Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2423: 165-177, 2022.
Article in English | MEDLINE | ID: mdl-34978698

ABSTRACT

Oncolytic virotherapy translational research in the current era is heavily focused on the interaction of the immune system and tumor microenvironment with oncolytic viruses. Preclinical xenograft studies using human cells in immunodeficient mouse models does not serve this purpose. As a consequence, developing syngeneic immunocompetent murine cancer models sensitive to infection and growth of specific oncolytic viruses is required. The group 3 subtype of medulloblastoma, among the four molecular subgroups-WNT, SHH, Group 3, and Group 4, has the worst prognosis and the poorest outcome. Sadly, current treatments cause long-term toxicity and morbidity to survivors adversely affecting their quality of life. Alternate effective therapy with less side effects is urgently needed. We have shown that oncolytic measles virus (MV) is effective against localized as well as CSF-disseminated medulloblastoma in immunodeficient mouse models. To study the interaction of immune system with oncolytic measles virotherapy, we have developed a murine group 3 medulloblastoma cell line (CSCG) that is infectible by MV, is killed by MV, allows replication of MV, and is tumorigenic in the brain of syngeneic transgenic immune-competent mice. Intratumoral injection of MV results in significant prolongation of survival in mice bearing CSCG tumors in the brain. This model provides the first suitable platform to examine therapeutic regimens of MV therapy for MB tumors in the presence of intact immune system. Here, we describe our lab protocols to develop this cell line and the mouse model.


Subject(s)
Cerebellar Neoplasms , Measles , Medulloblastoma , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Cell Line, Tumor , Cerebellar Neoplasms/therapy , Humans , Measles/therapy , Measles virus/genetics , Medulloblastoma/pathology , Medulloblastoma/therapy , Mice , Mice, Nude , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Quality of Life , Tumor Microenvironment , Xenograft Model Antitumor Assays
2.
Neuro Oncol ; 20(12): 1606-1615, 2018 11 12.
Article in English | MEDLINE | ID: mdl-29912438

ABSTRACT

Background: Oncolytic measles virus (MV) is effective in xenograft models of many tumor types in immune-compromised mice. However, no murine cell line exists that is tumorigenic, grows in immune-competent mice, and is killed by MV. The lack of such a model prevents an examination of the effect of the immune system on MV oncotherapy. Methods: Cerebellar stem cells from human CD46-transgenic immunocompetent mice were transduced to express Sendai virus C-protein, murine C-Myc, and Gfi1b proteins. The resultant cells were injected into the brain of NSG mice, and a cell line, called CSCG, was prepared from the resulting tumor. Results: CSCG cells are highly proliferative, and express stem cell markers. These cells are permissive for replication of MV and are killed by the virus in a dose- and time-dependent manner. CSCG cells form aggressive tumors that morphologically resemble medulloblastoma when injected into the brains of immune-competent mice. On the molecular level, CSCG tumors overexpress natriuretic peptide receptor 3 and gamma-aminobutyric acid type A receptor alpha 5, markers of Group 3 medulloblastoma. A single intratumoral injection of MV‒green fluorescent protein resulted in complete tumor regression and prolonged survival of animals compared with treatments with phosphate buffered saline (P = 0.0018) or heat-inactivated MV (P = 0.0027). Conclusions: This immune-competent model provides the first platform to test therapeutic regimens of oncolytic MV for Group 3 medulloblastoma in the presence of anti-measles immunity. The strategy presented here can be used to make MV-sensitive murine models of any human tumor for which the driving mutations are known.


Subject(s)
Cerebellar Neoplasms/therapy , Disease Models, Animal , Immunocompetence , Measles virus/genetics , Medulloblastoma/therapy , Oncolytic Virotherapy , Animals , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/virology , Humans , Measles/virology , Medulloblastoma/immunology , Medulloblastoma/metabolism , Medulloblastoma/virology , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Cells, Cultured , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Xenograft Model Antitumor Assays
3.
Mol Ther Oncolytics ; 7: 57-66, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29367943

ABSTRACT

Modified measles virus (MV) has effective oncolytic activity preclinically and is currently being investigated in clinical trials for various types of cancer. We investigated the use of cystine knot proteins (CKPs) to direct MV activity. CKPs are short polypeptides that bind their targets with high affinity. We used a CKP that binds αvß3, αvß5, and α5ß1 integrins with single-digit nanomolar affinity to retarget MV to the integrins (MV-CKPint). MV-CKPint infected, replicated in, and killed human glioblastoma, medulloblastoma, diffuse intrinsic pontine glioma (DIPG), and melanoma cancer cells in vitro, all of which express the target integrins. MV-CKPint activity was competitively blocked by echistatin, an integrin binding peptide. When the CKP was cleaved from the viral H protein at an included protease site, virus activity was abrogated. When delivered intravenously (i.v.), the retargeted virus reached a subcutaneous glioblastoma tumor bed and produced cytopathic effects similar to that shown by intratumoral injection of the virus. Because these target integrins are overexpressed by tumor vascular endothelium, MV-CKPint may allow for effective therapy with i.v. injection. These results indicate for the first time that CKPs can be used to retarget MV for a receptor of choice. In addition, MV-CKPint provides proof of principle for the use of a CKP of interest to retarget any enveloped virus for both oncolytic and gene therapy purposes.

4.
Hum Gene Ther Clin Dev ; 27(4): 145-151, 2016 12.
Article in English | MEDLINE | ID: mdl-27604429

ABSTRACT

The modified Edmonston vaccine strain of measles virus (MV) has shown potent oncolytic efficacy against various tumor types and is being investigated in clinical trials. Our laboratory showed that MV effectively kills medulloblastoma tumor cells in both localized disease and when tumor cells are disseminated through cerebrospinal fluid (CSF). Although the safety of repeated intracerebral injection of modified MV in rhesus macaques has been established, the safety of administering MV into CSF has not been adequately investigated. In this study, we assessed the safety of MV-NIS (MV modified to express the human sodium iodide symporter protein) injected into the CSF of measles-immunized and measles virus-susceptible transgenic (CD46, IFNαRko) mice. Treated animals were administered a single intraventricular injection of 1 × 105 or 1 × 106 TCID50 (50% tissue culture infective dose) of MV-NIS. Detailed clinical observation was performed over a 90-day period. Clinically, we did not observe any measles-related toxic effects or behavioral abnormality in animals of any treated cohort. The complete blood count and blood chemistry analysis results were found to be within normal range for all the cohorts. Histologic examination of brains and spinal cords revealed inflammatory changes, mostly related to the needle track; these resolved by day 21 postinjection. To assess viral biodistribution, quantitative RT-PCR to detect the measles virus N-protein was performed on blood and brain samples. Viral RNA was not detectable in the blood as soon as 2 days after injection, and virus cleared from the brain by 45 days postadministration in all treatment cohorts. In conclusion, our data suggest that a single injection of modified MV into the CSF is safe and can be used in future therapeutic applications.


Subject(s)
Measles virus/pathogenicity , Measles/therapy , Membrane Cofactor Protein/physiology , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Receptor, Interferon alpha-beta/physiology , Symporters/physiology , Animals , Female , Humans , Injections, Intraventricular , Male , Measles/immunology , Measles/virology , Mice , Mice, Transgenic
5.
Sci Rep ; 6: 27012, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27255663

ABSTRACT

Risk or presence of metastasis in medulloblastoma causes substantial treatment-related morbidity and overall mortality. Through the comparison of cytokines and growth factors in the cerebrospinal fluid (CSF) of metastatic medulloblastoma patients with factors also in conditioned media of metastatic MYC amplified medulloblastoma or leptomeningeal cells, we were led to explore the bioactivity of IGF1 in medulloblastoma by elevated CSF levels of IGF1, IGF-sequestering IGFBP3, IGFBP3-cleaving proteases (MMP and tPA), and protease modulators (TIMP1 and PAI-1). IGF1 led not only to receptor phosphorylation but also accelerated migration/adhesion in MYC amplified medulloblastoma cells in the context of appropriate matrix or meningothelial cells. Clinical correlation suggests a peri-/sub-meningothelial source of IGF-liberating proteases that could facilitate leptomeningeal metastasis. In parallel, studies of key factors responsible for cell autonomous growth in MYC amplified medulloblastoma prioritized IGF1R inhibitors. Together, our studies identify IGF1R as a high value target for clinical trials in high risk medulloblastoma.


Subject(s)
Cerebellar Neoplasms/cerebrospinal fluid , Medulloblastoma/cerebrospinal fluid , Meningeal Neoplasms/cerebrospinal fluid , Receptors, Somatomedin/metabolism , Adolescent , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/cerebrospinal fluid , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/pathology , Child , Drug Screening Assays, Antitumor , Female , Gene Expression , Humans , Inhibitory Concentration 50 , Insulin-Like Growth Factor Binding Protein 3/cerebrospinal fluid , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/cerebrospinal fluid , Insulin-Like Growth Factor I/genetics , Male , Matrix Metalloproteinase 9/cerebrospinal fluid , Matrix Metalloproteinase 9/genetics , Medulloblastoma/drug therapy , Medulloblastoma/secondary , Meningeal Neoplasms/drug therapy , Meningeal Neoplasms/secondary , Molecular Targeted Therapy , Plasminogen Activator Inhibitor 1/cerebrospinal fluid , Plasminogen Activator Inhibitor 1/genetics , Receptor, IGF Type 1 , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/genetics , Tissue Inhibitor of Metalloproteinase-1/cerebrospinal fluid , Tissue Inhibitor of Metalloproteinase-1/genetics
6.
PLoS One ; 10(7): e0131842, 2015.
Article in English | MEDLINE | ID: mdl-26222911

ABSTRACT

BACKGROUND: We tested the hypothesis that αv-integrin and the human epidermal growth factor receptor type 2 (HER2) interact with each other in brain trophic metastatic breast cancer cells and influence their invasive phenotype. METHODS: Clones of MDA-MB231BR human breast cancer cells with stable knock down of αv-integrin in combination with high or low levels of HER2 were created. The interactions of these two proteins and their combined effect on cell migration and invasion were investigated in vitro and in vivo. RESULTS: Knockdown of αv-integrin in MDA-MB231BR clones altered the actin cytoskeleton and cell morphology. HER2 co-precipitated with αv-integrin in three breast cancer cell lines in vitro, suggesting they complex in cells. Knockdown of αv-integrin altered HER2 localization from its normal membrane position to a predominantly lysosomal localization. When αv-integrin expression was decreased by 69-93% in HER2-expressing cells, cellular motility was significantly reduced. Deficiency of both αv-integrin and HER2 decreased cellular migration and invasion by almost 90% compared to cells expressing both proteins (P<0.01). After intracerebral inoculation, cells expressing high levels of both αv-integrin and HER2 showed a diffusely infiltrative tumor phenotype, while cells deficient in αv-integrin and/or HER2 showed a compact tumor growth phenotype. In the αv-integrin positive/HER2 positive tumors, infiltrative growth was 57.2 ± 19% of tumor volume, compared to only 5.8 ± 6.1% infiltration in the double deficient tumor cells. CONCLUSIONS: αv-integrin interacts with HER2 in breast cancer cells and may regulate HER2 localization. The combined impacts of αv-integrin and HER2 influence the invasive phenotype of breast cancer cells. Targeting αv-integrin in HER2-positive breast cancer may slow growth and decrease infiltration in the normal brain.


Subject(s)
Brain/metabolism , Breast Neoplasms/metabolism , Integrin alphaV/metabolism , Receptor, ErbB-2/metabolism , Animals , Brain/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Knockdown Techniques , Heterografts , Humans , Integrin alphaV/genetics , Neoplasm Invasiveness , Neoplasm Transplantation , Rats , Receptor, ErbB-2/genetics
7.
Biochem Biophys Res Commun ; 450(1): 555-60, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24928387

ABSTRACT

Leptomeningeal metastasis is a cause of morbidity and mortality in medulloblastoma, but the understanding of molecular mechanisms driving this process is nascent. In this study, we examined the secretory chemokine profile of medulloblastoma cells (DAOY) and a meningothelial cell line (BMEN1). Conditioned media (CM) of meningothelial cells increased adhesion, spreading and migration of medulloblastoma. VEGFA was identified at elevated levels in the CM from BMEN1 cells (as compared to DAOY CM); however, recombinant VEGFA alone was insufficient to enhance medulloblastoma cell migration. In addition, bevacizumab, the VEGFA scavenging monoclonal antibody, did not block the migratory phenotype induced by the CM. These results reveal that paracrine factors secreted by meningothelial cells can influence migration and adherence of medulloblastoma tumor cells, but VEGFA may not be a specific target for therapeutic intervention in this context.


Subject(s)
Chemokines/immunology , Chemokines/metabolism , Medulloblastoma/immunology , Meninges/immunology , Meninges/metabolism , Meninges/pathology , Vascular Endothelial Growth Factor A/immunology , Cell Communication/immunology , Cell Line, Tumor , Cell Movement/immunology , Humans , Medulloblastoma/pathology
8.
J Neurosci Res ; 90(4): 769-81, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22183788

ABSTRACT

Glioblastoma is an aggressive primary brain tumor with a 5-year survival rate of less than 5%. The ability of glioblastoma cells to invade surrounding brain tissue presents the primary challenge for the success of focal therapeutic approaches. We previously reported that the calcium-activated protease calpain 2 is critical for glioblastoma cell invasion in vitro. Here, we show that expression of calpain 2 is required for the dispersal of glioblastoma cells in a living brain microenvironment. Knockdown of calpain 2 resulted in a 2.9-fold decrease in the invasion of human glioblastoma cells in zebrafish brain. Control cells diffusely migrated up to 450 µm from the site of injection, whereas knockdown cells remained confined in clusters. The invasion study was repeated in organotypic mouse brain tissues, and calpain 2 knockdown cells demonstrated a 2.3-fold lower area of dispersal compared with control cells. In zebrafish brain, glioblastoma cells appeared to migrate in part along the blood vessels of the host. Furthermore, angiogenesis was detected in 27% of zebrafish injected with control cells, whereas only 12.5% of fish receiving knockdown cells showed the formation of new vessels, suggesting a role for calpain 2 in tumor cell angiogenesis. Consistent with the progression of glioblastoma in humans, transplanted tumor cells were not observed to metastasize outside the brain of zebrafish. This study demonstrates that calpain 2 expression is required for the dispersal of glioblastoma cells within the dynamic microenvironment of the brain, identifying zebrafish as a valuable orthotopic system for studying glioblastoma cell invasion.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Calpain/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Animals , Animals, Genetically Modified , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , In Vitro Techniques , Neoplasm Transplantation/methods , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/pathology , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured , Zebrafish
9.
J Pediatr Hematol Oncol ; 34(2): 116-21, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22146535

ABSTRACT

Overexpression of platelet-derived growth factor receptor alpha (PDGFR-A) has been documented in association with primary tumors and metastasis in medulloblastoma. Tumors from our genetically engineered sonic hedgehog-driven medulloblastoma mouse model overexpress PDGFR-A in primary tumors and thus this mouse model is a good platform with which to study the role of PDGFR-A in this central nervous system malignancy. We hypothesized that inhibition of PDGFR-A in medulloblastoma can slow or inhibit tumor progression in living individuals. To test our hypothesis, we targeted PDGFR-A mediated tumor growth in vitro and in vivo using the tyrosine kinase inhibitor, tandutinib (MLN-518), which strongly inhibits PDGFR-A. Although PDGFR-A inhibition by this agent resulted in reduced mouse tumor cell growth and increased apoptosis in vitro, and reduced tumor cell proliferation in vivo, tandutinib did reduce tumor volume at the doses tested (360 mg/kg) in vivo. Thus, tandutinib may be an agent of interest for sonic hedgehog-driven medulloblastoma if a synergistic drug combination can be identified.


Subject(s)
Antineoplastic Agents/pharmacology , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Piperazines/pharmacology , Quinazolines/pharmacology , Animals , Blotting, Western , Cell Separation , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Disease Models, Animal , Flow Cytometry , Immunohistochemistry , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Transgenic , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
10.
Neurochem Res ; 35(11): 1796-804, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20730561

ABSTRACT

Invasion of glioblastoma cells significantly reduces the effectiveness of current treatments, highlighting the importance of understanding dispersal mechanisms and characteristics of the invasive population. Induction of calcium fluxes into glioblastoma cells by autocrine glutamate is critical for invasion. However, the target(s) by which calcium acts to stimulate the dispersal of glioblastoma cells is not clear. In this study, we tested the hypothesis that the calcium-activated protease calpain 2 is required for glioblastoma cell invasion. Knockdown of calpain 2 expression using shRNA or chemical inhibition of calpain activity reduced glioblastoma cell invasion by 90%. Interestingly, decreased expression of calpain 2 did not influence morphology or migration, suggesting regulation of invasion specific mechanisms. Consistent with this idea, 39% less extracellular MMP2 was measured from knockdown cells identifying one mechanism by which calpain 2 mediates glioblastoma cell invasion. This is the first report demonstrating that calpain 2 is required for glioblastoma cell invasion.


Subject(s)
Calpain/metabolism , Glioblastoma/pathology , Matrix Metalloproteinase 2/biosynthesis , Calpain/genetics , Cell Movement/physiology , Gene Knockdown Techniques , Humans , Neoplasm Invasiveness , RNA, Small Interfering/pharmacology
11.
J Biol Chem ; 283(14): 9217-23, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18258589

ABSTRACT

Calpain-mediated proteolysis regulates cytoskeletal dynamics and is altered during aging and the progression of numerous diseases or pathological conditions. Although several cytoskeletal proteins have been identified as substrates, how localized calpain activity is regulated and the mechanisms controlling substrate recognition are not clear. In this study, we report that phosphoinositide binding regulates the susceptibility of the cytoskeletal adhesion protein alpha-actinin to proteolysis by calpains 1 and 2. At first, alpha-actinin did not appear to be a substrate for calpain 2; however, phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P(3)) binding to alpha-actinin resulted in nearly complete proteolysis of the full-length protein, producing stable breakdown products. Calpain 1 was able to cleave alpha-actinin in the absence of phosphoinositide binding; however, PtdIns(3,4,5)P(3) binding increased the rate of proteolysis, and phosphatidylinositol 4,5-diphosphate (PtdIns(4,5)P(2)) binding significantly inhibited cleavage. Phosphoinositide binding appeared to regulate calpain proteolysis of alpha-actinin by modulating the exposure of a highly sensitive cleavage site within the calponin homology 2 domain. In U87MG glioblastoma cells, which contain elevated levels of PtdIns(3,4,5)P(3), alpha-actinin colocalized with calpain within dynamic actin cytoskeletal structures. Furthermore, proteolysis of alpha-actinin producing stable breakdown products was observed in U87MG cells treated with calcium ionophore to activate the calcium-dependent calpains. Additional evidence of PtdIns(3,4,5)P(3)-mediated calpain proteolysis of alpha-actinin was observed in rat embryonic fibroblasts. These results suggest that PtdIns(3,4,5)P(3) binding is a critical determinant for alpha-actinin proteolysis by calpain. In conclusion, phosphoinositide binding to the substrate is a potential mechanism for regulating susceptibility to proteolysis by calpain.


Subject(s)
Actinin/metabolism , Calpain/metabolism , Cytoskeleton/pathology , Phosphatidylinositol Phosphates/metabolism , Actins/metabolism , Animals , Calcium/metabolism , Calpain/antagonists & inhibitors , Cell Line, Tumor , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Rats , Substrate Specificity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...