Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Pediatr Surg ; 44(1): 204-10; discussion 210-1, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19159744

ABSTRACT

PURPOSE: Osteoprotegerin (OPG) inhibits osteoclast activation and reduces osteolysis in bone tumors. We hypothesized that tumor-tropic neural progenitor cells (NPCs) engineered to express OPG would reduce neuroblastoma disease burden in the bone. METHODS: Stable expression of green fluorescent protein (NPC-GFP) and OPG (NPC-OPG) was established in human NPCs by lentivirus-mediated transduction. Bone disease was established by intrafemoral injection of luciferase-expressing human neuroblastoma (CHLA-255) cells into 20 SCID mice. Three weeks later, mice began receiving intravenous injection of 2 x 10(6) NPC-OPG or NPC-GFP (control) every 10 days x 3 doses. Disease was monitored with quantitative bioluminescence imaging and x-ray images, which were evaluated on a scale of 0 to 4. These studies were approved by the Institutional Animal Care and Use Committee. RESULTS: Osteoprotegerin treatment in vitro produced no direct toxicity to tumor cells. Coculture of tumor cells with bone marrow significantly increased activation of bone marrow-derived osteoclasts as assessed by tartrate-resistant acid phosphatase staining (156 +/- 10.8 osteoclasts per well) compared to bone marrow culture alone (91.67 +/- 4.7, P = .005). This increase was abrogated by adding OPG-containing media (68.3 +/- 2.8, P = .001). NPC-OPG slowed tumor progression (108-fold increase from pretreatment) compared to mice treated with NPC-GFP (538-fold), as judged by bioluminescence imaging. X-rays subjectively demonstrated less bone disease in NPC-OPG-treated mice (2.27 +/- 0.25) compared to NPC-GFP-treated mice (3.25 +/- 0.22, P = .04). CONCLUSIONS: Neural progenitor cell-mediated delivery of OPG slowed disease progression in a preclinical model of neuroblastoma bone metastasis. The decrease in bone disease was not from direct tumor cell toxicity but likely occurred indirectly through inhibition of osteoclast-directed bone resorption. Thus, targeted delivery of OPG by NPCs may be effective in the treatment of neuroblastoma bone metastasis.


Subject(s)
Bone Neoplasms/pathology , Neoplasm Metastasis/prevention & control , Neuroblastoma/pathology , Osteoprotegerin/pharmacology , Stem Cells/metabolism , Animals , Cell Line, Tumor , Disease Progression , Green Fluorescent Proteins/pharmacology , Humans , In Vitro Techniques , Mice , Mice, SCID , Statistics, Nonparametric , Transduction, Genetic
2.
PLoS One ; 3(11): e3629, 2008.
Article in English | MEDLINE | ID: mdl-18978946

ABSTRACT

The actin cytoskeleton is a primary determinant of tumor cell motility and metastatic potential. Motility and metastasis are thought to be regulated, in large part, by the interaction of membrane proteins with cytoplasmic linker proteins and of these linker proteins, in turn, with actin. However, complete membrane-to-actin linkages have been difficult to identify. We used co-immunoprecipitation and competitive peptide assays to show that intercellular adhesion molecule-2 (ICAM-2)/alpha-actinin/actin may comprise such a linkage in neuroblastoma cells. ICAM-2 expression limited the motility of these cells and redistributed actin fibers in vitro, and suppressed development of disseminated tumors in an in vivo model of metastatic neuroblastoma. Consistent with these observations, immunohistochemical analysis demonstrated ICAM-2 expression in primary neuroblastoma tumors exhibiting features that are associated with limited metastatic disease and more favorable clinical outcome. In neuroblastoma cell lines, ICAM-2 expression did not affect AKT activation, tumorigenic potential or chemosensitivity, as has been reported for some types of transfected cells. The observed ICAM-2-mediated suppression of metastatic phenotype is a novel function for this protein, and the interaction of ICAM-2/alpha-actinin/actin represents the first complete membrane-linker protein-actin linkage to impact tumor cell motility in vitro and metastatic potential in an in vivo model. Current work focuses on identifying specific protein domains critical to the regulation of neuroblastoma cell motility and metastasis and on determining if these domains represent exploitable therapeutic targets.


Subject(s)
Actins/metabolism , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Actinin/metabolism , Animals , Antigens, CD/physiology , Cell Adhesion Molecules/physiology , Cell Movement/physiology , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasm Staging , Phenotype , Protein Binding , Transplantation, Heterologous , Tumor Cells, Cultured
3.
Ann Surg Oncol ; 15(11): 3259-67, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18726131

ABSTRACT

BACKGROUND: We have shown that continuous systemic delivery of interferon beta (IFN-beta) remodels dysfunctional tumor vasculature, thereby improving tumor perfusion and enhancing delivery and efficacy of chemotherapeutic drugs. We hypothesized that because of their inherent tumor tropism, neural progenitor cells (NPCs) engineered to express IFN-beta could also effect maturation of tumor vasculature without generating high systemic levels of IFN-beta. METHODS: Mice with luciferase-expressing disseminated human neuroblastoma were divided into four groups of equal tumor burden by bioluminescence imaging: (1) untreated controls; (2) NPC-IFN-beta only; (3) cyclophosphamide (CTX) only; and (4) NPC-IFN-beta in combination with CTX. Two million NPC-IFN-beta cells were administered twice, 7 days apart, starting 21 days after tail vein administration of tumor cells. CTX was administered every 6 days for three doses. Mice were killed at 6 weeks, livers and kidneys weighed, and tumor removed for immunohistochemical staining for endothelial cells (CD34), pericytes (alpha-SMA), apoptosis (TUNEL [terminal deoxynucleotidyl transferase dUTP nick-end labeling]), and diI-labeled NPCs. RESULTS: Fluorescent-labeled NPCs confirmed localization of these cells to tumors. The alpha-SMA/CD34 ratio, a marker for vascular maturation, greatly increased in NPC-IFN-beta-treated tumors compared with controls. Bioluminescent signal from luciferase-expressing tumor cells, reflecting tumor burden, was lower with combination therapy than control or either monotherapy, and combination therapy resulted in far less tumor burden by weight in the kidneys and liver. CONCLUSIONS: Targeted delivery of IFN-beta with NPCs produced low circulating levels of IFN-beta, yet the maturing effect on the tumor vasculature and the enhanced efficacy of adjuvant therapy was maintained. Thus, combination therapy of NPC-IFN-beta with CTX warrants further investigation for the treatment of high-risk neuroblastoma patients.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Antiviral Agents/administration & dosage , Cyclophosphamide/therapeutic use , Interferon-beta/administration & dosage , Neuroblastoma/therapy , Neurons/physiology , Stem Cells/physiology , Adenoviridae/genetics , Animals , Cells, Cultured , Disease Models, Animal , Drug Delivery Systems/methods , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Injections, Intravenous , Male , Mice , Mice, SCID , Neuroblastoma/genetics , Neuroblastoma/pathology , Tumor Burden
4.
Cancer Res ; 67(1): 22-5, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17210679

ABSTRACT

Neural stem cells and progenitor cells migrate selectively to tumor loci in vivo. We exploited the tumor-tropic properties of HB1.F3.C1 cells, an immortalized cell line derived from human fetal telencephalon, to deliver the cDNA encoding a secreted form of rabbit carboxylesterase (rCE) to disseminated neuroblastoma tumors in mice. This enzyme activates the prodrug CPT-11 more efficiently than do human enzymes. Mice bearing multiple tumors were treated with rCE-expressing HB1.F3.C1 cells and schedules of administration of CPT-11 that produced levels of active drug (SN-38) tolerated by patients. Both HB1.F3.C1 cells and CPT-11 were given i.v. None of the untreated mice and 30% of mice that received only CPT-11 survived long term. In contrast, 90% of mice treated with rCE-expressing HB1.F3.C1 cells and 15 mg/kg CPT-11 survived for 1 year without detectable tumors. Plasma carboxylesterase activity and SN-38 levels in mice receiving both rCE-expressing HB1.F3.C1 cells (HB1.F3.C1/AdCMVrCE) and CPT-11 were comparable with those in mice receiving CPT-11 only. These data support the hypothesis that the antitumor effect of the described neural stem/progenitor cell-directed enzyme prodrug therapy (NDEPT) is mediated by production of high concentrations of active drug selectively at tumor sites, thereby maximizing the antitumor effect of CPT-11. NDEPT approaches merit further investigation as effective, targeted therapy for metastatic tumors. We propose that the described approach may have greatest use for eradicating minimum residual disease.


Subject(s)
Camptothecin/analogs & derivatives , Carboxylesterase/metabolism , Genetic Therapy/methods , Neuroblastoma/therapy , Prodrugs/pharmacology , Telencephalon/physiology , Adenoviridae/genetics , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Carboxylesterase/biosynthesis , Carboxylesterase/genetics , Cell Line, Tumor , Combined Modality Therapy , DNA, Complementary/administration & dosage , DNA, Complementary/genetics , Disease-Free Survival , Humans , Irinotecan , Mice , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Neuroblastoma/genetics , Prodrugs/pharmacokinetics , Telencephalon/cytology , Telencephalon/enzymology , Transduction, Genetic , Xenograft Model Antitumor Assays
5.
PLoS One ; 1: e23, 2006 Dec 20.
Article in English | MEDLINE | ID: mdl-17183650

ABSTRACT

BACKGROUND: Patients diagnosed with metastatic cancer have almost uniformly poor prognoses. The treatments available for patients with disseminated disease are usually not curative and have side effects that limit the therapy that can be given. A treatment that is selectively toxic to tumors would maximize the beneficial effects of therapy and minimize side effects, potentially enabling effective treatment to be administered. METHODS AND FINDINGS: We postulated that the tumor-tropic property of stem cells or progenitor cells could be exploited to selectively deliver a therapeutic gene to metastatic solid tumors, and that expression of an appropriate transgene at tumor loci might mediate cures of metastatic disease. To test this hypothesis, we injected HB1.F3.C1 cells transduced to express an enzyme that efficiently activates the anti-cancer prodrug CPT-11 intravenously into mice bearing disseminated neuroblastoma tumors. The HB1.F3.C1 cells migrated selectively to tumor sites regardless of the size or anatomical location of the tumors. Mice were then treated systemically with CPT-11, and the efficacy of treatment was monitored. Mice treated with the combination of HB1.F3.C1 cells expressing the CPT-11-activating enzyme and this prodrug produced tumor-free survival of 100% of the mice for >6 months (P<0.001 compared to control groups). CONCLUSIONS: The novel and significant finding of this study is that it may be possible to exploit the tumor-tropic property of stem or progenitor cells to mediate effective, tumor-selective therapy for metastatic tumors, for which no tolerated curative treatments are currently available.


Subject(s)
Camptothecin/analogs & derivatives , Neoplasm Metastasis/therapy , Animals , Base Sequence , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Cell Line , Cell Line, Tumor , DNA Primers/genetics , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/transplantation , Humans , Irinotecan , Mice , Mice, SCID , Multipotent Stem Cells/enzymology , Multipotent Stem Cells/transplantation , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Neuroblastoma/secondary , Neuroblastoma/therapy , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transduction, Genetic , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...