Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-17T19:40:02.083Z Has data issue: false hasContentIssue false

Gene therapy for β-thalassaemia: the continuing challenge

Published online by Cambridge University Press:  01 October 2010

Evangelia Yannaki*
Affiliation:
Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece.
David W. Emery
Affiliation:
Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA. Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
George Stamatoyannopoulos
Affiliation:
Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA. Department of Genome Sciences, University of Washington, Seattle, WA, USA.
*
*Corresponding author: Evangelia Yannaki, G. Papanicolaou Hospital, Gene and Cell Therapy Center, Hematology-BMT Unit, Thessaloniki 57010, Greece. E-mail: eyannaki@u.washington.edu

Abstract

The β-thalassaemias are inherited anaemias that form the most common class of monogenic disorders in the world. Treatment options are limited, with allogeneic haematopoietic stem cell transplantation offering the only hope for lifelong cure. However, this option is not available for many patients as a result of either the lack of compatible donors or the increased risk of transplant-related mortality in subjects with organ damage resulting from accumulated iron. The paucity of alternative treatments for patients that fall into either of these categories has led to the development of a revolutionary treatment strategy based on gene therapy. This approach involves replacing allogeneic stem cell transplantation with the transfer of normal globin genes into patient-derived, autologous haematopoietic stem cells. This highly attractive strategy offers several advantages, including bypassing the need for allogeneic donors and the immunosuppression required to achieve engraftment of the transplanted cells and to eliminate the risk of donor-related graft-versus-host disease. This review discusses the many advances that have been made towards this endeavour as well as the hurdles that must still be overcome before gene therapy for β-thalassaemia, as well as many other gene therapy applications, can be widely applied in the clinic.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2010

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

References

1Stamatoyannopoulos, G. et al. (2001). The Molecular Basis of Blood Diseases. W.B. Saunders, Philadelphia, PA, USAGoogle Scholar
2Weatherall, D.J. and Clegg, J.B. (2001) Inherited haemoglobin disorders: an increasing global health problem. Bulletin of the World Health Organization 79, 704-712Google ScholarPubMed
3Lucarelli, G., Andreani, M. and Angelucci, E. (2002) The cure of thalassemia by bone marrow transplantation. Blood Reviews 16, 81-85CrossRefGoogle ScholarPubMed
4Gaziev, D. et al. (1997) Graft-versus-host disease after bone marrow transplantation for thalassemia: an analysis of incidence and risk factors. Transplantation 63, 854-860CrossRefGoogle ScholarPubMed
5Lucarrelli, G. et al. (1999) Bone marrow transplantation in adult thalassemic patients. Blood 93, 1164-1167CrossRefGoogle Scholar
6La Nasa, G. et al. (2002) Unrelated donor bone marrow transplantation for thalassemia: the effect of extended haplotypes. Blood 15, 4350-4356CrossRefGoogle Scholar
7La Nasa, G. et al. (2005) Unrelated donor stem cell transplantation in adult patients with thalassemia. Bone Marrow Transplantation 36, 971-975CrossRefGoogle ScholarPubMed
8Gaziev, D. et al. (2000) Bone marrow transplantation from alternative donors for thalassemia: HLA-phenotypically identical relative and HLA-nonidentical sibling or parent transplants. Bone Marrow Transplantation 25, 815-821CrossRefGoogle ScholarPubMed
9Lucarelli, G. and Gaziev, J. (2008) Advances in the allogeneic transplantation for thalassemia. Blood Reviews 22, 53-63CrossRefGoogle ScholarPubMed
10Lisowski, L. and Sadelain, M. (2008) Current status of globin gene therapy for the treatment of beta-thalassaemia. British Journal of Haematology 141, 335-345CrossRefGoogle ScholarPubMed
11Emery, D.W. et al. (2002) Hematopoietic stem cell gene therapy. International Journal of Hematology 75, 228-236CrossRefGoogle ScholarPubMed
12Kohn, D.B. et al. (2003) American Society of Gene Therapy (ASGT) ad hoc subcommittee on retroviral-mediated gene transfer to hematopoietic stem cells. Molecular Therapy 8, 180-187CrossRefGoogle Scholar
13Berry, C. et al. (2006) Selection of target sites for mobile DNA integration in the human genome. PLoS Computation Biology 2, e157CrossRefGoogle ScholarPubMed
14Emery, D.W., Aker, M. and Stamatoyannopoulos, G. (2003) Chromatin insulators and position effects. In Gene Transfer and Expression in Mammalian Cells (Makrides, S.C., ed.), pp. 381-395, EIC Laboratories, Inc., Norwood, MA, USACrossRefGoogle Scholar
15Li, Q. et al. (2002) Locus control regions. Blood 100, 3077-3086CrossRefGoogle ScholarPubMed
16Grosveld, F. et al. (1987) Position-independent, high-level expression of the human beta-globin gene in transgenic mice. Cell 51, 975-985CrossRefGoogle ScholarPubMed
17Blom van Assendelft, G. et al. (1989) The beta-globin dominant control region activates homologous and heterologous promoters in a tissue-specific manner. Cell 56, 969-977CrossRefGoogle Scholar
18Karlsson, S. et al. (1987) Retroviral-mediated transfer of genomic globin genes leads to regulated production of RNA and protein. Proceedings of the National Academy of Sciences of the United States of America 84, 2411-2415CrossRefGoogle ScholarPubMed
19Dzierzak, E.A., Papayannopoulou, T. and Mulligan, R.C. (1988) Lineage-specific expression of a human beta-globin gene in murine bone marrow transplant recipients reconstituted with retrovirus-transduced stem cells. Nature 331, 35-41CrossRefGoogle ScholarPubMed
20Plavec, I. et al. (1993) 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 81, 1384-1392CrossRefGoogle Scholar
21Novak, U. et al. (1990) High-level beta-globin expression after retroviral transfer of locus activation region-containing human beta-globin gene derivatives into murine erythroleukemia cells. Proceedings of the National Academy of Sciences of the United States of America 87, 3386-3390CrossRefGoogle ScholarPubMed
22Emery, D.W. et al. (1998) Development of a condensed locus control region cassette and testing in retrovirus vectors for A gamma-globin. Blood Cells, Molecules, and Diseases 24, 322-339CrossRefGoogle ScholarPubMed
23Sadelain, M. et al. (1995) Generation of a high-titer retroviral vector capable of expressing high levels of the human beta-globin gene. Proceedings of the National Academy of Sciences of the United States of America 92, 6728-6732CrossRefGoogle ScholarPubMed
24Miller, A.D. et al. (1988) Design of retrovirus vectors for transfer and expression of the human beta-globin gene. Journal of Virology 62, 4337-4345CrossRefGoogle ScholarPubMed
25Leboulch, P. et al. (1994) 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 Journal 13, 3065-3076CrossRefGoogle ScholarPubMed
26Emery, D.W. et al. (2002) Development of virus vectors for gene therapy of beta chain hemoglobinopathies: flanking with a chromatin insulator reduces gamma-globin gene silencing in vivo. Blood 100, 2012-2019CrossRefGoogle ScholarPubMed
27Miyoshi, H. et al. (1999) Transduction of human CD34+ cells that mediate long-term engraftment of NOD/SCID mice by HIV vectors. Science 283, 682-686CrossRefGoogle ScholarPubMed
28Kumar, M. et al. (2001) Systematic determination of the packaging limit of lentiviral vectors. Human Gene Therapy 12, 1893-1905CrossRefGoogle ScholarPubMed
29May, C. et al. (2000) Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin. Nature 406, 82-86CrossRefGoogle ScholarPubMed
30Dull, T. et al. (1998) A third-generation lentivirus vector with a conditional packaging system. Journal of Virology 72, 8463-8471CrossRefGoogle ScholarPubMed
31Miyoshi, H. et al. (1998) Development of a self-inactivating lentivirus vector. Journal of Virology 72, 8150-8157CrossRefGoogle ScholarPubMed
32Zufferey, R. et al. (1998) Self-inactivating lentivirus vector for safe and efficient in vivo gene delivery. Journal of Virology 72, 9873-9880CrossRefGoogle ScholarPubMed
33Naldini, L. and Verma, I.M. (2000) Lentiviral vectors. Advances in Virus Research 55, 599-609CrossRefGoogle ScholarPubMed
34Kay, M.A., Glorioso, J.C. and Naldini, L. (2001) Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics. Nature Medicine 7, 33-40CrossRefGoogle ScholarPubMed
35Montini, E. et al. (2006) Hematopoietic stem cell gene transfer in a tumor-prone mouse model uncovers low genotoxicity of lentiviral vector integration. Nature Biotechnology 24, 687-696CrossRefGoogle Scholar
36Arumugam, P.I. et al. (2009) Genotoxic potential of lineage-specific lentivirus vectors carrying the beta-globin locus control region. Molecular Therapy 17, 1929-1937CrossRefGoogle ScholarPubMed
37Pawliuk, R. et al. (2001) Correction of sickle cell disease in transgenic mouse models by gene therapy. Science 294, 2368-2371CrossRefGoogle ScholarPubMed
38Imren, S. et al. (2002) Permanent and panerythroid correction of murine beta thalassemia by multiple lentiviral integration in hematopoietic stem cells. Proceedings of the National Academy of Sciences of the United States of America 99, 14380-14385CrossRefGoogle ScholarPubMed
39Persons, D.A. et al. (2003) 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 101, 2175-2183CrossRefGoogle ScholarPubMed
40Rivella, S. et al. (2003) A novel murine model of Cooley anemia and its rescue by lentiviral-mediated human beta-globin gene transfer. Blood 101, 2932-2939CrossRefGoogle ScholarPubMed
41Miccio, A. et al. (2008) In vivo selection of genetically modified erythroblastic progenitors leads to long-term correction of beta-thalassemia. Proceedings of the National Academy of Sciences of the United States of America 105, 10547-10552CrossRefGoogle ScholarPubMed
42Perumbeti, A. et al. (2009) A novel human gamma-globin gene vector for genetic correction of sickle cell anemia in a humanized sickle mouse model: critical determinants for successful correction. Blood 114, 1174-1185CrossRefGoogle Scholar
43Puthenveetil, G. et al. (2004) Successful correction of the human beta-thalassemia major phenotype using a lentiviral vector. Blood 104, 3445-3453CrossRefGoogle ScholarPubMed
44Aker, M. et al. (2007) Extended core sequences from the cHS4 insulator are necessary for protecting retroviral vectors from silencing position effects. Human Gene Therapy 18, 333-343CrossRefGoogle ScholarPubMed
45Arumugam, P.I. et al. (2007) Improved human beta-globin expression from self-inactivating lentiviral vectors carrying the chicken hypersensitive site-4 (cHS4) insulator element. Molecular Therapy 15, 1863-1871CrossRefGoogle ScholarPubMed
46May, C. et al. (2002) Successful treatment of murine beta-thalassemia intermedia by transfer of the human beta-globin gene. Blood 99, 1902-1908CrossRefGoogle ScholarPubMed
47Cavazzana-Calvo, M. et al. (2000) Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 288, 669-672CrossRefGoogle ScholarPubMed
48Hacein-Bey-Abina, S. et al. (2002) Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. New England Journal of Medicine 346, 1185-1193CrossRefGoogle ScholarPubMed
49Gaspar, H.B. et al. (2004) Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector. Lancet 364, 2181-2187CrossRefGoogle ScholarPubMed
50Aiuti, A. et al. (2009) Gene therapy for immunodeficiency due to adenosine deaminase deficiency. New England Journal of Medicine 360, 447-458CrossRefGoogle ScholarPubMed
51Hacein-Bey-Abina, S. et al. (2008) Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. Journal of Clinical Investigation 118, 3132-3142CrossRefGoogle ScholarPubMed
52Howe, S.J. et al. (2008) Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. Journal of Clinical Investigation 118, 3143-3150CrossRefGoogle ScholarPubMed
53Hacein-Bey-Abina, S. et al. (2003) LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 302, 415-419CrossRefGoogle ScholarPubMed
54Ott, M.G. et al. (2006) Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nature Medicine 12, 401-409CrossRefGoogle ScholarPubMed
55Stein, S. et al. (2010) Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease. Nature Medicine 16, 198-204CrossRefGoogle ScholarPubMed
56Aiuti, A. et al. (2007) Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy. Journal of Clinical Investigation 117, 2233-2240CrossRefGoogle ScholarPubMed
57Li, Z. et al. (2002) Murine leukemia induced by retroviral gene marking. Science 296, 497CrossRefGoogle ScholarPubMed
58Shou, Y. et al. (2006) Unique risk factors for insertional mutagenesis in a mouse model of XSCID gene therapy. Proceedings of the National Academy of Sciences of the United States of America 103, 11730-11735CrossRefGoogle Scholar
59Kustikova, O. et al. (2005) Clonal dominance of hematopoietic stem cells triggered by retroviral gene marking. Science 308, 1171-1174CrossRefGoogle ScholarPubMed
60Li, C.L. et al. (2009) Genomic and functional assays demonstrate reduced gammaretroviral vector genotoxicity associated with use of the cHS4 chromatin insulator. Molecular Therapy 17, 716-724CrossRefGoogle ScholarPubMed
61Seggewiss, R. et al. (2006) Acute myeloid leukemia is associated with retroviral gene transfer to hematopoietic progenitor cells in a rhesus macaque. Blood 107, 3865-3867CrossRefGoogle Scholar
62Sadelain, M. (2004) Insertional oncogenesis in gene therapy: how much of a risk? Gene Therapy 11, 569-573CrossRefGoogle ScholarPubMed
63Baum, C. et al. (2002) Side effects of retroviral gene transfer into hematopoietic stem cells. Blood 101, 2099-2114CrossRefGoogle Scholar
64Aker, M. et al. (2006) Integration bias of gammaretrovirus vectors following transduction and growth of primary mouse hematopoietic progenitor cells with and without selection. Molecular Therapy 14, 226-235CrossRefGoogle ScholarPubMed
65Poeschla, E.M. (2008) Integrase, LEDGF/p75 and HIV replication. Cellular and Molecular Life Sciences 65, 1403-1424CrossRefGoogle ScholarPubMed
66Montini, E. et al. (2009) The genotoxic potential of retroviral vectors is strongly modulated by vector design and integration site selection in a mouse model of HSC gene therapy. Journal of Clinical Investigation 119, 964-975CrossRefGoogle Scholar
67Schroder, A.R. et al. (2002) HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110, 521-529CrossRefGoogle ScholarPubMed
68Wu, W. et al. (2003) Transcription start regions in the human genome are favored targets for MLV integration. Science 300, 1749-1751CrossRefGoogle ScholarPubMed
69Zychlinski, D. et al. (2008) Physiological promoters reduce the genotoxic risk of integrating gene vectors. Molecular Therapy 16, 718-725CrossRefGoogle ScholarPubMed
70Chang, A.H. and Sadelain, M. (2007) The genetic engineering of hematopoietic stem cells: the rise of lentiviral vectors, the conundrum of the LTR, and the promise of lineage-restricted vectors. Molecular Therapy 15, 445-456CrossRefGoogle ScholarPubMed
71Hargrove, P.W. et al. (2008) Globin lentiviral vector insertions can perturb the expression of endogenous genes in beta-thalassemic hematopoietic cells. Molecular Therapy 16, 525-533CrossRefGoogle ScholarPubMed
72Gaszner, M. and Felsenfeld, G. (2006) Insulators: exploiting transcriptional and epigenetic mechanisms. Nature Reviews Genetics 7, 703-713CrossRefGoogle ScholarPubMed
73Wallace, J.A. and Felsenfeld, G. (2007) We gather together: insulators and genome organization. Current Opinion in Genetics and Development 17, 400-407CrossRefGoogle ScholarPubMed
74Chung, J.H., Bell, A.C. and Felsenfeld, G. (1997) Characterization of the chicken beta-globin insulator. Proceedings of the National Academy of Sciences of the United States of America 94, 575-580CrossRefGoogle ScholarPubMed
75Rivella, S. et al. (2000) The cHS4 insulator increases the probability of retroviral expression at random chromosomal integration sites. Journal of Virology 74, 4679-4687CrossRefGoogle ScholarPubMed
76Emery, D.W. et al. (2000) A chromatin insulator protects retrovirus vectors from chromosomal position effects. Proceedings of the National Academy of Sciences of the United States of America 97, 9150-9155CrossRefGoogle ScholarPubMed
77Yannaki, E. et al. (2002) Topological constraints governing the use of the chicken HS4 chromatin insulator in oncoretrovirus vectors. Molecular Therapy 5, 589-598CrossRefGoogle ScholarPubMed
78Evans-Galea, M.V. (2007) Suppression of clonal dominance in cultured human lymphoid cells by addition of the cHS4 insulator to a lentiviral vector. Molecular Therapy 15, 801-809CrossRefGoogle ScholarPubMed
79Ramezani, A., Hawley, T.S. and Hawley, R.G. (2008) Combinatorial incorporation of enhancer-blocking components of the chicken beta-globin 5′HS4 and human T-cell receptor alpha/delta BEAD-1 insulators in self-inactivating retroviral vectors reduces their genotoxic potential. Stem Cells 26, 3257-3266CrossRefGoogle Scholar
80Nishino, T. et al. (2006) Effects of human gamma-globin in murine beta-thalassaemia. British Journal of Haematology 134, 100-108CrossRefGoogle ScholarPubMed
81Neff, T. et al. (2003) Methylguanine methyltransferase-mediated in vivo selection and chemoprotection of allogeneic stem cells in a large-animal model. Journal of Clinical Investigation 112, 1581-1588CrossRefGoogle Scholar
82Ragg, S. et al. (2000) 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 Research 60, 5187-5195Google ScholarPubMed
83Jin, L. et al. (2000) In vivo selection using a cell-growth switch. Nature Genetics 26, 64-66CrossRefGoogle ScholarPubMed
84Emery, D.W. et al. (2005) Selection with a regulated cell growth switch increases the likelihood of expression for a linked gamma-globin gene. Blood Cells, Molecules, and Diseases 34, 235-247CrossRefGoogle ScholarPubMed
85Zhao, H. et al. (2009) Amelioration of murine beta-thalassemia through drug selection of hematopoietic stem cells transduced with a lentiviral vector encoding both gamma-globin and the MGMT drug-resistance gene. Blood 113, 5747-5756CrossRefGoogle ScholarPubMed
86Burroughs, L. and Storb, R. (2005) Low-intensity allogeneic hematopoietic stem cell transplantation for myeloid malignancies: separating graft-versus-leukemia effects from graft-versus-host disease. Current Opinion in Hematology 12, 45-54CrossRefGoogle ScholarPubMed
87Aiuti, A. et al. (2002) Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science 296, 2410-2413CrossRefGoogle ScholarPubMed
88Mardiney, M. and Malech, H.L. (1996) Enhanced engraftment of hematopoietic progenitor cells in mice treated with granulocyte colony-stimulating factor before low-dose irradiation: implications for gene therapy. Blood 87, 4049-4056Google ScholarPubMed
89Rosenzwieg, M. et al. (1999) Efficient and durable gene marking of hematopoietic progenitor cells in nonhuman primates after nonablative conditioning. Blood 94, 2271-2286CrossRefGoogle Scholar
90Bordignon, C. et al. (1995) Gene therapy in peripheral blood lymphocytes and bone marrow for ADA-immunodeficient patients. Science 270, 470-475CrossRefGoogle ScholarPubMed
91Kohn, D.B. et al. (1995) Engraftment of gene-modified umbilical cord blood cells in neonates with adenosine deaminase deficiency. Nature Medicine 1, 1017-1023CrossRefGoogle ScholarPubMed
92Malech, H.L. et al. (1997) Prolonged production of NADPH oxidase-corrected granulocytes after gene therapy of chronic granulomatous disease. Proceedings of the National Academy of Sciences of the United States of America 94, 12133-12138CrossRefGoogle ScholarPubMed
93Kohn, D.B. et al. (1998) T lymphocytes with a normal ADA gene accumulate after transplantation of transduced autologous umbilical cord blood CD34+ cells in ADA-deficient SCID neonates. Nature Medicine 4, 775-780CrossRefGoogle ScholarPubMed
94Dunbar, C.E. et al. (1998) Retroviral transfer of the glucocerebrosidase gene into CD34+ cells from patients with Gaucher disease: in vivo detection of transduced cells without myeloablation. Human Gene Therapy 9, 2629-2640CrossRefGoogle ScholarPubMed
95Schuening, F. et al. (1997) Retrovirus-mediated transfer of the cDNA for human glucocerebrosidase into peripheral blood repopulating cells of patients with Gaucher's disease. Human Gene Therapy 8, 2143-2160CrossRefGoogle ScholarPubMed
96Kelly, P. et al. (2007) Stem cell collection and gene transfer in Fanconi anemia. Molecular Therapy 15, 211-219CrossRefGoogle ScholarPubMed
97Kang, E.M. et al. (2010) Retrovirus gene therapy for X-linked chronic granulomatous disease can achieve stable long-term correction of oxidase activity in peripheral blood neutrophils. Blood 115, 783-791CrossRefGoogle ScholarPubMed
98Gaspar, H.B. et al. (2006) Successful reconstitution of immunity in ADA-SCID by stem cell gene therapy following cessation of PEG-ADA and use of mild preconditioning. Molecular Therapy 14, 505-513CrossRefGoogle ScholarPubMed
99Cartier, N. et al. (2009) Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 326, 818-823CrossRefGoogle ScholarPubMed
100Bank, A., Dorazio, R. and Leboulch, P. (2005) A phase I/II clinical trial of beta-globin gene therapy for beta-thalassemia. Annals of the New York Academy of Sciences, 1054, 308-316CrossRefGoogle ScholarPubMed
101Kaiser, J. (2010) Gene therapy. Beta-thalassemia treatment succeeds, with a caveat. Science 326, 1468-1469CrossRefGoogle Scholar
102Andreani, M. et al. (1996) Persistence of mixed chimerism in patients transplanted for the treatment of thalassemia. Blood 87, 3494-3499CrossRefGoogle ScholarPubMed
103Iannone, R. et al. (2003) Results of minimally toxic nonmyeloablative transplantation in patients with sickle cell anemia and beta-thalassemia. Biology of Blood and Marrow Transplantation 9, 519-528CrossRefGoogle ScholarPubMed
104Persons, D.A. et al. (2001) Functional requirements for phenotypic correction of murine beta-thalassemia: implications for human gene therapy. Blood 97, 3275-3282CrossRefGoogle ScholarPubMed
105Gratwohl, A. et al. (2006) EBMT activity survey 2004 and changes in disease indication over the past 15 years. Bone Marrow Transplantation 37, 1069-1085CrossRefGoogle ScholarPubMed
106To, L.B. et al. (1997) The biology and clinical uses of blood stem cells. Blood 89, 2233-2258CrossRefGoogle ScholarPubMed
107Dunbar, C.E. et al. (1996) Improved retroviral gene transfer into murine and Rhesus peripheral blood or bone marrow repopulating cells primed in vivo with stem cell factor and granulocyte colony-stimulating factor. Proceedings of the National Academy of Sciences of the United States of America 93, 11871-11876CrossRefGoogle ScholarPubMed
108Horn, P.A. et al. (2004) Efficient lentiviral gene transfer to canine repopulating cells using an overnight transduction protocol. Blood 103, 3710-3716CrossRefGoogle ScholarPubMed
109Thomasson, B. et al. (2003) Direct comparison of steady-state marrow, primed marrow, and mobilized peripheral blood for transduction of hematopoietic stem cells in dogs. Human Gene Therapy 14, 1683-1686CrossRefGoogle ScholarPubMed
110Hematti, P. et al. (2003) Retroviral transduction efficiency of G-CSF + SCF-mobilized peripheral blood CD34+ cells is superior to G-CSF or G-CSF + Flt3-L-mobilized cells in nonhuman primates. Blood 101, 2199-2205CrossRefGoogle ScholarPubMed
111Hematti, P. et al. (2004) Comparison of retroviral transduction efficiency in CD34+ cells derived from bone marrow versus G-CSF-mobilized or G-CSF plus stem cell factor-mobilized peripheral blood in nonhuman primates. Stem Cells 22, 1062-1069CrossRefGoogle ScholarPubMed
112Bodine, D.M. et al. (1994) Efficient retrovirus transduction of mouse pluripotent hematopoietic stem cells mobilized into the peripheral blood by treatment with granulocyte colony-stimulating factor and stem cell factor. Blood 84, 1482-1491CrossRefGoogle ScholarPubMed
113O'Malley, D.P., Whalen, M. and Banks, P.M. (2003) Spontaneous splenic rupture with fatal outcome following G-CSF administration for myelodysplastic syndrome. American Journal of Hematology 73, 294-295CrossRefGoogle ScholarPubMed
114Brown, S.L. and Dale, D.C. (1997) Spontaneous splenic rupture following administration of granulocyte colony-stimulating factor (G-CSF): occurrence in an allogeneic donor of peripheral blood stem cells. Biology of Blood and Marrow Transplantation 3, 341-343Google Scholar
115Becker, P.S. et al. (1997) Spontaneous splenic rupture following administration of granulocyte colony-stimulating factor (G-CSF): occurrence in an allogeneic donor of peripheral blood stem cells. Biology of Blood and Marrow Transplantation 3, 45-49Google Scholar
116Abboud, M., Laver, J. and Blau, C.A. (1998) Granulocytosis causing sickle-cell crisis. Lancet 351, 959CrossRefGoogle ScholarPubMed
117Adler, B.K. et al. (2001) Fatal sickle cell crisis after granulocyte colony-stimulating factor administration. Blood 97, 3313-3314CrossRefGoogle ScholarPubMed
118Broxmeyer, H.E. et al. (2005) Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. Journal of Experimental Medicine 201, 1307-1318CrossRefGoogle ScholarPubMed
119Larochelle, A. et al. (2006) AMD3100 mobilizes hematopoietic stem cells with long-term repopulating capacity in nonhuman primates. Blood 107, 3772-3778CrossRefGoogle ScholarPubMed
120Li, K. et al. (1999) Granulocyte colony-stimulating factor-mobilized peripheral blood stem cells in beta-thalassemia patients: kinetics of mobilization and composition of apheresis product. Experimental Hematology 27, 526-532CrossRefGoogle ScholarPubMed
121Yannaki, E. et al. (2010) Mobilization of hematopoietic stem cells in a thalassemic mouse model: implications for human gene therapy of thalassemia. Human Gene Therapy 21, 299-310CrossRefGoogle Scholar
122Yannaki, E. and Stamatoyannopoulos, G. (2010) Hematopoietic stem cell mobilization strategies for gene therapy of beta thalassemia and sickle cell disease. Annals of the New York Academy of Sciences 1202, 59-63CrossRefGoogle ScholarPubMed
123Sadelain, M. et al. (2007) Therapeutic options for patients with severe beta-thalassemia: the need for globin gene therapy. Human Gene Therapy 18, 1-9CrossRefGoogle ScholarPubMed
124Persons, D.A. (2009) Hematopoietic stem cell gene transfer for the treatment of hemoglobin disorders. Hematology/The Education Program of the American Society of Hematology 2009, 690-697CrossRefGoogle Scholar
125Pathak, V.K. and Temin, H.M. (1990) Broad spectrum of in vivo forward mutations, hypermutations, and mutational hotspots in a retroviral shuttle vector after a single replication cycle: deletions and deletions with insertions. Proceedings of the National Academy of Sciences of the United States of America 87, 6024-6028CrossRefGoogle Scholar
126Patel, M. and Yang, S. (2010) Advances in reprogramming somatic cells to induced pluripotent stem cells. Stem Cell Reviews 6, 367-380CrossRefGoogle ScholarPubMed
127Rashid, S.T. and Vallier, L. (2010) Induced pluripotent stem cells – alchemist's tale or clinical reality? Expert Reviews in Molecular Medicine 12, e25CrossRefGoogle ScholarPubMed
128Takahashi, K. and Yamanaka, S. (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663-676CrossRefGoogle ScholarPubMed
129Yu, J. et al. (2007) Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917-1920CrossRefGoogle ScholarPubMed
130Yu, J. et al. (2009) Human induced pluripotent stem cells free of vector and transgene sequences. Science 324, 797-801CrossRefGoogle ScholarPubMed
131Kim, D. et al. (2009) Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cells 4, 472-476CrossRefGoogle ScholarPubMed
132Ebert, A.D. et al. (2009) Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature 457, 277-280CrossRefGoogle ScholarPubMed
133Lee, G. et al. (2009) Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature 461, 402-406CrossRefGoogle ScholarPubMed
134Kiskinis, E. and Eggan, K. (2010) Progress toward the clinical application of patient-specific pluripotent stem cells. Journal of Clinical Investigation 120, 51-59CrossRefGoogle ScholarPubMed
135Park, I. et al. (2008) Disease-specific induced pluripotent stem cells. Cell 134, 877-886CrossRefGoogle ScholarPubMed
136Hanna, J. et al. (2007) Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science 318, 1920-1923CrossRefGoogle ScholarPubMed
137Rega, A. et al. (2009) Disease-corrected haematopoietic progenitors from Fanconi anaemia induced pluripotent stem cells. Nature 460, 53-49Google Scholar
138Ye, L. et al. (2009) Induced pluripotent stem cells offer new approach to therapy in thalassemia and sickle cell anemia and option in prenatal diagnosis in genetic diseases. Proceedings of the National Academy of Sciences of the United States of America 106, 9826-9830CrossRefGoogle ScholarPubMed
139Okita, K. and Yamanaka, S. (2010) Induction of pluripotency by defined factors. Experimental Cell Research 316, 2565-2570CrossRefGoogle ScholarPubMed
140Yamanaka, S. and Blau, H.M. (2010) Nuclear reprogramming to a pluripotent state by three approaches. Nature 465, 704-712CrossRefGoogle ScholarPubMed
141Aiuti, A. and Roncarolo, M.G. (2010) Ten years of gene therapy for primary immune deficiencies. Hematology/The Education Program of the American Society of Hematology 2009, 682-689CrossRefGoogle Scholar
142Gaspar, H.B. et al. (2009) How I treat ADA deficiency. Blood 114, 3524-3532CrossRefGoogle Scholar

Further reading, resources and contacts

The ClinicalTrials.gov website, maintained by the US National Institutes of Health, is a registry of federally and privately supported clinical trials that are being conducted internationally; it provides information on a trial's purpose, eligibility criteria, locations, and contact phone numbers:

Weatherall, D.J. (2010) The inherited diseases of hemoglobin are an emerging global health burden. Blood 115, 4331-4336CrossRefGoogle ScholarPubMed
Sadelain, M. et al. (2008) Stem cell engineering for the treatment of severe hemoglobinopathies. Current Molecular Medicine 8, 690-697CrossRefGoogle ScholarPubMed
Urbinati, F., Madigan, C. and Malik, P. (2006) Pathophysiology and therapy of haemoglobinopathies. Part II: thalassaemias. Expert Reviews in Molecular Medicine 8, 1-26CrossRefGoogle ScholarPubMed
Cohen, A.R. et al. (2004) Thalassemia. Hematology/American Society of Hematology Education Program 2004, 14-34CrossRefGoogle Scholar
Weatherall, D.J. (2001) The thalassemias. In the Molecular Basis of Blood Diseases (Stamatoyannopoulos, G. et al. , eds), pp. 183-226, W.B. Saunders Company, Philadelphia, PA, USAGoogle Scholar
Sorrentino, B.P. et al. (2001) Gene therapy for hematopoietic diseases. In the Molecular Basis of Blood Diseases (Stamatoyannopoulos, G. et al. , eds), pp. 969-1003, W.B. Saunders Company, Philadelphia, PA, USAGoogle Scholar
Weatherall, D.J. (2010) The inherited diseases of hemoglobin are an emerging global health burden. Blood 115, 4331-4336CrossRefGoogle ScholarPubMed
Sadelain, M. et al. (2008) Stem cell engineering for the treatment of severe hemoglobinopathies. Current Molecular Medicine 8, 690-697CrossRefGoogle ScholarPubMed
Urbinati, F., Madigan, C. and Malik, P. (2006) Pathophysiology and therapy of haemoglobinopathies. Part II: thalassaemias. Expert Reviews in Molecular Medicine 8, 1-26CrossRefGoogle ScholarPubMed
Cohen, A.R. et al. (2004) Thalassemia. Hematology/American Society of Hematology Education Program 2004, 14-34CrossRefGoogle Scholar
Weatherall, D.J. (2001) The thalassemias. In the Molecular Basis of Blood Diseases (Stamatoyannopoulos, G. et al. , eds), pp. 183-226, W.B. Saunders Company, Philadelphia, PA, USAGoogle Scholar
Sorrentino, B.P. et al. (2001) Gene therapy for hematopoietic diseases. In the Molecular Basis of Blood Diseases (Stamatoyannopoulos, G. et al. , eds), pp. 969-1003, W.B. Saunders Company, Philadelphia, PA, USAGoogle Scholar