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.
Cytotherapy ; 14(4): 451-60, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22242831

ABSTRACT

BACKGROUND AIMS: Drug-resistance genes have been explored as powerful in vivo selection markers in hematopoietic cell gene therapy, and cytidine deaminase (CDD) represents a particularly attractive candidate given the virtual absence of non-hematopoietic side-effects after low/intermediate dose application of the associated drug cytosine-arabinoside (Ara-C). METHODS: We investigated the in vivo selection potential of CDD overexpression and prolonged low/intermediate-dose Ara-C application in a murine model. Furthermore, non-transplanted mice were utilized to study Ara-C toxicity in different hematopoietic cell compartments. RESULTS: Significant protection of myelo- and thrombopoiesis and up to 6-fold in vivo enrichment of CDD-transduced hematopoietic cells was observed. Enrichment was most robust early after Ara-C application and was correlated with dosage and duration of chemotherapy. Enrichment remained significant for several weeks, indicating selection at the level of a progenitor population. This notion was supported by Ara-C toxicity studies, demonstrating profound hematotoxicity and a marked delay in hematopoietic recovery, specifically in the progenitor/stem cell compartment after low/intermediate-dose Ara-C. CONCLUSIONS: These data support the concept of CDD/Ara-C as a clinically applicable in vivo selection system in hematopoietic gene therapy. The data also demonstrate marked differences in hematotoxicity between alternative Ara-C dosing schemes and suggest thorough in vivo toxicity studies to optimize further Ara-C dosing en route to safe and stable enrichment of gene-corrected hematopoiesis.


Subject(s)
Cytarabine/pharmacology , Cytidine Deaminase/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Cytidine Deaminase/genetics , Female , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Mice , Mice, Inbred C57BL
2.
Mol Ther ; 16(4): 757-64, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18334984

ABSTRACT

Pulmonary alveolar proteinosis (PAP) due to deficiency of the common beta-chain (beta(c)) of the interleukin-3 (IL-3)/IL-5/granulocyte-macrophage colony-stimulating factor (GM-CSF) receptors is a rare monogeneic disease characterized by functional insufficiency of pulmonary macrophages. Hematopoietic stem cell gene therapy for restoring expression of beta(c)-protein in the hematopoietic system may offer a curative approach. Toward this end, we generated a retroviral construct expressing the murine beta(c) (mbeta(c)) gene and conducted investigations in a murine model of beta(c)-deficient PAP. Functional correction of mbeta(c) activity in mbeta(c)(-/-) bone marrow (BM) cells was demonstrated by restoration of in vitro colony formation in response to GM-CSF. In addition, in a murine in vivo model of mbeta(c)-deficient PAP mbeta(c) gene transfer to hematopoietic stem cells not only restored the GM-CSF-sensitivity of hematopoietic progenitor cells but also, within a period of 12 weeks, almost completely reversed the morphologic features of surfactant accumulation. These results were obtained despite modest transduction levels (10-20%) and, in comparison to wild-type mice, clearly reduced beta(c) expression levels were detected in hematopoietic cells. Therefore, our data demonstrating genetic and functional correction of mbeta(c)(-/-) deficiency in vitro as well as in a murine in vivo model of PAP strongly suggest gene therapy as a potential new treatment modality in beta(c)-deficient PAP.


Subject(s)
Cytokine Receptor Common beta Subunit/biosynthesis , Hematopoietic Stem Cells/metabolism , Pulmonary Alveolar Proteinosis/therapy , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Cells, Cultured , Colony-Forming Units Assay , Cytokine Receptor Common beta Subunit/genetics , Genetic Therapy , Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/pathology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Surfactants/metabolism , Retroviridae/genetics
3.
Mol Ther ; 16(4): 757-764, 2008 Apr.
Article in English | MEDLINE | ID: mdl-28178466

ABSTRACT

Pulmonary alveolar proteinosis (PAP) due to deficiency of the common ß-chain (ßc) of the interleukin-3 (IL-3)/IL-5/granulocyte-macrophage colony-stimulating factor (GM-CSF) receptors is a rare monogeneic disease characterized by functional insufficiency of pulmonary macrophages. Hematopoietic stem cell gene therapy for restoring expression of ßc-protein in the hematopoietic system may offer a curative approach. Toward this end, we generated a retroviral construct expressing the murine ßc (mßc) gene and conducted investigations in a murine model of ßc-deficient PAP. Functional correction of mßc activity in mßc-/- bone marrow (BM) cells was demonstrated by restoration of in vitro colony formation in response to GM-CSF. In addition, in a murine in vivo model of mßc-deficient PAP mßc gene transfer to hematopoietic stem cells not only restored the GM-CSF-sensitivity of hematopoietic progenitor cells but also, within a period of 12 weeks, almost completely reversed the morphologic features of surfactant accumulation. These results were obtained despite modest transduction levels (10-20%) and, in comparison to wild-type mice, clearly reduced ßc expression levels were detected in hematopoietic cells. Therefore, our data demonstrating genetic and functional correction of mßc-/- deficiency in vitro as well as in a murine in vivo model of PAP strongly suggest gene therapy as a potential new treatment modality in ßc-deficient PAP.

4.
Intervirology ; 50(3): 197-203, 2007.
Article in English | MEDLINE | ID: mdl-17283445

ABSTRACT

OBJECTIVE: Retroviral vectors represent one of the most robust technologies for in vivo expression of heterologous gene sequences and are still the most commonly used vectors in clinical gene therapy trials. The production of high titer retroviral preparations, however, can be a problematic procedure for certain constructs. METHODS: GALV- or RD114-pseudotyped retroviral particles carrying selectable fluorescence markers or drug resistance genes, such as the green fluorescent protein (GFP) or the O(6)-methylguanine-DNA-methyltransferase (MGMT) mutants, were used to stably transduce Phoenix-(FNX-)eco cells. Thereafter, a polyclonal population of producer cells was generated by enriching cells with high marker gene expression. In addition, single producer clones were selected by limiting dilution. RESULTS: Retroviral titers were increased 1-2 logs by enriching for a polyclonal population of producer cells, and selection of single producer clones allowed another 1- to 2-log increase in titers. Using this method, reproducibly high titer viral preparations allowing efficient transduction of hematopoietic stem cells were generated. CONCLUSION: A reliable and time-effective method to generate stable high titer producer cells based on the FNX-cell line for problematic retroviral vector constructs is described.


Subject(s)
Cell Line/virology , Genetic Therapy/methods , Genetic Vectors , Retroviridae/growth & development , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Hematopoietic Stem Cells , Humans , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Reproducibility of Results , Transduction, Genetic
5.
Blood ; 108(9): 2965-71, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16835384

ABSTRACT

Hematopoietic stem cell gene transfer of the drug-resistance gene cytidine deaminase (CDD) protecting cells from the cytotoxic cytidine analogs cytarabine and gemcitabine was investigated in a murine transplant model. Following transplantation of CDD-transduced cells and cytarabine application (500 mg/kg; days 1-4; intraperitoneally) significant myeloprotection was demonstrated with nadir counts of peripheral blood granulocytes and thrombocytes of 2.9 +/- 0.6/nL versus 0.7 +/- 0.1/nL (P < .001) and 509 +/- 147/nL versus 80 +/- 9/nL (P = .008), respectively (CDD versus control). Protection also was observed from otherwise lethal gemcitabine treatment (250 mg/kg; days 1-3). Stable levels of gene-marked cells in primary and secondary recipients were demonstrated for up to 9 months, and whereas CDD overexpression clearly reduced B- and T-lymphocyte numbers, no major toxicity was observed in the myeloid compartment. Despite the profound myeloprotective properties, however, CDD overexpression did not allow for pharmacologic enrichment of transduced hematopoiesis in our model. Thus, in summary, our data establish CDD as a drug-resistance gene highly suitable for myeloprotective purposes, which, given the lack of selection observed in our hands, might best be used in combination with selectable drug-resistance genes such as MGMT (P140K) or MDR1.


Subject(s)
Bone Marrow Transplantation/immunology , Cytarabine/toxicity , Cytidine Deaminase/genetics , Deoxycytidine/analogs & derivatives , 3T3 Cells , Animals , Bone Marrow Transplantation/mortality , Colony-Forming Units Assay , Cytidine Deaminase/metabolism , Deoxycytidine/toxicity , Gene Transfer Techniques , Graft Survival/physiology , Leukocyte Count , Mice , Models, Animal , Platelet Count , Recombinant Fusion Proteins/metabolism , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...