Publication:
Induced human pluripotent stem cells and advanced therapies Future perspectives for the treatment of haemophilia?

Loading...
Thumbnail Image
Full text at PDC
Publication Date
2011-07
Advisors (or tutors)
Editors
Journal Title
Journal ISSN
Volume Title
Publisher
Elsevier
Citations
Google Scholar
Research Projects
Organizational Units
Journal Issue
Abstract
Human induced pluripotent stem cells (iPSCs) have revolutionized the stem cell field. These iPSCs from somatic cells have been reprogrammed with the introduction of transcription factors and are capable to differentiate into cells from all three germ layers. These strategies require retrovirus transduction or transfection of plasmid vectors strategy without viral transduction. Another promising strategy is based on direct delivery of the reprogramming proteins, addition of signal transduction inhibitors and chemical promoter cell survival. The main advantages of iPSCs cells are that they have not included in the debate over the ethics of embryonic stem cell. Current therapy of haemophilia is based on factor VIII (FVIII) or factor IX (FIX) replacement therapy including prophylactic or demand protocols of fixed-dose, and as future alternative, gene and cell therapy. Gene therapy can be made by using viral vectors, mainly lentiviral (LVV) and adeno-associated viruses (AAV) in adult stem cells and autologous fibroblasts, platelets or haematopoietic stem cells, and transfer using non-viral vectors (NVV). Cell therapy for haemophilia is based, mainly, in transplantation of healthy cells to replace the deficient function, for example, the transplantation of liver sinusoidal endothelial cells or endothelial progenitor cells derived from iPSCs. Recently, as first time in haemophilia, endothelial progenitor cells derived from iPSCs cells express FVIII protein effectively, engraft within the hepatic parenchyma, and functionally integrate to provide the therapeutic benefit for a phenotypic correction in haemophilia. Advanced therapies, gene and cell therapy and tissue engineering or iPSCs technology, can provide a potential clinical application in the treatment of haemophilia and other monogenic disorders. Because to date there are not relevant results for phenotypic correction in haemophilia, iPSCs technology could represent a potential alternative based on cellular therapy.
Description
Keywords
Citation
[1] Yamanaka S. Strategies and new developments in the generation of patientspecific pluripotent stem cells. Cell Stem Cell 2007;1:39–49. [2] Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663–76. [3] Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007;318:1917–20. [4] Yu J, Hu K, Smuga-Otto K, et al. Human induced pluripotent stem cells free of vector and transgene sequences. Science 2009;324:797–801. [5] Park IH, Zhao R, West JA, et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature 2008;451:141–6. [6] Okita K, Hong H, Takahashi K, et al. Generation of mouse-induced pluripotent stem cells with plasmid vectors. Nat Protoc 2010;5:418–28. [7] Kaji K, Norrby K, Paca A, et al. Virus-free induction of pluripotency and subsequent excision of reprogramming factors. Nature 2009;458:771–5. [8] Woltjen K, Michael IP, Mohseni P, et al. PiggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature 2009;458:766–70. [9] Zhou H, Wu S, Joo JY, et al. Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell 2009;4:381–4. [10] Lin T, Ambasudhan R, Yuan X, et al. A chemical platform for improved induction of human iPSCs. Nat Methods 2009;6:805–8. [11] Lowry WE, Richter L, Yachechko R, et al. Generation of human induced pluripotent stem cells from dermal fibroblasts. Proc Natl Acad Sci USA 2008;105:2883–8. [12] Alternative sources of human pluripotent stem cells. . Available atWhite paper: The President's Council on Bioethics. Washington, D.C; 2005 http://bioethics. georgetown.edu/pcbe/reports/white_paper/alternative_sources_white_paper.pdf. (last accessed on 23 January 2011). [13] 2008 Amendments to the National Academies’guidelines for human embryonic stem cell research. Available at National Research Council and Institute of Medicine. National Academy Press; 2008 http://www.nap.edu/openbook.php?record_id=12260&page=1. (last accessed on 23 January 2011). [14] Mayshar Y, Ben-David U, Lavon N, et al. Identification and classification of chromosomal aberrations in human induced pluripotent stem cells. Cell Stem Cell 2010;7:521–31. [15] Rolletschek A, Wobus AM. Induced human pluripotent stem cells: promises and open questions. Biol Chem 2009;390:845–9. [16] Amabile G, Meissner A. Induced pluripotent stem cells: current progress and potential for regenerative medicine. Trends Mol Med 2009;15:59–68. [17] Hochedlinger K, Plath K. Epigenetic reprogramming and induced pluripotency. Development 2009;136:509–23. [18] Tateishi K, He J, Taranova O, et al. Generation of insulin-secreting islet-like clusters from human skin fibroblasts. J Biol Chem 2008;283:31601–7. [19] Dimos JT, Rodolfa KT, Niakan KK, et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 2008;321:1218–21. [20] Park IH, Arora N, Huo H, et al. Disease-specific induced pluripotent stem cells. Cell 2008;134:877–86. [21] Ebert AD, Yu J, Rose FF, et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature 2009;457:277–80. [22] Caspi O, Itzhaki I, Arbel G, et al. In vitro electrophysiological drug testing using human embryonic stem cell derived cardiomyocytes. Stem Cells Dev 2009;18: 161–72. [23] Wernig M, Zhao JP, Pruszak J, et al. Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson's disease. Proc Natl Acad Sci USA 2008;105:5856–61. [24] Hanna J, Wernig M, Markoulaki S, et al. Treatment of sickle cell anemia mouse model with iPSCs cells generated from autologous skin. Science 2007;318:1920–3. [25] Mauritz C, Schwanke K, Reppel M, et al. Generation of functional murine cardiac myocytes from induced pluripotent stem cells. Circulation 2008;118:507–17. [26] Zhang J, Wilson GF, Soerens AG, et al. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ Res 2009;104:e30–41. [27] Mason C, Manzotti E. Induced pluripotent stem cells: an emerging technology platform and the Gartner hype cycle. Regen Med 2009;4:329–31. [28] Nelson TJ, Terzic A. Induced pluripotent stem cells: reprogrammed without a trace. Regen Med 2009;4:333–55. [29] Nichols TC, Dillow AM, Franck HW, et al. Protein replacement therapy and gene transfer in canine models of haemophilia A, haemophilia B, von willebrand disease, and factor VII deficiency. ILAR J 2009;50:144–67. [30] Nienhuis AW. Development of gene therapy for blood disorders. Blood 2009;111:4431–44. [31] Liras A. Gene therapy for haemophilia: The end of a “royal pathology” in the third millennium? Haemophilia 2001;7:441–5. [32] Liras A, Olmedillas S. Gene therapy for haemophilia…yes, but…with non-viral vectors? Haemophilia 2009;15:811–6. [33] Viiala NO, Larsen SR, Rasko JE. Gene therapy for hemophilia: clinical trials and technical tribulations. Semin Thromb Hemost 2009;35:81–92. [34] Brown BD, Cantore A, Annoni A, et al. A microRNA-regulated lentiviral vector mediates stable correction of hemophilia B mice. Blood 2007;110:4144–52. [35] Jeon HJ, Oh TK, Kim OH, et al. Delivery of factor VIII gene into skeletal muscle cells using lentiviral vector. Yonsei Med J 2010;51:52–7. [36] Wang L, Herzog RW. AAV-mediated gene transfer for treatment of hemophilia.Curr Gene Ther 2005;5:349–60. [37] Arruda VR, Stedman HH, Haurigot V, et al. Peripheral transvenular delivery of adeno-associated viral vectors to skeletal muscle as a novel therapy for hemophilia B. Blood 2010;115:4678–88. [38] Lu H, Chen L, Wang J, et al. Complete correction of hemophilia A with adenoassociated viral vectors containing a full-size expression cassette. Hum Gene Ther 2008;19:648–54. [39] Jayandharan GR, Zhong L, Sack BK, et al. Optimized adeno-associated virus (AAV)-protein phosphatase-5 helper viruses for efficient liver transduction by singles-tranded AAV vectors: therapeutic expression of factor IX at reduced vector doses.Hum Gene Ther 2010;21:271–83. [40] Haurigot V, Mingozzi F, Buchlis G, et al. Safety of AAV Factor IX Peripheral Transvenular Gene Delivery to Muscle in Hemophilia B Dogs. Mol Ther 2010;18:1318–29. [41] Greene TK, Wang C, Hirsch JD, et al. In vivo efficacy of platelet-delivered, high specific activity factor VIII variants. Blood 2010;116:6114–22. [42] Shi Q, Wilcox DA, Fahs SA, et al. Lentivirus-ediated platelet-derived factor VIII gene therapy in murine haemophilia A. J Thromb Haemost 2007;5:352–61. [43] Zhang G, Shi Q, Fahs SA, et al. Factor IX ectopically expressed in platelets can be stored in alpha-granules and corrects the phenotype of hemophilia B mice. Blood 2010;116:1235–43. [44] Xu L, Nichols TC, Sarkar R, et al. Absence of a desmopressin response after therapeutic expression of factor VIII in hemophilia A dogs with liver-directed neonatal gene therapy. Proc Natl Acad Sci USA 2005;102:6080–5. [45] Xu L, Mei M, Haskins ME, et al. Immune response after neonatal transfer of a human factor IX-expressing retroviral vector in dogs, cats, and mice. Thromb Res 2007;120:269–80. [46] Schmidt M, Hacein-Bey-Abina S, Wissler M, et al. Clonal evidence for the transduction of CD34+ cells with lymphomyeloid differentiation potential and self-renewal capacity in the SCID-X1 gene therapy trial. Blood 2005;105:2699–706. [47] Hacein-Bey-Abina S, Hauer J, Lim A, et al. Efficacy of Gene Therapy for X-Linked Severe Combined Immunodeficiency. N Engl J Med 2010;363:355–64. [48] Aiuti A, Cattaneo F, Galimberti S, et al. Gene therapy for immunodeficiency due to adenosine deaminase deficiency. N Engl J Med 2009;360:447–58. [49] Ide LM, Iwakoshi NN, Gangadharan B, et al.Functional aspects of factor VIII expression after transplantation of genetically-modified hematopoietic stem cells for hemophilia A. J Gene Med 2010;12:333–44. [50] Sivalingam J, Krishnan S, Ng WH, et al. Biosafety assessment of site-directed transgene integration in human umbilical cord-lining cells. Mol Ther 2010;18:1346–56. [51] Aronovich A, Tchorsh D, Katchman H, et al. Correction of hemophilia as a proof of concept for treatment of monogenic diseases by fetal spleen transplantation. Proc Natl Acad Sci USA 2006;103:19075–80. [52] Follenzi A, Benten D, Novikoff P, et al.Transplanted endothelial cells repopulate the liver endothelium and correct the phenotype of hemophilia A mice. J Clin Invest 2008;118:935–45. [53] Xu D, Alipio Z, Fink LM, et al. Phenotypic correction of murine hemophilia A using an iPSCs cell-based therapy. Proc Natl Acad Sci USA 2009;106:808–13. [54] Yadav N, Kanjirakkuzhiyil S, Kumar S, et al. The therapeutic effect of bone marrow–derived liver cells in the phenotypic correction of murine hemophilia A. Blood 2009;114:4552–61. [55] Nelson TJ, Martinez-Fernandez A, Yamada S, et al. Induced pluripotent stem cells: advances to applications. Stem Cells Cloning Adv Appl 2010;3:29–37. [56] Zhou H, Ding S. Evolution of induced pluripotent stem cell technology. Curr Opin Hematol 2010;17:276–80. [57] Lengner CJ. iPS cell technology in regenerative medicine. Ann NY Acad Sci 2010;1192:38–44. [58] Phillips BW, Crook JM. Pluripotent human stem cells: A novel tool in drug discovery. BioDrugs 2010;24:99–108. [59] Lapillonne H, Kobari L, Mazurier C, et al. Red blood cells generation from human induced pluripotent stem cells: perspectives for transfusion medicine. Haematologica 2010;95:1651–9. [60] Wong GK, Chiu AT. Gene therapy, gene targeting and inducinduced pluripotent stemcells: Applications in monogenic disease treatment. Biotechnol Adv 2011;29:1–10.
Collections