Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Cereb Blood Flow Metab ; 38(10): 1741-1753, 2018 10.
Article in English | MEDLINE | ID: mdl-28627960

ABSTRACT

Neo-angiogenesis represents an important factor for the delivery of oxygen and nutrients to a growing tumour, and is considered to be one of the main pathodiagnostic features of glioblastomas (GBM). Anti-angiogenic therapy by vascular endothelial growth factor (VEGF) blocking agents has been shown to lead to morphological vascular normalisation resulting in a reduction of contrast enhancement as seen by magnetic resonance imaging (MRI). Yet the functional consequences of this normalisation and its potential for improved delivery of cytotoxic agents to the tumour are not known. The presented study aimed at determining the early physiologic changes following bevacizumab treatment. A time series of perfusion MRI and hypoxia positron emission tomography (PET) scans were acquired during the first week of treatment, in two human GBM xenograft models treated with either high or low doses of bevacizumab. We show that vascular morphology was normalised over the time period investigated, but vascular function was not improved, resulting in poor tumoural blood flow and increased hypoxia.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bevacizumab/pharmacology , Brain Neoplasms/pathology , Glioblastoma/pathology , Neovascularization, Pathologic/pathology , Animals , Female , Humans , Male , Mice, Nude , Xenograft Model Antitumor Assays
2.
Nat Cell Biol ; 17(12): 1556-68, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26595383

ABSTRACT

L-Glutamine (Gln) functions physiologically to balance the carbon and nitrogen requirements of tissues. It has been proposed that in cancer cells undergoing aerobic glycolysis, accelerated anabolism is sustained by Gln-derived carbons, which replenish the tricarboxylic acid (TCA) cycle (anaplerosis). However, it is shown here that in glioblastoma (GBM) cells, almost half of the Gln-derived glutamate (Glu) is secreted and does not enter the TCA cycle, and that inhibiting glutaminolysis does not affect cell proliferation. Moreover, Gln-starved cells are not rescued by TCA cycle replenishment. Instead, the conversion of Glu to Gln by glutamine synthetase (GS; cataplerosis) confers Gln prototrophy, and fuels de novo purine biosynthesis. In both orthotopic GBM models and in patients, (13)C-glucose tracing showed that GS produces Gln from TCA-cycle-derived carbons. Finally, the Gln required for the growth of GBM tumours is contributed only marginally by the circulation, and is mainly either autonomously synthesized by GS-positive glioma cells, or supplied by astrocytes.


Subject(s)
Brain Neoplasms/metabolism , Cell Proliferation , Glioblastoma/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamine/metabolism , Nucleotides/biosynthesis , Animals , Astrocytes/cytology , Astrocytes/metabolism , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Citric Acid Cycle , Coculture Techniques , Female , Glioblastoma/genetics , Glioblastoma/pathology , Glutamate-Ammonia Ligase/genetics , Glutamic Acid/metabolism , Humans , Male , Mice, Inbred NOD , Mice, SCID , Models, Biological , Neoplastic Stem Cells/metabolism , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
3.
J Transl Med ; 12: 278, 2014 Oct 04.
Article in English | MEDLINE | ID: mdl-25280402

ABSTRACT

BACKGROUND: Although several studies suggest that stromal fibroblasts mediate treatment resistance in several cancer types, little is known about how tumor-associated astrocytes modulate the treatment response in brain tumors. Since traditionally used metabolic assays do not distinguish metabolic activity between stromal and tumor cells, and since 2-dimensional co-culture system does not recreate the formidable complexity of the microenvironment within 3-dimensional structures such as solid tumor tissue, we instead established a glioblastoma (GBM) cell-specific bioluminescent assay for direct measurements of tumor cell viability in the treatment of clinical relevant drugs. METHODS: Using lentiviral transfection, we established a panel of human GBM cell lines constitutively expressing a fusion transgene encoding luciferase and the enhanced green fluorescence protein (eGFP). We then initiated co-cultures with immortalized astrocytes, TNC-1, and the eGFP/Luc GBM cell lines. Next, we treated all eGFP/Luc GBM cell lines with Temozolomide (TMZ) or Doxorubicin, comparing co-cultures of glioblastoma (GBM) cells and TNC-1 astrocytes with mono-cultures of eGFP/Luc GBM cells. Cell viability was quantitated by measuring the luciferase expression. RESULTS: Titration experiments demonstrated that luciferase expression was proportional to the number of eGFP/Luc GBM cells, whereas it was not influenced by the number of TNC-1 cells present. Notably, the presence of TNC-1 astrocytes mediated significantly higher cell survival after TMZ treatment in the U251, C6, A172 cell lines as well as the in vivo propagated primary GBM tumor cell line (P3). Moreover, TNC-1 astrocytes mediated significantly higher survival after Doxorubicin treatment in the U251, and LN18 glioma cell lines. CONCLUSION: Glioma cell-specific bioluminescent assay is a reliable tool for assessment of cell viability in the brain tumor cell compartment following drug treatment. Moreover, we have applied this assay to demonstrate that astrocytes can modulate chemo sensitivity of GBM tumor cells. These effects varied both with the cell line and cytotoxic drug that were used, suggesting that several mechanisms may be involved.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/pathology , Coculture Techniques/methods , Drug Resistance, Neoplasm , Glioblastoma/pathology , Luminescence , Models, Biological , Astrocytes/drug effects , Astrocytes/metabolism , Brain Neoplasms/drug therapy , Cell Compartmentation , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Glioblastoma/drug therapy , Humans , Reproducibility of Results , Spheroids, Cellular/pathology , Stromal Cells/drug effects , Stromal Cells/metabolism , Temozolomide
4.
PLoS One ; 8(3): e59773, 2013.
Article in English | MEDLINE | ID: mdl-23527265

ABSTRACT

Oligodendroglioma poses a biological conundrum for malignant adult human gliomas: it is a tumor type that is universally incurable for patients, and yet, only a few of the human tumors have been established as cell populations in vitro or as intracranial xenografts in vivo. Their survival, thus, may emerge only within a specific environmental context. To determine the fate of human oligodendroglioma in an experimental model, we studied the development of an anaplastic tumor after intracranial implantation into enhanced green fluorescent protein (eGFP) positive NOD/SCID mice. Remarkably after nearly nine months, the tumor not only engrafted, but it also retained classic histological and genetic features of human oligodendroglioma, in particular cells with a clear cytoplasm, showing an infiltrative growth pattern, and harboring mutations of IDH1 (R132H) and of the tumor suppressor genes, FUBP1 and CIC. The xenografts were highly invasive, exhibiting a distinct migration and growth pattern around neurons, especially in the hippocampus, and following white matter tracts of the corpus callosum with tumor cells accumulating around established vasculature. Although tumors exhibited a high growth fraction in vivo, neither cells from the original patient tumor nor the xenograft exhibited significant growth in vitro over a six-month period. This glioma xenograft is the first to display a pure oligodendroglioma histology and expression of R132H. The unexpected property, that the cells fail to grow in vitro even after passage through the mouse, allows us to uniquely investigate the relationship of this oligodendroglioma with the in vivo microenvironment.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Isocitrate Dehydrogenase/genetics , Oligodendroglioma/genetics , Repressor Proteins/genetics , Animals , Base Sequence , DNA Fingerprinting , DNA Primers/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mice, Inbred NOD , Mice, SCID , Microarray Analysis , Molecular Sequence Data , RNA-Binding Proteins , Sequence Analysis, DNA , Transplantation, Heterologous
5.
Acta Neuropathol ; 125(5): 683-98, 2013 May.
Article in English | MEDLINE | ID: mdl-23429996

ABSTRACT

Angiogenesis is regarded as a hallmark of cancer progression and it has been postulated that solid tumor growth depends on angiogenesis. At present, however, it is clear that tumor cell invasion can occur without angiogenesis, a phenomenon that is particularly evident by the infiltrative growth of malignant brain tumors, such as glioblastomas (GBMs). In these tumors, amplification or overexpression of wild-type (wt) or truncated and constitutively activated epidermal growth factor receptor (EGFR) are regarded as important events in GBM development, where the complex downstream signaling events have been implicated in tumor cell invasion, angiogenesis and proliferation. Here, we show that amplification and in particular activation of wild-type EGFR represents an underlying mechanism for non-angiogenic, invasive tumor growth. Using a clinically relevant human GBM xenograft model, we show that tumor cells with EGFR gene amplification and activation diffusely infiltrate normal brain tissue independent of angiogenesis and that transient inhibition of EGFR activity by cetuximab inhibits the invasive tumor growth. Moreover, stable, long-term expression of a dominant-negative EGFR leads to a mesenchymal to epithelial-like transition and induction of angiogenic tumor growth. Analysis of human GBM biopsies confirmed that EGFR activation correlated with invasive/non-angiogenic tumor growth. In conclusion, our results indicate that activation of wild-type EGFR promotes invasion and glioblastoma development independent of angiogenesis, whereas loss of its activity results in angiogenic tumor growth.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Genes, erbB-1/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Transcriptional Activation , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Cetuximab , ErbB Receptors/drug effects , ErbB Receptors/genetics , Gene Amplification , Humans , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic , Xenograft Model Antitumor Assays
6.
Cancer Res ; 70(11): 4274-9, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20460538

ABSTRACT

Although CD133 has been proposed as a marker for brain tumor-initiating cells, studies show that a tumorigenic potential exists among CD133(-) glioma cells as well. However, it is not established whether the ability of CD133(-) cells to form tumors is a property confined to a small subpopulation, rather than a common trait associated with most glioma cell types. Thus, we used lentiviral vectors expressing green fluorescent protein under lineage-specific promoters to identify CD133(-) glioma cells expressing Nestin, glial fibrillary acidic protein (GFAP), and neuron-specific enolase (NSE). Flow cytometry analysis showed the presence of CD133(-) subpopulations expressing these markers in glioma cell lines and in primary cultures from human glioblastoma (GBM) biopsies. Moreover, analysis of cell cycle distribution showed that subgroups expressing Nestin, GFAP, and NSE uniformly contained actively cycling cells, when cultured in serum-containing medium and stem cell medium. These subpopulations were fluorescence-activated cell sorted from CD133(-) U373 glioma cells and implanted intracerebrally in severe combined immunodeficient mice. Moreover, we implanted Nestin-, GFAP-, and NSE-positive glioma cells sorted from a human GBM biopsy, following removal of CD133-positive cells. All the CD133(-) subpopulations produced tumors, with no significant differences in survival or tumor take rates. However, there was a trend toward lower take rates for CD133(-) glioma subpopulations expressing GFAP and NSE. These findings suggest that the ability to form tumors may be a general trait associated with different glioma cell phenotypes, rather than a property limited to an exclusive subpopulation of glioma stem cells.


Subject(s)
Biomarkers, Tumor/biosynthesis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , AC133 Antigen , Animals , Antigens, CD/biosynthesis , Brain Neoplasms/classification , Cell Growth Processes/physiology , Cell Line, Tumor , Genetic Vectors , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Glioblastoma/classification , Glycoproteins/biosynthesis , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Intermediate Filament Proteins/biosynthesis , Intermediate Filament Proteins/genetics , Lentivirus/genetics , Mice , Mice, SCID , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nestin , Peptides , Phosphopyruvate Hydratase/biosynthesis , Phosphopyruvate Hydratase/genetics
7.
PLoS One ; 4(7): e6381, 2009 Jul 28.
Article in English | MEDLINE | ID: mdl-19636430

ABSTRACT

Tumor hypoxia is relevant for tumor growth, metabolism and epithelial-to-mesenchymal transition (EMT). We report that hyperbaric oxygen (HBO) treatment induced mesenchymal-to-epithelial transition (MET) in a dimethyl-alpha-benzantracene induced mammary rat adenocarcinoma model, and the MET was associated with extensive coordinated gene expression changes and less aggressive tumors. One group of tumor bearing rats was exposed to HBO (2 bar, pO(2) = 2 bar, 4 exposures à 90 minutes), whereas the control group was housed under normal atmosphere (1 bar, pO(2) = 0.2 bar). Treatment effects were determined by assessment of tumor growth, tumor vascularisation, tumor cell proliferation, cell death, collagen fibrils and gene expression profile. Tumor growth was significantly reduced (approximately 16%) after HBO treatment compared to day 1 levels, whereas control tumors increased almost 100% in volume. Significant decreases in tumor cell proliferation, tumor blood vessels and collagen fibrils, together with an increase in cell death, are consistent with tumor growth reduction and tumor stroma influence after hyperoxic treatment. Gene expression profiling showed that HBO induced MET. In conclusion, hyperoxia induced MET with coordinated expression of gene modules involved in cell junctions and attachments together with a shift towards non-tumorigenic metabolism. This leads to more differentiated and less aggressive tumors, and indicates that oxygen per se might be an important factor in the "switches" of EMT and MET in vivo. HBO treatment also attenuated tumor growth and changed tumor stroma, by targeting the vascular system, having anti-proliferative and pro-apoptotic effects.


Subject(s)
Adenocarcinoma/pathology , Epithelial Cells/cytology , Hyperoxia/drug therapy , Mesoderm/cytology , Models, Biological , Animals , Female , Immunohistochemistry , Rats , Rats, Sprague-Dawley
8.
APMIS ; 117(3): 212-21, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19245594

ABSTRACT

Lymphangiogenesis is an important event in progression of colorectal cancer (CRC), and the estimated lymphatic vascular density (LVD) probably indicates facilitated lymphatic tumor cell invasion and metastasis. However, at what time point during tumor progression this process is triggered, is unclear. The aim of this study was twofold. Firstly, to examine LVD in paired samples of CRC tissue and normal mucosa with specific emphasis on possible difference in LVD between tumors stages II and III, and secondly, the expression of the lymphangiogenic growth factor fibroblast growth factor-2 (FGF-2). Eighteen patients were studied. Immunostaining for podoplanin was performed to highlight lymphatic vessels. FGF-2 mRNA expression was determined by quantitative real-time RT-PCR, whereas protein expression was quantitatively assessed by densitometric analysis of Western blot signal intensity. The immunoblots were further validated by FGF-2 immunostaining of histological sections. LVD was significantly increased in tumor tissue compared with the normal mucosa but no changes in LVD between stages II and III CRC was observed. FGF-2 was found to be downregulated both at the mRNA and protein level in tumor tissues compared with normal mucosa. Lymphangiogenesis was triggered early in tumor development. An increased LVD was established before the tumor reached stage II. FGF-2 was downregulated in tumor tissue. The importance of this finding remains unclear.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Fibroblast Growth Factor 2/metabolism , Intestinal Mucosa/pathology , Lymphangiogenesis , Lymphatic Vessels/pathology , Adenocarcinoma/metabolism , Aged , Aged, 80 and over , Blotting, Western , Colorectal Neoplasms/metabolism , Disease Progression , Down-Regulation , Female , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/genetics , Humans , Intestinal Mucosa/metabolism , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
9.
BMC Cancer ; 8: 48, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18261235

ABSTRACT

BACKGROUND: It has been suggested that a small population of cells with unique self-renewal properties and malignant potential exists in solid tumors. Such "cancer stem cells" have been isolated by flow cytometry, followed by xenograft studies of their tumor-initiating properties. A frequently used sorting marker in these experiments is the cell surface protein CD133 (prominin-1). The aim of this work was to examine the distribution of CD133 in pancreatic exocrine cancer. METHODS: Fifty-one cases of pancreatic ductal adenocarcinomas were clinically and histopathologically evaluated, and immunohistochemically investigated for expression of CD133, cytokeratin 19 and chromogranin A. The results were interpreted on the background of CD133 expression in normal pancreas and other normal and malignant human tissues. RESULTS: CD133 positivity could not be related to a specific embryonic layer of organ origin and was seen mainly at the apical/endoluminal surface of non-squamous, glandular epithelia and of malignant cells in ductal arrangement. Cytoplasmic CD133 staining was observed in some non-epithelial malignancies. In the pancreas, we found CD133 expressed on the apical membrane of ductal cells. In a small subset of ductal cells and in cells in centroacinar position, we also observed expression in the cytoplasm. Pancreatic ductal adenocarcinomas showed a varying degree of apical cell surface CD133 expression, and cytoplasmic staining in a few tumor cells was noted. There was no correlation between the level of CD133 expression and patient survival. CONCLUSION: Neither in the pancreas nor in the other investigated organs can CD133 membrane expression alone be a criterion for "stemness". However, there was an interesting difference in subcellular localization with a minor cell population in normal and malignant pancreatic tissue showing cytoplasmic expression. Moreover, since CD133 was expressed in shed ductal cells of pancreatic tumors and was found on the surface of tumor cells in vessels, this molecule may have a potential as clinical marker in patients suffering from pancreatic cancer.


Subject(s)
Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Glycoproteins/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Peptides/metabolism , AC133 Antigen , Cell Membrane/metabolism , Chromogranin A/metabolism , Cytoplasm/metabolism , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Keratin-19/metabolism , Reference Values , Stem Cells/metabolism , Tissue Distribution
10.
Int J Cancer ; 121(7): 1401-9, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17640040

ABSTRACT

Colorectal cancer (CRC) represents the second most common cause of cancer mortality in the western world. The tumors frequently show metastatic spread which affects different organs such as lymph nodes, liver and lungs. Although the pattern of spread may vary, the initial step usually involves dissemination to regional lymph nodes. At present it is clear that neovessel formation, including lymphangiogenesis, represents key events in tumor progression. However, to what extent lymphangiogenesis contributes in the progression of CRC is unclear. This work focuses on recent progress within the field of tumor lymphangiogenesis with special reference to CRC, and on novel therapeutic strategies for anti-lymphangiogenic therapies. Inhibition of metastatic spread may be achieved by restriction of lymphatic vessel growth by using targeted therapeutic strategies towards molecules involved in lymphangiogenic signalling. Such adjuvant therapeutic approaches in addition to existing therapeutic strategies may represent a favourable treatment for CRCs with higher than average risk of disease recurrence and progression.


Subject(s)
Colorectal Neoplasms/pathology , Lymphangiogenesis , Colorectal Neoplasms/therapy , Disease Progression , Humans , Models, Biological , Prognosis , RNA Interference , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics
11.
Surg Neurol ; 68(2): 133-43; discussion 143-4, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17537489

ABSTRACT

BACKGROUND: The IF protein nestin and the RNA-binding protein musashi are expressed by neural progenitor cells during CNS development. Their expression in glial tumors was evaluated by immunohistochemistry, and the histopathological scores correlated with levels of cysteine cathepsins that are known prognostic markers in several tumors. METHODS: The levels of nestin, musashi, and cathepsins B and L were assessed by immunohistochemical analysis of biopsies from 87 patients with primary CNS tumors. To confirm the immunohistochemical data, nestin expression was analyzed by real-time PCR in 12 brain tumor biopsies. The exact location of nestin-positive cells was determined by mapping the distribution of nestin in a highly invasive human glioma xenograft model. RESULTS: Immunostaining revealed nestin to be expressed in 95.8% and musashi in 80% of the patient biopsies. The total IHC score for nestin was significantly higher in high- than in low-grade tumors (P < .0001). No difference was observed for musashi (P = .11). Real-time PCR of nestin expression confirmed the immunohistochemical data. Nestin expression was shown to be a strong prognostic marker for decreased overall survival (P = .0001), whereas musashi expression has no prognostic significance. Moreover, nestin was shown by Cox regression analysis to be a stronger prognostic marker than cathepsins B and L. IHC staining of nestin in a xenograft model showed that its expression is localized mainly in the invasive tumor cells at the tumor periphery. CONCLUSIONS: Nestin is shown to be a strong prognostic marker for glioma malignancy. The presented data links the invasive glioma cells to CNS precursor cells, indicating that the most malignant cells in the gliomas may well be closely related to the glioma stem cells.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Adolescent , Adult , Aged , Animals , Biomarkers, Tumor/genetics , Brain Neoplasms/mortality , Cathepsin B/metabolism , Cathepsin L , Cathepsins/metabolism , Child , Child, Preschool , Cysteine Endopeptidases/metabolism , Female , Glioma/mortality , Humans , Intermediate Filament Proteins/genetics , Male , Middle Aged , Nerve Tissue Proteins/genetics , Nestin , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Rats , Survival Rate
12.
J Neurosurg ; 104(1): 118-28, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16509155

ABSTRACT

OBJECT: Conflicting results have been reported concerning the antitumor efficacy of the angiogenesis inhibitor endostatin. This may be due to differences in the biological distribution of endostatin between studies or to the varying biological efficacies of the different protein forms that were examined. To address this issue, the authors used a local delivery approach in which each tumor cell secreted endostatin, providing uniform endostatin levels throughout the tumors. This allowed a direct assessment of the biological efficacy of soluble endostatin in vivo. METHODS: The authors genetically engineered BT4C gliosarcoma cells so that they would stably express and secrete either the human or murine form of endostatin. Endostatin-producing cells or mock-infected cells were implanted intracerebrally in syngeneic BD-IX rats. The antitumor efficacy of endostatin was evaluated on the basis of survival data and tumor volume comparisons. In addition, microvascular parameters were assessed. The authors confirmed the continuous release of endostatin by the BT4C cells. A magnetic resonance imaging-assisted comparison of tumor volumes revealed that local production of murine endostatin significantly inhibited tumor growth. Notably, 40% of the animals in this treatment group experienced long-term survival without histologically verifiable tumors 7 months after cell implantation. After local treatment with murine endostatin, tumor blood plasma volumes were reduced by 71%, microvessel density counts by 84%, and vascular area fractions by 75%. In contrast, human endostatin did not inhibit tumor growth significantly in this model. Centrally located regions of necrosis were present in tumors secreting both the human and the murine species-specific form of endostatin. CONCLUSIONS: The results suggest that endostatin inhibits tumor angiogenesis in vivo in a species-specific manner.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Brain Neoplasms/drug therapy , Cell Transplantation , Endostatins/administration & dosage , Endostatins/therapeutic use , Gliosarcoma/pathology , Amino Acid Sequence , Angiogenesis Inhibitors/metabolism , Animals , Endostatins/metabolism , Genetic Engineering , Gliosarcoma/genetics , Humans , Mice , Molecular Sequence Data , Neovascularization, Pathologic , Rats , Survival , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...