Skip to main content Accessibility help
×
Hostname: page-component-84b7d79bbc-fnpn6 Total loading time: 0 Render date: 2024-07-27T16:16:11.681Z Has data issue: false hasContentIssue false

33 - Prospects for Gene Therapy of Sickle Cell Disease and Thalassemia

from SECTION EIGHT - NEW APPROACHES TO THE TREATMENT OF HEMOGLOBINOPATHIES AND THALASSEMIA

Published online by Cambridge University Press:  03 May 2010

Martin H. Steinberg
Affiliation:
Boston University
Bernard G. Forget
Affiliation:
Yale University, Connecticut
Douglas R. Higgs
Affiliation:
MRC Institute of Molecular Medicine, University of Oxford
David J. Weatherall
Affiliation:
Albert Einstein College of Medicine, New York
Get access

Summary

INTRODUCTION

It has been four decades since the concept of gene therapy for human disease began to be considered and discussed in earnest. With the advent of recombinant DNA technology in the 1970s and the concurrent discovery that viruses could carry novel genetic information into target cells, gene therapy was seen as having significant future potential. A major step in the development of gene therapy for hematopoietic disorders occurred in 1985 when the laboratories of Bernstein and Anderson simultaneously showed that the genome of mouse hematopoietic stem cells (HSCs) could be permanently modified using a genetically engineered retrovirus capable of integrating into host cell DNA. These results suggested that a patient's own HSCs could be harvested, genetically modified ex vivo, and then returned, perhaps following administration of radiation or chemotherapy to eradicate the diseased HSCs. From that time, it took almost a decade before functional correction of human disease model was demonstrated using retroviral-mediated, HSC-targeted gene transfer. The first such description was that of Heard and colleagues when they demonstrated the successful treatment of a murine model of the lysosomal disorder, mucopolysaccharidosis type VII. Subsequently, murine models of two immunodeficiencies – chronic granulomatous disease and a form of severe combined immunodeficiency due to JAK3 deficiency – were also successfully treated using gene therapy. These successes seemed to fulfill the stated goals of the Orkin and Motulsky NIH panel, which called for more basic science research into gene therapy and the use of murine disease models to demonstrate efficacy prior to clinical trials.

Type
Chapter
Information
Disorders of Hemoglobin
Genetics, Pathophysiology, and Clinical Management
, pp. 791 - 814
Publisher: Cambridge University Press
Print publication year: 2009

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Dick, JE, Magli, MC, Huszar, D, Phillips, RA, Bernstein, A. Introduction of a selectable gene into primitive stem cells capable of long-term reconstitution of the hemopoietic system of W/Wv mice. Cell. 1985;42:71–79.CrossRefGoogle Scholar
Eglitis, MA, Kantoff, P, Gilboa, E, Anderson, WF. Gene expression in mice after high efficiency retroviral-mediated gene transfer. Science. 1985;230:1395–1398.CrossRefGoogle ScholarPubMed
Marechal, V, Naffakh, N, Danos, O, Heard, JM. Disappearance of lysosomal storage in spleen and liver of mucopolysaccharidosis VII mice after transplantation of genetically modified bone marrow cells. Blood. 1993;82:1358–1365.Google ScholarPubMed
Mardiney, M, Jackson, SH, Spratt, SK, Li, F, Holland, SM, Malech, HL. Enhanced host defense after gene transfer in the murine p47phox-deficient model of chronic granulomatous disease. Blood. 1997;89:2268–2275.Google ScholarPubMed
Bunting, KD, Sangster, MY, Ihle, JN, Sorrentino, BP. Restoration of lymphocyte function in Janus kinase 3-deficient mice by retroviral-mediated gene transfer. Nat Med. 1998;4:58–64.CrossRefGoogle ScholarPubMed
Orkin, SH, Motulsky, AG. Report and recommendations of the panel to assess the NIH investment in research on gene therapy. 1995.
Nienhuis, AW. Development of gene therapy for blood disorders. Blood. 2008;111:4431–4444.CrossRefGoogle ScholarPubMed
Karlsson, S, Bodine, DM, Perry, L, Papayannopoulou, T, Nienhuis, AW. Expression of the human beta-globin gene following retroviral-mediated transfer into multipotential hematopoietic progenitors of mice. Proc Natl Acad Sci USA. 1988;85:6062–6066.CrossRefGoogle Scholar
Dzierzak, EA, Papayannopoulou, T, Mulligan, RC. Lineage-specific expression of a human beta-globin gene in murine bone marrow transplant recipients reconstituted with retrovirus-transduced stem cells. Nature. 1988;331:35–41.CrossRefGoogle ScholarPubMed
Tuan, D, Solomon, W, Li, Q, London, IM. The “beta-like-globin” gene domain in human erythroid cells. Proc Natl Acad Sci USA. 1985;82:6384–6388.CrossRefGoogle ScholarPubMed
Grosveld, F, Assendelft, GB, Greaves, DR, Kollias, G. Position-independent, high-level expression of the human beta-globin gene in transgenic mice. Cell. 1987;51:975–985.CrossRefGoogle ScholarPubMed
Novak, U, Harris, EA, Forrester, W, Groudine, M, Gelinas, R. High-level beta-globin expression after retroviral transfer of locus activation region-containing human beta-globin gene derivatives into murine erythroleukemia cells. Proc Natl Acad Sci USA. 1990;87:3386–3390.CrossRefGoogle ScholarPubMed
Baum, C, Schambach, A, Bohne, J, Galla, M. Retrovirus vectors: toward the plentivirus?Mol Ther. 2006;13:1050–1063.CrossRefGoogle ScholarPubMed
Hargrove, PW, Vanin, EF, Kurtzman, GJ, Nienhuis, AW. High-level globin gene expression mediated by a recombinant adeno-associated virus genome that contains the 3′ γ-globin gene regulatory element and integrates as tandem copies in erythroid cells. Blood. 1997;89:2167–2175.Google ScholarPubMed
Miller, JL, Donahue, RE, Sellers, SE, Samulski, RJ, Young, NS, Nienhuis, AW. Recombinant adeno-associated virus (rAAV)-mediated expression of a human γ-globin gene in human progenitor-derived erythroid cells. Proc Natl Acad Sci USA. 1995;91:10183–10187.CrossRefGoogle Scholar
Nathwani, AC, Hanawa, H, Vandergriff, J, et al. Efficient gene transfer into human cord blood CD34+ cells and the CD34+CD38– subset using highly purified recombinant adeno-associated viral vector preparations that are free of helper virus and wild-type AAV. Gene Ther. 2000;7:183–195.CrossRefGoogle ScholarPubMed
Zhong, L, Zhao, W, Wu, J, et al. Adeno-associated virus-mediated gene transfer in hematopoietic stem/progenitor cells as a therapeutic tool. Curr Gene Ther. 2006;6:683–698.CrossRefGoogle ScholarPubMed
Ryu, BY, Evans-Galea, MV, Gray, JT, et al. An experimental system for the evaluation of retroviral vector design to diminish the risk for proto-oncogene activation. Blood. 2008;111:1866–1875.CrossRefGoogle ScholarPubMed
Wu, Z, Asokan, A, Samulski, RJ. Adeno-associated virus serotypes:vector toolkit for human gene therapy. Mol Ther. 2006;14:316–327.CrossRefGoogle ScholarPubMed
Douglas, JT. Adenoviral vectors for gene therapy. Mol Biotechnol. 2007;36:71–80.CrossRefGoogle ScholarPubMed
Wang, Y. HSV-1 amplicon vectors are an efficient gene transfer system for skeletal muscle cells. Curr Gene Ther. 2006;6:371–381.CrossRefGoogle Scholar
Berges, BK, Wolfe, JH, Fraser, NW. Transduction of brain by herpes simplex virus vectors. Mol Ther. 2007;15:20–29.CrossRefGoogle ScholarPubMed
Kimchi-Sarfaty, C, Gottesman, MM. SV40 pseudovirions as highly efficient vectors for gene transfer and their potential application in cancer therapy. Curr Pharm Biotechnol. 2004;5:451–458.CrossRefGoogle ScholarPubMed
Sova, P, Wang, H, Bomsztyk, K, Stamatoyannopoulos, G, Lieber, A. Role of chromatin structure in integration of helper-dependent adenoviral vectors containing the beta-globin locus control region. Hum Gene Ther. 2008;19:153–166.CrossRefGoogle ScholarPubMed
Baltimore, D. RNA-dependent DNA polymerase in virions of RNA tumour viruses. Nature. 1970;226:1209–1211.CrossRefGoogle ScholarPubMed
Temin, HM, Mizutani, S. RNA-dependent DNA polymerase in virions of Rous sarcoma virus. Nature. 1970;226:1211–1213.CrossRefGoogle ScholarPubMed
Varmus, H. Retroviruses. Science. 1988;240:1427–1435.CrossRefGoogle ScholarPubMed
Mann, R, Mulligan, RC, Baltimore, D. Construction of a retrovirus packaging mutant and its use to produce helper-free defective retrovirus. Cell. 1983;33:153–159.CrossRefGoogle ScholarPubMed
Watanabe, S, Temin, HM. Construction of a helper cell line for avian reticuloendotheliosis virus cloning vectors. Mol Cell Biol. 1983;3:2241–2249.CrossRefGoogle ScholarPubMed
Cone, RD, Mulligan, RC. High-efficiency gene transfer into mammalian cells: generation of helper-free recombinant retrovirus with broad mammalian host range. Proc Natl Acad Sci USA. 1984;81:6349–6635.CrossRefGoogle ScholarPubMed
Markowitz, D, Goff, S, Bank, A. A safe packaging line for gene transfer: Separating viral genes on two different plasmids. J Virol. 1988;62:1120–1124.Google ScholarPubMed
Markowitz, D, Goff, S, Bank, A. Construction and use of a safe and efficient amphotropic packaging cell line. Virology. 1988;167:400–406.CrossRefGoogle ScholarPubMed
Bender, MA, Palmer, TD, Gelinas, RE, Miller, AD. Evidence that the packaging signal of Moloney murine leukemia virus extends into the gag region. J Virol. 1987;61:1639–1646.Google ScholarPubMed
Miller, AD, Rosman, GJ. Improved retroviral vectors for gene transfer and expression. Biotechniques. 1989;7:980–990.Google ScholarPubMed
Ory, DS, Neugeboren, BA, Mulligan, RC. A stable human-derived packaging cell line for production of high titer retrovirus/vesicular stomatitis virus G pseudotypes. Proc Natl Acad Sci USA. 1996;93:11400–11406.CrossRefGoogle ScholarPubMed
Davis, BM, Reese, JS, Koc, ON, et al. Selection for G156A O6-methylguanine DNA methyltransferase gene-transduced hematopoietic progenitors and protection from lethality in mice treated with O6-benzylguanine and 1,3-Bis(2-chloroethyl)-1-nitrosourea. Cancer Res. 1997;57:5093–5099.Google ScholarPubMed
Kim, JW, Closs, EI, Albritton, LM, Cunningham, JM. Transport of cationic amino acids by the mouse ecotropic retrovirus receptor. Nature. 1991;352:725–728.CrossRefGoogle ScholarPubMed
Wang, H, Kavanaugh, MP, North, RA, Kabat, D. Cell-surface receptor for ecotropic murine retroviruses is a basic amino-acid transporter. Nature. 1991;352:729–731.CrossRefGoogle ScholarPubMed
Kavanaugh, MP, Miller, DG, Zhang, W, et al. Cell-surface receptors for gibbon ape leukemia virus and amphotropic murine retrovirus are inducible sodium-dependent phosphate symporters. Proc Natl Acad Sci USA. 1994;91:7071–7075.CrossRefGoogle ScholarPubMed
Miller, DG, Edwards, RH, Miller, AD. Cloning of the cellular receptor for amphotropic murine retroviruses reveals homology to that for gibbon ape leukemia virus. Proc Natl Acad Sci USA. 1994;91:78–82.CrossRefGoogle ScholarPubMed
Miller, AD, Garcia, JV, Subh, N, et al. Construction and properties of retrovirus packaging cells based on gibbon ape leukemia virus. J Virol. 1991;65:2220–2224.Google ScholarPubMed
Kelly, PF, Vandergriff, J, Nathwani, A, Nienhuis, AW, Vanin, EF. Highly efficient gene transfer into cord blood nonobese diabetic/severe combined immunodeficiency repopulating cells by oncoretroviral vector particles pseudotyped with the feline endogenous retrovirus (RD114) envelope protein. Blood. 2000;96:1206–1214.Google ScholarPubMed
Lucas, ML, Seidel, NE, Porada, CD, et al. Improved transduction of human sheep repopulating cells by retrovirus vectors pseudotyped with feline leukemia virus type C or RD114 envelopes. Blood. 2005;106:51–58.CrossRefGoogle ScholarPubMed
Sandrin, V, Russell, SJ, Cosset, FL. Targeting retroviral and lentiviral vectors. Curr Top Microbiol Immunol. 2003;281:137–178.Google ScholarPubMed
Anderson, JL, Hope, TJ. Intracellular trafficking of retroviral vectors: obstacles and advances. Gene Ther. 2005;12:1667–1678.CrossRefGoogle ScholarPubMed
Naghavi, MH, Goff, SP. Retroviral proteins that interact with the host cell cytoskeleton. Curr Opin Immunol. 2007;19:402–407.CrossRefGoogle ScholarPubMed
Miller, DG, Adam, MA, Miller, AD. Gene transfer by retrovirus vectors occurs only in cells that are actively replicating at the time of infection [published erratum appears in Mol Cell Biol. 1992;12:433]. Mol Cell Biol. 1990;10:4239–4242.CrossRefGoogle Scholar
Lewis, PF, Emerman, M. Passage through mitosis is required for oncoretroviruses but not for the human immunodeficiency virus. J Virol. 1994;68:510–516.Google Scholar
Goff, SP. Retrovirus restriction factors. Mol Cell. 2004;16:849–859.CrossRefGoogle ScholarPubMed
Bieniasz, PD. Intrinsic immunity: a front-line defense against viral attack. Nat Immunol. 2004;5:1109–1115.CrossRefGoogle ScholarPubMed
Harris, RS, Liddament, MT. Retroviral restriction by APOBEC proteins. Nat Rev Immunol. 2004;4:868–877.CrossRefGoogle ScholarPubMed
Challita, PM, Kohn, DB. Lack of expression from a retroviral vector after transduction of murine hematopoietic stem cells is associated with methylation in vivo. Proc Natl Acad Sci USA. 1994;91:2567–2571.CrossRefGoogle ScholarPubMed
Chen, WY, Bailey, EC, McCune, SL, et al. Reactivation of silenced, virally transduced genes by inhibitors of histone deacetylase. Proc Natl Acad Sci USA. 1997;94:5798–5803.CrossRefGoogle ScholarPubMed
Cheng, L, Du, C, Lavau, C, et al. Sustained gene expression in retrovirally transduced, engrafting human hematopoietic stem cells and their lympho-myeloid progeny. Blood. 1998;92:83–92.Google ScholarPubMed
Halene, S, Wang, L, Cooper, RM, et al. Improved expression in hematopoietic and lymphoid cells in mice after transplantation of bone marrow transduced with a modified retroviral vector. Blood. 1999;94:3349–3357.Google ScholarPubMed
Ramezani, A, Hawley, TS, Hawley, RG. Stable gamma retroviral vector expression during embryonic stem cell-derived in vitro hematopoietic development. Mol Ther. 2006;14:245–254.CrossRefGoogle Scholar
Schambach, A, Mueller, D, Galla, M, et al. Overcoming promoter competition in packaging cells improves production of self-inactivating retroviral vectors. Gene Ther. 2006;13:1524–1533.CrossRefGoogle ScholarPubMed
Thornhill, SI, Schambach, A, Howe, SJ, et al. Self-inactivating gammaretroviral vectors for gene therapy of X-linked severe combined immunodeficiency. Mol Ther. 2008;16:590–598.CrossRefGoogle ScholarPubMed
Emerman, M, Temin, HM. Genes with promoters in retrovirus vectors can be independently suppressed by an epigenetic mechanism. Cell. 1984;39:459–467.CrossRefGoogle ScholarPubMed
Morgan, RA, Couture, L, Elroy-Stein, O, et al. Retroviral vectors containing putative internal ribosome entry sites: development of a polycistronic gene transfer system and applications to human gene therapy. Nucl Acids Res. 1992;20:1293–1299.CrossRefGoogle ScholarPubMed
Persons, DA, Allay, JA, Allay, ER, 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;90:1777–1786.Google ScholarPubMed
Persons, DA, Allay, JA, Riberdy, JM, et al. Utilization of the green flourescent protein gene as a marker to identify and track genetically-modified hematopoietic cells. Nat Med. 1998;4:1201–1205.CrossRefGoogle Scholar
Szymczak, AL, Workman, CJ, Wang, Y, et al. Correction of multi-gene deficiency in vivo using a single ‘self-cleaving’ 2A peptide-based retroviral vector. Nat Biotechnol. 2004;22:589–594.CrossRefGoogle ScholarPubMed
Hantzopoulos, PA, Sullenger, BA, Ungers, G, Gilboa, E. Improved gene expression upon transfer of the adenosine deaminase minigene outside the transcriptional unit of a retroviral vector. Proc Natl Acad Sci USA. 1989;86:3519–3523.CrossRefGoogle ScholarPubMed
Plavec, I, Papayannopoulou, T, Maury, C, Meyer, F. A human beta-globin gene fused to the human beta-globin locus control region is expressed at high levels in erythroid cells of mice engrafted with retrovirus-transduced hematopoietic stem cells. Blood. 1993;81:1384–1392.Google ScholarPubMed
Leboulch, P, Huang, GM, Humphries, RK, et al. Mutagenesis of retroviral vectors transducing human beta-globin gene and beta-globin locus control region derivatives results in stable transmission of an active transcriptional structure. EMBO J. 1994;13:3065–3076.Google ScholarPubMed
Sadelain, M, Wang, CH, Antoniou, M, et al. Generation of a high-titer retroviral vector capable of expressing high levels of the human beta-globin gene. Proc Natl Acad Sci USA. 1995;92:6728–6732.CrossRefGoogle ScholarPubMed
Ren, S, Wong, BY, Li, J, et al. Production of genetically stable high-titer retroviral vectors that carry a human γ-globin gene under the control of the alpha-globin locus control region. Blood. 1996;87:2518–2524.Google ScholarPubMed
May, C, Rivella, S, Callegari, J, et al. Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin. Nature. 2000;406:82–86.CrossRefGoogle ScholarPubMed
Bukrinsky, MI, Haggerty, S, Dempsey, MP, et al. A nuclear localization signal within HIV-1 matrix protein that governs infection of non-dividing cells. Nature. 1993;365:666–669.CrossRefGoogle ScholarPubMed
Heinzinger, NK, Bukinsky, MI, Haggerty, SA, et al. The Vpr protein of human immunodeficiency virus type 1 influences nuclear localization of viral nucleic acids in nondividing host cells. Proc Natl Acad Sci USA. 1994;91:7311–7315.CrossRefGoogle ScholarPubMed
Zennou, V, Petit, C, Guetard, D, et al. HIV-1 genome nuclear import is mediated by a central DNA flap. Cell. 2000;101:173–185.CrossRefGoogle ScholarPubMed
Sirven, A, Pflumino, F, Zennou, V, et al. The human immunodeficiency virus type-1 central DNA flap is a crucial determinant for lentiviral vector nuclear import and gene transduction of human hematopoietic stem cells. Blood. 2000;96:4103–4110.Google ScholarPubMed
Yamashita, M, Emerman, M. Retroviral infection of non-dividing cells: old and new perspectives. Virology. 2006;334:88–93.CrossRefGoogle Scholar
Caron, MC, Caruso, M. A nuclear localization signal in the matrix of spleen necrosis virus (SNV) does not allow efficient gene transfer into quiescent cells with SNV-derived vectors. Virology. 2005;338:292–296.CrossRefGoogle Scholar
Yamashita, M, Emerman, M. The cell cycle independence of HIV infections is not determined by known karyophilic viral elements. PLoS. Pathog. 2005;1:e18.CrossRefGoogle Scholar
Yamashita, M, Emerman, M. Capsid is a dominant determinant of retrovirus infectivity in nondividing cells. J Virol. 2004;78:5670–5678.CrossRefGoogle ScholarPubMed
Yamashita, M, Perez, O, Hope, TJ, Emerman, M. Evidence for direct involvement of capsid protein in Hiv infection of nondividing cells. PLoS Pathog. 2007;3:1502–1510.CrossRefGoogle ScholarPubMed
Parolin, C, Taddeo, B, Palu, G, Sodroski, J. Use of cis- and trans-acting viral regulatory sequences to improve expression of human immunodeficiency virus vectors in human lymphocytes. Virology. 1996;222:415–422.CrossRefGoogle ScholarPubMed
Akkina, RK, Walton, RM, Chen, ML, et al. High-efficiency gene transfer into CD34+ cells with a human immunodeficiency virus type 1-based retroviral vector pseudotyped with vesicular stomatitis virus envelope glycoprotein G. J Virol. 1996;70:2581–2585.Google ScholarPubMed
Naldini, L, Blomer, U, Gally, P, et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science. 1996;272:263–267.CrossRefGoogle ScholarPubMed
Hanawa, H, Kelly, PF, Nathwani, AC, et al: Comparison of various envelope proteins for their ability to pseudotype lentiviral vectors and transduce primitive hematopoietic cells from human blood. Mol. Ther. 2002;5:242–251.CrossRefGoogle ScholarPubMed
Charneau, P, Marambeau, G, Roux, P, et al. Hiv-1 reverse transcription. A termination step at the center of the genome. J Mol Biol. 1994;241:651–662.CrossRefGoogle ScholarPubMed
Zufferey, R, Nagy, D, Mandel, RJ, et al. Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo. Nat. Biotechnol. 1997;15:871–875.CrossRefGoogle ScholarPubMed
Fuller, M, Anson, DS. Helper plasmids for production of HIV-1-derived vectors. Hum Gene Ther. 2001;12:2081–2093.CrossRefGoogle ScholarPubMed
Westerman, KA, Ao, Z, Cohen, EA, Leboulch, P. Design of a trans protease lentiviral packaging system that produces high titer virus. Retrovirology. 2007;4:96–110.CrossRefGoogle ScholarPubMed
Cockrell, AS, Kafri, T. Gene delivery by lentivirus vectors. Mol Biotechnol. 2007;36:184–204.CrossRefGoogle ScholarPubMed
Kafri, T, Praag, H, Ouyang, L et al. A packaging cell line for lentivirus vectors. J Virol. 1999;73:576–584.Google ScholarPubMed
Kim, VN, Mitrophanous, K, Kingsman, SM, Kingsman, AJ. Minimal requirement for a lentivirus vector based on human immunodeficiency virus type 1. J Virol. 1998;72:811–816.Google ScholarPubMed
Ikeda, Y, Takeuchi, Y, Martin, F, et al. Continuous high-titer HIV-1 vector production. Nat Biotechnol. 2003;21:569–572.CrossRefGoogle ScholarPubMed
Strang, BL, Ikeda, Y, Cosset, FL, Collins, MK, Takeuchi, U. Characterization of HIV-1 vectors with gammaretrovirus envelope glycoproteins produced from stable packaging cells. Gene Ther. 2004;11:591–598.CrossRefGoogle Scholar
Hwang, LH, Gilboa, E. Expression of genes introduced into cells by retroviral infection is more efficient than that of genes introduced into cells by DNA transfection. J Virol. 1984;50:417–424.Google ScholarPubMed
Broussau, S, Jabbour, N, Lachapelle, G, et al. Inducible packaging cells for large-scale production of lentiviral vectors in serum-free suspension culture. Mol Ther. 2008;16:500–507.CrossRefGoogle ScholarPubMed
Schambach, A, Galla, M, Maetzig, T, Loew, R, Baum, C. Improving transcriptional termination of self-inactivating gamma-retroviral and lentiviral vectors. Mol Ther. 2007;15:1167–1173.CrossRefGoogle ScholarPubMed
Gaszner, M, Felsenfeld, G. Insulators: exploiting transcriptional and epigenetic mechanisms. Nat Rev Genet. 2006;7:703–713.CrossRefGoogle ScholarPubMed
Ramezani, A, Hawley, TS, Hawley, RG. Performance-and safety-enhanced lentiviral vectors containing the human interferon-beta scaffold attachment region and the chicken beta-globin insulator. Blood. 2003;101:4717–4724.CrossRefGoogle ScholarPubMed
Evans-Galea, MV, Wielgosz, MM, Hanawa, H, et al. Suppression of clonal dominance in cultured human lymphoid cells by addition of the cHS4 insulator to a lentiviral vector. Mol Ther. 2007;15:801–809.CrossRefGoogle ScholarPubMed
Charrier, S, Dupre, L, Scaramuzza, S, et al. Lentiviral vectors targeting WASp expression to hematopoietic cells, efficiently transduce and correct cells from WAS patients. Gen Ther. 2007;14:415–428.CrossRefGoogle ScholarPubMed
Frecha, C, Toscano, MG, Costa, C, et al. Improved lentiviral vectors for Wiskott-Aldrich syndrome gene therapy mimic endogenous expression profiles throughout haematopoiesis. Gene Ther. 2008;Mar 6;[Epub ahead of print].
Kafri, T, Blomer, U, Peterson, DA, et al. Sustained expression of genes delivered directly into liver and muscle by lentiviral vectors. Nat Genet. 1997;17:314–317.CrossRefGoogle ScholarPubMed
Naldini, L, Blomer, U, Gage, FH et al. Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proc Natl Acad Sci USA. 1996; 93:11382–11388.CrossRefGoogle ScholarPubMed
Blomer, U, Naldini, L, Kafri, T, et al. Highly efficient and sustained gene transfer in adult neurons with a lentivirus vector. J Virol. 1997;71:6641–6649.Google ScholarPubMed
Miyoshi, H, Takahashi, M, Gage, FH, Verma, IM. Stable and efficient gene transfer into the retina using an HIV-based lentiviral vector. Proc Natl Acad Sci USA. 1997;94:10319–10323.CrossRefGoogle ScholarPubMed
Miyoshi, H, Smith, KA, Mosier, , et al. Efficient transduction of human CD34+ cells that mediate long-term engraftment of NOD/SCID mice by HIV vectors. Science. 1999;283:682–686.CrossRefGoogle ScholarPubMed
Brass, AL, Dykxhoorn, DM, Benita, Y, et al. Identification of host proteins required for HIV infection through a functional genomic screen. Science. 2008;319:921–926.CrossRefGoogle ScholarPubMed
Goff, SP. Host factors exploited by retroviruses. Nat Rev Microbiol. 2007;5:253–263.CrossRefGoogle ScholarPubMed
Brennan, G, Kozyrev, Y, Kodama, T, Hu, SL. Novel TRIM5 isoforms expressed by Macaca nemestrina. J Virol. 2007;81:12210–12217.CrossRefGoogle ScholarPubMed
Russell, DW, Miller, AD. Foamy virus vectors. J Virol. 1996;70:217–222.Google ScholarPubMed
Rethwilm, A. Foamy virus vectors: an awaited alternative to gammaretro- and lentiviral vectors. Curr Gene Ther. 2007;7:261–271.CrossRefGoogle ScholarPubMed
Switzer, WM, Bhullar, V, Shanmugam, V, et al. Frequent simian foamy virus infection in persons occupationally exposed to nonhuman primates. J Virol. 2004;78:2780–2789.CrossRefGoogle ScholarPubMed
Hirata, RK, Miller, AD, Andrews, RG, Russell, DW. Transduction of hematopoietic cells by foamy virus vectors. Blood. 1996;88:3654–3661.Google ScholarPubMed
Josephson, NC, Trobridge, G, Russel, DW. Transduction of long-term and mobilized peripheral blood-derived NOD/SCID repopulating cells by foamy virus vectors. Hum Gene Ther. 2004;15:87–92.CrossRefGoogle ScholarPubMed
Leurs, C, Jansen, M, Pollok, KE, et al. Comparison of three retroviral vector systems for transduction of nonobese diabetic/severe combined immunodeficiency mice repopulating human CD34+ cord blood cells. Hum Gene Ther. 2003;14:509–519.CrossRefGoogle ScholarPubMed
Laer, D, Lindemann, D, Roscher, S, et al. Low-level expression of functionally foamy virus receptor on hematopoietic progenitor cells. Virology. 2001;288:139–144.CrossRefGoogle Scholar
Kiem, HP, Allen, J, Trobridge, G, et al. Foamy-virus-mediated gene transfer to canine repopulating cells. Blood. 2007;109:65–70.CrossRefGoogle ScholarPubMed
Bauer, TR, Allen, JM, Hai, M, et al. Successful treatment of canine leukocyte adhesion deficiency by foamy virus vectors. Nat Med. 2008;14:93–97.CrossRefGoogle ScholarPubMed
Trobridge, G, Josephson, N, Vassilopoulos, G, Mac, J, Russell, DW. Improved foamy virus vectors with minimal viral sequences. Mol Ther. 2002;6:321–328.CrossRefGoogle ScholarPubMed
Heinkelein, M, Dressler, M, Jarmy, G, et al. Improved primate foamy virus vectors and packaging constructs. J Virol. 2002;76:3774–3783.CrossRefGoogle ScholarPubMed
Bastone, P, Romen, F, Liu, W, et al. Construction and characterization of efficient, stable and safe replication-deficient foamy virus vectors. Gene Ther. 2007;14:613–620.CrossRefGoogle ScholarPubMed
Trobridge, G, Russell, DW. Cell cycle requirements for transduction by foamy virus vectors compared to those of oncovirus and lentivirus vectors. J Virol. 2004;78:2327–2335.CrossRefGoogle Scholar
Gaspar, HB, Thrasher, AJ. Gene therapy for severe combined immunodeficienies. Expert Opin Biol Ther. 2005;5:1175–1182.CrossRefGoogle Scholar
Fischer, A, Nacein-Bey-Albina, S, Cavazzana-Calvo, M. Gene therapy for immunodeficiency diseases. Semin Hematol. 2004;41:272–278.CrossRefGoogle ScholarPubMed
Stephan, V, Wahn, V, Deist, F, et al. Atypical Xlinked severe combined immunodeficiency due to possible spontaneous reversion of the genetic defect in T cells. N Engl J Med. 1996;335:1563–1567.CrossRefGoogle Scholar
Hacein-Bey-Abina, S, Deist, F, Carlier, F, et al. Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. N Engl J Med. 2002;346:1185–1193.CrossRefGoogle ScholarPubMed
Persons, DA, Allay, ER, Sabatino, , Kelly, P, Bodine, DM, Nienhuis, AW: Functional requirements for phenotypic correction of murine beta-thalassemia: implications for human gene therapy. Blood. 2001;97:3275–3282.CrossRefGoogle ScholarPubMed
Walters, MC, Storb, R, Patience, M, et al. Impact of bone marrow transplantation for symptomatic sickle cell disease: an interim report. Multicenter investigation of bone marrow transplantation for sickle cell disease. Blood. 2000;95:1918–1924.Google ScholarPubMed
Hanawa, H, Hematti, P, Keyvanfar, K, et al. Efficient gene transfer into rhesus repopulating hematopoietic stem cells using a simian immunodeficiency virus-based lentiviral vector system. Blood. 2004;103:4062–4069.CrossRefGoogle ScholarPubMed
Orlic, D, Girard, LJ, Jordan, CT, Anderson, SM, Cline, AP, Bodine, DM. The level of mRNA encoding the amphotropic retrovirus receptor in mouse and human hematopoietic stem cells is low and correlates with the efficiency of retrovirus transduction. Proc Natl Acad Sci USA. 1996;93:11097–11102.CrossRefGoogle ScholarPubMed
Cheshier, SH, Morrison, SJ, Liao, X, Weissman, IL. In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells. Proc Natl Acad Sci USA. 1999;96:3120–3125.CrossRefGoogle ScholarPubMed
Mahmud, N, Devine, SM, Weller, KP, et al. The relative quiescence of hematopoietic stem cells in nonhuman primates. Blood. 2001;97:3061–3068.CrossRefGoogle ScholarPubMed
Zhang, S, Joseph, G, Pollok, K, et al. G2 cell cycle arrest and cyclophilin A in lentiviral gene transfer. Mol Ther. 2006;14:546–454.CrossRefGoogle ScholarPubMed
Conneally, E, Cashman, J, Petzer, A, Eaves, C. Expansion in vitro of transplantable human cord blood stem cells demonstrated using a quantitative assay of their lympho-myeloid repopulating activity in nonobese diabetic-scid/scid mice. Proc Natl Acad Sci USA. 1997;94:9836–9841.CrossRefGoogle ScholarPubMed
Sorrentino, BP. Clinical strategies for expansion of haematopoietic stem cells. Nat Rev Immunol. 2004;4:878–888.CrossRefGoogle ScholarPubMed
Amsellem, S, Pflumio, F, Bardinet, D, et al. Ex vivo expansion of human hematopoietic stem cells by direct delivery of the HOXB4 homeoprotein. Nat Med. 2003;9:1423–1427.CrossRefGoogle ScholarPubMed
Willert, K, Brown, JD, Daneberg, E, et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature. 2003;423:448–452.CrossRefGoogle ScholarPubMed
Zhang, CC, Kaba, M, Ge, G, et al. Angiopoietin-like proteins stimulate ex vivo expansion of hematopoietic stem cells. Nat Med. 2006;12:240–245.CrossRefGoogle ScholarPubMed
Zhang, CC, Kaba, M, Iizuka, S, Huynh, H, Lodish, HF. Angiopoietin-like 5 and IGFBP2 stimulate ex vivo expansion of human cord blood hematopoietic stem cells as assayed by NOD/SCID transplantation. Blood. 2008 Jan 17 [Epub ahead of print]
Ito, K, Hirao, A, Arai, F, et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med. 2006;12:446–451.CrossRefGoogle ScholarPubMed
Ito, K, Hirao, A, Arai, F, et al. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature. 2004;431:997–1002.CrossRefGoogle ScholarPubMed
Tothova, Z, Kollipara, R, Huntly, BJ, et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. 2007;128:325–333.CrossRefGoogle ScholarPubMed
Lambert, JF, Liu, M, Colvin, GA, et al. Marrow stem cells shift gene expression and engraftment phenotype with cell cycle transit. J Exp Med. 2003;197:1563–1572.CrossRefGoogle ScholarPubMed
Takatoku, M, Sellers, S, Agricola, BA, et al. Avoidance of stimulation improves engraftment of cultured and retrovirally transduced hematopoietic cells in primates. J Clin Invest. 2001;108:447–455.CrossRefGoogle ScholarPubMed
Christopherson, KW 2nd, Hangoc, G, Mantel, CR, Broxmeyer, HE. Modulation of hematopoietic stem cell homing and engraftment by CD26. Science. 2004;305(5686):1000–1003CrossRefGoogle ScholarPubMed
Christopherson, KW 2nd, Paanessi, , Napier, S, Porecha, NK. CD26 inhibition on CD34+ or lineage-human umbilical cord blood donor hematopoietic stem cells/hematopoietic progenitor cells improves long-term engraftment into NOD/SCID/beta2-null immunodeficient mice. Stem Cells Dev. 2007;16:355–360.CrossRefGoogle ScholarPubMed
Puthenveetil, G, Scholes, J, Carbonell, D, et al. Successful correction of the human beta-thalassemia major phenotype using a lentiviral vector. Blood. 2004;104:3445–3453.CrossRefGoogle ScholarPubMed
Neff, T, Beard, BC, Kiem, HP. Survival of the fittest: in vivo selection and stem cell gene therapy. Blood. 2006;107:1751–1760.CrossRefGoogle ScholarPubMed
Allay, JA, Persons, DA, Galipeau, J, et al. In vivo selection of retrovirally transduced hematopoietic stem cells. Nat Med. 1998;4:1136–1143.CrossRefGoogle ScholarPubMed
Persons, DA, Allay, JA, Bonifacino, A, et al. Transient in vivo selection of transduced peripheral blood cells using antifolate drug selection in rhesus macaques that received transplants with hematopoietic stem cells expressing dihydrofolate reductase vectors. Blood. 2004;103:796–803.CrossRefGoogle ScholarPubMed
Liu, L, Gerson, SL. Targeted modulation of MGMT: clinical implications. Clin Cancer Res. 2006;12:328–331.CrossRefGoogle ScholarPubMed
Ragg, S, Xu-Welliver, M, Bailey, J, et al. Direct reversal of DNA damage by mutant methyltransferase protein protects mice against dose-intensified chemotherapy and leads to in vivo selection of hematopoietic stem cells. Cancer Res. 2000;60:5187–5195.Google ScholarPubMed
Sawai, N, Zhou, S, Vanin, EF, Houghton, P, Brent, TP, Sorrentino, BP. Protection and in vivo selection of hematopoietic stem cells using temozolomide, O6-benzylguanine, and an alkyltransferase-expressing retroviral vector. Mol Ther. 2001;3:78–87.CrossRefGoogle ScholarPubMed
Persons, DA, Allay, ER, Sawai, N, et al. Successful treatment of murine beta-thalassemia using in vivo selection of genetically modified, drug-resistant hematopoietic stem cells. Blood. 2003;101:506–513.CrossRefGoogle Scholar
Neff, T, Horn, PA, Peterson, LJ, et al. Methylguanine methyltransferase-mediated in vivo selection and chemoprotection of allogeneic stem cells in a large-animal model. J Clin Invest. 2003;112:1581–1588.CrossRefGoogle Scholar
Neff, T, Beard, BC, Peterson, LJ, Anandakumar, P, Thompson, J, Kiem, HP. Polyclonal chemoprotection against temozolomide in a large-animal model of drug resistance gene therapy. Blood. 2005;105:997–1002.CrossRefGoogle Scholar
Williams, DA, Lemischka, IR, Nathan, DG, Mulligan, RC. Introduction of new genetic material into pluripotent haematopoietic stem cells of the mouse. Nature. 1984;310:476–480.CrossRefGoogle ScholarPubMed
Ingram, VM. Abnormal human haemoglobins. III. The chemical difference between normal and sickle cell haemoglobins. Biochim Biophys Acta. 1959;36:402–411.CrossRefGoogle ScholarPubMed
Maniatis, T, Kee, SG, Efstratiadis, A, Kafatos, FC. Amplification and characterization of a beta-globin gene synthesized in vitro. Cell. 1976;8:163–182.CrossRefGoogle ScholarPubMed
Lawn, RM, Fritsch, EF, Parker, RC, Blake, G, Maniatis, T. The isolation and characterization of linked delta- and beta-globin genes from a cloned library of human DNA. Cell. 1978;15:1157–1174.CrossRefGoogle ScholarPubMed
Blattner, FR, Blechl, AE, Denniston-Thompson, K, et al. Cloning human fetal gamma globin and mouse alpha-type globin DNA: preparation and screening of shotgun collections. Science. 1978;202:1279–1284.CrossRefGoogle ScholarPubMed
Smithies, O, Blechl, AE, Denniston-Thompson, K, et al. Cloning human fetal gamma globin and mouse alpha-type globin DNA: characterization and partial sequencing. Science. 1978;202:1284–1289.CrossRefGoogle ScholarPubMed
Efstratiadis, A, Posakony, JW, Maniatis, T, et al. The structure and evolution of the human beta-globin gene family. Cell. 1980;21:653–668.CrossRefGoogle ScholarPubMed
Tiemeier, DC, Tilghman, SM, Polsky, FI, et al. A comparison of two cloned mouse beta-globin genes and their surrounding and intervening sequences. Cell. 1978;14:237–245.CrossRefGoogle ScholarPubMed
Ramirez, F, Natta, C, O'Donnell, JV, et al. Relative numbers of human globin genes assayed with purified alpha and beta complementary human DNA. Proc Natl Acad Sci USA. 1975;72:1550–1554.CrossRefGoogle ScholarPubMed
Ottolenghi, S, Lanyon, WG, Williamson, R, Weatherall, DJ, Clegg, JB, Pitcher, CS. Human globin gene analysis for a patient with beta-o/delta beta-thalassemia. Proc Natl Acad Sci USA. 1975;72:2294–2299.CrossRefGoogle ScholarPubMed
Fritsch, EF, Lawn, RM, Maniatis, T. Molecular cloning and characterization of the human beta-like globin gene cluster. Cell. 1980;19:959–972.CrossRefGoogle ScholarPubMed
Bender, MA, Gelinas, RE, Miller, AD. A majority of mice show long-term expression of a human beta-globin gene after retrovirus transfer into hematopoietic stem cells. Mol Cell Biol. 1989;9:1426–1434.CrossRefGoogle ScholarPubMed
Tuan, D, London, IM. Mapping of DNase I-hypersensitive sites in the upstream DNA of human embryonic epsilon-globin gene in K562 leukemia cells. Proc Natl Acad Sci USA. 1984;81:2718–2722.CrossRefGoogle ScholarPubMed
Forrester, WC, Novak, U, Gelinas, R, Groudine, M. Molecular analysis of the human beta-globin locus activation region. Proc Natl Acad Sci USA. 1989;86:5439–5443.CrossRefGoogle ScholarPubMed
Skow, LC, Burkhart, BA, Johnson, FM, et al. A mouse model for beta-thalassemia. Cell. 1983;34:1043–1052.CrossRefGoogle ScholarPubMed
Yang, B, Kirby, S, Lewis, J, Detloff, PJ, Maeda, N, Smithies, O. A mouse model for beta 0-thalassemia. Proc Natl Acad Sci USA. 1995;92:11608–11612.CrossRefGoogle ScholarPubMed
Persons, DA, Hargrove, PW, Allay, ER, Hanawa, H, Nienhuis, AW. The degree of phenotypic correction of murine beta-thalassemia intermedia following lentiviral-mediated transfer of a human gamma-globin gene is influenced by chromosomal position effects and vector copy number. Blood. 2003;101:2175–2183.CrossRefGoogle ScholarPubMed
Hanawa, H, Hargrove, PW, Kepes, S, Srivastava, DK, Nienhuis, AW, Persons, DA. Extended beta-globin locus control region elements promote consistent therapeutic expression of a gamma-globin lentiviral vector in murine beta-thalassemia. Blood. 2004;104:2281–2290.CrossRefGoogle ScholarPubMed
Imren, S, Payen, E, Westerman, KA, et al. Permanent and panerythroid correction of murine beta thalassemia by multiple lentiviral integration in hematopoietic stem cells. Proc Natl Acad Sci USA. 2002;99:14380–14385.CrossRefGoogle ScholarPubMed
Greaves, DR, Fraser, P, Vidal, MA, et al. A transgenic mouse model of sickle cell disorder. Nature. 1990;343:183–185.CrossRefGoogle ScholarPubMed
Ryan, TM, Townes, TM, Reilly, MP, et al. Human sickle hemoglobin in transgenic mice. Science. 1990;247:566–568.CrossRefGoogle ScholarPubMed
Trudel, M, Saadane, N, Garel, MC, et al. Towards a transgenic mouse model of sickle cell disease: hemoglobin SAD. EMBO J. 1991;10:3157–3165.Google ScholarPubMed
Fabry, ME, Costantini, F, Pachnis, A, et al. High expression of human beta S- and alpha-globins in transgenic mice: erythrocyte abnormalities, organ damage, and the effect of hypoxia. Proc Natl Acad Sci USA. 1992;89:12155–12159.CrossRefGoogle Scholar
Roy, RP, Nagel, RL, Acharya, AS. Molecular basis of the inhibition of beta s-chain-dependent polymerization by mouse alpha-chain. Semisynthesis of chimeras of human and mouse alpha-chains. J Biol Chem. 1993;268:16406–16412.Google ScholarPubMed
Paszty, C, Mohandas, N, Stevens, ME, et al. Lethal alpha-thalassaemia created by gene targeting in mice and its genetic rescue. Nat Genet. 1995;11:33–39.CrossRefGoogle ScholarPubMed
Chang, J, Lu, RH, Xu, SM, et al. Inactivation of mouse alpha-globin gene by homologous recombination: mouse model of hemoglobin H disease. Blood. 1996;88:1846–1851.Google ScholarPubMed
Ryan, TM, Ciavatta, DJ, Townes, TM. Knockout-transgenic mouse model of sickle cell disease. Science. 1997;278:873–876.CrossRefGoogle ScholarPubMed
Paszty, C, Brion, CM, Manci, E, et al. Transgenic knockout mice with exclusively human sickle hemoglobin and sickle cell disease. Science. 1997;278:876–878.CrossRefGoogle ScholarPubMed
Pawliuk, R, Westerman, KA, Fabry, ME, et al. Correction of sickle cell disease in transgenic mouse models by gene therapy. Science. 2001;294:2368–2371.CrossRefGoogle ScholarPubMed
Kaul, DK, Hebbel, RP. Hypoxia/reoxygenation causes inflammatory response in transgenic sickle mice but not in normal mice. J Clin Invest. 2000;106:411–420.CrossRefGoogle ScholarPubMed
Dasgupta, T, Hebbel, RP, Kaul, DK. Protective effect of arginine on oxidative stress in transgenic sickle mouse models. Free Radic Biol Med. 2006;41:1771–1780.CrossRefGoogle ScholarPubMed
Belcher, JD, Mahaseth, H, Welch, TE, Otterbein, , Hebbel, RP, Vercellotti, GM. Heme oxygenase-1 is a modulator of inflammation and vaso-occlusion in transgenic sickle mice. J Clin Invest. 2006;116:808–816.CrossRefGoogle ScholarPubMed
Turhan, A, Weiss, , Mohandas, N, Coller, BS, Frenette, PS. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. Proc Natl Acad Sci USA. 2002;99:3047–3051.CrossRefGoogle ScholarPubMed
Chang, JC, Lu, R, Lin, C, et al. Transgenic knockout mice exclusively expressing human hemoglobin S after transfer of a 240-kb betas-globin yeast artificial chromosome: A mouse model of sickle cell anemia. Proc Natl Acad Sci USA. 1998;95:14886–14890.CrossRefGoogle ScholarPubMed
Fabry, ME, Suzuka, SM, Weinberg, RS, et al. Second generation knockout sickle mice: the effect of HbF. Blood. 2001;97:410–418.CrossRefGoogle ScholarPubMed
Kaul, DK, Liu, XD, Chang, HY, Nagel, RL, Fabry, ME. Effect of fetal hemoglobin on microvascular regulation in sickle transgenic-knockout mice. J Clin Invest. 2004;114:1136–1145.CrossRefGoogle ScholarPubMed
May, C, Rivella, S, Chadburn, A, Sadelain, M. Successful treatment of murine beta-thalassemia intermedia by transfer of the human beta-globin gene. Blood. 2002;99:1902–1908.CrossRefGoogle ScholarPubMed
Rivella, S, May, C, Chadburn, A, Riviere, I, Sadelain, M. A novel murine model of Cooley anemia and its rescue by lentiviral-mediated human beta -globin gene transfer. Blood. 2003;101:2932–2939.CrossRefGoogle ScholarPubMed
Levasseur, DN, Ryan, TM, Pawlik, KM, Townes, TM. Correction of a mouse model of sickle cell disease: lentiviral/antisickling beta-globin gene transduction of unmobilized, purified hematopoietic stem cells. Blood. 2003;102:4312–4329.CrossRefGoogle ScholarPubMed
Sabatino, , Seidel, NE, Aviles-Mendoza, GJ, et al. Long-term expression of gamma-globin mRNA in mouse erythrocytes from retrovirus vectors containing the human gamma-globin gene fused to the ankyrin-1 promoter. Proc Natl Acad Sci USA. 2000;97:13294–13299.CrossRefGoogle ScholarPubMed
Emery, DW, Yannaki, E, Tubb, J, Nishino, T, Li, Q, Stamatoyannopoulos, G. Development of virus vectors for gene therapy of beta chain hemoglobinopathies: flanking with a chromatin insulator reduces gamma-globin gene silencing in vivo. Blood. 2002;100:2012–2019.CrossRefGoogle ScholarPubMed
Pestina, TI, Hargrove, PW, Jay, D, Gray, JT, Boyd, KM, and Persons, DA. Correction of murine sickle cell disease using a novel γ-globin lentiviral vector to mediate high level expression of fetal hemoglobin. Accepted for publication. Molecular Therapy, 2008.
Chung, JH, Bell, AC, Felsenfeld, G. Characterization of the chicken beta-globin insulator. Proc Natl Acad Sci USA. 1997;94:575–580.CrossRefGoogle ScholarPubMed
Arumugam, PI, Scholes, J, Perelman, N, Xia, P, Yee, JK, Malik, P. Improved human beta-globin expression from self-inactivating lentiviral vectors carrying the chicken hypersensitive site-4 (cHS4) insulator element. Mol Ther. 2007;15:1863–1871.CrossRefGoogle ScholarPubMed
Aker, M, Tubb, J, Groth, AC, et al. Extended core sequences from the cHS4 insulator are necessary for protecting retroviral vectors from silencing position effects. Hum Gene Ther. 2007;18:333–343.CrossRefGoogle ScholarPubMed
Weinberg, KI, Kohn, DB. Gene therapy for congenital lymphoid immunodeficiency diseases. Semin Hematol. 1998;35:354–366.Google ScholarPubMed
Blaese, RM, Culver, KW, Miller, AD, et al. T lymphocyte-directed gene therapy for ADA- SCID: initial trial results after 4 years. Science. 1995;270:475–480.CrossRefGoogle Scholar
Kohn, DB, Weinberg, KI, Nolta, JA, et al. Engraftment of gene-modified umbilical cord blood cells in neonates with adenosine deaminase deficiency. Nat Med. 1995;1:1017–1023.CrossRefGoogle ScholarPubMed
Kohn, DB, Hershfield, MS, Carbonaro, D, et al. T lymphocytes with a normal ADA gene accumulate after transplantation of transduced autologous umbilical cord blood CD34+ cells in ADA-deficient SCID neonates. Nat Med. 1998;4:775–780.CrossRefGoogle ScholarPubMed
Cavazzana-Calvo, M, Hacein-Bey, S, Saint Basile, G, et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science. 2000;288:669–672.CrossRefGoogle ScholarPubMed
Otsu, M, Sugamura, K, Candotti, F. In vivo competitive studies between normal and common gamma chain-defective bone marrow cells: implications for gene therapy. Hum Gene Ther. 2000;11:2051–2056.CrossRefGoogle ScholarPubMed
Gaspar, HB, Parsley, KL, Howe, S, et al. Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gamma retroviral vector. Lancet. 2004;364:2181–2187.CrossRefGoogle Scholar
Aiuti, A, Slavin, S, Aker, M, et al. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science. 2002;296:2410–2413.CrossRefGoogle ScholarPubMed
Aiuti, A, Cassani, B, Andolfi, G, et al. Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy. J Clin Invest. 2007;117:2233–2240.CrossRefGoogle ScholarPubMed
Hacein-Bey-Abina, S, Kalle, C, Schmidt, M, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-XI. Science. 2003;301:2526–2527.Google Scholar
Fischer, A, Abina, SH, Thrasher, A, Kalle, C, Cavazzana-Calvo, M. LMO2 and gene therapy for severe combined immunodeficiency. N Engl J Med. 2004;350(24):2526–2527.Google ScholarPubMed
McCormack, MP, Rabbitts, TH. Activation of the T-cell oncogene LMO2 after gene therapy for X-linked severe combined immunodeficiency. N Engl J Med. 2004;350:913–922.CrossRefGoogle ScholarPubMed
Larson, RC, Fisch, P, Larson, TA, et al. T cell tumours of disparate phenotype in mice transgenic for Rbtn-2. Oncogene. 1994;9:3675–3681.Google ScholarPubMed
Wu, X, Li, Y, Crise, B, Burgess, SM. Transcription start regions in the human genome are favored targets for MLV integration. Science. 2003;300:1749–1751.CrossRefGoogle ScholarPubMed
Cattoglio, C, Facchini, G, Sartori, D, et al. Hot spots of retroviral integration in human CD34+ hematopoietic cells. Blood. 2007;110:1770–1778.CrossRefGoogle ScholarPubMed
Mitchell, RS, Beitzel, BF, Schroder, AR, et al. Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLoS Biol. 2004 Aug;2:E234.CrossRefGoogle ScholarPubMed
Lo, M, Bloom, ML, Imada, K, et al. Activation of the T-cell oncogene LMO2 after gene therapy for X-linked severe combined immunodeficiency. N Engl J Med. 2004;350:913–922.Google Scholar
Soudais, C, Shiho, T, Sharara, LI, et al. Stable and functional lymphoid reconstitution of common cytokine receptor gamma chain deficient mice by retroviral-mediated gene transfer. Blood. 2000;95:3071–3077.Google ScholarPubMed
Shou, Y, Ma, Z, Lu, T, Sorrentino, BP. Unique risk factors for insertional mutagenesis in a mouse model of XSCID gene therapy. Proc Natl Acad Sci USA. 2006;10:11730–11735.CrossRefGoogle Scholar
Gardie, B, Cayuela, JM, Martini, S, Sigaux, F. Genomic alterations of the p19ARF encoding exons in T-cell acute lymphoblastic leukemia. Blood. 1998;91:1016–1020.Google ScholarPubMed
Kiem, HP, Sellers, S, Thomasson, B, et al. Long-term clinical and molecular follow-up of large animals receiving retrovirally transduced stem and progenitor cells: no progression to clonal hematopoiesis or leukemia. Mol Ther. 2004;9:389–395.CrossRefGoogle ScholarPubMed
Seggewiss, R, Pittaluga, S, Adler, RL, et al. Acute myeloid leukemia is associated with retroviral gene transfer to hematopoietic progenitor cells in a rhesus macaque. Blood. 2006;107:3865–3867.CrossRefGoogle Scholar
Ott, MG, Schmidt, M, Schwarzwaelder, K, et al. Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nat Med. 2006;12:401–409.CrossRefGoogle ScholarPubMed
Kustikova, O, Fehse, B, Modlich, U, et al. Clonal dominance of hematopoietic stem cells triggered by retroviral gene marking. Science. 2005;308:1171–1174.CrossRefGoogle ScholarPubMed
Calmels, B, Ferguson, C, Laukkanen, MO, et al. Recurrent retroviral vector integration at the Mds1/Evi1 locus in nonhuman primate hematopoietic cells. Blood. 2005;106:2530–2533.CrossRefGoogle ScholarPubMed
Hematti, P, Hong, BK, Ferguson, C, et al. Distinct genomic integration of MLV and SIV vectors in primate hematopoietic stem and progenitor cells. PLoS Biol. 2004;2:e423.CrossRefGoogle ScholarPubMed
Montini, E, Cesana, D, Schmidt, M, et al. Hematopoietic stem cell gene transfer in a tumor-prone mouse model uncovers low genotoxicity of lentiviral vector integration. Nat Biotechnol. 2006;24:687–696.CrossRefGoogle Scholar
Akagi, K, Suzuki, T, Stephens, RM, Jenkins, NA, Copeland, NG. RTCGD: retroviral tagged cancer gene database. Nucl Acids Res. 2004;32(Database issue):D523–527.CrossRefGoogle ScholarPubMed
Hargrove, PW, Kepes, S, Hanawa, H, Obenauer, JC, et al. Globin lentiviral vector insertions can perturb the expression of endogenous genes in beta-thalassemic hematopoietic cells. Mol Ther. 2008;16:525–533.CrossRefGoogle ScholarPubMed
Evans-Galea, MV, Wielgosz, MM, Hanawa, H, Srivastava, DK, Nienhuis, AW. Suppression of clonal dominance in cultured human lymphoid cells by addition of the cHS4 insulator to a lentiviral vector. Mol Ther. 2007;15:801–809.CrossRefGoogle ScholarPubMed
Hanna, J, Wernig, M, Markoulaki, S, Sun, CW, et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science. 2007;318:1920–3.CrossRefGoogle ScholarPubMed
Persons, DA, Tisdale, JF. Gene therapy for hemoglobin disorders. Semin Hematol. 2004;41:279–286.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×