)
Bevacizumab/therapeutic use , Models, Biological , Neuroblastoma/pathology , Neuroblastoma/therapy , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Bevacizumab/administration & dosage , Bevacizumab/pharmacokinetics , Cell Line, Tumor , Humans , Mathematical Concepts , Mice , Neuroblastoma/blood supply , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays
5.
Invest New Drugs ; 34(6): 685-692, 2016 12.
Article in English | MEDLINE | ID: mdl-27586230

ABSTRACT

Background High-risk neuroblastoma has poor outcomes with high rates of relapse despite aggressive treatment, and novel therapies are needed to improve these outcomes. Ponatinib is a multi-tyrosine kinase inhibitor that targets many pathways implicated in neuroblastoma pathogenesis. We hypothesized that ponatinib would be effective against neuroblastoma in preclinical models. Methods We evaluated the effects of ponatinib on survival and migration of human neuroblastoma cells in vitro. Using orthotopic xenograft mouse models of human neuroblastoma, we analyzed tumors treated with ponatinib for growth, gross and histologic appearance, and vascularity. Results Ponatinib treatment of neuroblastoma cells resulted in decreased cell viability and migration in vitro. In mice with orthotopic xenograft neuroblastoma tumors, treatment with ponatinib resulted in decreased growth and vascularity. Conclusions Ponatinib reduces neuroblastoma cell viability in vitro and reduces tumor growth and vascularity in vivo. The antitumor effects of ponatinib suggest its potential as a novel therapeutic agent for neuroblastoma, and further preclinical testing is warranted.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Movement/drug effects , Imidazoles/pharmacology , Neovascularization, Pathologic/prevention & control , Neuroblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Mice , Mice, Nude , Neuroblastoma/blood supply , Neuroblastoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
J Neurosurg Sci ; 58(2): 113-6, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24819488

ABSTRACT

AIM: The surgical resection rate in childhood neuroblastoma (Stage III and IV) is relatively low and influences the prognosis greatly. This study analyzes the primary resection rate neuroblastoma in children. METHODS: The tumors are shrunk with pre-operative chemotherapy and surgical resection beginning from the iliac vessels is performed to skeletalize the large retroperitoneal vascular. Using this method, 22 cases of childhood neuroblastoma received resections and the outcomes were analyzed. RESULTS: The tumors were completely removed in 21 out of the 22 cases (95.45% in total). There were no serious complications and perioperative deaths. CONCLUSION: Using proper surgical methods and skeletalizing the large retroperitoneal vessels significantly increases the resection rate.


Subject(s)
Adrenal Gland Neoplasms , Chemotherapy, Adjuvant/methods , Adrenal Gland Neoplasms/blood supply , Adrenal Gland Neoplasms/drug therapy , Adrenal Gland Neoplasms/surgery , Celiac Artery/surgery , Child , Child, Preschool , Feasibility Studies , Female , Ganglia, Sympathetic/surgery , Humans , Iliac Artery/surgery , Infant , Male , Neuroblastoma/blood supply , Neuroblastoma/drug therapy , Neuroblastoma/surgery , Preoperative Care/methods , Retroperitoneal Space/blood supply , Treatment Outcome
7.
Angiogenesis ; 16(1): 85-100, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22956186

ABSTRACT

Despite existing aggressive treatment modalities, the prognosis for advanced stage neuroblastoma remains poor with significant long-term illness in disease survivors. Advance stage disease features are associated with tumor vascularity, and as such, angiogenesis inhibitors may prove useful along with current therapies. The matricellular protein, secreted protein acidic and rich in cysteine (SPARC), is known to inhibit proliferation and migration of endothelial cells stimulated by growth factors. Here, we sought to determine the effect of SPARC on neuroblastoma tumor cell-induced angiogenesis and to decipher the molecular mechanisms involved in angiogenesis inhibition. Conditioned medium from SPARC-overexpressed neuroblastoma cells (pSPARC-CM) inhibited endothelial tube formation, cell proliferation, induced programmed cell death and suppressed expression of pro-angiogenic molecules such as VEGF, FGF, PDGF, and MMP-9 in endothelial cells. Further analyses revealed that pSPARC-CM-suppressed expression of growth factors was mediated by inhibition of the Notch signaling pathway, and cells cultured on conditioned medium from tumor cells that overexpress both Notch intracellular domain (NICD-CM) and SPARC resumed the pSPARC-CM-suppressed capillary tube formation and growth factor expression in vitro. Further, SPARC overexpression in neuroblastoma cells inhibited neo-vascularization in vivo in a mouse dorsal air sac model. Furthermore, SPARC overexpression-induced endothelial cell death was observed by co-localization studies with TUNEL assay and an endothelial marker, CD31, in xenograft tumor sections from SPARC-overexpressed mice. Our data collectively suggest that SPARC overexpression induces endothelial cell apoptosis and inhibits angiogenesis both in vitro and in vivo.


Subject(s)
Neovascularization, Pathologic/metabolism , Neuroblastoma/blood supply , Neuroblastoma/metabolism , Osteonectin/metabolism , Receptors, Notch/metabolism , Signal Transduction , Angiogenesis Inducing Agents/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Mice , Mice, Nude , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
Br J Cancer ; 108(12): 2485-94, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23695022

ABSTRACT

BACKGROUND: The use of ß-blockers for the management of hypertension has been recently associated with significant clinical benefits in cancer patients. Herein, we investigated whether ß-blockers could be used in combination with chemotherapy for the treatment of neuroblastoma. METHODS: Seven ß-blockers were tested for their antiproliferative and anti-angiogenic properties alone, and in combination with chemotherapy in vitro; the most potent drug combinations were evaluated in vivo in the TH-MYCN mouse model of neuroblastoma. RESULTS: Three ß-blockers (i.e., carvedilol, nebivolol and propranolol) exhibited potent anticancer properties in vitro and interacted synergistically with vincristine, independently of P-glycoprotein expression. ß-blockers potentiated the anti-angiogenic, antimitochondrial, antimitotic and ultimately pro-apoptotic effects of vincristine. In vivo, ß-blockers alone transiently slowed tumour growth as compared with vehicle only (P<0.01). More importantly, when used in combination, ß-blockers significantly increased the tumour regression induced by vincristine (P<0.05). This effect was associated with an increase in tumour angiogenesis inhibition (P<0.001) and ultimately resulted in a four-fold increase in median survival, as compared with vincristine alone (P<0.01). CONCLUSION: ß-blockers can increase treatment efficacy against neuroblastoma, and their combination with chemotherapy may prove beneficial for the treatment of this disease and other drug-refractory cancers.


Subject(s)
Abdominal Neoplasms/drug therapy , Adrenergic beta-Antagonists/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neuroblastoma/drug therapy , Abdominal Neoplasms/blood supply , Abdominal Neoplasms/pathology , Angiogenesis Inhibitors/administration & dosage , Animals , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Humans , Mice , Mice, Transgenic , Neovascularization, Pathologic/drug therapy , Neuroblastoma/blood supply , Neuroblastoma/pathology
9.
Am J Pathol ; 180(4): 1370-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22285670

ABSTRACT

The influences of cytotoxic drugs on endothelial cells remain incompletely understood. Herein, we examined the effects of chemotherapeutic agents in experimental angiogenesis models and analyzed vessel densities in clinical neuroblastoma tumor samples. Cisplatin (20 to 500 ng/mL), doxorubicin (4 to 100 ng/mL), and vincristine (0.5 to 4 ng/mL), drugs commonly involved in neuroblastoma therapy protocols, induced pro-angiogenic effects in different angiogenesis models. They enhanced endothelial cell tube formation, endothelial cell sprouting from spheroids, formation of tip cells in the sprouting assay, expression of αvß3 integrin, and vitronectin binding. All three drugs increased global cellular kinase phosphorylation levels, including the angiogenesis-relevant molecules protein kinase Cß and Akt. Pharmacological inhibition of protein kinase Cß or Akt upstream of phosphatidylinositol 3-kinase reduced chemotherapy-induced endothelial cell tube formation. Moreover, the investigated chemotherapeutics dose dependently induced vessel formation in the chick chorioallantoic membrane assay. Tumor samples from seven high-risk patients with neuroblastoma were analyzed for vessel density by IHC. Results revealed that neuroblastoma samples taken after chemotherapy consistently showed an enhanced microvessel density compared with the corresponding samples taken before chemotherapy. In conclusion, our data show that chemotherapy can activate endothelial cells by inducing multiple pro-angiogenic signaling pathways and exert pro-angiogenic effects in vitro and in vivo. Moreover, we report a previously unrecognized clinical phenomenon that might, in part, be explained by our experimental observations: chemotherapy-associated enhanced vessel formation in tumors from patients with neuroblastoma.


Subject(s)
Antineoplastic Agents/adverse effects , Neovascularization, Pathologic/chemically induced , Neuroblastoma/blood supply , Animals , Antineoplastic Agents/pharmacology , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Cisplatin/adverse effects , Cisplatin/pharmacology , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Endothelial Cells/drug effects , Humans , Neovascularization, Pathologic/pathology , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Signal Transduction/drug effects , Tumor Cells, Cultured , Vincristine/adverse effects , Vincristine/pharmacology
10.
Biochem Biophys Res Commun ; 422(2): 339-43, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22580000

ABSTRACT

Lysophosphatidic acid (LPA) receptors belong to G protein-coupled transmembrane receptors (LPA receptors; LPA(1) to LPA(6)). They indicate a variety of cellular response by the interaction with LPA, including cell proliferation, migration and differentiation. Recently, we have reported that constitutive active mutated LPA(1) induced the strong biological effects of rat neuroblastoma B103 cells. In the present study, we examined the effects of mutated LPA(1) on the interaction between B103 cells and endothelial F-2 cells. Each LPA receptor expressing B103 cells were maintained in serum-free DMEM and cell motility assay was performed with a Cell Culture Insert. When F-2 cells were cultured with conditioned medium from Lpar1 and Lpar3-expressing cells, the cell motility of F-2 cells was significantly higher than control cells. Interestingly, the motile activity of F-2 cells was strongly induced by mutated LPA(1) than other cells, correlating with the expression levels of vascular endothelial growth factor (Vegf)-A and Vegf-C. Pretreatment of LPA signaling inhibitors inhibited F-2 cell motility stimulated by mutated LPA(1). These results suggest that activation of LPA signaling via mutated LPA(1) may play an important role in the promotion of angiogenesis in rat neuroblastoma cells.


Subject(s)
Cell Movement , Endothelium, Vascular/pathology , Neovascularization, Pathologic/pathology , Neuroblastoma/blood supply , Receptors, Lysophosphatidic Acid/metabolism , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Mutation , Neovascularization, Pathologic/genetics , Rats , Receptors, Lysophosphatidic Acid/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor C/genetics
11.
Histopathology ; 60(6): 911-23, 2012 May.
Article in English | MEDLINE | ID: mdl-22372545

ABSTRACT

AIMS: As new biomarkers are urgently needed to identify children with high-risk neuroblastoma (NB), we studied the contribution of angiogenin (ANG) to angiogenesis and its association with clinicopathological and biological features and patient outcome in NB. METHODS AND RESULTS: Ninety NBs and 12 ganglioneuromas (GNs) were immunostained for ANG and CD31. ANG expression in NB tumoral cells (ANG scores) and vessels [ANG microvascular density (MVD)] and total MVD (CD31 MVD) were determined. The ANG score was significantly greater in NBs than in GNs (P = 0.015) and in NBs from children with stage 4 tumours, high-risk disease, unfavourable pathology (P < 0.001 for each), MYCN amplification (P = 0.003), and 1p deletion (P = 0.002). ANG scores correlated with ANG MVD and CD31 MVD (P < 0.001 for each). Total ANG and CD31 protein levels, measured with a sensitive enzyme-linked immunosorbent assay, were highly correlated (P = 0.003). High ANG scores were associated with decreased overall and event-free survival (log-rank test, P = 0.025 and P = 0.018, respectively). High ANG MVD was associated with decreased overall and event-free survival (log-rank test, P = 0.009 and P = 0.026, respectively). High CD31 MVD was associated with decreased event-free survival (P = 0.045). CONCLUSIONS: The strong correlation of ANG up-regulation with total MVD and adverse clinicopathological and biological factors indicates that ANG supports growth and progression in NB.


Subject(s)
Abdominal Neoplasms/diagnosis , Angiogenesis Inducing Agents/metabolism , Neuroblastoma/diagnosis , Ribonuclease, Pancreatic/metabolism , Up-Regulation , Abdominal Neoplasms/blood supply , Abdominal Neoplasms/mortality , Adolescent , Biomarkers, Tumor/metabolism , Child , Child, Preschool , Ganglioneuroma/blood supply , Ganglioneuroma/diagnosis , Ganglioneuroma/mortality , Humans , Infant , Infant, Newborn , Microvessels/metabolism , Microvessels/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neuroblastoma/blood supply , Neuroblastoma/mortality , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Prognosis , Survival Rate , United Kingdom/epidemiology
12.
Pediatr Res ; 71(2): 168-78, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22258128

ABSTRACT

INTRODUCTION: We investigated the use of dietary omega-3 (ω-3) polyunsaturated fatty acids (PUFAs) in the treatment of neuroblastoma both as a sole agent and in combination with sunitinib, a broad-spectrum tyrosine kinase receptor inhibitor. RESULTS: Substitution of all dietary fat with menhaden oil (ω-3 PUFA rich) resulted in a 40-70% inhibition of tumor growth and a statistically significant difference in the levels of several PUFAs (18:2 ω-6, 20:4 ω-6, 22:4 ω-6, 20:5 ω-3) as compared with a control diet. Furthermore, tumors from animals on the ω-3 fatty acid (FA)-enriched diet had an elevated triene/tetraene ratio suggestive of a change in local eicosanoid metabolism in these tissues similar to that seen with essential fatty acid deficiency. The ω-3 FA-enriched diet also decreased tumor-associated inflammatory cells and induced mitochondrial changes suggestive of mitochondrial damage. Combination treatment with sunitinib resulted in further reduction in tumor proliferation and microvessel density. DISCUSSION: These findings suggest a potential role for ω-3 PUFAs in the combination treatment of neuroblastoma. METHODS: We used a murine model of orthotopic and subcutaneous human neuroblastoma and diets that differ in the FA content to define the optimal dietary ω-3/omega-6 (ω-6) FA ratio required for the inhibition of these tumors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Proliferation/drug effects , Diet , Fatty Acids, Omega-3/administration & dosage , Fish Oils/administration & dosage , Indoles/pharmacology , Neuroblastoma/diet therapy , Neuroblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Animals , Arachidonic Acid/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Fatty Acids, Omega-3/metabolism , Fish Oils/metabolism , Humans , Lipid Metabolism , Male , Mice , Mice, SCID , Microvessels/drug effects , Microvessels/pathology , Mitochondria/drug effects , Mitochondria/pathology , Neuroblastoma/blood supply , Neuroblastoma/enzymology , Neuroblastoma/pathology , Sunitinib , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Pediatr Blood Cancer ; 59(4): 642-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22147414

ABSTRACT

BACKGROUND: More effective therapy for children with high-risk neuroblastoma is desperately needed. Preclinical studies have shown that neuroblastoma tumor growth can be inhibited by agents that block angiogenesis. We hypothesized that drugs which target both neuroblastoma cells and tumor angiogenesis would have potent anti-tumor activity. In this study we tested the effects of sorafenib, a multi-kinase inhibitor, on neuroblastoma cell proliferation and signaling, and in mice with subcutaneous human neuroblastoma xenografts or orthotopic adrenal tumors. PROCEDURE: Mice with subcutaneous neuroblastoma xenografts or orthotopic adrenal tumors were treated with sorafenib, and tumor growth rates were measured. Blood vessel architecture and vascular density were evaluated histologically in treated and control neuroblastoma tumors. The in vitro effects of sorafenib on neuroblastoma proliferation, cell cycle, and signaling were also evaluated. RESULTS: Sorafenib inhibited tumor growth in mice with subcutaneous and orthotopic adrenal tumors. Decreased numbers of cycling neuroblastoma cells and tumor blood vessels were seen in treated versus control tumors, and the blood vessels in the treated tumors had more normal architecture. Sorafenib treatment also decreased neuroblastoma cell proliferation, attenuated ERK signaling, and enhanced G(1) /G(0) cell cycle arrest in vitro. CONCLUSIONS: Our results demonstrate that sorafenib inhibits the growth of neuroblastoma tumors by targeting both neuroblastoma cells and tumor blood vessels. Single agent sorafenib should be evaluated in future phase II neuroblastoma studies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacology , Cell Proliferation/drug effects , MAP Kinase Signaling System/drug effects , Neuroblastoma/pathology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Female , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Neuroblastoma/blood supply , Neuroblastoma/physiopathology , Niacinamide/analogs & derivatives , Phenylurea Compounds , Sorafenib
14.
Cancer Cell ; 6(4): 347-60, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15488758

ABSTRACT

We identify a novel alternative TrkA splice variant, TrkAIII, with deletion of exons 6, 7, and 9 and functional extracellular IG-C1 and N-glycosylation domains, that exhibits expression restricted to undifferentiated early neural progenitors, human neuroblastomas (NBs), and a subset of other neural crest-derived tumors. This NGF-unresponsive isoform is oncogenic in NIH3T3 cells and promotes tumorigenic NB cell behavior in vitro and in vivo (cell survival, xenograft growth, angiogenesis) resulting from spontaneous tyrosine kinase activity and IP3K/Akt/NF-kappaB but not Ras/MAPK signaling. TrkAIII antagonizes NGF/TrkAI signaling, which is responsible for NB growth arrest and differentiation through Ras/MAPK, and its expression is promoted by hypoxia at the expense of NGF-responsive receptors, providing a mechanism for converting NGF/TrkA/Ras/MAPK antioncogenic signals to TrkAIII/IP3K/Akt/NF-kappaB tumor-promoting signals during tumor progression.


Subject(s)
Alternative Splicing/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Receptor, trkA/genetics , Receptor, trkA/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Base Sequence , Cell Line , Cloning, Molecular , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/genetics , Mice , Molecular Sequence Data , NF-kappa B/metabolism , Neovascularization, Pathologic , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Neuroblastoma/blood supply , Neuroblastoma/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma , Protein Binding , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/chemistry , Shc Signaling Adaptor Proteins , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1 , Type C Phospholipases/metabolism
15.
Mol Ther ; 19(12): 2201-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21829174

ABSTRACT

The anaplastic lymphoma kinase (ALK) is a tyrosine kinase receptor that is involved in the pathogenesis of different types of human cancers, including neuroblastoma (NB). In NB, ALK overexpression, or point mutations, are associated with poor prognosis and advanced stage disease. Inhibition of ALK kinase activity by small-molecule inhibitors in lung cancers carrying ALK translocations has shown therapeutic potential. However, secondary mutations may occur that, generate tumor resistance to ALK inhibitors. To overcome resistance to ALK inhibitors in NB, we adopted an alternative RNA interference (RNAi)-based therapeutic strategy that is able to knockdown ALK, regardless of its genetic status [mutated, amplified, wild-type (WT)]. NB cell lines, transduced by lentiviral short hairpin RNA (shRNA), showed reduced proliferation and increased apoptosis when ALK was knocked down. In mice, a nanodelivery system for ALK-specific small interfering RNA (siRNA), based on the conjugation of antibodies directed against the NB-selective marker GD(2) to liposomes, showed strong ALK knockdown in vivo in NB cells, which resulted in cell growth arrest, apoptosis, and prolonged survival. ALK knockdown was associated with marked reductions in vascular endothelial growth factor (VEGF) secretion, blood vessel density, and matrix metalloproteinases (MMPs) expression in vivo, suggesting a role for ALK in NB-induced neoangiogenesis and tumor invasion, confirming this gene as a fundamental oncogene in NB.


Subject(s)
Apoptosis , Mutation/genetics , Neovascularization, Pathologic/prevention & control , Neuroblastoma/blood supply , Neuroblastoma/therapy , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Animals , Blotting, Western , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Gangliosides/immunology , Gangliosides/metabolism , HeLa Cells , Humans , Immunoenzyme Techniques , Liposomes , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Mice, SCID , Neuroblastoma/mortality , Phosphorylation , RNA Interference , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Survival Rate
16.
Carcinogenesis ; 32(7): 1005-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21572098

ABSTRACT

The MYCN oncogene is frequently amplified in neuroblastoma. It is one of the most consistent markers of bad prognosis for this disease. Dickkopf-3 (DKK3) is a secreted protein of the DKK family of Wnt regulators. It functions as a tumor suppressor in a range of cancers, including neuroblastoma. MYCN was recently found to downregulate DKK3 mRNA. In this study, we show that MYCN knockdown in MYCN-amplified (MNA) neuroblastoma cell lines increases secretion of endogenous DKK3 to the culture media. MicroRNAs (miRNAs) are ∼20 nt long single-stranded RNA molecules that downregulate messenger RNAs by targeting the 3' untranslated region (3'UTR). Many miRNAs regulate genes involved in the pathogenesis of cancer and are extensively deregulated in different tumors. Using miRNA target prediction software, we found several MYCN-regulated miRNAs that could target the 3'UTR sequence of DKK3, including mir-92a, mir-92b and let-7e. Luciferase expression from a reporter vector containing the DKK3-3'UTR was decreased when this construct was cotransfected with mir-92a, mir-92b or let-7e in HEK293 cells. Mutation of the mir-92 seed sequence in the 3'UTR completely rescued the observed decrease in reporter expression when cotransfected with mir-92a and mir-92b. Antagomir and miRNA-mimic transfections in neuroblastoma cell lines confirmed that DKK3 secretion to the culture media is regulated by mir-92. Consistent with reports from other cancers, we found DKK3 to be expressed in the endothelium of primary neuroblastoma samples and to be absent in tumors with MYCN amplification. Our data demonstrate that MYCN-regulated miRNAs are able to modulate the expression of the tumor suppressor DKK3 in neuroblastoma.


Subject(s)
Genes, Tumor Suppressor , Intercellular Signaling Peptides and Proteins/metabolism , MicroRNAs/physiology , Neuroblastoma/metabolism , Nuclear Proteins/physiology , Oncogene Proteins/physiology , Oncogenes , 3' Untranslated Regions , Adaptor Proteins, Signal Transducing , Blood Vessels/metabolism , Cell Line, Tumor , Chemokines , DNA Methylation , Gene Knockdown Techniques , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , N-Myc Proto-Oncogene Protein , Neuroblastoma/blood supply , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
17.
Int J Cancer ; 128(11): 2748-58, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-20715103

ABSTRACT

Tumor angiogenesis in childhood neuroblastoma is an important prognostic factor suggesting a potential role for antiangiogenic agents in the treatment of high-risk disease. Within the KidsCancerKinome project, we evaluated the new oral selective pan-VEGFR tyrosine kinase inhibitor axitinib (AG-013736) against neuroblastoma cell lines and the subcutaneous and orthotopic xenograft model IGR-N91 derived from a primary bone marrow metastasis. Axitinib reduced cell proliferation in a dose-dependent manner with IC(50) doses between 274 and >10,000 nmol/l. Oral treatment with 30 mg/kg BID for 2 weeks in advanced tumors yielded significant tumor growth delay, with a median time to reach five times initial tumor volume of 11.4 days compared to controls (p = 0.0006) and resulted in significant reduction in bioluminescence. Simultaneous inhibition of VEGFR downstream effector mTOR using rapamycin 20 mg/kg q2d×5 did not statistically enhance tumor growth delay compared to single agent activities. Axitinib downregulated VEGFR-2 phosphorylation resulting in significantly decreased microvessel density (MVD) and overall surface fraction of tumor vessels (OSFV) in all xenografts as measured by CD34 immunohistochemical staining (mean MVD ± SD and OSFV at 14 days 21.27 ± 10.03 in treated tumors vs. 48.79 ± 17.27 in controls and 0.56% vs. 1.29%; p = 0.0006, respectively). We further explored the effects of axitinib on circulating mature endothelial cells (CECs) and endothelial progenitor cells (CEPs) measured by flow cytometry. While only transient modification was observed for CECs, CEP counts were significantly reduced during and up to 14 days after end of treatment. Axitinib has potent antiangiogenic properties that may warrant further evaluation in neuroblastoma.


Subject(s)
Bone Marrow Neoplasms/drug therapy , Imidazoles/therapeutic use , Indazoles/therapeutic use , Neuroblastoma/drug therapy , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Apoptosis , Axitinib , Blotting, Western , Bone Marrow Neoplasms/secondary , Child , Drug Synergism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Male , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neuroblastoma/blood supply , Neuroblastoma/pathology , Phosphorylation , Sirolimus/pharmacology , Stem Cells/drug effects , Tumor Cells, Cultured
18.
J Pathol ; 222(2): 138-47, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20662003

ABSTRACT

Neuroblastoma (NB) is the second most common extracranial tumour of childhood. Angiogenesis plays a crucial role in the growth and development of NB and vascular endothelial growth factor (VEGF), one of the most potent stimuli of angiogenesis, has been studied extensively in vitro. VEGF(165) has been shown to be the predominant angiogenic isoform expressed in NB cell lines and tumours. In this study, we investigated the anti-angiogenic isoform of VEGF-A, generated from distal splice site selection in the terminal exon of VEGF (VEGF(165)b) and shown to be down-regulated in epithelial malignancies. The expression of both the pro- (VEGF(xxx)) and the anti-angiogenic (VEGF(xxx)b) isoforms was compared in a range of NB and ganglioneuroma (GN) tumours. Whereas VEGF(xxx)b and VEGF(xxx) were both expressed in GN, specific up-regulation of the VEGF(xxx) isoforms was seen in NB at RNA and protein levels. Highly tumourigenic NB cell lines also showed up-regulation of the angiogenic isoforms relative to VEGF(xxx)b compared to less tumourigenic cell lines, and the isoforms were differentially secreted. These results indicate that VEGF(165) is up-regulated in NB and that there is a difference in the balance of isoform expression from anti-angiogenic VEGF(165)b to angiogenic VEGF(165). Treatment with recombinant human VEGF(165)b significantly reduced the growth rate of established xenografts of SK-N-BE(2)-C cells (4.24 +/- 1.01 fold increase in volume) compared with those treated with saline (9.76 +/- 3.58, p < 0.01). Microvascular density (MVD) was significantly decreased in rhVEGF(165)b-treated tumours (19.4 +/- 1.9 vessels/mm(3)) in contrast to the saline-treated tumours (45.5 +/- 8.6 vessels/mm(3)). VEGF(165)b had no significant effect on the proliferative or apoptotic activity, viability or cytotoxicity of SK-N-BE(2)-C cells after 48 h. In conclusion, VEGF(165)b is an effective inhibitor of NB growth. These findings provide the rationale for further investigation of VEGF(165)b in NB and other paediatric malignancies.


Subject(s)
Neovascularization, Pathologic/metabolism , Neuroblastoma/metabolism , Vascular Endothelial Growth Factor A/physiology , Angiogenesis Inducing Agents/metabolism , Angiogenesis Inhibitors/physiology , Animals , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neuroblastoma/blood supply , Neuroblastoma/pathology , Protein Isoforms/physiology , Recombinant Proteins/therapeutic use , Transplantation, Heterologous , Tumor Cells, Cultured , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/therapeutic use , Xenograft Model Antitumor Assays
19.
Coll Antropol ; 35(4): 1071-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22397241

ABSTRACT

Although the role of angiogenesis in tumor progression and response to treatment is generally well-characterized, for neuroblastomas clinical data regarding the contribution of angiogenesis and its predictive capacity remain unclear. The aim of this study was to evaluate whether tumor vascularity in the combination with expression of vascular endothelial growth factor (VEGF) represent prognostic factors for patients with neuroblastoma. Immunohistochemistry using anti-CD34 and anti-VEGF antibodies was used to analyze paraffin-embedded primary tumor tissues from 56 patients diagnosed with neuroblastoma. Tumor vascularity was estimated by calculating the tumor vascular volume fraction (TVVF), and VEGF expression was determined using semi-quantitative scoring. Statistical analyses including multivariate analysis were performed and compared with these two factors. Tumor vascularity had impact on survival of high VEGF expression neuroblastoma patients. Combination of high VEGF expression and TVVF value < or = 5% was independent predictor of overall survival (p-value = 0.0041, odds ratio (OR) (95% CI) = 8.67 (1.99-37.69) by the Cox proportional hazards model). This study revealed for the first time a group of extremely high-risk neuroblastoma with both high VEGF expression and poor vascularity. For these patients reduced rates of survival were observed (37% vs. 92.5%) (p < 0.0001). These patients did not experience a significant improvement following hematopoietic stem cell transplantation, and could be candidates for receiving novel therapies. These results indicate the importance of the mutual relationship between tumor vascularity and VEGF, because it gives better insight into the prognosis of patients with neuroblastoma.


Subject(s)
Neovascularization, Pathologic , Neuroblastoma/blood supply , Neuroblastoma/mortality , Vascular Endothelial Growth Factor A/analysis , Antigens, CD34/analysis , Child , Child, Preschool , Female , Humans , Infant , Male , Prognosis , Survival Rate
20.
Mol Cancer ; 9: 138, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20525313

ABSTRACT

BACKGROUND: New, more effective strategies are needed to treat highly aggressive neuroblastoma. Our laboratory has previously shown that full-length Secreted Protein Acidic and Rich in Cysteine (SPARC) and a SPARC peptide corresponding to the follistatin domain of the protein (FS-E) potently block angiogenesis and inhibit the growth of neuroblastoma tumors in preclinical models. Peptide FS-E is structurally complex and difficult to produce, limiting its potential as a therapeutic in the clinic. RESULTS: In this study, we synthesized two smaller and structurally more simple SPARC peptides, FSEN and FSEC, that respectively correspond to the N-and C-terminal loops of peptide FS-E. We show that both peptides FSEN and FSEC have anti-angiogenic activity in vitro and in vivo, although FSEC is more potent. Peptide FSEC also significantly inhibited the growth of neuroblastoma xenografts. Histologic examination demonstrated characteristic features of tumor angiogenesis with structurally abnormal, tortuous blood vessels in control neuroblastoma xenografts. In contrast, the blood vessels observed in tumors, treated with SPARC peptides, were thin walled and structurally more normal. Using a novel method to quantitatively assess blood vessel abnormality we demonstrated that both SPARC peptides induced changes in blood vessel architecture that are consistent with blood vessel normalization. CONCLUSION: Our results demonstrate that SPARC peptide FSEC has potent anti-angiogenic and anti-tumorigenic effects in neuroblastoma. Its simple structure and ease of production indicate that it may have clinical utility in the treatment of high-risk neuroblastoma and other types of pediatric and adult cancers, which depend on angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neuroblastoma/drug therapy , Osteonectin/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Disease Progression , Endothelial Cells/drug effects , Fluorescent Antibody Technique , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neuroblastoma/blood supply , Peptides , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL