Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
Mol Cancer Ther ; 5(6): 1577-84, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16818517

ABSTRACT

Enzyme-prodrug approaches to cancer therapy, theoretically, have the potential to mediate tumor-selective cytotoxicity. However, even if tumor-specific prodrug activation is achieved, enzyme-prodrug systems investigated thus far comprised a single enzyme and a specific prodrug. Although targeted, such systems constitute single-agent therapy, which may be ineffective and/or may promote development of drug resistance. Therefore, a goal of our laboratories was to design and characterize a novel dipiperidinyl derivative of etoposide [1,4'-dipiperidine-1'-carboxylate-etoposide (dp-VP16)] that would act as a prodrug. We envisioned that dp-VP16 would be converted to the active chemotherapeutic agent VP-16 by the same rabbit carboxylesterase (rCE) that we have previously shown to efficiently activate the prodrug irinotecan (CPT-11). This dp-VP16 prodrug might then be used in combination with CPT-11, with both drugs activated by a single enzyme. We evaluated the ability of pure rCE and two human carboxylesterases, hCE1 and hiCE (hCE2), to activate dp-VP16 in vitro, and in neuroblastoma cell lines designed to express/overexpress each enzyme. In SK-N-AS neuroblastoma cell transfectants, expression of rCE or hiCE decreased the IC50 of dp-VP16 as a single agent by 8.3- and 3.4-fold, respectively, in growth inhibition assays. Purified hCE1 did not metabolize dp-VP16 in vitro and did not affect its IC50 in intact cells. The combination indices of sequential exposure to CPT-11 followed by dp-VP16 ranged from approximately 0.4 to 0.6, suggesting that this combination produced greater-than-additive cytotoxicity in neuroblastoma cells expressing rCE. These data provide proof-of-principle that enzyme-prodrug therapy approaches comprised of prodrugs with complementary mechanisms of cytotoxicity that are activated by a single enzyme can be developed.


Subject(s)
Carboxylesterase/antagonists & inhibitors , Etoposide/therapeutic use , Neuroblastoma/drug therapy , Prodrugs/therapeutic use , Animals , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Carboxylesterase/metabolism , Catalysis , Cell Division/drug effects , Humans , Inhibitory Concentration 50 , Irinotecan , Molecular Structure , Neuroblastoma/enzymology , Rabbits , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...