Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Gene Ther ; 21(2): 148-57, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24257347

ABSTRACT

Reduced intensity conditioning (RIC) is desirable for hematopoietic stem cell (HSC) targeted gene therapy; however, RIC may be insufficient for efficient engraftment and inducing immunological tolerance to transgenes. We previously established long-term gene marking in our rhesus macaque autologous HSC transplantation model following 10 Gy total body irradiation (TBI). In this study, we evaluated RIC transplantation with 4 Gy TBI in two rhesus macaques that received equal parts of CD34(+) cells transduced with green fluorescent protein (GFP)-expressing lentiviral vector and empty vector not expressing transgenes. In both animals, equivalently low gene marking between GFP and empty vectors was observed 6 months post-transplantation, even with efficient transduction of CD34(+) cells in vitro. Autologous lymphocyte infusion with GFP marking resulted in an increase of gene marking in lymphocytes in a control animal with GFP tolerance, but not in the two RIC-transplanted animals. In vitro assays revealed strong cellular and humoral immune responses to GFP protein in the two RIC-transplanted animals, but this was not observed in controls. In summary, 4 Gy TBI is insufficient to permit engraftment of genetically modified HSCs and induce immunological tolerance to transgenes. Our findings should help in the design of conditioning regimens in gene therapy trials.


Subject(s)
Antigens, CD34/metabolism , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Graft Survival/immunology , Graft Survival/radiation effects , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Whole-Body Irradiation/methods , Animals , Cells, Cultured , Combined Modality Therapy , Dose-Response Relationship, Radiation , Hematopoietic Stem Cells/radiation effects , Lentivirus/genetics , Macaca mulatta , Models, Animal , Transduction, Genetic , Transgenes , Transplantation Conditioning
2.
Oral Dis ; 16(3): 269-77, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20374510

ABSTRACT

OBJECTIVES: Salivary glands are useful target organs for local and systemic gene therapeutics. For such applications, the regulation of transgene expression is important. Previous studies by us in murine submandibular glands showed that a rapamycin transcriptional regulation system in a single serotype 2, adeno-associated viral (AAV2) vector was effective for this purpose. This study evaluated if such a vector was similarly useful in rhesus macaque parotid glands. METHODS: A recombinant AAV2 vector (AAV-TF-RhEpo-2.3w), encoding rhesus erythropoietin (RhEpo) and a rapamycin-inducible promoter, was constructed. The vector was administered to macaques at either of two doses [1.5 x 10(11) (low dose) or 1.5 x 10(12) (high dose) vector genomes] via cannulation of Stensen's duct. Animals were followed up for 12-14 weeks and treated at intervals with rapamycin (0.1 or 0.5 mg kg(-1)) to induce gene expression. Serum chemistry, hematology, and RhEpo levels were measured at interval. RESULTS: AAV-TF-RhEpo-2.3w administration led to low levels of rapamycin-inducible RhEpo expression in the serum of most macaques. In five animals, no significant changes were seen in serum chemistry and hematology values over the study. One macaque, however, developed pneumonia, became anemic and subsequently required euthanasia. After the onset of anemia, a single administration of rapamycin led to significant RhEpo production in this animal. CONCLUSION: Administration of AAV-TF-RhEpo-2.3w to macaque parotid glands was generally safe, but led only to low levels of serum RhEpo in healthy animals following rapamycin treatment.


Subject(s)
Gene Expression Regulation/drug effects , Genetic Vectors/administration & dosage , Parotid Gland/metabolism , Sirolimus/pharmacology , Transduction, Genetic , Adenoviridae/genetics , Animals , Dose-Response Relationship, Drug , Erythropoietin/blood , Erythropoietin/genetics , Erythropoietin/metabolism , Macaca mulatta , Male , Promoter Regions, Genetic , Recombinant Proteins , Transgenes
3.
Gene Ther ; 17(1): 50-60, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19759566

ABSTRACT

Salivary glands are potentially useful target sites for multiple clinical applications of gene transfer. Previously, we have shown that serotype 2 adeno-associated viral (AAV2) vectors lead to stable gene transfer in the parotid glands of rhesus macaques. As AAV5 vectors result in considerably greater transgene expression in murine salivary glands than do AAV2 vectors, herein we have examined the use of AAV5 vectors in macaques at two different doses (n = 3 per group; 10(10) or 3 x 10(11) particles per gland). AAV5 vector delivery, as with AAV2 vectors, led to no untoward clinical, hematological or serum chemistry responses in macaques. The extent of AAV5-mediated expression of rhesus erythropoietin (RhEpo) was dose-dependent and similar to that seen with an AAV2 vector. However, unlike results with the AAV2 vector, AAV5 vector-mediated RhEpo expression was transient. Maximal expression peaked at day 56, was reduced by approximately 80% on day 84 and thereafter remained near background levels until day 182 (end of experiment). Quantitative PCR studies of high-dose vector biodistribution at this last time point showed much lower AAV5 copy numbers in the targeted parotid gland (approximately 1.7%) than found with the same AAV2 vector dose. Molecular analysis of the conformation of vector DNA indicated a markedly lower level of concatamerization for the AAV5 vector compared with that of a similar AAV2 vector. In addition, cellular immunological studies suggest that host response differences may occur with AAV2 and AAV5 vector delivery at this mucosal site. The aggregate data indicate that results with AAV5 vectors in murine salivary glands apparently do not extend to macaque glands.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Parotid Gland/metabolism , Animals , Genetic Therapy/methods , Macaca mulatta , Parotid Gland/virology , Transduction, Genetic , Transgenes
4.
Hum Gene Ther ; 13(17): 2041-50, 2002 Nov 20.
Article in English | MEDLINE | ID: mdl-12489999

ABSTRACT

Adult marrow-derived cells have been shown to contribute to various nonhematologic tissues and, conversely, primitive cells isolated from nonhematopoietic tissues have been shown to reconstitute hematopoiesis. Circulating endothelial progenitor cells (EPCs) have been reported to be at least partially donor derived after allogeneic bone marrow transplantation, and shown to contribute to neovascularization in murine ischemia models. However, it is unknown whether these EPCs are actually clonally derived from the same population of stem and progenitor cells that reconstitute hematopoiesis, or from another cell population found in the marrow or mobilized blood that is transferred during transplantation. To approach this question, we characterized circulating EPCs and also endothelial cells from large vessels harvested at autopsy from rhesus macaques previously transplanted with retrovirally transduced autologous CD34-enriched peripheral blood stem cells (PBSCs). Endothelial cells were grown in culture for 21-28 days and were characterized as CD31(+) CD14(-) via flow cytometry, as acLDL(+) UEA-1(+) via immunohistochemistry, and as Flk-1(+) by reverse transcriptase-polymerase chain reaction (RT-PCR). Animals had stable vector marking in hematopoietic lineages of 2-15%. Neither cultured circulating EPCs collected in steady state (n = 3), nor endothelial cells grown from large vessels (n = 2), had detectable retroviral marking. EPCs were CD34(+) and could be mobilized into the circulation with granulocyte colony-stimulating factor. Under ex vivo culture conditions, in which CD34(+) cells were optimized to transduce hematopoietic progenitor and stem cells, there was a marked depletion of EPCs. Transduction of EPCs was much more efficient under conditions supporting endothelial cell growth. Further elucidation of the origin and in vivo behavior of EPCs may be possible, using optimized transduction conditions and a vascular injury model.


Subject(s)
Endothelium, Vascular/metabolism , 3T3 Cells , Analysis of Variance , Animals , Antigens, CD34/genetics , Bacterial Proteins/metabolism , Cell Lineage , Cells, Cultured , Clone Cells , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Erythroid Precursor Cells/metabolism , Genetic Vectors , Granulocyte Colony-Stimulating Factor/pharmacology , Green Fluorescent Proteins , Hematopoietic Stem Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Luminescent Proteins/metabolism , Macaca mulatta , Mice , Models, Animal , Retroviridae/genetics , Transduction, Genetic
5.
Transfusion ; 41(11): 1438-44, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11724992

ABSTRACT

BACKGROUND: Defining the optimum regimen and time for repeat peripheral blood progenitor cell mobilization would have important clinical applications. STUDY DESIGN AND METHODS: Remobilization with SCF and G-CSF at 2 weeks after an initial mobilization in mice and at 2 or 4 weeks after an initial mobilization in nonhuman primates was examined. In mice, competitive repopulation assays were used to measure long-term progenitor cell-repopulating activity. In monkeys, mobilization of hematopoietic progenitor CFUs was used as a surrogate marker for progenitor cell-repopulating ability. RESULTS: Efficacy of progenitor cell remobilization differed in the two animal species. In mice, peripheral blood progenitor cell-repopulating ability with repeat mobilization at 2 weeks was 70 percent of that with the initial mobilization. In monkeys, there was no significant difference in peripheral blood progenitor cell mobilization between the initial and the repeat mobilizations at 2 weeks. In mobilizations separated by 4 weeks, however, peripheral blood progenitor cell mobilization was higher than that with initial mobilizations. CONCLUSION: In animal models, mobilization of peripheral blood progenitor cells with remobilization after a 2-week interval is similar to or moderately decreased from that with the initial mobilization. Progenitor cell collection at this time point may be useful in certain clinical circumstances. A 4-week interval between remobilizations may be preferable. Clinical trials in humans would be useful to clarify these issues.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Macaca mulatta/blood , Mice/blood , Stem Cell Factor/pharmacology , Animals , Cell Division/drug effects , Female , Mice, Inbred C57BL , Time Factors
6.
J Clin Invest ; 108(3): 447-55, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489938

ABSTRACT

Recent reports suggest that cells in active cell cycle have an engraftment defect compared with quiescent cells. We used nonhuman primates to investigate this finding, which has direct implications for clinical transplantation and gene therapy applications. Transfer of rhesus CD34(+) cells to culture in stem cell factor (SCF) on the CH-296 fibronectin fragment (FN) after 4 days of culture in stimulatory cytokines maintained cell viability but decreased cycling. Using retroviral marking with two different gene transfer vectors, we compared the engraftment potential of cytokine-stimulated cells versus those transferred to nonstimulatory conditions (SCF on FN alone) before reinfusion. In vivo competitive repopulation studies showed that the level of marking originating from the cells continued in culture for 2 days with SCF on FN following a 4-day stimulatory transduction was significantly higher than the level of marking coming from cells transduced for 4 days and reinfused without the 2-day culture under nonstimulatory conditions. We observed stable in vivo overall gene marking levels of up to 29%. This approach may allow more efficient engraftment of transduced or ex vivo expanded cells by avoiding active cell cycling at the time of reinfusion.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Animals , Antigens, CD34/metabolism , Base Sequence , Cell Cycle/drug effects , Cell Transformation, Viral , Cells, Cultured , Cytokines/pharmacology , DNA Primers/genetics , Fibronectins/pharmacology , Genetic Therapy , Genetic Vectors , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Macaca mulatta , Peptide Fragments/pharmacology , Retroviridae/genetics , Stem Cell Factor/pharmacology , Transduction, Genetic
7.
Ann N Y Acad Sci ; 938: 236-44; discussion 244-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11458513

ABSTRACT

The impairment of engraftment ability after ex vivo or in vivo stimulation of hematopoietic stem cells, potentially related to induction of active cell cycling, has recently been a topic of intense interest. Our group has used the non-human primate autologous transplantation model and genetic marking to investigate a number of questions in hematopoiesis with direct relevance to human clinical applications. The issue of a potential reversible engraftment defect would have many implications for gene therapy and allogeneic or autologous transplantation. Initial in vitro studies with rhesus CD34+ cells indicated that after 4 days of stimulatory culture in stem cell factor (SCF), megakaryocyte growth and development factor (MDGF), and flt3 ligand (FLT), transfer of the cells to SCF alone on retronectin (FN) support resulted in decreased active cycling and a halt to proliferation, without a loss of viability or induction of apoptosis. We then directly compared the engraftment potential of cytokine-stimulated cells versus those transferred to SCF on FN alone before reinfusion, SCF/G-CSF mobilized CD34+ cells from three animals were split into two parts and transduced with either of two retroviral marking vectors for 4 days in the presence of SCF/FLT/MGDF on FN. One aliquot was cryopreserved, and the other was continued in culture without transduction for 2 days in the presence of SCF alone on FN. After total body irradiation, both aliquots were thawed and reinfused into each animal. In all animals, the level of marking from the fraction continued in culture for 2 days with SCF on FN was significantly higher than the level of marking from the aliquot transduced for 4 days without the 2-day period in SCF alone. This approach may allow more efficient engraftment of successfully transduced or ex vivo expanded cells by avoiding active cell cycling at the time of reinfusion.


Subject(s)
Growth Substances/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Intercellular Signaling Peptides and Proteins , Membrane Proteins/pharmacology , Stem Cell Factor/pharmacology , Animals , Cell Cycle/drug effects , Cell Movement , Cells, Cultured/drug effects , Fibronectins , Graft Survival , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Macaca mulatta , Radiation Chimera , Transfection , Transplantation, Autologous
8.
Hum Gene Ther ; 12(6): 607-17, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11426461

ABSTRACT

Transfer of genes into hematopoietic stem cells or primary lymphocytes has been a primary focus of the gene therapy field for more than a decade because of the wide variety of congenital and acquired diseases that potentially could be cured by successful gene transfer into these cell populations. However, despite success in murine models and in vitro, progress has been slow, and early clinical trials were disappointing due to inefficient gene transfer into long-term repopulating cells. The unique predictive value of nonhuman primate or other large animal models has become more apparent, and major advances in gene transfer efficiency have been made by utilizing these powerful but expensive and complex systems. This review summarizes more recent findings from nonhuman primate investigations focusing on hematopoietic stem cells or lymphocytes as target populations, and highlights specific preclinical issues, including safety. Results from studies using standard retroviral vectors, lentiviral vectors, adenoviral vectors, and adeno-associated viral vectors are discussed. Judicious application of these models should continue to be a priority, and advances should now be tested in proof-of-concept clinical trials.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Hematopoietic Stem Cells/physiology , Macaca/physiology , Models, Animal , Animals , Drug Evaluation, Preclinical , Genetic Vectors , Hematopoietic Stem Cells/virology , Humans , T-Lymphocytes/physiology , T-Lymphocytes/virology
9.
J Virol ; 75(8): 3547-55, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11264344

ABSTRACT

Nonhuman primate model systems of autologous CD34+ cell transplant are the most effective means to assess the safety and capabilities of lentivirus vectors. Toward this end, we tested the efficiency of marking, gene expression, and transplant of bone marrow and peripheral blood CD34+ cells using a self-inactivating lentivirus vector (CS-Rh-MLV-E) bearing an internal murine leukemia virus long terminal repeat derived from a murine retrovirus adapted to replicate in rhesus macaques. In vitro cytokine stimulation was not required to achieve efficient transduction of CD34+ cells resulting in marking and gene expression of the reporter gene encoding enhanced green fluorescent protein (EGFP) following transplant of the CD34+ cells. Monkeys transplanted with mobilized peripheral blood CD34+ cells resulted in EGFP expression in 1 to 10% of multilineage peripheral blood cells, including red blood cells and platelets, stable for 15 months to date. The relative level of gene expression utilizing this vector is 2- to 10-fold greater than that utilizing a non-self-inactivating lentivirus vector bearing the cytomegalovirus immediate-early promoter. In contrast, in animals transplanted with autologous bone marrow CD34+ cells, multilineage EGFP expression was evident initially but diminished over time. We further tested our lentivirus vector system by demonstrating gene transfer of the human common gamma-chain cytokine receptor gene (gamma(c)), deficient in X-linked SCID patients and recently successfully used to treat disease. Marking was 0.42 and.001 HIV-1 vector DNA copy per 100 cells in two animals. To date, all EGFP- and gamma(c)-transplanted animals are healthy. This system may prove useful for expression of therapeutic genes in human hematopoietic cells.


Subject(s)
Cytokines/metabolism , Gene Transfer Techniques , Genetic Vectors/genetics , HIV-1/genetics , Hematopoietic Stem Cells/metabolism , Macaca mulatta/metabolism , Receptors, Cell Surface/genetics , Animals , Antigens, CD34/metabolism , Biomarkers , Flow Cytometry , Gene Expression , Granulocyte Colony-Stimulating Factor/pharmacology , Green Fluorescent Proteins , Hematopoietic Stem Cell Transplantation , Humans , Leukapheresis , Luminescent Proteins , Lymphocytes/metabolism , Macaca mulatta/genetics , Polymerase Chain Reaction , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Stem Cell Factor/pharmacology , Time Factors , Transduction, Genetic
10.
Mol Ther ; 3(3): 359-67, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11273778

ABSTRACT

The fibronectin fragment CH-296 improved gene transfer to cytokine-mobilized nonhuman primate CD34+ cells irrespective of tropism to the MoMLV, GaLV, and VSV-G envelope proteins using murine stem cell virus (MSCV) and human immunodeficiency virus-1 (HIV-1)-based retrovirus vectors. For the HIV-1 lentivirus vector, CH-296 enhanced gene transfer in the absence of added hematopoietic growth factors necessary for ex vivo stem cell expansion. In the presence of CH-296, apoptosis of CD34+ cells was inhibited, and in mobilized peripheral blood CD34+ cells, cell division was stimulated as measured by cell history/tracking experiments.


Subject(s)
Fibronectins/genetics , Gammaretrovirus/genetics , Gene Transfer Techniques , Genetic Vectors , HIV-1/genetics , Hematopoietic Stem Cells/metabolism , Animals , Antigens, CD34 , Apoptosis , Cell Line , Fibronectins/metabolism , Genetic Therapy , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Macaca mulatta , Moloney murine leukemia virus/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Transduction, Genetic , Tropism , Viral Envelope Proteins/genetics , Virus Replication
11.
Blood ; 97(6): 1888-91, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238136

ABSTRACT

Transduction of murine stem cells with a multidrug-resistance 1 gene (MDR1) retrovirus results in dramatic ex vivo and in vivo expansion of repopulating cells accompanied by a myeloproliferative disorder. Given the use of MDR1-containing vectors in human trials, investigations have been extended to nonhuman primates. Peripheral blood stem cells from 2 rhesus monkeys were collected, CD34-enriched, split into 2 portions, and transduced with either MDR1 vectors or neo vectors and continued in culture for a total of 10 days before reinfusion. At engraftment, the copy number in granulocytes was extremely high from both MDR vectors and neo vectors, but the copy number fell to 0.01 to 0.05 for both. There were no perturbations of the leukocyte count or differential noted. After 3 cycles of stem cell factor/granulocyte colony-stimulating factor, there were no changes in the levels of MDR1 vector- or neo vector-containing cells. There was no evidence for expansion of MDR1 vector-transduced cells. Long-term engraftment with MDR1 vector- and neo vector-transduced cells occurred despite prolonged culture.


Subject(s)
Drug Resistance, Microbial/genetics , Genes, MDR/genetics , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Animals , Cell Culture Techniques , Cell Division/drug effects , Gene Dosage , Genetic Therapy/standards , Genetic Vectors/adverse effects , Genetic Vectors/standards , Hematopoietic Stem Cell Transplantation/standards , Hematopoietic Stem Cells/drug effects , Humans , Macaca mulatta , Models, Animal , Neomycin , Transduction, Genetic/methods , Transduction, Genetic/standards
12.
Mol Ther ; 1(3): 285-93, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10933944

ABSTRACT

Low-level retroviral transduction and engraftment of hematopoietic long-term repopulating cells in large animals and humans remain primary obstacles to the successful application of hematopoietic stem cell (HSC) gene transfer in humans. Recent studies have reported improved efficiency by including stromal cells (STR), or the fibronectin fragment CH-296 (FN), and various cytokines such as flt3 ligand (FLT) during ex vivo culture and transduction in nonhuman primates. In this work, we extend our studies using the rhesus competitive repopulation model to further explore optimal and clinically feasible peripheral blood (PB) progenitor cell transduction methods. First, we compared transduction in the presence of either preformed autologous STR or immobilized FN. Long-term clinically relevant gene marking levels in multiple hematopoietic lineages from both conditions were demonstrated in vivo by semiquantitative PCR, colony PCR, and genomic Southern blotting, suggesting that FN could replace STR in ex vivo transduction protocols. Second, we compared transduction on FN in the presence of IL-3, IL-6, stem cell factor (SCF), and FLT (our best cytokine combination in prior studies) with a combination of megakaryocyte growth and development factor (MGDF), SCF, and FLT. Gene marking levels were equivalent in these animals, with no significant effect on retroviral gene transfer efficiency assessed in vivo by the replacement of IL-3 and IL-6 with MGDF. Our results indicate that SCF/G-CSF-mobilized PB CD34+ cells are transduced with equivalent efficiency in the presence of either STR or FN, with stable long-term marking of multiple lineages at levels of 10-15% and transient marking as high as 54%. These results represent an advance in the field of HSC gene transfer using methods easily applied in the clinical setting.


Subject(s)
Antigens, CD34/genetics , Gene Transfer Techniques , Hematopoietic Stem Cells/metabolism , Retroviridae/genetics , Animals , Blotting, Southern , Cells, Cultured , Colony-Forming Units Assay , Fibronectins/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Macaca mulatta , Membrane Proteins/pharmacology , Polymerase Chain Reaction , Stem Cell Factor/pharmacology , Stromal Cells/metabolism , Thrombopoietin/pharmacology
13.
Mol Ther ; 1(6): 533-44, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10933978

ABSTRACT

Host immune responses against foreign transgenes may be a major obstacle to successful gene therapy. To clarify the impact of an immune response to foreign transgene products on the survival of genetically modified cells, we studied the in vivo persistence of cells transduced with a vector expressing a foreign transgene compared to cells transduced with a nonexpressing vector in the clinically predictive rhesus macaque model. We constructed retroviral vectors containing the neomycin phosphotransferase gene (neo) sequences modified to prevent protein expression (nonexpressing vectors). Rhesus monkey lymphocytes or hematopoietic stem cells (HSCs) were transduced with nonexpressing and neo-expressing vectors followed by reinfusion, and their in vivo persistence was studied. While lymphocytes transduced with a nonexpressing vector could be detected for more than 1 year, lymphocytes transduced with a neo-expressing vector were no longer detectable within several weeks of infusion. However, five of six animals transplanted with HSCs transduced with nonexpression or neo-expression vectors, and progeny lymphocytes marked with either vector persisted for more than 2 years. Furthermore, in recipients of transduced HSCs, infusion of mature lymphocytes transduced with a second neo-expressing vector did not result in elimination of the transduced lymphocytes. Our data show that introduction of a xenogeneic gene via HSCs induces tolerance to the foreign gene products. HSC gene therapy is therefore suitable for clinical applications where long-term expression of a therapeutic or foreign gene is required.


Subject(s)
Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Immune Tolerance , Animals , Base Sequence , Blood Transfusion, Autologous , DNA Primers/genetics , Gene Expression , Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cell Transplantation , Kanamycin Kinase/genetics , Kanamycin Kinase/immunology , Lymphocyte Transfusion , Lymphocytes/immunology , Lymphocytes/metabolism , Macaca mulatta , Models, Biological , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transduction, Genetic , Transplantation, Autologous
14.
Blood ; 96(1): 1-8, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10891424

ABSTRACT

Retroviral insertion site analysis was used to track the contribution of retrovirally transduced primitive progenitors to hematopoiesis after autologous transplantation in the rhesus macaque model. CD34-enriched mobilized peripheral blood cells were transduced with retroviral marking vectors containing the neo gene and were reinfused after total body irradiation. High-level gene transfer efficiency allowed insertion site analysis of individual myeloid and erythroid colony-forming units (CFU) and of highly purified B- and T-lymphoid populations in 2 animals. At multiple time points up to 1 year after transplantation, retroviral insertion sites were identified by performing inverse polymerase chain reaction and sequencing vector-containing CFU or more than 99% pure T- and B-cell populations. Forty-eight unique insertion sequences were detected in the first animal and also in the second animal, and multiple clones contributed to hematopoiesis at 2 or more time points. Multipotential clones contributing to myeloid and lymphoid lineages were identified. These results support the concept that hematopoiesis in large animals is polyclonal and that individual multipotential stem or progenitor cells can contribute to hematopoiesis for prolonged periods. Gene transfer to long-lived, multipotent clones is shown and is encouraging for human gene therapy applications.


Subject(s)
B-Lymphocytes/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , T-Lymphocytes/cytology , Animals , Antigens, CD34/blood , Cell Differentiation , Colony-Forming Units Assay , Gene Transfer Techniques , Genes, Reporter , Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cell Mobilization , Humans , Kanamycin Kinase/genetics , Macaca mulatta , Retroviridae , Transfection , Whole-Body Irradiation
15.
Virology ; 272(1): 168-76, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10873759

ABSTRACT

A-2 plaque virus (A2 virus) was originally isolated from the icteric-phase sera of US servicemen with viral hepatitis in the 1960s, but apart from a preliminary characterization little is known about the agent. We have now successfully cloned and sequenced the complete viral genome. A2 viral RNA consists of 7312 nucleotides, excluding the 62 nucleotide poly(A) tract at the 3' end, with one large open reading frame. Although clearly a member of the Picornaviridae, there is low homology to the available sequences, suggesting it is only loosely related to the classic rhino/enterovirus genus. In addition, there was no reactivity with group specific monoclonal antibody blends against polioviruses, enteroviruses 70 and 71, coxsackievirus B, and echoviruses. Two tamarins were inoculated with A2 virus to study viral pathogenesis. Both animals that received A2 virus became transiently viremic 1 week after the infection, as determined by RT-PCR, and they developed an antibody response to A2 virus. However, no physical signs or biochemical abnormalities, including elevated liver transaminases, were observed. In addition, no liver samples from patients with fulminant hepatitis (n = 7) or controls (n = 7) were positive for A2 viral RNA nor was anti-A2 neutralizing antibody detected in sera from hepatitis patients (n = 14), healthy laboratory donors (n = 14), or US blood donors (n = 33); however, most sera contained antibodies reactive with A2 virus proteins. These results suggest that A2 virus is a new member of the Picornaviridae but that its pathogenicity in nonhuman primates and association with human disease still need to be determined.


Subject(s)
Genome, Viral , Picornaviridae/genetics , Picornaviridae/immunology , Anemia, Aplastic/immunology , Anemia, Aplastic/virology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antibody Specificity/immunology , Chlorocebus aethiops , Cloning, Molecular , Enterovirus/genetics , Enterovirus/immunology , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Hepatitis, Viral, Human/immunology , Hepatitis, Viral, Human/virology , Hepatovirus/genetics , Hepatovirus/immunology , Humans , Liver/pathology , Liver/virology , Molecular Sequence Data , Neutralization Tests , Phylogeny , Picornaviridae/chemistry , Picornaviridae/classification , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , RNA, Viral/analysis , RNA, Viral/genetics , Saguinus/immunology , Saguinus/virology , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
16.
Blood ; 95(2): 445-52, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10627448

ABSTRACT

We have used a murine retrovirus vector containing an enhanced green fluorescent protein complimentary DNA (EGFP cDNA) to dynamically follow vector-expressing cells in the peripheral blood (PB) of transplanted rhesus macaques. Cytokine mobilized CD34(+) cells were transduced with an amphotropic vector that expressed EGFP and a dihydrofolate reductase cDNA under control of the murine stem cell virus promoter. The transduction protocol used the CH-296 recombinant human fibronectin fragment and relatively high concentrations of the flt-3 ligand and stem cell factor. Following transplantation of the transduced cells, up to 55% EGFP-expressing granulocytes were obtained in the peripheral circulation during the early posttransplant period. This level of myeloid marking, however, decreased to 0.1% or lower within 2 weeks. In contrast, EGFP expression in PB lymphocytes rose from 2%-5% shortly following transplantation to 10% or greater by week 5. After 10 weeks, the level of expression in PB lymphocytes continued to remain at 3%-5% as measured by both flow cytometry and Southern blot analysis, and EGFP expression was observed in CD4(+), CD8(+), CD20(+), and CD16/56(+) lymphocyte subsets. EGFP expression was only transiently detected in red blood cells and platelets soon after transplantation. Such sustained levels of lymphocyte marking may be therapeutic in a number of human gene therapy applications that require targeting of the lymphoid compartment. The transient appearance of EGFP(+) myeloid cells suggests that transduction of a lineage-restricted myeloid progenitor capable of short-term engraftment was obtained with this protocol. (Blood. 2000;95:445-452)


Subject(s)
Cytokines/pharmacology , Granulocytes/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Luminescent Proteins/genetics , Tetrahydrofolate Dehydrogenase/genetics , Transfection/methods , Animals , DNA, Complementary/administration & dosage , Fibronectins/pharmacology , Green Fluorescent Proteins , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Humans , Luminescent Proteins/biosynthesis , Lymphocyte Subsets/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Macaca mulatta , Peptide Fragments/pharmacology , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/pharmacology , Tetrahydrofolate Dehydrogenase/biosynthesis
17.
J Virol ; 74(3): 1286-95, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10627539

ABSTRACT

Recently, gene delivery vectors based on human immunodeficiency virus (HIV) have been developed as an alternative mode of gene delivery. These vectors have a number of advantages, particularly in regard to the ability to infect cells which are not actively dividing. However, the use of vectors based on human immunodeficiency virus raises a number of issues, not the least of which is safety; therefore, further characterization of marking and gene expression in different hematopoietic lineages in primate animal model systems is desirable. We use two animal model systems for gene therapy to test the efficiency of transduction and marking, as well as the safety of these vectors. The first utilizes the rhesus animal model for cytokine-mobilized autologous peripheral blood CD34(+) cell transplantation. The second uses the SCID-human (SCID-hu) thymus/liver chimeric graft animal model useful specifically for human T-lymphoid progenitor cell reconstitution. In the rhesus macaques, detectable levels of vector were observed in granulocytes, lymphocytes, monocytes, and, in one animal with the highest levels of marking, erythrocytes and platelets. In transplanted SCID-hu mice, we directly compared marking and gene expression of the lentivirus vector and a murine leukemia virus-derived vector in thymocytes. Marking was observed at comparable levels, but the lentivirus vector bearing an internal cytomegalovirus promoter expressed less efficiently than did the murine retroviral vector expressed from its own long terminal repeats. In assays for infectious HIV type 1 (HIV-1), no replication-competent HIV-1 was detected in either animal model system. Thus, these results indicate that while lentivirus vectors have no apparent deleterious effects and may have advantages over murine retroviral vectors, further study of the requirements for optimal use are warranted.


Subject(s)
Antigens, CD34/analysis , Gene Expression , Genetic Vectors , HIV-1/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cytomegalovirus/genetics , Green Fluorescent Proteins , Hematopoietic Stem Cells/virology , Humans , Leukemia Virus, Murine/genetics , Leukopoiesis , Liver Transplantation/immunology , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Lymphocyte Activation , Macaca mulatta , Mice , Mice, SCID , Promoter Regions, Genetic , T-Lymphocytes/immunology , T-Lymphocytes/virology , Thymus Gland/immunology , Thymus Gland/transplantation , Transduction, Genetic , Virus Replication
18.
Blood ; 94(12): 3968-75, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10590040

ABSTRACT

We used a first-generation adenovirus vector (AVC3FIX5) to assess whether human factor IX could be expressed and detected in the rhesus macaque, which we have shown does not make high-titer antibodies to human factor IX protein. Three animals received 1 x 10(10) to 1 x 10(11) plaque-forming units per kilogram by intravenous injection. Human factor IX was present within 24 hours of vector administration and peaked 4 days later at 4,000 ng/mL in the high-dose recipient, and lower levels were seen in the intermediate-dose recipient. No human factor IX was detected in the low-dose recipient's plasma. Serum cytokine analysis and early hypoferremia suggested a dose-dependent acute-phase response to the vector. Human factor IX was detectable in rhesus plasma for 2 to 3 weeks for the high- and intermediate-dose recipients, but disappeared concomitant with high-titer antihuman factor IX antibody development. There was substantial, dose-dependent, dose-limiting liver toxicity that was manifest as elevated serum transaminase levels, hyperbilirubinemia, hypoalbuminemia, and prolongation of clotting times. Of particular interest was prolongation of the thrombin clotting time, an indicator of decreased fibrinogen or fibrinogen dysfunction. All evidence of liver toxicity resolved except for persistent hypofibrinogenemia in the high-dose recipient, indicating possible permanent liver damage. Our data suggest a narrow therapeutic window for first-generation adenovirus-mediated gene transfer. The development of antihuman factor IX antibodies and abnormalities of fibrinogen in the rhesus macaque is of concern for application of adenovirus (or other viral) vectors to hemophilia gene therapy.


Subject(s)
Adenoviridae , Factor IX/genetics , Gene Transfer Techniques , Genetic Vectors , Animals , Dose-Response Relationship, Drug , Factor IX/biosynthesis , Factor IX/toxicity , Gene Transfer Techniques/adverse effects , Genetic Vectors/adverse effects , Humans , Macaca mulatta
19.
Blood ; 94(7): 2263-70, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10498598

ABSTRACT

We have compared adeno-associated virus (AAV)-based and retrovirus-based vectors for their ability to transduce primary T lymphocytes in vitro and then tracked the persistence of these genetically marked lymphocytes in vivo, using the rhesus monkey model. To avoid the complication of immune rejection of lymphocytes transduced with xenogeneic genes in tracking studies primarily designed to investigate transduction efficiency and in vivo kinetics, the vectors were designed without expressed genes. All vectors contained identically mutated beta-galactosidase gene (beta-gal) and neomycin resistance gene (neo) DNA sequences separated by different length polylinkers, allowing simple differentiation by polymerase chain reaction (PCR). Each of 2 aliquots of peripheral blood lymphocytes from 4 rhesus monkeys were transduced with either AAV or retroviral vectors. The in vitro transduction efficiency (mean vector copy number/cell) after the ex vivo culture was estimated by PCR at 0.015 to 3.0 for AAV, varying depending on the multiplicity of infection (MOI) used for transduction, and 0.13 to 0.19 for the retroviral transductions. Seven days after transduction, Southern blot analysis of AAV-transduced lymphocytes showed double-stranded and head-to-tail concatemer forms but failed to show integration of the AAV vector. AAV and retroviral aliquots were reinfused concurrently into each animal. Although the retrovirally marked lymphocytes could be detected for much longer after infusion, AAV transduction resulted in higher short-term in vivo marking efficiency compared with retroviral vectors, suggesting possible clinical applications of AAV vectors in lymphocyte gene therapy when long-term vector persistence is not required or desired.


Subject(s)
Dependovirus/genetics , Genetic Vectors , Retroviridae/genetics , T-Lymphocytes/cytology , Animals , Drug Resistance, Microbial/genetics , Genes, Reporter , Lymphocyte Transfusion , Macaca mulatta , Neomycin , Polymerase Chain Reaction , T-Lymphocytes/immunology , Transfection , beta-Galactosidase/genetics
20.
Hum Gene Ther ; 10(11): 1783-90, 1999 Jul 20.
Article in English | MEDLINE | ID: mdl-10446918

ABSTRACT

The ability to engraft significant numbers of genetically modified hematopoietic stem and progenitor cells without the requirement for fully myeloablative conditioning therapy is a highly desirable goal for the treatment of many nonmalignant hematologic disorders. The aims of this study were to examine, in nonhuman primates (rhesus), (1) the effects of pretreatment of host animals with cytokines (G-CSF and SCF), i.e., before nonmyeloablative irradiation, on the degree and duration of neo gene marking of circulating leukocytes after autologous cell reinfusion and (2) to compare transduction of primitive hematopoietic target cells in the presence of our standard transduction cytokine combination of IL-3, IL-6, and stem cell factor (SCF) and in the presence of an alternative combination containing SCF, G-CSF, and the thrombopoietin analog MGDF. Cytokine-mobilized rhesus peripheral blood progenitor/stem cells (PBSCs) were enriched for CD34+ cells and transduced with neo vectors (either G1Na or LNL6) for 96 hr in cultures containing rhIL-3, rhIL-6, and rhSCF or MGDF, rhSCF, and rhG-CSF and cryopreserved. Four animals underwent minimal myeloablative conditioning with 500 cGy irradiation with or without pretreatment with SCF and G-CSF, followed by reinfusion of the cryopreserved cells on the subsequent day. Neutrophil nadirs (< or =500/mm3) were 0-3 days in duration; there were no significant periods of severe thrombocytopenia. Marking of circulating granulocytes and mononuclear cells was extensive and durable in all animals (exceeding 12% in the mononuclear cells of one animal) and persisted beyond the final sampling time in all animals (up to 33 weeks). No difference in extent or duration of marking was attributable to either cytokine presensitization of recipients prior to irradiation, or to the substitution of MDGF and G-CSF for IL-3 and IL-6 during transduction.


Subject(s)
Graft Survival/drug effects , Graft Survival/radiation effects , Hematopoietic Stem Cell Transplantation , Retroviridae/genetics , Transplantation Conditioning , Animals , Anti-Bacterial Agents/pharmacology , Antigens, CD34/metabolism , Drug Resistance/genetics , Gene Transfer Techniques , Genetic Markers , Genetic Vectors , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/immunology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Hematopoietic Stem Cells/virology , Leukocytes, Mononuclear/immunology , Macaca mulatta , Neomycin/pharmacology , Polymerase Chain Reaction , Radiation Dosage , Stem Cell Factor/pharmacology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...