Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Neuropathol Appl Neurobiol ; 37(2): 189-205, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20819169

ABSTRACT

AIMS: brain metastasis is a common cause of mortality in cancer patients, and associated with poor prognosis. Our objective was to develop a clinically relevant animal model by transplanting human biopsy spheroids derived from metastatic lesions into brains of immunodeficient rats. METHODS: nine different patient brain metastases from four different primary cancers were implanted into brains of immunodeficient rats. The xenografts were compared with patient tumours by magnetic resonance imaging, histochemistry, immunohistochemistry and DNA copy number analysis. RESULTS: after transplantation, tumour growth was achieved in seven out of nine human brain metastases. Spheroids derived from four of the metastases initiated in the rat brains were further serially transplanted into new animals and a 100% tumour take was observed during second passage. Three of the biopsies were implanted subcutaneously, where no tumour take was observed. The animal brain metastases exhibited similar radiological features as observed clinically. Histological comparisons between the primary tumours from the patients, the patient brain metastases and the derived xenografts showed striking similarities in histology and growth patterns. Also, immunohistochemistry showed a strong marker expression similarity between the patient tumours and the corresponding xenografts. DNA copy number analysis between the brain metastases, and the corresponding xenografts revealed strong similarities in gains and losses of chromosomal content. CONCLUSION: we have developed a representative in vivo model for studying the growth of human metastatic brain cancers. The model described represents an important tool to assess responses to new treatment modalities and for studying mechanisms behind metastatic growth in the central nervous system.


Subject(s)
Brain Neoplasms/secondary , Disease Models, Animal , Xenograft Model Antitumor Assays , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Gene Dosage , Humans , Immunohistochemistry , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Rats , Tissue Array Analysis , Tumor Cells, Cultured
2.
Gene Ther ; 17(2): 202-16, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19829315

ABSTRACT

The objective of the present study was to evaluate the cellular effects of the oncolytic HSV-1 based vector, G207, on the tumor microenvironment. We established progressively growing intracerebral xenografts in athymic nude rats generated from three different human GBM surgical specimens. The lesions were identified by MRI and subsequently injected with a concentrated vector stock. The animals were killed 10 or 30 days after G207 injection and the tumors were quantitatively evaluated for virus-induced changes in proliferation, apoptosis and vascularity. Moreover, we assessed vector spread as well as the infiltration pattern of CD68-positive inflammatory cells. In all G207-injected lesions, immunostaining identified widespread regions of viral infection and replication (plaques). Proliferation indices were significantly lower, whereas apoptotic counts were significantly elevated in plaques as compared with that in non-infected areas of the same lesions, as well as in corresponding control xenografts. Furthermore, there was a significant decline in the number of blood vessels in the plaques and the vascular area fractions were reduced. CD68-positive inflammatory cells accumulated in the plaques. The present study highlights the favorable cellular responses to G207 treatment seen from a clinical viewpoint, such as reduced tumor cell proliferation, more frequent events of tumor cell death and a strongly attenuated tumor vascular compartment. However, our results suggest that transduction of a significant volume of tumor tissue is essential, as these beneficial changes were only observed in areas of active viral replication, leaving non-transduced tumor tissues unaffected.


Subject(s)
Brain Neoplasms/blood supply , Brain Neoplasms/therapy , Glioblastoma/therapy , Herpesvirus 1, Human/genetics , Oncolytic Virotherapy/methods , Animals , Cell Death , Cell Line, Tumor , Genetic Vectors , Glioblastoma/blood supply , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Virus Replication
3.
Neuropathol Appl Neurobiol ; 35(4): 380-93, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19508445

ABSTRACT

AIMS: Cancer stem-like cells might have important functions in chemoresistance. We have developed a model where highly infiltrative brain tumours with a stem-like phenotype were established by orthotopic transplantation of human glioblastomas to immunodeficient rats. Serial passaging gradually transformed the tumours into a less invasive and more angiogenic phenotype (high-generation tumours). The invasive phenotype (low-generation tumours) was characterized by an increase in stem cell markers and increased phosphorylation of kinases in the phosphatidylinositol 3-kinase (PI3K)/AKT pathway. These markers were reduced in the serially passaged vascular tumours. The present study was aimed at investigating how the two phenotypes responded in vitro to doxorubicin, a clinically potent cytotoxic drug for solid tumours. METHODS: Biopsy spheroids were implanted and passaged intracranially in nude rats. Gene expression and protein analyses were performed, and drug sensitivity was assessed. RESULTS: Microarray analysis revealed gene ontology categories connected to developmental aspects and negative regulators of differentiation, especially in the infiltrative stem cell-like tumours. The highly invasive stem-like phenotype was chemoresistant compared with the angiogenic phenotype. By interfering with the PI3K it was possible to sensitize tumour spheroids to chemotherapy. Real-time quantitative polymerase chain reaction showed downregulation of the stem cell markers Nestin and Musashi-1 in low-generation biopsy spheroids following PI3K inhibition. CONCLUSIONS: Highly invasive tumours with a stem-like phenotype are more chemoresistant than angiogenic tumours derived from the same patients. We suggest that treatment resistance in glioblastomas can be related to PI3K/AKT activity in stem-like tumour cells, and that targeted interference with the PI3K/AKT pathway might differentiate and sensitize this subpopulation to chemotherapy.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/physiopathology , Glioblastoma/drug therapy , Glioblastoma/physiopathology , Stem Cells/physiology , Animals , Antineoplastic Agents/pharmacology , Brain/drug effects , Brain/physiopathology , Chromones/pharmacology , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/metabolism , Humans , Intermediate Filament Proteins/metabolism , Morpholines/pharmacology , Neoplasm Transplantation , Nerve Tissue Proteins/metabolism , Nestin , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Nude
4.
Oncogene ; 27(39): 5182-94, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18469852

ABSTRACT

Chemoresistance represents a major problem in the treatment of many malignancies. Overcoming this obstacle will require improved understanding of the mechanisms responsible for this phenomenon. The progenitor cell marker NG2/melanoma proteoglycan (MPG) is aberrantly expressed by various tumors, but its role in cell death signaling and its potential as a therapeutic target are largely unexplored. We have assessed cytotoxic drug-induced cell death in glioblastoma spheroids from 15 patients, as well as in five cancer cell lines that differ with respect to NG2/MPG expression. The tumors were treated with doxorubicin, etoposide, carboplatin, temodal, cisplatin and tumor necrosis factor (TNF)alpha. High NG2/MPG expression correlated with multidrug resistance mediated by increased activation of alpha3beta1 integrin/PI3K signaling and their downstream targets, promoting cell survival. NG2/MPG knockdown with shRNAs incorporated into lentiviral vectors attenuated beta1 integrin signaling revealing potent antitumor effects and further sensitized neoplastic cells to cytotoxic treatment in vitro and in vivo. Thus, as a novel regulator of the antiapoptotic response, NG2/MPG may represent an effective therapeutic target in several cancer subtypes.


Subject(s)
Antigens/physiology , Brain Neoplasms/metabolism , Drug Resistance, Neoplasm , Glioma/metabolism , Integrins/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proteoglycans/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Apoptosis/physiology , Brain Neoplasms/pathology , Enzyme Activation , Glioma/pathology , Humans , Tumor Necrosis Factor-alpha/physiology
5.
J Neurooncol ; 85(2): 191-202, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17557137

ABSTRACT

This study describes the biological effects of hyperoxic treatment on BT4C rat glioma xenografts in vivo with special reference to tumor growth, angiogenesis, apoptosis, general morphology and gene expression parameters. One group of tumor bearing animals was exposed to normobaric hyperoxia (1 bar, pO(2) = 1.0) and another group was exposed to hyperbaric hyperoxia (2 bar, pO(2) = 2.0), whereas animals housed under normal atmosphere (1 bar, pO(2) = 0.2) served as controls. All treatments were performed at day 1, 4 and 7 for 90 min. Treatment effects were determined by assessment of tumor growth, vascular morphology (immunostaining for von Willebrand factor), apoptosis by TUNEL staining and cell proliferation by Ki67 staining. Moreover, gene expression profiles were obtained and verified by real time quantitative PCR. Hyperoxic treatment caused a approximately 60% reduction in tumor growth compared to the control group after 9 days (p < 0.01). Light microscopy showed that the tumors exposed to hyperoxia contained large "empty spaces" within the tumor mass. Moreover, hyperoxia induced a significant increase in the fraction of apoptotic cells ( approximately 21%), with no significant change in cell proliferation. After 2 bar treatment, the mean vascular density was reduced in the central parts of the tumors compared to the control and 1 bar group. The vessel diameters were significantly reduced (11-24%) in both parts of the tumor tissue. Evidence of induced cell death and reduced angiogenesis was reflected by gene expression analyses.Increased pO(2)-levels in experimental gliomas, using normobaric and moderate hyperbaric oxygen therapy, caused a significant reduction in tumor growth. This process is characterized by enhanced cell death, reduced vascular density and changes in gene expression corresponding to these effects.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Hyperbaric Oxygenation , Hyperoxia/metabolism , Neovascularization, Pathologic/prevention & control , Animals , Apoptosis/physiology , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/physiology , Glioma/blood supply , Glioma/pathology , Hyperoxia/pathology , Male , Neoplasms, Experimental , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Oxygen/metabolism , Oxygen/therapeutic use , RNA, Neoplasm/analysis , Random Allocation , Rats , Rats, Nude
6.
Neuropathol Appl Neurobiol ; 32(4): 419-27, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16866987

ABSTRACT

Tumour spheroids initiated from glioma biopsy specimens provide a valuable three-dimensional cell culture system that share several biological features of malignant brain tumours in situ. Upon xenotransplantation in immunodeficient rats, tumours derived from such spheroids exhibit a highly infiltrative growth. Successful cryopreservation of spheroid specimens therefore represents an excellent tool for future comparative studies of tumour growth and progression. Thus, if frozen stocks of human glioma spheroids can be established, similar to those obtained from cancer cell lines, it would ease the planning of biopsy-based experiments. In this context, it is crucial that cryopreservation does not alter the biological behaviour of the tumour spheroids. The biopsy spheroids were frozen to -40 degrees Celsius, stored for 1 week at -196 degrees Celsius, thawed rapidly and cultured for 1 week. The viability of the spheroids was compared against controls using a two-colour fluorescence assay, which demonstrated that cryopreservation was well tolerated. Using an in vitro invasion assay, it is shown that the freezing procedures did not affect the spheroids ability to invade a collagen gel. Cryopreserved and control tumour spheroids were equally tumourogenic, and produced overlapping survival curves when transplanted into the brains of immunocompromised rats. Immunohistochemical analyses showed no significant changes regarding microvessel density or proliferation index. Furthermore, gene expression profiling using a macroarray system detected no significant changes following cryopreservation. The present data show that cryopreservation is well tolerated, and represent a methodologically reliable storage method for biopsy spheroids that can be used in experimental studies at later time points.


Subject(s)
Brain Neoplasms/pathology , Cryopreservation , Glioma/pathology , Spheroids, Cellular , Animals , Biopsy , Brain Neoplasms/genetics , Cell Survival , Cryopreservation/methods , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Glioma/genetics , Humans , Immunohistochemistry , Neoplasm Invasiveness/pathology , Neoplasm Transplantation/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Phenotype , Rats
7.
Oncogene ; 25(37): 5103-12, 2006 Aug 24.
Article in English | MEDLINE | ID: mdl-16636676

ABSTRACT

Invasion of brain tumor cells has made primary malignant brain neoplasms among the most recalcitrant to therapeutic strategies. We tested whether the secreted protein Slit2, which guides the projection of axons and developing neurons, could modulate brain tumor cell invasion. Slit2 inhibited the invasion of medulloblastoma cells in a variety of in vitro models. The effect of Slit2 was inhibited by the Robo ectodomain. Time-lapse videomicroscopy indicated that Slit2 reduced medulloblastoma invasion rate without affecting cell direction or proliferation. Both medulloblastoma and glioma tumors express Robo1 and Slit2, but only medulloblastoma invasion is inhibited by recombinant Slit2 protein. Downregulation of activated Cdc42 may contribute to this differential response. Our findings reinforce the concept that neurodevelopmental cues such as Slit2 may provide insights into brain tumor invasion.


Subject(s)
Medulloblastoma/pathology , Neoplasm Invasiveness/prevention & control , Nerve Tissue Proteins/physiology , Animals , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/pathology , Cell Division/drug effects , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Coculture Techniques , Culture Media, Conditioned , Glioma/pathology , Humans , Intercellular Signaling Peptides and Proteins , Kinetics , Medulloblastoma/genetics , Mice , Microscopy, Video , Nerve Tissue Proteins/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Receptors, Immunologic/genetics , Receptors, Immunologic/physiology , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Roundabout Proteins
8.
Neuroimage ; 29(3): 965-76, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16253523

ABSTRACT

Tumour angiogenesis is a tightly regulated process involving cross-talk between tumour cells and the host tissue. The underlying mechanisms that regulate such interactions remain largely unknown. NG2 is a transmembrane proteoglycan whose presence on transformed cells has been demonstrated to increase proliferation in vitro and angiogenesis in vivo. To study the effects of NG2 during tumour growth and progression, we engineered an NG2 positive human glioma cell line (U251-NG2) from parental NG2 negative cells (U251-WT) and implanted both cell types stereotactically into immunodeficient nude rat brains. The tumours were longitudinally monitored in vivo using multispectral MRI employing two differently sized contrast agents (Gd-DTPA-BMA and Gadomer) to assess vascular leakiness, vasogenic oedema, tumour volumes and necrosis. Comparisons of Gd-DTPA-BMA and Gadomer revealed differences in their spatial distribution in the U251-NG2 and U251-WT tumours. The U251-NG2 tumours exhibited a higher leakiness of the larger molecular weight Gadomer and displayed a stronger vasogenic oedema (69.9 +/- 15.2, P = 0.018, compared to the controls (10.7 +/- 7.7). Moreover, immunohistochemistry and electron microscopy revealed that the U251-NG2 tumours had a higher microvascular density (11.81 +/- 0.54; P = 0.0010) compared to controls (5.76 +/- 0.87), with vessels that displayed larger gaps between the endothelial cells. Thus, tumour cells can regulate both the function and structure of the host-derived tumour vasculature through NG2 expression, suggesting a role for NG2 in the cross-talk between tumour-host compartments.


Subject(s)
Antigens/biosynthesis , Antigens/physiology , Brain Neoplasms/pathology , Neovascularization, Pathologic/pathology , Proteoglycans/biosynthesis , Proteoglycans/physiology , Animals , Antigens/genetics , Brain Neoplasms/physiopathology , Capillaries/pathology , Cell Line, Tumor , Gene Expression/physiology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Microscopy, Electron, Transmission , Neoplasm Transplantation , Neovascularization, Pathologic/physiopathology , Proteoglycans/genetics , Rats , Rats, Nude , Transplantation, Heterologous
9.
Cell Transplant ; 13(1): 35-44, 2004.
Article in English | MEDLINE | ID: mdl-15040603

ABSTRACT

The potential benefit of continuous local administration of antiangiogenic proteins to CNS tumors in vivo has recently been demonstrated using endostatin-producing recombinant cells encapsulated in alginate beads. Due to the treatment potential of transplanted alginate-encapsulated cells producing therapeutic proteins, we describe a successful method of cryopreservation (CP) of such beads, in which cellular viability, alginate structure, and protein secretion were maintained. Alginate beads containing human embryonic kidney cells (HEK 293 cells) stably transfected with the gene encoding for endostatin were cryopreserved in dimethyl sulfoxide (DMSO) using a slow freezing procedure. Briefly, the DMSO concentration was gradually increased prior to the freezing procedure. The cryotubes were further supercooled to -7.5 degrees C and nucleated. Thereafter, the samples were cooled at a rate of 0.25 degrees C/min and stored in liquid nitrogen. The viability of the encapsulated cells was assessed using confocal microscopy quantification (CLSM) technique and a MTS assay. The cell cycle distribution inside the beads was assessed by DNA flow cytometry and endostatin production was determined by an endostatin-specific ELISA assay, both prior to and after CP. CLSM measurements showed sustained esterase activity in the beads after thawing, with only a slight transient decrease 24 h after CP. The MTS assay verified these findings by displaying similar variations of intracellular dehydrogenase activity. Flow cytometric analyses revealed no cryorelated disturbances in cellular ploidy. Furthermore, ELISA measurements showed a well-preserved endostatin production after CP. In conclusion, this work describes the successful CP of alginate-encapsulated recombinant cells secreting a therapeutic protein. Together with previous published reports, these results further substantiate the feasibility and potential of cell encapsulation therapy in the treatment of malignant tumors.


Subject(s)
Alginates , Cell Transplantation/methods , Glucuronic Acid , Hexuronic Acids , Angiogenesis Inhibitors , Capsules , Cell Line , Cryopreservation/methods , Dimethyl Sulfoxide , Endostatins/genetics , Humans , Image Processing, Computer-Assisted , Kidney/cytology , Microscopy, Confocal , Transfection
10.
Acta Neurochir Suppl ; 88: 137-41, 2003.
Article in English | MEDLINE | ID: mdl-14531571

ABSTRACT

Despite aggressive surgery and post-operative radiation and chemotherapy, the prognosis is poor for glioblastoma patients. Anti-angiogenic therapy with compounds such as endostatin could delay the onset of relapse. However, the short systemic half-life of this proteins as well as the blood-brain barrier makes the use of this therapy difficult for brain cancer patients. The aim of this project is to develop and implant genetically engineered producer cells secreting endostatin that are encapsulated in calcium cross-linked alginate gel beads. Encapsulation of cells within alginate gels has a potential as a sustained release system in addition to the fact that the encapsulation technology protects the cells from rejection by the immune system. Human embryonal kidney 293 cells have been transfected with the gene for endostatin. These cells have been encapsulated in calcium cross-linked alginate gels and optimized for the secretion of endostatin. Alginate gel beads implanted into rat brain have shown only a moderate loss in cell viability but extended endostatin release for periods of up to 12 months. Visualization of the anti-angiogenic effect on C6 rat glioma growth, tumor vasculature and microhemodynamics has been demonstrated by using intravital video microscopy. The data indicates that endostatin greatly affects tumor-associated microcirculation but does not appear to affect normal microcirculation. The local delivery of endostatin seems to specifically affect tumor-associated microvessels by reduction of the vessel density, diameter and functionality. Tumor cell migration and invasion was greatly reduced in the endostatin treated animals.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Brain Neoplasms/drug therapy , Cell Transplantation , Endostatins/administration & dosage , Gene Targeting , Glioma/drug therapy , Neovascularization, Pathologic/drug therapy , Alginates , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Animals , Brain Neoplasms/blood supply , Capsules , Cell Line , Drug Delivery Systems , Endostatins/genetics , Endostatins/metabolism , Glioma/blood supply , Humans , Microcirculation/drug effects , Rats , Transfection
11.
J Clin Oncol ; 21(8): 1624-36, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12697889

ABSTRACT

Tumors of glial origin consist of a core mass and a penumbra of invasive, single cells, decreasing in numbers towards the periphery and still detectable several centimeters away from the core lesion. Several decades ago, the diffuse nature of malignant gliomas was recognized by neurosurgeons when super-radical resections using hemispherectomies failed to eradicate these tumors. Local invasiveness eventually leads to regrowth of a recurrent tumor predominantly adjacent to the resection cavity, which is not significantly altered by radiation or chemotherapy. This raises the question of whether invasive glioma cells activate cellular programs that render these cells resistant to conventional treatments. Clinical and experimental data demonstrate that glioma invasion is determined by several independent mechanisms that facilitate the spread of these tumors along different anatomic and molecular structures. A common denominator of this cellular behavior may be cell motility. Gene-expression profiling showed upregulation of genes related to motility, and functional studies demonstrated that cell motility contributes to the invasive phenotype of malignant gliomas. There is accumulating evidence that invasive glioma cells show a decreased proliferation rate and a relative resistance to apoptosis, which may contribute to chemotherapy and radiation resistance. Interestingly, interference with cell motility by different strategies results in increased susceptibility to apoptosis, indicating that this dynamic relationship can potentially be exploited as an anti-invasive treatment paradigm. In this review, we discuss mechanisms of glioma invasion, characteristics of the invasive cell, and consequences of this cellular phenotype for surgical resection, oncologic treatments, and future perspectives for anti-invasive strategies.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/therapy , Glioma/pathology , Glioma/therapy , Brain Neoplasms/genetics , Brain Neoplasms/surgery , Cell Movement , Glioma/genetics , Glioma/surgery , Humans , Neoplasm Invasiveness , Phenotype
12.
Neuropathol Appl Neurobiol ; 28(5): 367-80, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12366818

ABSTRACT

Glial precursor cells express NG2 and GD3 in the developing brain. These antigens are both over-expressed during neoplasia, which suggests they may have specific functions in the malignant progression of human brain tumours. This study describes the expression of NG2 and GD3 in 28 paediatric and adult brain tumours. Glioblastoma biopsy spheroids were also implanted into nude rats to assess the regional distribution of the molecules within the tumour. These xenografts showed extensive infiltration and growth that mimicked the growth patterns of human gliomas in situ. NG2 was identified in 20 out of 28 brain tumours, where the expression was confined to the main mass of the tumour, and was reduced towards the tumour periphery. NG2 was mainly associated with blood vessels on both the pericyte and basement membrane components of the tumour vasculature. Ki67 (MIB-1) labelling indicated that NG2 expression was associated with areas of high cellular proliferation. Conversely, all the tumours expressed GD3, which was present both in the tumour main mass and throughout the periphery. Thus, the expression of NG2 may be indicative of tumour progression and might be an amenable target for future therapeutic interventions.


Subject(s)
Antigens/metabolism , Brain Neoplasms/blood supply , Glioma/blood supply , Neovascularization, Pathologic/metabolism , Pericytes/metabolism , Proteoglycans/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antigens/analysis , Biopsy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Child , Child, Preschool , Female , Glioma/metabolism , Glioma/pathology , Humans , Male , Meningioma/blood supply , Meningioma/metabolism , Meningioma/pathology , Neoplasm Staging , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Pericytes/pathology , Proteoglycans/analysis , Rats , Rats, Nude , Sensitivity and Specificity , Spheroids, Cellular , Tumor Cells, Cultured
13.
Radiother Oncol ; 61(3): 313-20, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11731002

ABSTRACT

BACKGROUND AND PURPOSE: Combretastatin A-4 disodium phosphate (CA-4) enhances thermal damage in s.c. BT(4)An rat gliomas. We currently investigated how CA-4 and hyperthermia affect the tumor microenvironment and neovasculature to disclose how the two treatment modalities interact to produce tumor response. METHODS: By confocal microscopy and immunostaining for von Willebrand factor, we examined the extent of vascular damage subsequent to CA-4 (50 mg/kg) and hyperthermia (waterbath 44 degrees C, 60 min). The influence on tumor oxygenation was assessed using interstitial pO(2)-probes (Licox system) and by immunostaining for pimonidazole. We examined the direct effect of CA-4 on the tumor cell population by flow cytometry (cell cycle distribution) and immunostaining for beta-tubulin (cytoskeletal damage). RESULTS: Whereas slight vascular damage was produced by CA-4 in the BT(4)An tumors, local hyperthermia exhibited moderate anti-vascular activity. In tumors exposed to CA-4 3 h before hyperthermia, massive vascular damage ensued. CA-4 reduced the pO(2) from 36.1 to 17.6 mmHg (P=0.01) in the tumor base, and tumor hypoxia increased slightly in the tumor center (pimonidazole staining). Extensive tumor hypoxia developed subsequent to hyperthermia or combination therapy. Despite a profound influence on beta-tubulin organization in vitro, CA-4 had no significant effect on the cell cycle distribution in vivo. CONCLUSION: Our results indicate that the anti-vascular activity exhibited by local hyperthermia can be augmented by previous exposure to CA-4.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Glioma/blood supply , Glioma/therapy , Hyperthermia, Induced/adverse effects , Stilbenes/therapeutic use , Animals , Combined Modality Therapy , Female , Male , Rats , Rats, Inbred Strains
14.
Int J Radiat Oncol Biol Phys ; 51(2): 535-44, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567830

ABSTRACT

PURPOSE: The antivascular and antitumor activity of vinblastine and hyperthermia at different tumor volumes were examined in the subcutaneous (s.c.) BT(4)An rat glioma model. METHODS AND MATERIALS: The influence of vinblastine (3 mg/kg) and hyperthermia (44 degrees C/60 min) on tumor growth was assessed in small (100 mm(3)) and large (200 mm(3)) BT(4)An tumors. To disclose how vinblastine and hyperthermia interacted in the neoplasms, tumor blood flow and the extent of vascular damage, hypoxia, cell proliferation, and apoptosis were assessed after treatment. The content of smooth muscle cells/pericytes in the tumor vasculature was examined in small and large tumors to assess how the vascular phenotype changed during tumor growth. RESULTS: In the large tumors, vinblastine reduced the blood flow, but the tumor growth was not affected. The combination of drug and local heating yielded massive vascular damage and a significant tumor response. The small neoplasms had a higher content of smooth muscle cells/pericytes in the vessel walls (host vasculature), and the tumor vasculature displayed a higher resistance to vascular damage than the large neoplasms. Yet, vinblastine alone exhibited a potent antiproliferative activity and induced massive apoptosis in the small tumors, and the drug significantly inhibited tumor growth. The addition of hyperthermia yielded no additional growth delay in the small tumors. CONCLUSION: The antivascular properties of vinblastine and hyperthermia can be exploited to facilitate vascular damage in BT(4)An solid tumors with a low content of host vasculature.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/therapy , Glioma/therapy , Hyperthermia, Induced , Neovascularization, Pathologic/therapy , Vinblastine/therapeutic use , Animals , Apoptosis , Brain Neoplasms/blood supply , Cell Division , Cell Hypoxia , Combined Modality Therapy , Female , Glioma/blood supply , In Situ Nick-End Labeling , Male , Phenotype , Rats
15.
Cancer Res ; 61(18): 6830-7, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559558

ABSTRACT

The current study describes new, antivascular, and antitumor effects of human endostatin. A novel system for continuous, localized delivery of antiangiogenic compounds to brain tumors was used. The delivery system was composed of endostatin-producing 293 cells encapsulated into immuno-isolating sodium alginate. Intravital multifluorescence microscopy was used to assess vascular and antitumor effects of endostatin in C6 glioma spheroids implanted into an ectopic as well as an orthotopic setting. Analysis of total and functional vascular density, microvascular diameters, vessel perfusion, tumor growth, and tumor cell migration were performed repetitively. Tumor growth was reduced by 35% in treated animals. It was of interest that tumor cell invasion into the surrounding tissue was also inhibited. The total vascular density was reduced by 67.6%, perfusion by 67%, and vessel diameters by 37%. This resulted in a significant reduction in tumor perfusion, although the vessel permeability was not influenced. We have demonstrated that human endostatin not only reduces total vascular density, as shown previously, but also greatly reduces the functionality and the diameters of the vessels. Furthermore, we show that this therapeutic approach also inhibits tumor cell invasion, thus supporting the hypothesis that tumor angiogenesis and invasion represent two interrelated processes. Finally, this work further confirms the new therapeutic concept using alginate cell-encapsulation technology for the localized delivery of therapeutic compounds to central nervous system malignancies.


Subject(s)
Alginates/administration & dosage , Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Collagen/administration & dosage , Drug Delivery Systems/methods , Glioma/drug therapy , Peptide Fragments/administration & dosage , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Division/drug effects , Cell Movement/drug effects , Collagen/genetics , Drug Carriers/administration & dosage , Endostatins , Female , Glioma/blood supply , Glioma/pathology , Glucuronic Acid , Hexuronic Acids , Humans , Male , Mice , Mice, Nude , Microscopy, Fluorescence , Microscopy, Video , Neoplasm Invasiveness , Neovascularization, Pathologic/drug therapy , Peptide Fragments/genetics , Rats , Spheroids, Cellular , Transfection , Xenograft Model Antitumor Assays
16.
Radiother Oncol ; 60(2): 147-54, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11439209

ABSTRACT

BACKGROUND AND PURPOSE: Attacking tumor vasculature is a promising approach for the treatment of solid tumors. The tubulin inhibitor combretastatin A-4 disodium phosphate (CA-4) is a new vascular targeting drug which displays a low toxicity profile. We wanted to investigate how CA-4 influences tumor perfusion in the BT4An rat glioma and how the vascular targeting properties of CA-4 could be exploited to augment hyperthermic damage towards tumor vasculature. MATERIAL AND METHODS: We used the (86)RbCl extraction technique to assess how CA-4 influences tumor perfusion, and the tumor endothelium was examined for morphological changes induced by the drug. We combined CA-4 (50 mg/kg i.p.) with hyperthermia (44 degrees C, 60 min) at different time intervals to evaluate how therapy should be designed to affect tumor growth, and we studied the tumors histologically to assess tissue viability. RESULTS: We found that CA-4 induced a profound, but transient reduction in tumor perfusion 3-6 h postinjection. If hyperthermia was administered 3-6 h after injecting CA-4, massive hemorrhagic necrosis developed, and tumor response was significantly enhanced compared to simultaneous administration of the two treatment modalities (P<0.005). CA-4 alone had no influence on tumor growth and failed to disrupt the vasculature of the BT4An solid tumors. Interestingly though, a mild endothelial edema was observed in some tumor areas 3 h after injecting CA-4. CONCLUSIONS: We conclude that the combination of CA-4 and hyperthermia is a potent therapeutic option for BT4An tumors, but the selection of adequate time intervals between CA-4 and hyperthermia are imperative to obtain tumor response.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Glioma/therapy , Hyperthermia, Induced , Stilbenes/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Combined Modality Therapy , Female , Glioma/blood supply , Glioma/pathology , Hyperthermia, Induced/adverse effects , Male , Neoplasm Transplantation , Rats , Rats, Inbred Strains , Stilbenes/administration & dosage
17.
Neuro Oncol ; 3(3): 201-10, 2001 07.
Article in English | MEDLINE | ID: mdl-11465401

ABSTRACT

Gene therapy using viral vectors has to date failed to reveal its definitive clinical usefulness. Cell encapsulation technology represents an alternative, nonviral approach for the delivery of biologically active compounds to tumors. This strategy involves the use of genetically engineered producer cells that secrete a protein with therapeutic potential. The cells are encapsulated in an immunoisolating material that makes them suitable for transplantation. The capsules, or bioreactors, permit the release of recombinant proteins that may assert their effects in the tumor microenvironment. During the last decades, there has been significant progress in the development of encapsulation technologies that comprise devices for both macro- and microencapsulation. The polysaccharide alginate is the most commonly used material for cell encapsulation and is well tolerated by various tissues. A wide spectrum of cells and tissues has been encapsulated and implanted, both in animals and humans, indicating the general applicability of this approach for both research and medical purposes, including CNS malignancies. Gliomas most frequently recur at the resection site. To provide local and sustained drug delivery, the bioreactors can be implanted in the brain parenchyma or in the ventricular system. The development of comprehensive analyses of geno- and phenotypic profiles of a tumor (genomics and proteomics) may provide new and important guidelines for choosing the optimal combination of bioreactors and recombinant proteins for therapeutic use.


Subject(s)
Alginates/administration & dosage , Biocompatible Materials/administration & dosage , Bioreactors , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Genetic Therapy/methods , Cerebral Ventricles , Delayed-Action Preparations , Drug Delivery Systems , Glucuronic Acid , Hexuronic Acids , Humans , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use
18.
Nat Biotechnol ; 19(1): 29-34, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135548

ABSTRACT

We describe a technique for the treatment of malignant brain tumors based on local delivery of the anti-angiogenic protein endostatin from genetically engineered cells encapsulated in ultrapure sodium alginate. Alginate consists of L-guluronic and D-mannuronic acid, which in the presence of divalent cations forms an extended gel network, in which cells reside and remain immunoisolated, when implanted into the rat brain. Here, we show that endostatin-transfected cells encapsulated in alginate maintain endostatin secretion for at least four months after intracerebral implantation in rats. During the implantation period 70% of the encapsulated cells remained viable, as opposed to 85% in in vitro-cultured capsules. Rats that received transplants of BT4C glioma cells, together with endostatin-producing capsules (0.2 microg/ml per capsule), survived 84% longer than the controls. The endostatin released from the capsules led to an induction of apoptosis, hypoxia, and large necrotic avascular areas within 77% of the treated tumors, whereas all the controls were negative. The encapsulation technique may be used for many different cell lines engineered to potentially interfere with the complex microenvironment in which tumor and normal cells reside. The present work may thus provide the basis for new therapeutic approaches toward brain tumors.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Brain Neoplasms/therapy , Collagen/therapeutic use , Glioma/therapy , Peptide Fragments/therapeutic use , Alginates , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/toxicity , Animals , Apoptosis , Biocompatible Materials , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Capsules , Cell Hypoxia , Cell Line , Cell Survival/drug effects , Cell Transplantation , Collagen/genetics , Collagen/toxicity , Endostatins , Glioma/blood supply , Glioma/pathology , Glucuronic Acid , Hexuronic Acids , Humans , Magnetic Resonance Imaging , Peptide Fragments/genetics , Peptide Fragments/toxicity , Rats , Transfection , Transplantation, Heterologous
19.
Neuropathol Appl Neurobiol ; 27(6): 451-64, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11903928

ABSTRACT

This work describes the in vivo expression and distribution of glioma-associated gangliosides (GD3, GM2, 3'-isoLM1) in a novel human brain tumour nude rat xenograft model. In this model, the tumours, which are established directly from human glioblastoma biopsies, show extensive infiltrative growth within the rat brain. This model therefore provides an opportunity to study ganglioside expression not only within the macroscopic tumour, but also in brain areas with tumour cell infiltration. The ganglioside expression was studied by confocal microscopy of immunostained brain sections using antiganglioside monoclonal antibodies. Xenografts from four human glioblastoma multiformes were established in rats and the brains removed after 3-4 months. Ganglioside GD3 was expressed in the tumour parenchyma while ganglioside 3'-isoLM1 was more abundantly expressed in the periphery of the tumour associated with areas of tumour cell invasion. GM2 expression was only seen in one tumour, where it was located within the main tumour mass. Double staining with a pan antihuman monoclonal antibody (3B4) and the antiganglioside monoclonal antibodies confirmed that the ganglioside expression was associated with tumour cells. This work supports the concept of different biological roles for individual gangliosides and indicates that antibodies or ligands directed against GD3 and 3'-isoLM1 might be complementary when applied in the treatment of human glioblastomas.


Subject(s)
Brain Neoplasms/metabolism , Gangliosides/analysis , Glioblastoma/metabolism , Animals , Antibodies, Monoclonal , Antigens, Tumor-Associated, Carbohydrate/analysis , Antigens, Tumor-Associated, Carbohydrate/biosynthesis , Brain Injuries/metabolism , Brain Neoplasms/chemistry , Disease Models, Animal , Fluorescent Antibody Technique , Gangliosides/biosynthesis , Gangliosides/immunology , Glioblastoma/chemistry , Humans , Microscopy, Confocal , Neoplasm Transplantation , Rats , Rats, Nude , Transplantation, Heterologous , Wounds, Stab/metabolism
20.
Neoplasia ; 3(6): 469-79, 2001.
Article in English | MEDLINE | ID: mdl-11774029

ABSTRACT

Bromelain is an aqueous extract from pineapple stem that contains proteinases and exhibits pleiotropic therapeutic effects, i.e., antiedematous, antiinflammatory, antimetastatic, antithrombotic, and fibrinolytic activities. In this study, we tested bromelain's effects on glioma cells to assess whether bromelain could be a potential contributor to new antiinvasive strategies for gliomas. Several complementary assays demonstrated that bromelain significantly and reversibly reduced glioma cell adhesion, migration, and invasion without affecting cell viability, even after treatment periods extending over several months. Immunohistochemistry and immunoblotting experiments demonstrated that alpha3 and beta1 integrin subunits and hyaluronan receptor CD44 protein levels were reduced within 24 hours of bromelain treatment. These effects were not reflected at the RNA level because RNA profiling did not show any significant effects on gene expression. Interestingly, metabolic labelling with 35-S methionine demonstrated that de novo protein synthesis was greatly attenuated by bromelain, in a reversible manner. By using a transactivating signaling assay, we found that CRE-mediated signaling processes were suppressed. These results indicate that bromelain exerts its antiinvasive effects by proteolysis, signaling cascades, and translational attenuation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/pathology , Bromelains/pharmacology , Glioma/pathology , Animals , Antigens, CD/analysis , Brain/cytology , Brain/embryology , Cell Adhesion/drug effects , Cell Aggregation , Cell Movement/drug effects , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Extracellular Matrix/metabolism , Gene Expression Profiling , Genes, Reporter , Green Fluorescent Proteins , Humans , Hyaluronan Receptors/analysis , Integrin alpha3 , Integrin beta1/analysis , Integrins/analysis , Luminescent Proteins/biosynthesis , Neoplasm Invasiveness/prevention & control , Neoplasm Proteins/analysis , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Inbred Strains , Recombinant Fusion Proteins/biosynthesis , Signal Transduction/drug effects , Spheroids, Cellular/cytology , Transcriptional Activation/drug effects , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...