Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Clinical strategies for expansion of haematopoietic stem cells

Key Points

  • Expansion of haematopoietic stem cells (HSCs) would be useful for developing new strategies for clinical transplantation and gene therapy. Through an increased understanding of the molecular mechanisms that control HSC self-renewal, HSC expansion has been achieved in several experimental systems.

  • Three general mechanisms can be modulated to achieve HSC expansion: cell-signalling pathways, transcription factors that are involved in HSC replication and genes that regulate cell-cycle progression.

  • WNT proteins, homeobox B4 (HOXB4) and Notch ligands have been generated as soluble factors. Each has resulted in the expansion of HSCs in vitro when added to the culture medium.

  • Retroviral vectors that express HOXB4 alone or together with a gene that confers drug resistance might also be useful for the selective expansion of transduced HSCs. The safety of this approach is not yet established and might be improved by creating a pharmacologically regulated HOXB4 fusion protein.

  • Analysis of mice with homozygous disruptions of cell-cycle genes has identified p18 and p21 as potential targets for HSC expansion. Downregulation of these genes by antisense oligonucleotides or small interfering RNA molecules could be used for expanding HSCs in culture.

  • HSC expansion could be used to increase the rate of immune reconstitution in patients who receive allogeneic transplants, to broaden the use of transplantation of cord blood and to simplify the collection of HSCs from normal donors. HSC expansion would also improve the outcome of gene therapy by potentially improving transduction rates and by limiting the 'dose' of insertion events to which the patient is exposed.

Abstract

Haematopoietic stem cells (HSCs) give rise to all blood and immune cells and are used in clinical transplantation protocols to treat a wide variety of diseases. The ability to increase the number of HSCs either in vivo or in vitro would provide new treatment options, but the amplification of HSCs has been difficult to achieve. Recent insights into the mechanisms of HSC self-renewal now make the amplification of HSCs a plausible clinical goal. This article reviews the molecular mechanisms that control HSC numbers and discusses how these can be modulated to increase the number of HSCs. Clinical applications of HSC expansion are then discussed for their potential to address the current limitations of HSC transplantation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Basics of haematopoiesis.
Figure 2: Functional effects of enforced HOXB4 expression in transplanted mouse HSCs.
Figure 3: Synergistic effects of Hoxb4 and a drug-resistance gene for in vivo selection of transduced HSCs.
Figure 4: The WNT-signalling pathway.
Figure 5: Cell-cycle genes in the regulation of HSCs.

Similar content being viewed by others

References

  1. Osawa, M., Hanada, K., Hamada, H. & Nakauchi, H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 273, 242–245 (1996).

    Article  CAS  PubMed  Google Scholar 

  2. Thomas, E. D., Lochte, H. L. Jr, Cannon, J. H., Sahler, O. D. & Ferrebee, J. W. Supralethal whole body irradiation and isologous marrow transplantation in man. J. Clin. Invest. 38, 1709–1716 (1959).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gaziev, J. & Lucarelli, G. Stem cell transplantation for hemoglobinopathies. Curr. Opin. Pediatr. 15, 24–31 (2003).

    Article  PubMed  Google Scholar 

  4. Cashman, J. D. et al. Kinetic evidence of the regeneration of multilineage hematopoiesis from primitive cells in normal human bone marrow transplanted into immunodeficient mice. Blood 89, 4307–4316 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Hiramatsu, H. et al. Complete reconstitution of human lymphocytes from cord blood CD34+ cells using the NOD/SCID/γcnull mice model. Blood 102, 873–880 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Bodine, D. M., Karlsson, S. & Nienhuis, A. W. Combination of interleukins 3 and 6 preserves stem cell function in culture and enhances retrovirus-mediated gene transfer into hematopoietic stem cells. Proc. Natl Acad. Sci. USA 86, 8897–8901 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bodine, D. M., Orlic, D., Birkett, N. C., Seidel, N. E. & Zsebo, K. M. Stem cell factor increases colony-forming unit-spleen number in vitro in synergy with interleukin-6, and in vivo in Sl/Sld mice as a single factor. Blood 79, 913–919 (1992).

    Article  CAS  PubMed  Google Scholar 

  8. Petzer, A. L., Zandstra, P. W., Piret, J. M. & Eaves, C. J. Differential cytokine effects on primitive (CD34+CD38) human hematopoietic cells: novel responses to Flt3-ligand and thrombopoietin. J. Exp. Med. 183, 2551–2558 (1996).

    Article  CAS  PubMed  Google Scholar 

  9. Bhatia, M. et al. Quantitative analysis reveals expansion of human hematopoietic repopulating cells after short-term ex vivo culture. J. Exp. Med. 186, 619–624 (1997). This paper shows that human HSCs can be moderately expanded for short periods of time when cultured in an appropriate combination of cytokines, but longer periods of culture result in a progressive loss of repopulating HSCs, as defined using an immunodeficient mouse model of human HSC engraftment.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Glimm, H. & Eaves, C. J. Direct evidence for multiple self-renewal divisions of human in vivo repopulating hematopoietic cells in short-term culture. Blood 94, 2161–2168 (1999).

    Article  CAS  PubMed  Google Scholar 

  11. Gerber, H. P. et al. VEGF regulates haematopoietic stem cell survival by an internal autocrine loop mechanism. Nature 417, 954–958 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. de Haan, G. et al. In vitro generation of long-term repopulating hematopoietic stem cells by fibroblast growth factor-1. Dev. Cell 4, 241–251 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Karanu, F. N. et al. The Notch ligand Jagged-1 represents a novel growth factor of human hematopoietic stem cells. J. Exp. Med. 192, 1365–1372 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Calvi, L. M. et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425, 841–846 (2003).

    CAS  PubMed  Google Scholar 

  15. Zhang, J. et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 425, 836–841 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Arai, F. et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell 118, 149–161 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Varnum-Finney, B. et al. Pluripotent, cytokine-dependent, hematopoietic stem cells are immortalized by constitutive Notch1 signaling. Nature Med. 6, 1278–1281 (2000).

    Article  CAS  PubMed  Google Scholar 

  18. Stier, S., Cheng, T., Dombkowski, D., Carlesso, N. & Scadden, D. T. Notch1 activation increases hematopoietic stem cell self-renewal in vivo and favors lymphoid over myeloid lineage outcome. Blood 99, 2369–2378 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Kumano, K. et al. Notch1 inhibits differentiation of hematopoietic cells by sustaining GATA-2 expression. Blood 98, 3283–3289 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Magli, M. C., Largman, C. & Lawrence, H. J. Effects of HOX homeobox genes in blood cell differentiation. J. Cell. Physiol. 173, 168–177 (1997).

    Article  CAS  PubMed  Google Scholar 

  21. Sauvageau, G. et al. Overexpression of HOXB4 in hematopoietic cells causes the selective expansion of more primitive populations in vitro and in vivo. Genes Dev. 9, 1753–1765 (1995). This is the first description of HSC expansion in transplanted mice using a Hoxb4 -containing retroviral vector. HSC expansion is still responsive to normal control mechanisms and is limited after normal HSC numbers have been regenerated.

    Article  CAS  PubMed  Google Scholar 

  22. Krosl, J. et al. Cellular proliferation and transformation induced by HOXB4 and HOXB3 proteins involves cooperation with PBX1. Oncogene 16, 3403–3412 (1998).

    Article  CAS  PubMed  Google Scholar 

  23. DiMartino, J. F. et al. The Hox cofactor and proto-oncogene Pbx1 is required for maintenance of definitive hematopoiesis in the fetal liver. Blood 98, 618–626 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Krosl, J., Beslu, N., Mayotte, N., Humphries, R. K. & Sauvageau, G. The competitive nature of HOXB4-transduced HSC is limited by PBX1: the generation of ultra-competitive stem cells retaining full differentiation potential. Immunity 18, 561–571 (2003).

    Article  CAS  PubMed  Google Scholar 

  25. Beslu, N. et al. Molecular interactions involved in HOXB4-induced activation of HSC self-renewal. Blood 29 June 2004 (doi:10. 1182/blood-2004-04-1653).

  26. Brun, A. C. et al. Hoxb4-deficient mice undergo normal hematopoietic development but exhibit a mild proliferation defect in hematopoietic stem cells. Blood 103, 4126–4133 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Antonchuk, J., Sauvageau, G. & Humphries, R. K. HOXB4-induced expansion of adult hematopoietic stem cells ex vivo. Cell 109, 39–45 (2002). This paper describes a 2,000-fold expansion of mouse HSCs in culture after transduction with a Hoxb4 -containing retroviral vector. This degree of HSC expansion far exceeds the levels that have been obtained using any other method.

    Article  CAS  PubMed  Google Scholar 

  28. Krosl, J. et al. In vitro expansion of hematopoietic stem cells by recombinant TAT–HOXB4 protein. Nature Med. 9, 1428–1432 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Amsellem, S. et al. Ex vivo expansion of human hematopoietic stem cells by direct delivery of the HOXB4 homeoprotein. Nature Med. 9, 1423–1427 (2003). This paper shows that a recombinant HOXB4 protein that can penetrate the cell membrane can be used to expand human HSCs in culture. This paper supports the rationale that transcription factors that increase self-renewal can be used as growth factors in culture media.

    Article  CAS  PubMed  Google Scholar 

  30. Antonchuk, J., Sauvageau, G. & Humphries, R. K. HOXB4 overexpression mediates very rapid stem cell regeneration and competitive hematopoietic repopulation. Exp. Hematol. 29, 1125–1134 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Malech, H. L. et al. Prolonged production of NADPH oxidase-corrected granulocytes after gene therapy of chronic granulomatous disease. Proc. Natl Acad. Sci. USA 94, 12133–12138 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Dunbar, C. E. et al. Retroviral transfer of the glucocerebrosidase gene into CD34+ cells from patients with Gaucher disease: in vivo detection of transduced cells without myeloablation. Hum. Gene Ther. 9, 2629–2640 (1998).

    Article  CAS  PubMed  Google Scholar 

  33. Mardiney, M. et al. Enhanced host defense after gene transfer in the murine p47phox- deficient model of chronic granulomatous disease. Blood 89, 2268–2275 (1997).

    Article  CAS  PubMed  Google Scholar 

  34. Bjorgvinsdottir, H. et al. Retroviral-mediated gene transfer of gp91phox into bone marrow cells rescues defect in host defense against Aspergillus fumigatus in murine X-linked chronic granulomatous disease. Blood 89, 41–48 (1997).

    Article  CAS  PubMed  Google Scholar 

  35. Allay, J. A. et al. In vivo selection of retrovirally transduced hematopoietic stem cells. Nature Med. 4, 1136–1143 (1998). This paper provides definitive proof, using a mouse model, that drug-resistance genes can be used to select HSCs in vivo.

    Article  CAS  PubMed  Google Scholar 

  36. Persons, D. A. et al. Successful treatment of murine β-thalassemia using in vivo selection of genetically modified, drug-resistant hematopoietic stem cells. Blood 102, 506–513 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Zielske, S. P. & Gerson, S. L. Lentiviral transduction of P140K MGMT into human CD34+ hematopoietic progenitors at low multiplicity of infection confers significant resistance to BG/BCNU and allows selection in vitro. Mol. Ther. 5, 381–387 (2002).

    Article  CAS  PubMed  Google Scholar 

  38. Davis, B. M., Koc, O. N. & Gerson, S. L. Limiting numbers of G156A O6-methylguanine-DNA methyltransferase-transduced marrow progenitors repopulate nonmyeloablated mice after drug selection. Blood 95, 3078–3084 (2000).

    Article  CAS  PubMed  Google Scholar 

  39. Sorrentino, B. P. Gene therapy to protect haematopoietic cells from cytotoxic cancer drugs. Nature Rev. Cancer 2, 431–441 (2002).

    Article  CAS  Google Scholar 

  40. Sawai, N., Persons, D. A., Zhou, S., Lu, T. & Sorrentino, B. P. Reduction in hematopoietic stem cell numbers with in vivo drug selection can be partially abrogated by HOXB4 gene expression. Mol. Ther. 8, 376–384 (2003). This paper shows that in vivo selection of HSCs with cytotoxic drugs results in a decrease in the overall number of HSCs. This reduction can be overcome by co-expression of HOXB4, which also results in an increase in the efficiency of the selection process.

    Article  CAS  PubMed  Google Scholar 

  41. Szymczak, A. L. et al. Correction of multi-gene deficiency in vivo using a single 'self-cleaving' 2A peptide-based retroviral vector. Nature Biotechnol. 22, 589–594 (2004).

    Article  CAS  Google Scholar 

  42. Perkins, A. C. & Cory, S. Conditional immortalization of mouse myelomonocytic, megakaryocytic and mast cell progenitors by the Hox-2.4 homeobox gene. EMBO J. 12, 3835–3846 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Thorsteinsdottir, U. et al. Overexpression of HOXA10 in murine hematopoietic cells perturbs both myeloid and lymphoid differentiation and leads to acute myeloid leukemia. Mol. Cell. Biol. 17, 495–505 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Brun, A. C., Fan, X., Bjornsson, J. M., Humphries, R. K. & Karlsson, S. Enforced adenoviral vector-mediated expression of HOXB4 in human umbilical cord blood CD34+ cells promotes myeloid differentiation but not proliferation. Mol. Ther. 8, 618–628 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. Schiedlmeier, B. et al. High-level ectopic HOXB4 expression confers a profound in vivo competitive growth advantage on human cord blood CD34+ cells, but impairs lymphomyeloid differentiation. Blood 101, 1759–1768 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Giles, R. H., van Es, J. H. & Clevers, H. Caught up in a Wnt storm: Wnt signaling in cancer. Biochim. Biophys. Acta 1653, 1–24 (2003).

    CAS  PubMed  Google Scholar 

  47. Muller-Tidow, C. et al. Translocation products in acute myeloid leukemia activate the Wnt signaling pathway in hematopoietic cells. Mol. Cell. Biol. 24, 2890–2904 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Austin, T. W., Solar, G. P., Ziegler, F. C., Liem, L. & Matthews, W. A role for the Wnt gene family in hematopoiesis: expansion of multilineage progenitor cells. Blood 89, 3624–3635 (1997).

    Article  CAS  PubMed  Google Scholar 

  49. van den Berg, D. J., Sharma, A. K., Bruno, E. & Hoffman, R. Role of members of the Wnt gene family in human hematopoiesis. Blood 92, 3189–3202 (1998).

    Article  CAS  PubMed  Google Scholar 

  50. Murdoch, B. et al. Wnt-5A augments repopulating capacity and primitive hematopoietic development of human blood stem cells in vivo. Proc. Natl Acad. Sci. USA 100, 3422–3427 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Reya, T. et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature 423, 409–414 (2003). This study shows that mouse HSCs can be expanded in culture after transduction with an activated β-catenin gene. It also shows that the WNT-signalling pathway is used by HSCs.

    Article  CAS  PubMed  Google Scholar 

  52. Zhu, A. J. & Watt, F. M. β-catenin signalling modulates proliferative potential of human epidermal keratinocytes independently of intercellular adhesion. Development 126, 2285–2298 (1999).

    Article  CAS  PubMed  Google Scholar 

  53. Korinek, V. et al. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nature Genet. 19, 379–383 (1998).

    Article  CAS  PubMed  Google Scholar 

  54. Willert, K. et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature 423, 448–452 (2003). This study tested whether soluble WNT3A is an HSC growth factor in culture. By culturing single cells in the presence of stem-cell factor and WNT3A, considerable expansion of mouse HSCs was obtained.

    Article  CAS  PubMed  Google Scholar 

  55. Sherr, C. J. & Roberts, J. M. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 13, 1501–1512 (1999).

    Article  CAS  PubMed  Google Scholar 

  56. Cheng, T. et al. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 287, 1804–1808 (2000). This paper shows that the HSC pool size is increased in p21-deficient mice owing to an increase in HSC proliferation. These data provide clear evidence that cyclin-dependent kinase inhibitors regulate HSC kinetics.

    Article  CAS  PubMed  Google Scholar 

  57. Stier, S. et al. Ex vivo targeting of p21Cip1/Waf1 permits relative expansion of human hematopoietic stem cells. Blood 102, 1260–1266 (2003).

    Article  CAS  PubMed  Google Scholar 

  58. Cheng, T., Rodrigues, N., Dombkowski, D., Stier, S. & Scadden, D. T. Stem cell repopulation efficiency but not pool size is governed by p27kip1. Nature Med. 6, 1235–1240 (2000).

    Article  CAS  PubMed  Google Scholar 

  59. Dao, M. A., Taylor, N. & Nolta, J. A. Reduction in levels of the cyclin-dependent kinase inhibitor p27kip-1 coupled with transforming growth factor β neutralization induces cell-cycle entry and increases retroviral transduction of primitive human hematopoietic cells. Proc. Natl Acad. Sci. USA 95, 13006–13011 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Dao, M. A., Hwa, J. & Nolta, J. A. Molecular mechanism of transforming growth factor β-mediated cell-cycle modulation in primary human CD34+ progenitors. Blood 99, 499–506 (2002).

    Article  CAS  PubMed  Google Scholar 

  61. Yuan, Y., Shen, H., Franklin, D. S., Scadden, D. T. & Cheng, T. In vivo self-renewing divisions of haematopoietic stem cells are increased in the absence of the early G1-phase inhibitor, p18INK4C. Nature Cell Biol. 6, 436–442 (2004). This paper shows that bone marrow from p18-deficient mice contains more HSCs owing to increased self-renewal divisions. Unlike loss of p21 expression, loss of p18 expression does not lead to the exhaustion of HSCs with time.

    Article  CAS  PubMed  Google Scholar 

  62. Park, I. K., Morrison, S. J. & Clarke, M. F. Bmi1, stem cells, and senescence regulation. J. Clin. Invest. 113, 175–179 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Lessard, J. & Sauvageau, G. Polycomb group genes as epigenetic regulators of normal and leukemic hemopoiesis. Exp. Hematol. 31, 567–585 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. Jacobs, J. J., Kieboom, K., Marino, S., DePinho, R. A. & van Lohuizen, M. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature 397, 164–168 (1999).

    Article  CAS  PubMed  Google Scholar 

  65. Lowe, S. W. & Sherr, C. J. Tumor suppression by Ink4a–Arf: progress and puzzles. Curr. Opin. Genet. Dev. 13, 77–83 (2003).

    Article  CAS  PubMed  Google Scholar 

  66. Park, I. K. et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature 423, 302–305 (2003). This paper shows that, in maturing adult mice, loss of the BMI1 polycomb repressor results in the progressive loss of HSCs with time. This phenotype occurs because of the activation of p16 and p19ARF expression by HSCs.

    Article  CAS  PubMed  Google Scholar 

  67. Molofsky, A. V. et al. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature 425, 962–967 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lessard, J. & Sauvageau, G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature 423, 255–260 (2003).

    Article  CAS  PubMed  Google Scholar 

  69. Roux, E. et al. Analysis of T-cell repopulation after allogeneic bone marrow transplantation: significant differences between recipients of T-cell depleted and unmanipulated grafts. Blood 87, 3984–3992 (1996).

    Article  CAS  PubMed  Google Scholar 

  70. Dumont-Girard, F. et al. Reconstitution of the T-cell compartment after bone marrow transplantation: restoration of the repertoire by thymic emigrants. Blood 92, 4464–4471 (1998).

    Article  CAS  PubMed  Google Scholar 

  71. Douek, D. C. et al. Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution. Lancet 355, 1875–1881 (2000).

    Article  CAS  PubMed  Google Scholar 

  72. Roux, E. et al. Recovery of immune reactivity after T-cell-depleted bone marrow transplantation depends on thymic activity. Blood 96, 2299–2303 (2000).

    Article  CAS  PubMed  Google Scholar 

  73. Lang, P. et al. Clinical scale isolation of highly purified peripheral CD34+ progenitors for autologous and allogeneic transplantation in children. Bone Marrow Transplant. 24, 583–589 (1999).

    Article  CAS  PubMed  Google Scholar 

  74. Handgretinger, R. et al. Megadose transplantation of purified peripheral blood CD34+ progenitor cells from HLA-mismatched parental donors in children. Bone Marrow Transplant. 27, 777–783 (2001). This paper describes the importance of using high doses of CD34+ stem cells when using parental donors for paediatric transplants.

    Article  CAS  PubMed  Google Scholar 

  75. Lang, P. et al. Transplantation of highly purified peripheral-blood CD34+ progenitor cells from related and unrelated donors in children with nonmalignant diseases. Bone Marrow Transplant. 33, 25–32 (2004).

    Article  CAS  PubMed  Google Scholar 

  76. Gluckman, E. et al. Hematopoietic reconstitution in a patient with Fanconi's anemia by means of umbilical-cord blood from an HLA-identical sibling. N. Engl. J. Med. 321, 1174–1178 (1989).

    Article  CAS  PubMed  Google Scholar 

  77. Gluckman, E. Hematopoietic stem-cell transplants using umbilical-cord blood. N. Engl. J. Med. 344, 1860–1861 (2001).

    Article  CAS  PubMed  Google Scholar 

  78. Wagner, J. E. et al. Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival. Blood 100, 1611–1618 (2002).

    Article  CAS  PubMed  Google Scholar 

  79. Shpall, E. J. et al. Transplantation of ex vivo expanded cord blood. Biol. Blood Marrow Transplant. 8, 368–376 (2002).

    Article  PubMed  Google Scholar 

  80. Fernandez, M. N. et al. Cord blood transplants: early recovery of neutrophils from co-transplanted sibling haploidentical progenitor cells and lack of engraftment of cultured cord blood cells, as ascertained by analysis of DNA polymorphisms. Bone Marrow Transplant. 28, 355–363 (2001).

    Article  CAS  PubMed  Google Scholar 

  81. Miller, 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. 10, 4239–4242 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Moscow, J. A. et al. Engraftment of MDR1 and NeoR gene-transduced hematopoietic cells after breast cancer chemotherapy. Blood 94, 52–61 (1999).

    Article  CAS  PubMed  Google Scholar 

  83. Naldini, L. et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science 272, 263–267 (1996).

    Article  CAS  PubMed  Google Scholar 

  84. Chinnasamy, D. et al. Lentiviral-mediated gene transfer into human lymphocytes: role of HIV-1 accessory proteins. Blood 96, 1309–1316 (2000).

    Article  CAS  PubMed  Google Scholar 

  85. Hacein-Bey-Abina, S. et al. Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. N. Engl. J. Med. 346, 1185–1193 (2002).

    Article  CAS  PubMed  Google Scholar 

  86. Aiuti, A. et al. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science 296, 2410–2413 (2002).

    Article  CAS  PubMed  Google Scholar 

  87. Bunting, K. D., Sangster, M. Y., Ihle, J. N. & Sorrentino, B. P. Restoration of lymphocyte function in Janus kinase 3-deficient mice by retroviral-mediated gene transfer. Nature Med. 4, 58–64 (1998).

    Article  CAS  PubMed  Google Scholar 

  88. Hacein-Bey-Abina, S. et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N. Engl. J. Med. 348, 255–256 (2003).

    Article  PubMed  Google Scholar 

  89. Hacein-Bey-Abina, S. et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 302, 415–419 (2003). This paper describes a molecular analysis of two cases of T-cell leukaemia that occurred in a human trial of gene therapy for XSCID. Both showed insertional activation of the LMO2 oncogene by the retroviral vector.

    Article  CAS  PubMed  Google Scholar 

  90. Kiem, H. P. 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. 9, 389–395 (2004).

    Article  CAS  PubMed  Google Scholar 

  91. Jin, L. et al. In vivo selection using a cell-growth switch. Nature Genet. 26, 64–66 (2000).

    Article  CAS  PubMed  Google Scholar 

  92. Jin, L. et al. Targeted expansion of genetically modified bone marrow cells. Proc. Natl Acad. Sci. USA 95, 8093–8097 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Kohn, D. B. et al. American Society of Gene Therapy (ASGT) ad hoc subcommittee on retroviral-mediated gene transfer to hematopoietic stem cells. Mol. Ther. 8, 180–187 (2003).

    Article  CAS  PubMed  Google Scholar 

  94. Baum, C. et al. Side effects of retroviral gene transfer into hematopoietic stem cells. Blood 101, 2099–2114 (2003). An excellent review of the safety issues associated with HSC-targeted gene therapy. There is a detailed consideration of how safety can be improved through modified gene-therapy vectors and protocols.

    Article  CAS  PubMed  Google Scholar 

  95. Kondo, M., Weissman, I. L. & Akashi, K. Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell 91, 661–672 (1997).

    Article  CAS  PubMed  Google Scholar 

  96. Akashi, K., Traver, D., Miyamoto, T. & Weissman, I. L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404, 193–197 (2000).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

I thank C. Sherr for his critical reading of this manuscript and for his many useful suggestions. This work was supported by grants from the National Institutes of Health (Bethesda, United States), a Program Project Grant (United States) and the American Lebanese Syrian Associated Charities (Memphis, United States).

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The author declares no competing financial interests.

Related links

Related links

DATABASES

Entrez Gene

ARF

β-catenin

BMI1

CD34

GATA2

HOXB4

Notch-1

p16

p18

p21

p27

Glossary

AUTOLOGOUS HSCs

A transplant with autologous haematopoietic stem cells (HSCs) is a treatment in which transplanted HSCs are obtained directly from the patient. This is typically used to support intensive treatment with cytotoxic drugs, but it can also be used for gene therapy of genetic disorders.

GENE THERAPY

In the context of haematopoietic disorders, this is a strategy for transducing autologous haematopoietic stem cells or lymphocyte progenitors with genetic vectors that express a therapeutic transgene. Genetically modified cells are then re-infused to reconstitute the haematopoietic system. The goal can be either to replace a defective gene, such as for treatment of sickle-cell anaemia, or to confer a new property to blood cells, such as resistance to cytotoxic drugs.

ALLOGENEIC HSCs

A transplant with allogeneic haematopoietic stem cells (HSCs) is a treatment in which transplanted HSCs are obtained from a normal donor. This approach can be used to treat either malignant or non-malignant disorders. Mismatches between the histocompatibility antigens of the donor and patient can lead to adverse events, such as rejection of the transplanted graft or pathological immune responses to normal tissues in the patient.

MATCHED SIBLING TRANSPLANTS

Transplants for which the donor is a sibling who has all of the same paternal and maternal major histocompatibility alleles as the patient. These cases are usually associated with the least transplant-related complications.

MISMATCHED UNRELATED-DONOR TRANSPLANTS

Allogeneic transplants for which the donor is an unrelated individual. The donors are screened for phenotypic similarities in histocompatibility antigens; however, adverse immunological reactions are more problematic than for matched sibling transplants.

β-THALASSAEMIA

An inherited disorder of erythrocytes caused by decreased or absent expression of β-globin and resulting in chronic anaemia. The most severe form of β-thalassaemia, sometimes called Cooley's anaemia, is characterized by the requirement for regular blood transfusions to sustain life. β-thalassaemia is a relatively common cause of anaemia in Africa, Europe and Asia.

SICKLE-CELL ANAEMIA

An inherited disorder of erythrocytes, with a high prevalence in African and African American populations, that is caused by a mutation in the β-globin gene. A single nucleotide substitution (and the resultant amino-acid substitution) leads to the polymerization of haemoglobin when it is deoxygenated, ultimately resulting in the occlusion of small blood vessels. Disease manifestations include chronic anaemia, multiple painful crises, organ damage and increased susceptibility to bacterial infections.

ACUTE MYELOID LEUKAEMIA

A clonal, malignant disease of blood cells that is characterized by proliferation of abnormal leukemic blast cells in the bone marrow. These blast cells express myeloid cell-surface markers and are usually present in high concentrations in the blood and bone marrow. Non-random cytogenetic abnormalities are usually present and define the causal genetic lesion underlying the malignancy.

CHRONIC GRANULOMATOUS DISEASE

An inherited disorder caused by defective oxidase activity in the respiratory burst of phagocytes. It results from mutations in any of the four genes that are necessary to generate the superoxide radicals required for normal neutrophil function. Affected patients suffer from increased susceptibility to recurrent infections.

LYSOSOMAL-STORAGE DISORDERS

A group of inherited disorders in which one or more tissues become progressively engorged with lipid. Mutations in lysosomal enzymes result in an accumulation of lipid-degradation products, which typically occurs in monocytes and macrophages derived from the bone marrow. Many of these disorders result in damage to the spleen, liver, brain and bone marrow.

VIRAL 2A SEQUENCES

Peptide sequences found in picornaviruses and other viruses that mediate protein cleavage through a ribosomal skip mechanism. These relatively small sequences have been used in gene-therapy vectors to express multiple proteins from a single mRNA transcript.

COMPETITIVE-REPOPULATION ASSAYS

Functional assays for measuring the number of mouse haematopoietic stem cells (HSCs). The HSC content of an undefined population of cells is determined by mixing the cells with a defined number of fresh bone-marrow cells from another source. After transplantation into lethally irradiated mice, genetic markers are used to distinguish progeny from the two HSC sources that are present in the blood and haematopoietic organs. These measurements allow a quantitative assessment of HSC activity.

SMALL INTERFERING RNA (siRNA) MOLECULES

Synthetic double-stranded RNA molecules of 19–23 nucleotides, which are used to 'knockdown' (silence the expression of) a specific gene. This is known as RNA interference (RNAi) and is mediated by the sequence-specific degradation of mRNA.

T-CELL-DEPLETED GRAFT

The removal of T cells from an allogeneic graft to prevent immunological complications, such as graft-versus-host disease. This is typically carried out for transplants from haploidentical and mismatched unrelated donors.

GRAFT-VERSUS-HOST DISEASE

(GVHD). A potentially serious complication that arises when donor-derived T cells attack host tissues, typically resulting in hepatic, dermatological and gastrointestinal damage. Acute GVHD occurs within the first 100 days after transplantation, whereas chronic GVHD occurs later and has a different pathophysiology.

HAPLOIDENTICAL TRANSPLANT

An allogeneic transplant in which the donor is matched for half of the major histocompatibility alleles of the recipient and is typically one of the parents of the patient. Because as many as half of the alleles are mismatched, specific treatment of the patient and processing of the haematopoietic stem-cell graft are required to avoid severe immunological consequences.

CD34+ CELLS

Human haematopoietic cells that are immunopurified based on their expression of the CD34 antigen. These cells typically comprise 5% of the total bone-marrow cell population. Although this population is considerably enriched for haematopoietic stem cells (HSCs), most CD34+ cells are not HSCs.

PERIPHERAL-BLOOD HSCs

Haematopoietic stem cells (HSCs) collected from the peripheral blood of the donor, usually after treatment with granulocyte colony-stimulating factor, which mobilizes HSCs to migrate from the bone marrow to the blood.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sorrentino, B. Clinical strategies for expansion of haematopoietic stem cells. Nat Rev Immunol 4, 878–888 (2004). https://doi.org/10.1038/nri1487

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri1487

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing