Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Pediatr Blood Cancer ; 66(9): e27835, 2019 09.
Article in English | MEDLINE | ID: mdl-31136074

ABSTRACT

Recent studies in mouse models of cancer have shown that exercise improves tumor vascular function, thereby improving chemotherapy delivery and efficacy. However, the mechanisms underlying this improvement remain unclear and the effect of exercise on Ewing sarcoma (ES), a pediatric bone and soft tissue cancer, is unknown. The effect of exercise on tumor vascular hyperpermeability, which inversely correlates with drug delivery to the tumor, has also not been evaluated. We hypothesized that exercise improves chemotherapy efficacy by enhancing its delivery through improving tumor vascular permeability. We treated ES-bearing mice with doxorubicin with or without moderate treadmill exercise. Exercise did not significantly alter ES tumor vessel morphology. However, compared to control mice, tumors of exercised mice had significantly reduced hyperpermeability, significantly decreased hypoxia, and higher doxorubicin penetration. Compared to doxorubicin alone, doxorubicin plus exercise inhibited tumor growth more efficiently. We evaluated endothelial cell sphingosine-1-phosphate receptors 1 and 2 (S1PR1 and S1PR2) as potential mediators of the improved vascular permeability and increased function afforded by exercise. Relative to tumors from control mice, vessels in tumors from exercised mice had increased S1PR1 and decreased S1PR2 expression. Our results support a model in which exercise remodels ES vasculature to reduce vessel hyperpermeability, potentially via modulation of S1PR1 and S1PR2, thereby improving doxorubicin delivery and inhibiting tumor growth more than doxorubicin alone does. Our data suggest moderate aerobic exercise should be tested in clinical trials as a potentially useful adjuvant to standard chemotherapy for patients with ES.


Subject(s)
Bone Neoplasms , Capillary Permeability , Doxorubicin/pharmacology , Physical Conditioning, Animal , Sarcoma, Ewing , Animals , Bone Neoplasms/blood supply , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Cell Line, Tumor , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Male , Mice , Mice, Nude , Neoplasm Proteins/biosynthesis , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Sarcoma, Ewing/therapy , Sphingosine-1-Phosphate Receptors/biosynthesis , Xenograft Model Antitumor Assays
2.
Mol Med Rep ; 17(3): 4291-4298, 2018 03.
Article in English | MEDLINE | ID: mdl-29328472

ABSTRACT

Ciclopirox (CPX) is a synthetic antifungal drug that is mainly used to treat dermatomycoses. The aim of the present study was to determine whether CPX could influence Ewing sarcoma progression. The present study suggested that CPX treatment may inhibit Ewing sarcoma (ES) progression through Ewing sarcoma breakpoint region 1­Friend leukemia integration 1 (EWS­FLI1), a common fusion transcript structure in patients with ES. To determine the underlying mechanisms of ES progression, cross analysis was conducted on three high­throughput genome or transcript me datasets from the Gene Expression Omnibus. The results indicated that CPX may inhibit ES growth by affecting vasculature development and DNA replication. A combination of genome­wide expression and binding profiles revealed several potential targets for CPX in ES, including collagen type I α2 chain, N­myc proto­oncogene and transforming growth factor ß1, which contained significantly enriched binding peaks of FLI1. In addition, network analysis, including a protein­protein interaction network and a transcription regulatory network, provided further detailed information about the roles of CPX in ES. This study may provide a novel solution for ES treatment and may also aid in improving its prognosis.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Genome, Human , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , Pyridones/therapeutic use , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/drug therapy , Antifungal Agents/therapeutic use , Bone Neoplasms/blood supply , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Ciclopirox , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Databases, Genetic , Drug Repositioning , Gene Expression Profiling , Gene Ontology , Gene Regulatory Networks , Humans , Molecular Sequence Annotation , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/metabolism , Protein Binding , Protein Interaction Mapping , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
3.
Ann R Coll Surg Engl ; 99(6): e162-e164, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28660821

ABSTRACT

A 65-year-old man presented with a right supraclavicular neck mass and right arm pain. Magnetic resonance imaging revealed a 96mm lesion in the upper thoracic paraspinal region extending into the deep supraclavicular fossa. The presentation was consistent with a sarcoma or lymphoma but fine needle aspiration was inconclusive. During open biopsy of the lesion, the patient had a rapid intraoperative haemorrhage of 1l from the tumour. Haemostasis could only be achieved by transarterial embolisation of the feeding vessel and the biopsy result confirmed Ewing's sarcoma. Open biopsy is considered the gold standard in the diagnosis of certain tumour types; however, the morbidity from haemorrhage must be considered. This case highlights the key role that transarterial embolisation can play in achieving haemostasis in the neck.


Subject(s)
Blood Loss, Surgical , Embolization, Therapeutic , Head and Neck Neoplasms , Sarcoma, Ewing , Aged , Biopsy, Fine-Needle , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/surgery , Humans , Male , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/diagnostic imaging , Sarcoma, Ewing/surgery , Tomography, X-Ray Computed
4.
Oncotarget ; 6(23): 19634-46, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26189059

ABSTRACT

Aggressive tumor cells can obtain the ability to transdifferentiate into cells with endothelial features and thus form vasculogenic networks. This phenomenon, called vasculogenic mimicry (VM), is associated with increased tumor malignancy and poor clinical outcome. To identify novel key molecules implicated in the process of vasculogenic mimicry, microarray analysis was performed to compare gene expression profiles of aggressive (VM+) and non-aggressive (VM-) cells derived from Ewing sarcoma and breast carcinoma. We identified the CD44/c-Met signaling cascade as heavily relevant for vasculogenic mimicry. CD44 was at the center of this cascade, and highly overexpressed in aggressive tumors. Both CD44 standard isoform and its splice variant CD44v6 were linked to increased aggressiveness in VM. Since VM is most abundant in Ewing sarcoma tumors functional analyses were performed in EW7 cells. Overexpression of CD44 allowed enhanced adhesion to its extracellular matrix ligand hyaluronic acid. CD44 expression also facilitated the formation of vasculogenic structures in vitro, as CD44 knockdown experiments repressed migration and vascular network formation. From these results and the observation that CD44 expression is associated with vasculogenic structures and blood lakes in human Ewing sarcoma tissues, we conclude that CD44 increases aggressiveness in tumors through the process of vasculogenic mimicry.


Subject(s)
Bone Neoplasms/metabolism , Breast Neoplasms/metabolism , Cell Transdifferentiation , Endothelial Cells/metabolism , Hyaluronan Receptors/metabolism , Neovascularization, Pathologic , Sarcoma, Ewing/metabolism , Biological Mimicry , Bone Neoplasms/blood supply , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion , Cell Movement , Endothelial Cells/pathology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Regulatory Networks , Humans , Hyaluronan Receptors/genetics , Hyaluronic Acid/metabolism , MCF-7 Cells , Phenotype , Protein Interaction Mapping , Protein Isoforms , RNA Interference , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Signal Transduction , Transfection
5.
Oncotarget ; 5(9): 2436-49, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24810959

ABSTRACT

Angiogenesis is required for tumor growth. WT1, a protein that affects both mRNA transcription and splicing, has recently been shown to regulate expression of vascular endothelial growth factor (VEGF), one of the major mediators of angiogenesis. In the present study, we tested the hypothesis that WT1 is a key regulator of tumor angiogenesis in Ewing sarcoma. We expressed exogenous WT1 in the WT1-null Ewing sarcoma cell line, SK-ES-1, and we suppressed WT1 expression using shRNA in the WT1-positive Ewing sarcoma cell line, MHH-ES. Suppression of WT1 in MHH-ES cells impairs angiogenesis, while expression of WT1 in SK-ES-1 cells causes increased angiogenesis. Different WT1 isoforms result in vessels with distinct morphologies, and this correlates with preferential upregulation of particular VEGF isoforms. WT1-expressing tumors show increased expression of pro-angiogenic molecules such as VEGF, MMP9, Ang-1, and Tie-2, supporting the hypothesis that WT1 is a global regulator of angiogenesis. We also demonstrate that WT1 regulates the expression of a panel of pro-angiogenic molecules in Ewing sarcoma cell lines. Finally, we found that WT1 expression is correlated with VEGF expression, MMP9 expression, and microvessel density in samples of primary Ewing sarcoma. Thus, our results demonstrate that WT1 expression directly regulates tumor angiogenesis by controlling the expression of a panel of pro-angiogenic genes.


Subject(s)
Bone Neoplasms/blood supply , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/metabolism , Sarcoma, Ewing/blood supply , Vascular Endothelial Growth Factor A/metabolism , WT1 Proteins/metabolism , Animals , Apoptosis , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Movement , Cell Proliferation , Chromatin Immunoprecipitation , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics , WT1 Proteins/genetics , Xenograft Model Antitumor Assays
6.
Oncotarget ; 4(12): 2487-501, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24318733

ABSTRACT

Ewing sarcoma (ES) is an aggressive malignancy driven by an oncogenic fusion protein, EWS-FLI1. Neuropeptide Y (NPY), and two of its receptors, Y1R and Y5R are up-regulated by EWS-FLI1 and abundantly expressed in ES cells. Paradoxically, NPY acting via Y1R and Y5R stimulates ES cell death. Here, we demonstrate that these growth-inhibitory actions of NPY are counteracted by hypoxia, which converts the peptide to a growth-promoting factor. In ES cells, hypoxia induces another NPY receptor, Y2R, and increases expression of dipeptidyl peptidase IV (DPPIV), an enzyme that cleaves NPY to a shorter form, NPY3-36. This truncated peptide no longer binds to Y1R and, therefore, does not stimulate ES cell death. Instead, NPY3-36 acts as a selective Y2R/Y5R agonist. The hypoxia-induced increase in DPPIV activity is most evident in a population of ES cells with high aldehyde dehydrogenase (ALDH) activity, rich in cancer stem cells (CSCs). Consequently, NPY, acting via Y2R/Y5Rs, preferentially stimulates proliferation and migration of hypoxic ALDHhigh cells. Hypoxia also enhances the angiogenic potential of ES by inducing Y2Rs in endothelial cells and increasing the release of its ligand, NPY3-36, from ES cells. In summary, hypoxia acts as a molecular switch shifting NPY activity away from Y1R/Y5R-mediated cell death and activating the Y2R/Y5R/DPPIV/NPY3-36 axis, which stimulates ES CSCs and promotes angiogenesis. Hypoxia-driven actions of the peptide such as these may contribute to ES progression. Due to the receptor-specific and multifaceted nature of NPY actions, these findings may inform novel therapeutic approaches to ES.


Subject(s)
Cell Hypoxia/physiology , Neuropeptide Y/metabolism , Sarcoma, Ewing/metabolism , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Dipeptidyl Peptidase 4/metabolism , Heterografts , Humans , Mice , Mice, Nude , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptors, Neuropeptide Y/antagonists & inhibitors , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology
7.
Neoplasia ; 14(10): 964-73, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23097630

ABSTRACT

One of the challenges of tailored antiangiogenic therapy is the ability to adequately monitor the angiogenic activity of a malignancy in response to treatment. The α(v)ß(3) integrin, highly overexpressed on newly formed tumor vessels, has been successfully used as a target for Arg-Gly-Asp (RGD)-functionalized nanoparticle contrast agents. In the present study, an RGD-functionalized nanocarrier was used to image ongoing angiogenesis in two different xenograft tumor models with varying intensities of angiogenesis (LS174T > EW7). To that end, iron oxide nanocrystals were included in the core of the nanoparticles to provide contrast for T(2)*-weighted magnetic resonance imaging (MRI), whereas the fluorophore Cy7 was attached to the surface to enable near-infrared fluorescence (NIRF) imaging. The mouse tumor models were used to test the potential of the nanoparticle probe in combination with dual modality imaging for in vivo detection of tumor angiogenesis. Pre-contrast and post-contrast images (4 hours) were acquired at a 9.4-T MRI system and revealed significant differences in the nanoparticle accumulation patterns between the two tumor models. In the case of the highly vascularized LS174T tumors, the accumulation was more confined to the periphery of the tumors, where angiogenesis is predominantly occurring. NIRF imaging revealed significant differences in accumulation kinetics between the models. In conclusion, this technology can serve as an in vivo biomarker for antiangiogenesis treatment and angiogenesis phenotyping.


Subject(s)
Bone Neoplasms/diagnosis , Colorectal Neoplasms/diagnosis , Magnetic Resonance Imaging , Molecular Imaging , Nanoparticles , Neovascularization, Pathologic , Sarcoma, Ewing/diagnosis , Spectroscopy, Near-Infrared , Animals , Bone Neoplasms/blood supply , Bone Neoplasms/metabolism , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/metabolism , Contrast Media , Disease Models, Animal , Fluorescence , Humans , Immunoenzyme Techniques , Integrin alphaVbeta3/metabolism , Mice , Oligopeptides/antagonists & inhibitors , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/metabolism
8.
Angiogenesis ; 15(4): 685-95, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22914877

ABSTRACT

Increasing experimental evidence suggests that IGF-1 may modulate tumor angiogenesis via activation of the expression of VEGF in Ewing sarcomas and rhabdomyosarcomas. This study investigates the effects of the PEGylated Adnectins™ CT-322, a VEGFR2-inhibitor and AT580Peg40, an IGF-1R inhibitor, as monotherapy and in combination in a murine A673 xenograft tumor model. The combination of Adnectins CT-322 and AT580Peg40 revealed a 83% reduction in tumor growth, a nearly 5 times lower vessel density, less necrotic areas and less appearance of intussusceptive angiogenesis. Monotherapy with IGF-1R or CT-322 revealed equally a significant inhibition of tumor and vessel growth. Combinatory inhibition of IGF-1R and VEGFR2 shows a downregulation of IGF-binding protein 2 and a compensatory upregulation of VEGF levels. Immunohistological analysis showed remodeling vascular effects of CT-322-treatment or combination therapy. The vascular architecture in Adnectin-treated tumors was characterized by a strong normalization of vasculature. 3D-evaluation in microvascular corrosion casts showed significantly higher intervascular and interbranching distances in Adnectin-treated tumors. CT-322-treatment and combinatory inhibition reveal a significant reduction of intussusceptive angiogenesis. These pronounced effects on tumor vasculature suggest potential therapeutic benefit of combinatorial IGF1- and VEGF-pathways inhibition in Ewing's sarcoma.


Subject(s)
Sarcoma, Ewing/pathology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Cell Line, Tumor , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Nude , Receptor, IGF Type 1/metabolism , Sarcoma, Ewing/blood supply , Transplantation, Heterologous
9.
J Surg Res ; 173(1): 1-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21658718

ABSTRACT

BACKGROUND: Expression of epidermal growth factor receptor (EGFR), a potent regulator of cellular homeostasis, is associated with aggressive tumor behavior. The mechanism by which EGFR inhibition functions is unclear, with controversial results demonstrating an effect on the tumor cells, endothelial cells, or pericytes. EGFR activation has been linked to the expression of vascular endothelial growth factor (VEGF), a known mitogen of angiogenesis, but the relationship between these factors and their effect on tumor vessel development is vague. We hypothesized that using an EGFR inhibitor on a human Ewing's sarcoma model would inhibit tumor growth by suppressing vessel proliferation. METHODS: A cell proliferation assay was performed on the Ewing's sarcoma (SK-NEP-1) cell line. Tumor cells were implanted intrarenally in athymic mice. Animals received daily gavage with vehicle or gefitinib 1 wk following implantation. Mice (n = 12/cohort) were euthanized 6 wk following implantation. Remaining mice were maintained without treatment for 2 wk. Vascular changes were assessed by angiography and immunohistochemically. EGFR and vascular endothelial growth factor (VEGF) expression were quantified using quantitative polymerase chain reaction (qPCR). RESULTS: Gefitinib suppressed in vitro cell growth with an IC(50) = 1.36 µM. Minimal tumor growth suppression was noted at 6 wk (6.01 ± 1.2 g in control versus 4.61 ± 0.9 g treated, P = 0.36). After cessation of gefitinib, tumor growth was increased in both groups (7.37 ± 1.62 g versus 6.77 ± 1.53 g, P = 0.79). Microvessel density was unchanged despite EGFR inhibition (161,000 ± 16,000 pixels versus 135,000 ± 18,000 pixels, P = 0.31). At 6 wk, the vascular maturity index was similar in both groups (3.63 ± 1.12 versus 4.09 ± 1.71, P = 0.83). A downward trend in EGFR expression (49% of control) and an upward trend in VEGF levels (50% of control) occurred in the treated group. CONCLUSIONS: EGFR expression was suppressed in cultured cells and xenograft tumors. Despite a cytotoxic effect on cell lines, gefitinib had little effect on tumor growth. No effects on the tumor vasculature were noted in the setting of EGFR suppression, suggesting that angiogenesis induced by SK-NEP-1 cells is refractory to EGFR inhibition. Interestingly, the resulting increase in VEGF expression following EGFR blockade, provides an alternative pro-angiogenic pathway promoting tumor survival.


Subject(s)
Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , Neovascularization, Pathologic/physiopathology , Quinazolines/pharmacology , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/pathology , Angiography , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Neoplasms/blood supply , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Female , Gefitinib , Humans , In Vitro Techniques , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Quinazolines/therapeutic use , Sarcoma, Ewing/drug therapy , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/metabolism
10.
Ann Saudi Med ; 31(2): 174-82, 2011.
Article in English | MEDLINE | ID: mdl-21422656

ABSTRACT

Ewing/PNET (peripheral neuroepithelioma) tumors are rare aggressive bone sarcomas occurring in young people. Rare-disease clinical trials can require global collaborations and many years. In vivo models that as accurately as possible reflect the clinical disease are helpful in selecting therapeutics with the most promise of positive clinical impact. Human Ewing/PNET sarcoma cell lines developed over the past 45 years are described. Several of these have undergone genetic analysis and have been confirmed to be those of Ewing/PNET sarcoma. The A673 Ewing sarcoma line has proven to be particularly useful in understanding the biology of this disease in the mouse. The chromosomal translocation producing the EWS/FLI1 fusion transcript characterizes clinical Ewing sarcoma. Cell lines that express this genetic profile are confirmed to be those of Ewing sarcoma. The A673 Ewing sarcoma line grows in culture and as a xenograft in immunodeficient mice. The A673 model has been used to study Ewing sarcoma angiogenesis and response to antiangiogenic agents. Many Ewing sarcoma clinical specimens express the cell surface protein endosialin. Several Ewing sarcoma cell lines, including the A673 line, also express cell surface endosialin when grown as subcutaneous tumor nodules and as disseminated disease; thus the A673 is a useful model for the study of endosialin biology and endosialin-directed therapies. With the advent of tools that allow characterization of clinical disease to facilitate optimal treatment, it becomes imperative, especially for rare tumors, to develop preclinical models reflecting disease subsets. Ewing PNET sarcomas are a rare disease where models are available.


Subject(s)
Disease Models, Animal , Neuroectodermal Tumors, Primitive/pathology , Sarcoma, Ewing/pathology , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Humans , Mice , Neuroectodermal Tumors, Primitive/blood supply , Neuroectodermal Tumors, Primitive/drug therapy , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/drug therapy
11.
Blood ; 117(2): 719-26, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20944072

ABSTRACT

Delta-like ligand 4 (DLL4) is essential for the formation of mature vasculature. However, the role of DLL4-Notch signaling in pericyte/vascular smooth muscle cell (vSMC) development is poorly understood. We sought to determine whether DLL4-Notch signaling is involved in pericyte/vSMC formation in vitro and during vasculogenesis in vivo using 2 Ewing sarcoma mouse models. Inhibition of DLL4 with the antibody YW152F inhibited pericyte/vSMC marker expression by bone marrow (BM) cells in vitro. Conversely, transfection of 10T1/2 cells with the active domains of Notch receptors led to increased expression of pericyte/vSMC markers. Furthermore, the blood vessels of Ewing sarcoma tumors from mice treated with YW152F had reduced numbers of BM-derived pericytes/vSMCs, fewer open lumens, and were less functional than the vessels in tumors of control-treated mice. Tumor growth was also inhibited. These data demonstrate a specific role for DLL4 in the formation of BM-derived pericytes/vSMCs and indicate that DLL4 may be a novel therapeutic target for the inhibition of vasculogenesis.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Neovascularization, Physiologic/physiology , Pericytes/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Animals , Bone Marrow Cells/metabolism , Bone Neoplasms/blood supply , Bone Neoplasms/metabolism , Calcium-Binding Proteins , Cell Differentiation , Disease Models, Animal , Immunohistochemistry , Mice , Microscopy, Confocal , Muscle, Smooth, Vascular/cytology , Neovascularization, Pathologic/metabolism , Pericytes/cytology , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/metabolism
12.
Khirurgiia (Mosk) ; (11): 41-4, 2011.
Article in Russian | MEDLINE | ID: mdl-22408800

ABSTRACT

Results of observation were analyzed in 68 patients with Ewing's sarcoma. Reagents made by DAKO (France) such as CD-31, endothelial cell, clone JC 70A, isotype: Lg GI, kappa 02/1 ml were used in the study. It was estimated, that CD31 expression activity in endothelial elements was inversely proportional and reliably correlated with malignant process by the Ewing's sarcoma. Indices of lymphoid infiltration in pathological focus were inversely proportional to the expression of CD-31 positive cells. Statistically reliable inversely proportional relationship was revealed between CD-31 expression of endothelial cells and spontaneous tissue necrotization in the Ewing's sarcoma. Reliable direct correlation was ascertained between tumor angiogenesis and amount of CD-31.


Subject(s)
Bone Neoplasms/blood supply , Sarcoma, Ewing/blood supply , Disease Progression , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Immunohistochemistry , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis
13.
Angiogenesis ; 14(2): 125-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21184173

ABSTRACT

An MHC-mismatch bone marrow (BM) transplant Ewing's sarcoma mouse model was used to investigate whether BM cells participate in the vessel formation that support Ewing's sarcoma lung metastasis. BM cells from H-2K(b/d) donor mice were transplanted into sublethally irradiated H-2K(d) recipient mice. Donor BM cells were identified using the H-2K(b) marker. Engraftment was confirmed by identifying the H-2K(b) IL-1ß-type specific polymorphism. After engraftment highly lung metastatic TC71-PM4 cells were injected intravenously. Mice were sacrificed 10 weeks after tumor cell injection. Hematoxylin-and-eosin staining was performed to identify lung metastatic foci. These tumors were then evaluated using immunohistochemical analysis. H-2K(b)-positive cells were found in lung metastases but not in normal lung, liver or spleen tissues. Injection of CM-Dil-labeled BM cells into tumor bearing and control mice showed that nonspecific organ migration occurred at 24 h, but that these cells were absent 1 week later in control mice. These data suggest that the migration of the H-2K(b) BM cells to lung nodules was specific because these cells were observed 14 weeks after transplantation. Co-localization of H-2K(b) and CD31 or VE-Cadherin demonstrated that some endothelial cells were BM-derived. Co-localization of H-2K(b) and Desmin, smooth muscle actin (α-SMA) or PDGFR-ß indicated that a fraction of pericytes was also BM-derived. These results suggest that BM cells participate in the vascular formation that supports the growth of Ewing's sarcoma lung metastases. BM cells migrated to the metastatic tumor and differentiated into endothelial cells and pericytes. These data indicated that targeting this process may have therapeutic potential.


Subject(s)
Bone Marrow Cells/pathology , Bone Marrow Transplantation , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Neovascularization, Pathologic/pathology , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/pathology , Animals , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Endothelial Cells/pathology , H-2 Antigens/metabolism , Humans , Interleukin-1beta/genetics , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Pericytes/pathology , Polymorphism, Genetic , Reproducibility of Results , Species Specificity
14.
Cancer Res ; 70(4): 1334-43, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20124484

ABSTRACT

The role of vasculogenesis as opposed to angiogenesis in tumor formation has been little explored genetically. Endothelial cells that lack the MEK kinase MEKK3 cannot form vessels. In this study, we employed mice with hematopoietic deletions of the Mekk3 gene to evaluate the importance of vasculogenesis in the formation of Ewing's sarcoma tumors. Bone marrow cells (BM) from LacZ(+) Mekk3-deficient conditional knockout mice (Mekk3(Deltaflox/-) mice) were transplanted into irradiated nude mice before injection of Ewing's sarcoma cells. Because the grafted Mekk3(Deltaflox/-) BM cells cannot contribute to vessel development in the same way as the host Mekk3(+/+) endothelial cells, angiogenesis is normal in the model whereas vasculogenesis is impaired. Four weeks after BM transplant, Ewing's sarcoma TC71 or A4573 cells were injected, and tumor growth and vessel density were compared. Strikingly, chimeric mice transplanted with Mekk3(Deltaflox/-) BM exhibited a reduction in tumor growth and vessel density compared with mice transplanted with Mekk3(Deltaflox/+) BM cells. Mekk3(Deltaflox/-) cells that were LacZ positive were visualized within the tumor; however, few of the LacZ(+) cells colocalized with either CD31(+) endothelial cells or desmin(+) pericytes. Quantification of double-positive LacZ(+) and CD31(+) endothelial cells or LacZ(+) and desmin(+) pericytes confirmed that chimeric mice transplanted with Mekk3(Deltaflox/-) BM were impaired for tumor vessel formation. In contrast, siRNA-mediated knockdown of Mekk3 in TC71 Ewing's sarcoma cells had no effect on tumor growth or vessel density. Our findings indicate that vasculogenesis is critical in the expansion of the tumor vascular network.


Subject(s)
Bone Marrow Cells/physiology , Bone Neoplasms/blood supply , Bone Neoplasms/pathology , Neovascularization, Pathologic/pathology , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/pathology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/physiology , Cell Proliferation , Embryo, Mammalian , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , MAP Kinase Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase Kinase 3/genetics , MAP Kinase Kinase Kinase 3/metabolism , MAP Kinase Kinase Kinase 3/physiology , Male , Mice , Mice, Nude , Mice, Transgenic , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , RNA, Small Interfering/pharmacology , Tumor Burden/genetics , Tumor Cells, Cultured
15.
FASEB J ; 24(6): 1689-99, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20075195

ABSTRACT

High density lipoprotein (HDL), an endogenous nanoparticle, transports fat throughout the body and is capable of transferring cholesterol from atheroma in the vessel wall to the liver. In the present study, we utilized HDL as a multimodal nanoparticle platform for tumor targeting and imaging via nonspecific accumulation and specific binding to angiogenically activated blood vessels. We reconstituted HDL (rHDL) with amphiphilic gadolinium chelates and fluorescent dyes. To target angiogenic endothelial cells, rHDL was functionalized with alphavbeta3-integrin-specific RGD peptides (rHDL-RGD). Nonspecific RAD peptides were conjugated to rHDL nanoparticles as a control (rHDL-RAD). It was observed in vitro that all 3 nanoparticles were phagocytosed by macrophages, while alphavbeta3-integrin-specific rHDL-RGD nanoparticles were preferentially taken up by endothelial cells. The uptake of nanoparticles in mouse tumors was evaluated in vivo using near infrared (NIR) and MR imaging. All nanoparticles accumulated in tumors but with very different accumulation/binding kinetics as observed by NIR imaging. Moreover, confocal microscopy revealed rHDL-RGD to be associated with tumor endothelial cells, while rHDL and rHDL-RAD nanoparticles were mainly found in the interstitial space. This study demonstrates the ability to reroute HDL from its natural targets to tumor blood vessels and its potential for multimodal imaging of tumor-associated processes.


Subject(s)
Lipoproteins, HDL , Molecular Probes , Nanoparticles , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/diagnosis , Oligopeptides , Sarcoma, Ewing/blood supply , Animals , Cells, Cultured , Endothelium, Vascular/metabolism , Humans , Immunoenzyme Techniques , Integrin alphaVbeta3/metabolism , Lipoproteins, HDL/chemistry , Macrophages/metabolism , Magnetic Resonance Imaging , Mice , Mice, Nude , Microscopy, Confocal , Molecular Imaging , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Oligopeptides/chemistry , Oligopeptides/metabolism , Phagocytosis , Sarcoma, Ewing/metabolism , Spectroscopy, Near-Infrared , Tissue Distribution , Umbilical Veins/cytology
16.
Clin Cancer Res ; 16(3): 848-56, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20103680

ABSTRACT

PURPOSE: Bone marrow (BM) cells contribute to tumor vessel formation that supports the growth of Ewing's sarcoma. These BM cells migrate into the tumor and differentiate into endothelial cells and pericytes. We investigated whether delta-like ligand 4 (DLL4) played a role in the formation of BM-derived pericytes/vascular smooth muscle cells (vSMC) during tumor vessel formation. EXPERIMENTAL DESIGN: Using immunohistochemistry, we examined the expression pattern of DLL4 in 14 patient samples and two xenograft mouse models of Ewing's sarcoma. We then used intratumor injections of short hairpin RNA to inhibit DLL4 expression in Ewing's sarcoma tumors in mice, and evaluated the effect on BM-derived pericytes/vSMCs. RESULTS: DLL4 was expressed by perivascular cells in 12 of 14 human samples and in BM-derived pericytes/vSMCs in both A4573 and TC71 xenograft tumors. Inhibition of DLL4 expression by short hairpin RNA correlated with the decreased numbers of BM-derived cells in tumor vessels and the decreased numbers of alpha-SMA(+), desmin(+), and NG2(+) pericytes/vSMCs, as well as increased tumor hypoxia. CONCLUSIONS: DLL4 is important for the formation of BM-derived pericytes/vSMCs during vasculogenesis in Ewing's sarcoma. DLL4 may be a therapeutic target for treatment of Ewing's sarcoma by inhibition of blood vessel formation.


Subject(s)
Bone Marrow Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Muscle, Smooth, Vascular/physiology , Neovascularization, Pathologic , Pericytes/physiology , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Marrow Transplantation , Calcium-Binding Proteins , Cell Differentiation , Cell Line , Down-Regulation , Humans , Mice , Mice, Nude , Mice, Transgenic , Receptor, Notch1/metabolism
17.
Cancer ; 116(3): 749-57, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20029966

ABSTRACT

Ewing sarcoma is the second most common type of bone cancer in children and young adults. In recent years, the mechanisms by which these tumors develop and maintain their vascular supply have been elucidated. Additional work has demonstrated that inhibition of angiogenic pathways or disruption of established vasculature can attenuate the growth of Ewing sarcoma mouse xenografts. Early clinical data suggest that these results also may extend to patients with Ewing sarcoma who are treated with antiangiogenic or antivascular therapies. For the current review, the authors summarized the available data supporting this approach.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/drug therapy , Animals , Humans , Neoplasms/drug therapy
18.
Eur J Cancer ; 45(4): 713-22, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19136249

ABSTRACT

The effects of the tubulin-binding vascular-disrupting agents (VDAs), combretastatin A4 phosphate (CA4P), OXi4503/CA1P and OXi8007, in subcutaneous mouse models of the Ewing's sarcoma family of tumours (ESFTs) have been investigated alone and in combination with doxorubicin. Delay in subcutaneous tumour growth was observed following treatment of mice with multiple doses of OXi4503/CA1P but not with CA4P or OXi8007. A single dose of OXi4503/CA1P caused complete shutdown of vasculature by 24h and extensive haemorrhagic necrosis by 48h. However, a viable rim of proliferating cells remained, which repopulated the tumour within 10 days following the withdrawal of treatment. Combined treatment with doxorubicin 1h prior to administration of OXi4503/CA1P enhanced the effects of OXi4503/CA1P causing a synergistic delay in tumour growth (p<0.001). This study demonstrates that OXi4503/CA1P is a potent VDA in ESFT and in combination with conventional cytotoxic agents represents a promising treatment strategy for this tumour group.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bibenzyls/therapeutic use , Bone Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Sarcoma, Ewing/drug therapy , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Bone Neoplasms/blood supply , Bone Neoplasms/pathology , Cell Proliferation/drug effects , Diphosphates/administration & dosage , Diphosphates/therapeutic use , Disease Models, Animal , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Drug Evaluation, Preclinical/methods , Mice , Mice, Nude , Necrosis , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/pathology , Stilbenes/administration & dosage , Stilbenes/therapeutic use
19.
Int J Oncol ; 34(1): 79-87, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19082480

ABSTRACT

Approval of the anti-vascular endothelial growth factor (VEGF) antibody bevacizumab by the FDA in 2004 reflected the success of this vascular targeting strategy in extending survival in patients with advanced cancers. However, consistent with previous reports that experimental tumors can grow or recur during VEGF blockade, it has become clear that many patients treated with VEGF inhibitors will ultimately develop progressive disease. Previous studies have shown that disruption of VEGF signaling in tumors induces remodeling in surviving vessels, and link increased expression of angiopoietin-1 (Ang-1) with this process. However, overexpression of Ang-1 in different tumors has yielded divergent results, restricting angiogenesis in some systems while promoting it in others. These data raise the possibility that effects of Ang-1/Tie-2 may be context-dependent. Expression of an Ang-1 construct (Ang1*) did not significantly change tumor growth in our model prior to treatment, although vessels exhibited changes consistent with increased Tie-2 signaling. During inhibition of VEGF, however, both overexpression of Ang1* and administration of an engineered Ang-1 agonist (Bow-Ang1) strikingly protected tumors and vasculature from regression. In this context, Ang-1/Tie-2 activation limited tumor hypoxia, increased vessel caliber, and promoted recruitment of mural cells. Thus, these studies support a model in which activation of Tie-2 is important for tumor and vessel survival when VEGF-dependent vasculature is stressed. Understanding such mechanisms of adaptation to this validated form of therapy may be important in designing regimens that make the best use of this approach.


Subject(s)
Angiopoietin-1/genetics , Gene Expression Regulation/physiology , Kidney Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Receptor, TIE-2/genetics , Sarcoma, Ewing/blood supply , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Kidney Neoplasms/pathology , Mice , Mice, Nude , Phosphorylation , Polymerase Chain Reaction , Sarcoma, Ewing/pathology , Transfection , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/metabolism
20.
Histol Histopathol ; 23(10): 1229-40, 2008 10.
Article in English | MEDLINE | ID: mdl-18712675

ABSTRACT

Sarcomas display varied degrees of karyotypic abnormality, vascularity and mesenchymal differentiation. We have reported that a strain of telomerized adult human bone marrow mesenchymal stem cells (hMSC-TERT20) spontaneously evolved a tumorigenic phenotype after long-term continuous culture. We asked to what extent our hMSC-TERT20 derived tumors reflected events found in human sarcomas using routine histopathological procedures. Early versus late passage hMSC-TERT20 cultures persistently expressed mesenchymal lineage proteins e.g. CD105, CD44, CD99 and vimentin. However, late passage cultures, showed increased immunohistochemical staining for CyclinD1 and p21WAF1/Cip1, whereas p27Kip1 staining was reduced. Notably, spectral karyotyping showed that tumorigenic hMSC-TERT20 cells retained a normal diploid karyotype, with no detectable chromosome abnormalities. Consistent with the bone-forming potential of early passage hMSC-TERT20 cells, tumors derived from late passage cells expressed early biomarkers of osteogenesis. However, hMSC-TERT20 cells were heterogeneous for alpha smooth muscle actin (ASMA) expression and one out of six hMSC-TERT20 derived single cell clones was strongly ASMA positive. Tumors from this ASMA+ clone had distinctive vascular qualities with hot spots of high CD34+ murine endothelial cell density, together with CD34- regions with a branching periodic acid Schiff reaction pattern. Such clone-specific differences in host vascular response provide novel models to explore interactions between mesenchymal stem and endothelial cells. Despite the lack of a characteristic chromosomal translocation, the histomorphology, biomarkers and oncogenic changes were similar to those prevalent for Ewing's sarcomas. The phenotype and ontogenesis of hMSC-TERT20 tumors was consistent with the hypothesis that sarcomas may arise from hMSC, providing a unique diploid model for exploring human sarcoma biology.


Subject(s)
Bone Neoplasms/pathology , Cell Differentiation , Cell Lineage , Cell Transformation, Neoplastic/pathology , Mesenchymal Stem Cells/pathology , Sarcoma, Ewing/pathology , Actins/metabolism , Animals , Biomarkers, Tumor/metabolism , Bone Neoplasms/blood supply , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Karyotyping , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Osteogenesis , Phenotype , Sarcoma, Ewing/blood supply , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Telomerase/genetics , Telomerase/metabolism , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL