Abstract

The tremendous potential of stem cell-directed gene therapy has recently been demonstrated with the cure of children suffering from X-linked severe combined immunodeficiency (X-SCID) and adenosine deaminase deficiency [1Aiuti A. et al.Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning.Science. 2002; 296: 2410-2413Crossref PubMed Scopus (786) Google Scholar, 2Cavazzana-Calvo M. et al.Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease.Science. 2000; 288: 669-672Crossref PubMed Scopus (1815) Google Scholar, 3Hacein-Bey-Abina S. et al.Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy.N. Engl. J. Med. 2002; 346: 1185-1193Crossref PubMed Scopus (779) Google Scholar]. A new era of medicine appeared to be on the horizon, but enthusiasm was quickly dampened after it became evident that this clinical success had not come without severe adverse events (SAEs). Unfortunately, 2 of 10 children treated with gene therapy developed leukemia [4Hacein-Bey-Abina S. et al.LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1.Science. 2003; 302: 415-419Crossref PubMed Scopus (2213) Google Scholar, 5Hacein-Bey-Abina S. et al.A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency.N. Engl. J. Med. 2003; 348: 255-256Crossref PubMed Scopus (1259) Google Scholar]. Thus, the very study that was initially regarded as the breakthrough for stem cell gene therapy is now considered by some as the breakdown for the field. To reach the point where both the great promise and the potential risk are equally apparent has taken over 20 years of developing and refining gene transfer techniques. A major obstacle has been the lack of tissue culture or rodent models predictive for stem cell gene transfer in humans. In this review we discuss the important contributions of large animal studies to the enormous progress in hematopoietic stem cell gene transfer and how large animal models can help to further improve efficiency and safety of stem cell gene therapy. Genetic modification of hematopoietic stem cells (HSCs) has the potential to cure a wide variety of genetic and malignant diseases affecting the hematopoietic system because HSCs provide a lifelong supply of circulating blood cells of all lineages. Genetic diseases such as Fanconi anemia, hemoglobinopathies, and immunodeficiencies, as well as acquired diseases such as AIDS and cancer, could potentially be treated by stem cell gene therapy. Although most of these diseases are curable by allogeneic stem cell transplantation, many patients lack a suitable donor, and the side effects of allogeneic stem cell transplantation such as graft-versus-host-disease make the transplantation of genetically modified autologous cells an attractive alternative. Furthermore,recent studies suggest that gene therapy may also enhance allogeneic stem cell transplantation [6Neff T. et al.Methylguanine methyltransferase-mediated in vivo selection and chemoprotection of allogeneic stem cells in a large-animal model.J. Clin. Invest. 2003; 112: 1581-1588Crossref PubMed Scopus (88) Google Scholar, 7Bank A. Hematopoietic stem cell gene therapy: selecting only the best.J. Clin. Invest. 2003; 112: 1478-1480Crossref PubMed Scopus (0) Google Scholar]. Hematopoietic stem cells have been targeted for genetic modification for over 2 decades. In the 1980s it was shown that mouse hematopoietic stem cells could be genetically modified with a degree of efficiency predicted to be therapeutic for many human diseases [8Joyner A. Keller G. Phillips R.A. Bernstein A. Retrovirus transfer of a bacterial gene into mouse haematopoietic progenitor cells.Nature. 1983; 305: 556-558Crossref PubMed Google Scholar, 9Williams D.A. Lemischka I.R. Nathan D.G. Mulligan R.C. Introduction of new genetic material into pluripotent haematopoietic stem cells of the mouse.Nature. 1984; 310: 476-480Crossref PubMed Scopus (0) Google Scholar]. These results prompted a high level of enthusiasm until it became apparent that the transducibility of hematopoietic stem cells is just another feature to distinguish mice from men. Because stem cell transduction protocols developed in the mouse did not translate into efficient gene transfer protocols in human studies, many investigators started to study stem cell transduction in dogs [10Kwok W.W. Storb R. Miller A.D. Canine model for gene therapy: inefficient gene expression in dogs reconstituted with autologous marrow infected with retroviral vectors.Blood. 1988; 71: 742-747PubMed Google Scholar, 11Schuening F.G. et al.Retrovirus-mediated gene transduction into long-term repopulating marrow cells of dogs.Blood. 1991; 78: 2568-2576Crossref PubMed Google Scholar, 12Carter R.F. et al.Autologous transplantation of canine long-term marrow culture cells genetically marked by retroviral vectors.Blood. 1992; 79: 356-364PubMed Google Scholar, 13Kiem H.-P. et al.Retrovirus-mediated gene transduction into canine peripheral blood repopulating cells.Blood. 1994; 83: 1467-1473Crossref PubMed Google Scholar, 14Bienzle D. et al.Gene transfer into hematopoietic stem cells: long-term maintenance of in vitro activated progenitors without marrow ablation.Proc. Natl. Acad. Sci. USA. 1994; 91: 350-354Crossref PubMed Scopus (0) Google Scholar] and nonhuman primates [15Donahue R.E. Dunbar C.E. Update on the use of nonhuman primate models for preclinical testing of gene therapy approaches targeting hematopoietic cells.Hum. Gene Ther. 2001; 12: 607-617Crossref PubMed Scopus (57) Google Scholar, 16Kantoff P.W. et al.Expression of human adenosine deaminase in nonhuman primates after retrovirus-mediated gene transfer.J. Exp. Med. 1987; 166: 219-234Crossref PubMed Scopus (0) Google Scholar, 17Bodine D.M. et al.Development of a high-titer retrovirus producer cell line capable of gene transfer into rhesus monkey hematopoietic stem cells.Proc. Natl. Acad. Sci. USA. 1990; 87: 3738-3742Crossref PubMed Scopus (116) Google Scholar, 18Van Beusechem V.W. Bart-Baumeister J.A. Bakx T.A. Kaptein L.C. Levinsky R.J. Valerio D. Gene transfer into nonhuman primate CD34+CD11b− bone marrow progenitor cells capable of repopulating lymphoid and myeloid lineages.Hum. Gene Ther. 1994; 5: 295-305Crossref PubMed Google Scholar, 19Van Beusechem V.W. et al.Retrovirus-mediated gene transfer into rhesus monkey hematopoietic stem cells—the effect of viral titers on transduction efficiency.Hum. Gene Ther. 1993; 4: 239-247Crossref PubMed Google Scholar, 20Bodine D.M. et al.Long-term in vivo expression of a murine adenosine deaminase gene in rhesus monkey hematopoietic cells of multiple lineages after retroviral mediated gene transfer into CD34+ bone marrow cells.Blood. 1993; 82: 1975-1980Crossref PubMed Google Scholar, 21Van Beusechem V.W. Kukler A. Heidt P.J. Valerio D. Long-term expression of human adenosine deaminase in rhesus monkeys transplanted with retrovirus-infected bone-marrow cells.Proc. Natl. Acad. Sci. USA. 1992; 89: 7640-7644Crossref PubMed Google Scholar]. Similar to the initial clinical trials, these early studies in large animal models yielded disappointingly low gene transfer rates. These initial difficulties in achieving high-level stem cell transduction demonstrated that results from large animal models were much more predictive of human stem cell gene therapy studies than mouse studies and suggested that these models might be more appropriate assays for the optimization of clinical gene therapy protocols. Two technical advances have substantially increased the information gained from large animal studies, namely the development of fluorescent marker proteins and the use of competitive repopulation assays: The competitive repopulation assay allows for efficient evaluation of different gene transfer conditions within one animal and reduces the otherwise substantial interanimal variability in outbred large animals. The cells to be used for transplantation are split into two or more equal aliquots, which are transduced under different conditions using distinguishable vectors. After transduction, all cells are pooled and reinfused into the autologous host, where gene marking derived from the different experimental arms is monitored in the marrow and/or peripheral blood after engraftment (Fig. 1A). A drawback of this assay is that it cannot distinguish between the efficiency of gene transfer to stem cells during ex vivo transduction and the subsequent engraftment of transduced stem cells. Early [10Kwok W.W. Storb R. Miller A.D. Canine model for gene therapy: inefficient gene expression in dogs reconstituted with autologous marrow infected with retroviral vectors.Blood. 1988; 71: 742-747PubMed Google Scholar, 11Schuening F.G. et al.Retrovirus-mediated gene transduction into long-term repopulating marrow cells of dogs.Blood. 1991; 78: 2568-2576Crossref PubMed Google Scholar, 12Carter R.F. et al.Autologous transplantation of canine long-term marrow culture cells genetically marked by retroviral vectors.Blood. 1992; 79: 356-364PubMed Google Scholar, 13Kiem H.-P. et al.Retrovirus-mediated gene transduction into canine peripheral blood repopulating cells.Blood. 1994; 83: 1467-1473Crossref PubMed Google Scholar, 14Bienzle D. et al.Gene transfer into hematopoietic stem cells: long-term maintenance of in vitro activated progenitors without marrow ablation.Proc. Natl. Acad. Sci. USA. 1994; 91: 350-354Crossref PubMed Scopus (0) Google Scholar, 17Bodine D.M. et al.Development of a high-titer retrovirus producer cell line capable of gene transfer into rhesus monkey hematopoietic stem cells.Proc. Natl. Acad. Sci. USA. 1990; 87: 3738-3742Crossref PubMed Scopus (116) Google Scholar, 22Kiem H.-P. et al.Long-term persistence of canine hematopoietic cells genetically marked by retrovirus vectors.Hum. Gene Ther. 1996; 7: 89-96Crossref PubMed Google Scholar, 23Kiem H.-P. et al.Gene transfer into marrow repopulating cells: comparison between amphotropic and gibbon ape leukemia virus pseudotyped retroviral vectors in a competitive repopulation assay in baboons.Blood. 1997; 90: 4638-4645Crossref PubMed Google Scholar] and also more recent [24Wu T. et al.Prolonged high-level detection of retrovirally marked hematopoietic cells in nonhuman primates after transduction of CD34+ progenitors using clinically feasible methods.Mol. Ther. 2000; 1: 285-293Abstract Full Text Full Text PDF PubMed Scopus (96) Google Scholar, 25Kim H.J. et al.Many multipotential gene-marked progenitor or stem cell clones contribute to hematopoiesis in nonhuman primates.Blood. 2000; 96: 1-8PubMed Google Scholar, 26Hu J. et al.Direct comparison of RD114-pseudotyped versus amphotropic-pseudotyped retroviral vectors for transduction of rhesus macaque long-term repopulating cells.Mol. Ther. 2003; 8: 611-617Abstract Full Text Full Text PDF PubMed Scopus (13) Google Scholar, 27Kluge K.A. Bonifacino A.C. Sellers S. Agricola B.A. Donahue R.E. Dunbar C.E. Retroviral transduction and engraftment ability of primate hematopoietic progenitor and stem cells transduced under serum-free versus serum-containing conditions.Mol. Ther. 2002; 5: 316-322Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar, 28Kurre P. Morris J. Horn P.A. Harkey M.A. Andrews R.G. Kiem H.-P. Gene transfer into baboon repopulating cells: a comparison of flt-3 ligand and megakaryocyte growth and development factor versus IL-3 during ex vivo transduction.Mol. Ther. 2001; 3: 920-927Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar] gene transfer studies in large animals have utilized the neomycin resistance gene (neo) as a marker to track the progeny of transduced stem cells by PCR. The green fluorescent protein (GFP) gene from the jellyfish Aequorea victoria [[29]Chalfie M. Tu Y. Euskirchen G. Ward W.W. Prasher D.C. Green fluorescent protein as a marker for gene expression.Science. 1994; 263: 802-805Crossref PubMed Google Scholar] was described 10 years ago, and subsequently improved versions with mutations designed to increase fluorescence and optimize codon usage have been developed [[30]Zhang G. Gurtu V. Kain S.R. An enhanced green fluorescent protein allows sensitive detection of gene transfer in mammalian cells.Biochem. Biophys. Res. Commun. 1996; 227: 707-711Crossref PubMed Scopus (218) Google Scholar]. Since then, multiple fluorescent proteins have become available in different colors (reviewed in [[31]Hawley T.S. Telford W.G. Hawley R.G. “Rainbow” reporters for multispectral marking and lineage analysis of hematopoietic stem cells.Stem Cells. 2001; 19: 118-124Crossref PubMed Google Scholar]). The potential of the fluorescent proteins for gene marking studies was quickly realized in murine [32Bierhuizen M.F. Westerman Y. Visser T.P. Dimjati W. Wognum A.W. Wagemaker G. Enhanced green fluorescent protein as selectable marker of retroviral-mediated gene transfer in immature hematopoietic bone marrow cells.Blood. 1997; 90: 3304-3315PubMed Google Scholar, 33Persons D.A. et al.Retroviral-mediated transfer of the green fluorescent protein gene into murine hematopoietic cells facilitates scoring and selection of transduced progenitors in vitro and identification of genetically modified cells in vivo.Blood. 1997; 90: 1777-1786PubMed Google Scholar] and large animal studies [34Donahue R.E. et al.High levels of lymphoid expression of enhanced green fluorescent protein in nonhuman primates transplanted with cytokine-mobilized peripheral blood CD34(+) cells.Blood. 2000; 95: 445-452PubMed Google Scholar, 35Hanazono Y. et al.Introduction of the green fluorescent protein gene into hematopoietic stem cells results in prolonged discrepancy of in vivo transduction levels between bone marrow progenitors and peripheral blood cells in nonhuman primates.J. Gene Med. 2002; 4: 470-477Crossref PubMed Scopus (14) Google Scholar, 36Hanawa H. et al.Efficient gene transfer into rhesus repopulating hematopoietic stem cells using a simian immunodeficiency virus-based lentiviral vector system.Blood. 2004; 103: 4062-4069Crossref PubMed Scopus (124) Google Scholar, 37Kurre P. Morris J. Andrews R.G. Kohn D.B. Kiem H.-P. Kinetics of fluorescence expression in nonhuman primates transplanted with GFP retrovirus-modified CD34 cells.Mol. Ther. 2002; 6: 83-90Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar]. The use of these fluorescent markers allows not only quantitation on the DNA level by quantitative real-time PCR, but also rapid identification of transduced cells by flow cytometry (Fig. 1B) or fluorescence microscopy or culture in real time without additional staining steps. The ability to distinguish different fluorescent markers by flow cytometry has greatly facilitated the analysis of competitive repopulation assays. Fluorescent proteins also allow for easy purification of live transduced populations by FACS. Furthermore, they allow for the assessment of gene expression in nonnucleated cells such as circulating red blood cells and platelets. Since large animal models are expensive and very labor intensive, investigators have developed surrogate assays such as xenotransplantation assays to study human candidate stem cells [38McCune J.M. Namikawa R. Kaneshima H. Shultz L.D. Lieberman M. Weissman I.L. The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function.Science. 1988; 241: 1632-1639Crossref PubMed Google Scholar, 39Nolta J.A. Hanley M.B. Kohn D.B. Sustained human hematopoiesis in immunodeficient mice by cotransplantation of marrow stroma expressing human interleukin-3: analysis of gene transduction of long-lived progenitors.Blood. 1994; 83: 3041-3051Crossref PubMed Google Scholar, 40Zanjani E.D. Pallavicini M.G. Harrison M.R. Tavassoli M. Successful stable xenograft of human fetal hemopoietic cells in preimmune fetal sheep.Trans. Assoc. Am. Physicians. 1991; 104: 181-186PubMed Google Scholar]. The most frequently used xenotransplantation system for the study of human candidate stem cells is the nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse [41Larochelle A. et al.Identification of primitive human hematopoietic cells capable of repopulating NOD/SCID mouse bone marrow: implications for gene therapy.Nat. Med. 1996; 2: 1329-1337Crossref PubMed Scopus (587) Google Scholar, 42Guenechea G. Gan O.I. Dorrell C. Dick J.E. Distinct classes of human stem cells that differ in proliferative and self-renewal potential.Nat. Immunol. 2001; 2: 75-82Crossref PubMed Scopus (235) Google Scholar, 43Josephson N.C. et al.Transduction of human NOD/SCID-repopulating cells with both lymphoid and myeloid potential by foamy virus vectors.Proc. Natl. Acad. Sci. USA. 2002; 99: 8295-8300Crossref PubMed Scopus (73) Google Scholar]. The NOD/SCID model is attractive because it allows one to query human cells in vivo. However, this assay is restricted by the limited proliferative demand placed on the transplanted cells, the short life span of the recipients, and the inability to support differentiation into all hematopoietic lineages. Generally, higher gene transfer rates have been reported for SCID repopulating cells (SRCs) than for cells capable of long-term repopulation in large animals. We recently performed a direct comparison between hematopoietic repopulation in NOD/SCID mice and autologous reconstitution in nonhuman primates and found substantially higher gene marking levels in SRCs than in cells repopulating nonhuman primates [[44]Horn P.A. Thomasson B.M. Wood B.L. Andrews R.G. Morris J.C. Kiem H.-P. Distinct hematopoietic stem/progenitor cell populations are responsible for repopulating NOD/SCID mice compared with nonhuman primates.Blood. 2003; 102: 4329-4335Crossref PubMed Scopus (0) Google Scholar]. In addition, integration site analysis revealed common proviral integrants in SRCs and in the baboon at 6 weeks but not at 6 months after transplantation [[44]Horn P.A. Thomasson B.M. Wood B.L. Andrews R.G. Morris J.C. Kiem H.-P. Distinct hematopoietic stem/progenitor cell populations are responsible for repopulating NOD/SCID mice compared with nonhuman primates.Blood. 2003; 102: 4329-4335Crossref PubMed Scopus (0) Google Scholar]. Thus, while the NOD/SCID model has provided useful data for the transduction of early human progenitors available data suggest that the NOD/SCID model assays short-term rather than long-term repopulating cells. Studies in the NOD/SCID model may therefore not translate directly into clinical stem cell gene therapy trials and studies in large animals and in NOD/SCID mice should probably be viewed as complementary approaches to optimize gene transfer to true multilineage long-term repopulating stem cells. The majority of clinical gene therapy trials and preclinical gene transfer studies have utilized oncoretroviral vectors based on the Moloney murine leukemia virus (MoMLV) to transfer genes stably to hematopoietic stem cells. One of the major disadvantages of oncoretroviral vectors is their need for target cell division for transduction [45Miller D.G. Adam M.A. Miller A.D. Gene transfer by retrovirus vectors occurs only in cells that are actively replicating at the time of infection.Mol. Cell. Biol. 1990; 10: 4239-4242Crossref PubMed Google Scholar, 46Lewis P.F. Emerman M. Passage through mitosis is required for oncoretroviruses but not for the human immunodeficiency virus.J. Virol. 1994; 68: 510-516Crossref PubMed Google Scholar, 47Andreadis S. Fuller A.O. Palsson B.O. Cell cycle dependence of retroviral transduction: an issue of overlapping time scales.Biotechnol. Bioeng. 1998; 58: 272-281Crossref PubMed Scopus (0) Google Scholar, 48Roe T. Reynolds T.C. Yu G. Brown P.O. Integration of murine leukemia virus DNA depends on mitosis.EMBO J. 1993; 12: 2099-2108Crossref PubMed Scopus (762) Google Scholar], which in turn requires cytokine stimulation and can lead to cell differentiation and diminished engraftment. In contrast to oncoretroviral vectors, lentiviral vectors based on the human immunodeficiency virus (HIV) genome are able to transduce nondividing cells [[46]Lewis P.F. Emerman M. Passage through mitosis is required for oncoretroviruses but not for the human immunodeficiency virus.J. Virol. 1994; 68: 510-516Crossref PubMed Google Scholar] since the HIV-1 preintegration complex can transit intact nuclear membranes [[49]Naldini L. et al.In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector.Science. 1996; 272: 263-267Crossref PubMed Google Scholar]. The development of vectors based on HIV-1 (reviewed in [[50]Federico M. Lentiviruses as gene delivery vectors.Curr. Opin. Biotechnol. 1999; 10: 448-453Crossref PubMed Scopus (26) Google Scholar]) was therefore thought to overcome the most significant obstacle to stem cell gene therapy, the need to induce cycling of stem cells to allow transduction. Stable transduction of nondividing human hematopoietic cells by HIV-1-based lentiviral vectors has been reported [51Uchida N. et al.HIV, but not murine leukemia virus, vectors mediate high efficiency gene transfer into freshly isolated G0/G1 human hematopoietic stem cells.Proc. Natl. Acad. Sci. USA. 1998; 95: 11939-11944Crossref PubMed Scopus (240) Google Scholar, 52Case S.S. et al.Stable transduction of quiescent CD34(+)CD38(−) human hematopoietic cells by HIV-1-based lentiviral vectors.Proc. Natl. Acad. Sci. USA. 1999; 96: 2988-2993Crossref PubMed Scopus (0) Google Scholar]; however, whether lentiviral vectors are capable of transducing truly quiescent, metabolically inactive cells, is still a matter of debate [[53]Emerman M. Learning from lentiviruses.Nat. Genet. 2000; 24: 8-9Crossref PubMed Scopus (0) Google Scholar]. Enthusiasm for lentiviral vectors rose when highly efficient gene transfer into human NOD/SCID repopulating cells using lentiviral vectors was reported [54Miyoshi H. Smith K.A. Mosier D.E. Verma I.M. Torbett B.E. Transduction of human CD34+ cells that mediate long-term engraftment of NOD/SCID mice by HIV vectors.Science. 1999; 283: 682-686Crossref PubMed Scopus (522) Google Scholar, 55Woods N.B. et al.Lentiviral gene transfer into primary and secondary NOD/SCID repopulating cells.Blood. 2000; 96: 3725-3733PubMed Google Scholar]. Surprisingly, results from two different laboratories using lentiviral vectors in primates suggest that they are in fact inferior to oncoretroviral vectors with respect to gene transfer to long-term repopulating cells [56An D.S. et al.Marking and gene expression by a lentivirus vector in transplanted human and nonhuman primate CD34(+) cells.J. Virol. 2000; 74: 1286-1295Crossref PubMed Scopus (86) Google Scholar, 57An D.S. et al.Lentivirus vector-mediated hematopoietic stem cell gene transfer of common gamma-chain cytokine receptor in rhesus macaques.J. Virol. 2001; 75: 3547-3555Crossref PubMed Scopus (64) Google Scholar, 58Horn P.A. et al.Lentivirus-mediated gene transfer into hematopoietic repopulating cells in baboons.Gene Ther. 2002; 9: 1464-1471Crossref PubMed Scopus (0) Google Scholar]. This is now thought to be a species-specific effect that will likely not apply to human clinical trials. An early replication block of HIV in monkey cells was described as early as 1995 [[59]Shibata R. Sakai H. Kawamura M. Tokunaga K. Adachi A. Early replication block of human immunodeficiency virus type 1 in monkey cells.J. Gen. Virol. 1995; 76: 2723-2730Crossref PubMed Google Scholar]. Species restriction of SIV- and HIV-based vectors has been analyzed systematically, and it has been shown that the postentry HIV-1 replication block occurs mainly in Old World monkeys [[60]Hofmann W. et al.Species-specific, postentry barriers to primate immunodeficiency virus infection.J. Virol. 1999; 73: 10020-10028PubMed Google Scholar]. Furthermore it has been shown that the early replication block in Old World monkeys is capsid-dependent [[61]Owens C.M. Yang P.C. Gottlinger H. Sodroski J. Human and simian immunodeficiency virus capsid proteins are major viral determinants of early, postentry replication blocks in simian cells.J. Virol. 2003; 77: 726-731Crossref PubMed Scopus (0) Google Scholar] and can be circumvented by defined mutations in the HIV capsid protein [[62]Kootstra N.A. Munk C. Tonnu N. Landau N.R. Verma I.M. Abrogation of postentry restriction of HIV-1-based lentiviral vector transduction in simian cells.Proc. Natl. Acad. Sci. USA. 2003; 100: 1298-1303Crossref PubMed Scopus (0) Google Scholar]. Recently, a mechanistic explanation has been provided by the demonstration that the simian TRIM5α protein is necessary and sufficient for the postentry replication block of HIV-1 and that overexpression of simian TRIM5α confers this replication block to human cells [[63]Stremlau M. Owens C.M. Perron M.J. Kiessling M. Autissier P. Sodroski J. The cytoplasmic body component TRIM5alpha restricts HIV-1 infection in Old World monkeys.Nature. 2004; 427: 848-853Crossref PubMed Scopus (1140) Google Scholar]. In keeping with these findings, recent studies using SIV-derived lentiviral vectors in rhesus (Old World) monkeys have resulted in very encouraging results [[36]Hanawa H. et al.Efficient gene transfer into rhesus repopulating hematopoietic stem cells using a simian immunodeficiency virus-based lentiviral vector system.Blood. 2004; 103: 4062-4069Crossref PubMed Scopus (124) Google Scholar]. In addition, studies in the dog model using HIV-1-based vectors have shown relatively efficient gene transfer after an overnight transduction culture with in vivo gene marking levels of up to 12% [[64]Horn P.A. et al.Efficient lentiviral gene transfer to canine repopulating cells using an overnight transduction protocol.Blood. 2004; 103: 3710-3716Crossref PubMed Scopus (56) Google Scholar]. In summary, species-specific effects impact on the effectiveness of lentiviral vectors in large animal studies and it appears that clinical trials will be necessary to determine definitively the success of these vectors in stem cell gene therapy. Of note, inborn antiviral defense mechanisms against HIV have been described in human cells [[65]Turelli P. et al.Cytoplasmic recruitment of INI1 and PML on incoming HIV preintegration complexes: interference with early steps of viral replication.Mol. Cell. 2001; 7: 1245-1254Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar]. How effectively these mechanisms operate in stem cells and if they will have an impact on lentiviral vector-mediated gene transfer is currently unknown. Overall, the development of lentiviral vectors resembles the development of the oncoretroviral vector system, for which 10 to 12 years of refinement were necessary to achieve efficient gene transfer levels in large animal models. Lentiviral vectors were first described in 1996 [[49]Naldini L. et al.In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector.Science. 1996; 272: 263-267Crossref PubMed Google Scholar], and only now are we beginning to see reliable and efficient marking in canine and nonhuman primate transplantation studies [36Hanawa H. et al.Efficient gene transfer into rhesus repopulating hematopoietic stem cells using a simian immunodeficiency virus-based lentiviral vector system.Blood. 2004; 103: 4062-4069Crossref PubMed Scopus (124) Google Scholar, 64Horn P.A. et al.Efficient lentiviral gene transfer to canine repopulating cells using an overnight transduction protocol.Blood. 2004; 103: 3710-3716Crossref PubMed Scopus (56) Google Scholar]. Available preclinical data therefore suggest that lentiviral vectors will be an efficient means for clinical gene transfer to hematopoietic stem cells. Foamy viruses (or spumaviruses) have been proposed as gene therapy vectors [[66]Hirata R.K. Miller A.D. Andrews R.G. Russell D.W. Transduction of hematopoietic cells by foamy virus vectors.Blood. 1996; 88: 3654-3661PubMed Google Scholar]. They have a wide tissue tropism [[67]Linial M.L. Foamy viruses are unconventional retroviruses.J. Virol. 1999; 73: 1747-1755Crossref PubMed Google Scholar], can be concentrated by centrifugation [[68]Vassilopoulos G. Trobridge G. Josephson N.C. Russell D.W. Gene transfer into murine hematopoietic stem cells with helper-free foamy virus vectors.Blood. 2001; 98: 604-609Crossref PubMed Scopus (60) Google Scholar], are nonpathogenic to humans [69Schweizer M. et al.Markers of foamy virus infections in monkeys, apes, and accidentally infected humans: appropriate testing fails to confirm suspected foamy virus prevalence in humans.AIDS Res. Hum. Retroviruses. 1995; 11: 161-170Crossref PubMed Google Scholar, 70Heneine W. et al.Identification of a human population infected with simian foamy viruses.Nat. Med. 1998; 4: 403-407Crossref PubMed Scopus (162) Google Scholar], are not inactivated by human serum, and have a large packaging capacity [71Russell D.W. Miller A.D. Foamy virus vectors.J. Virol. 1996; 70: 217-222Crossref PubMed Scopus (150) Google Scholar, 72Trobridge G. Josephson N. Vassilopoulos G. Mac J. Russell D.W. Improved foamy virus vectors with minimal viral sequences.Mol. Ther. 2002; 6: 321-328Abstract Full Text Full Text PDF PubMed Scopus (71) Google Scholar]. While foamy virus vectors are not capable of transducing nondividing cells, the persistence of a stable transduction intermediate allows efficient ex vivo transduction of (stem) cells that are quiescent during culture but divide following transplantation [[73]Trobridge G. Russell D.W. Cell cycle requirements for transduction by foamy virus vectors compared to those of oncovirus and lentivirus vectors.J. Virol. 2004; 78: 2327-2335Crossref PubMed Scopus (80) Google Scholar]. Foamy virus vectors have been shown to transduce human NOD/SCID repopulating cells efficiently [43Josephson N.C. et al.Transduction of human NOD/SCID-repopulating cells with both lymphoid and myeloid potential by foamy virus vectors.Proc. Natl. Acad. Sci. USA. 2002; 99: 8295-830

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call