1932

Abstract

The genetic basis of sickle cell disease (SCD) was elucidated >60 years ago, yet current therapy does not rely on this knowledge. Recent advances raise prospects for improved, and perhaps curative, treatment. First, transcription factors, BCL11A and LRF/ZBTB7A, that mediate silencing of the β-like fetal (γ-) globin gene after birth have been identified and demonstrated to act at the γ-globin promoters, precisely at recognition sequences disrupted in rare individuals with hereditary persistence of fetal hemoglobin. Second, transformative advances in gene editing and progress in lentiviral gene therapy provide diverse opportunities for genetic strategies to cure SCD. Approaches include hematopoietic gene therapy by globin gene addition, gene editing to correct the SCD mutation, and genetic manipulations to enhance fetal hemoglobin production, a potent modifier of the clinical phenotype. Clinical trials may soon identify efficacious and safe genetic approaches to the ultimate goal of cure for SCD.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-041817-125507
2019-01-27
2024-04-23
Loading full text...

Full text loading...

/deliver/fulltext/med/70/1/annurev-med-041817-125507.html?itemId=/content/journals/10.1146/annurev-med-041817-125507&mimeType=html&fmt=ahah

Literature Cited

  1. 1.  Pauling L, Itano H, Singer S et al. 1949. Sickle cell anaemia, a molecular disease. Science 110:2865543–48
    [Google Scholar]
  2. 2.  Ingram VM 1957. Gene mutations in human hæmoglobin: the chemical difference between normal and sickle cell hæmoglobin. Nature 180:4581326–28
    [Google Scholar]
  3. 3.  Antonarakis SE, Boehm CD, Serjeant GR et al. 1984. Origin of the beta S-globin gene in blacks: the contribution of recurrent mutation or gene conversion or both. PNAS 81:Feb.853–56
    [Google Scholar]
  4. 4.  Shriner D, Rotimi CN 2018. Whole-genome-sequence-based haplotypes reveal single origin of the sickle allele during the Holocene wet phase. Am. J. Hum. Genet. 102:4547–56
    [Google Scholar]
  5. 5.  Piel FB, Hay SI, Gupta S et al. 2013. Global burden of sickle cell anaemia in children under five, 2010–2050: modelling based on demographics, excess mortality, and interventions. PLOS Med 10:7e1001484
    [Google Scholar]
  6. 6.  Platt OS, Orkin SH, Dover G et al. 1984. Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia. J. Clin. Investig. 74:2652–56
    [Google Scholar]
  7. 7.  Charache S, Terrin ML, Moore RD et al. 1995. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. N. Engl. J. Med. 332:201317–22
    [Google Scholar]
  8. 8.  Cerami A, Manning JM 1971. Potassium cyanate as an inhibitor of the sickling of erythrocytes in vitro. PNAS 68:61180–83
    [Google Scholar]
  9. 9.  Oksenberg D, Dufu K, Patel MP et al. 2016. GBT440 increases haemoglobin oxygen affinity, reduces sickling and prolongs RBC half-life in a murine model of sickle cell disease. Br. J. Haematol. 175:1141–53
    [Google Scholar]
  10. 10.  Dong AC, Rivella S 2017. Gene addition strategies for β-thalassemia and sickle cell anemia. Adv. Exp. Med. Biol. 1013:155–76
    [Google Scholar]
  11. 11.  Dever DP, Bak RO, Reinisch A et al. 2016. CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells. Nature 539:7629384–89
    [Google Scholar]
  12. 12.  Hoban MD, Orkin SH, Bauer DE 2016. Genetic treatment of a molecular disorder: gene therapy approaches to sickle cell disease. Blood 127:839–48
    [Google Scholar]
  13. 13.  Manolio TA 2010. Genomewide association studies and assessment of the risk of disease. N. Engl. J. Med. 363:166–76
    [Google Scholar]
  14. 14.  Doudna JA, Charpentier E 2014. The new frontier of genome engineering with CRISPR-Cas9. Science 346:1258096
    [Google Scholar]
  15. 15.  Watson J 1948. The significance of the paucity of sickle cells in newborn negro infants. Am. J. Med. Sci. 215:419–23
    [Google Scholar]
  16. 16.  Forget BG 1998. Molecular basis of hereditary persistence of fetal hemoglobin. Ann. N. Y. Acad. Sci. 850:38–44
    [Google Scholar]
  17. 17.  Menzel S, Garner C, Gut I et al. 2007. A QTL influencing F cell production maps to a gene encoding a zinc-finger protein on chromosome 2p15. Nat. Genet. 39:101197–9917, 18. These papers report the first genome-wide association studies implicating BCL11A in control of HbF level.
    [Google Scholar]
  18. 18.  Uda M, Galanello R, Sanna S et al. 2008. Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of beta-thalassemia. PNAS 105:51620–2517, 18. These papers report the first genome-wide association studies implicating BCL11A in control of HbF level.
    [Google Scholar]
  19. 19.  Gardner K, Fulford T, Silver N et al. 2018. g(HbF): a genetic model of fetal hemoglobin in sickle cell disease. Blood Adv 2:3235–39
    [Google Scholar]
  20. 20.  Galarneau G, Palmer CD, Sankaran VG et al. 2010. Fine-mapping at three loci known to affect fetal hemoglobin levels explains additional genetic variation. Nat. Genet. 42:121049–51
    [Google Scholar]
  21. 21.  Nakamura T, Yamazaki Y, Saiki Y et al. 2000. Evi9 encodes a novel zinc finger protein that physically interacts with BCL6, a known human B-cell proto-oncogene product. Mol. Cell. Biol. 20:93178–86
    [Google Scholar]
  22. 22.  Liu P, Keller JR, Ortiz M et al. 2003. Bcl11a is essential for normal lymphoid development. Nat. Immunol. 4:6525–32
    [Google Scholar]
  23. 23.  Sankaran VG, Menne TF, Xu J et al. 2008. Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science 322:59091839–4223, 24. These studies demonstrate in cells and mice that targeting BCL11A could be therapeutic for SCD.
    [Google Scholar]
  24. 24.  Xu J, Peng C, Sankaran VG et al. 2011. Correction of sickle cell disease in adult mice by interference with fetal hemoglobin silencing. Science 334:6058993–9623, 24. These studies demonstrate in cells and mice that targeting BCL11A could be therapeutic for SCD.
    [Google Scholar]
  25. 25.  Bauer DE, Kamran SC, Lessard S et al. 2013. An erythroid enhancer of BCL11A subject to genetic variation determines fetal hemoglobin level. Science 342:6155253–57Characterizes the BCL11A enhancer as erythroid specific and as the site of HbF-associated genetic variation.
    [Google Scholar]
  26. 26.  Smith E, Luc S, Croney D et al. 2016. Strict in vivo specificity of the Bcl11a erythroid enhancer. Blood 128:192338–42
    [Google Scholar]
  27. 27.  Kurita R, Suda N, Sudo K et al. 2013. Establishment of immortalized human erythroid progenitor cell lines able to produce enucleated red blood cells. PLOS ONE 8:3e59890
    [Google Scholar]
  28. 28.  Canver MC, Smith EC, Sher F et al. 2015. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature 527:7577192–97Elucidates a therapeutic genome editing strategy employing NHEJ to disrupt the BCL11A enhancer (also Reference 71).
    [Google Scholar]
  29. 29.  Basak A, Hancarova M, Ulirsch JC et al. 2015. BCL11A deletions result in fetal hemoglobin persistence and neurodevelopmental alterations. J. Clin. Investig. 125:62363–68
    [Google Scholar]
  30. 30.  Funnell PW, Prontera P, Ottaviani V et al. 2015. 2p15–p16.1 microdeletions encompassing and proximal to BCL11A are associated with elevated HbF in addition to neurologic impairment. Blood 126:189–93
    [Google Scholar]
  31. 31.  Dias C, Estruch SB, Graham SA et al. 2016. BCL11A haploinsufficiency causes an intellectual disability syndrome and dysregulates transcription. Am. J. Hum. Genet. 99:1–22
    [Google Scholar]
  32. 32.  Luc S, Huang J, McEldoon JL et al. 2016. Bcl11a deficiency leads to hematopoietic stem cell defects with an aging-like phenotype. Cell Rep 16:123181–94
    [Google Scholar]
  33. 33.  Brendel C, Guda S, Renella R et al. 2016. Lineage-specific BCL11A knockdown circumvents toxicities and reverses sickle phenotype. J. Clin. Investig. 126:103868–78Describes preclinical evidence supporting a lentiviral trial using erythroid-specific shRNA to induce HbF.
    [Google Scholar]
  34. 34.  Masuda T, Wang X, Maeda M et al. 2016. Transcription factors LRF and BCL11A independently repress expression of fetal hemoglobin. Science 351:6270285–89
    [Google Scholar]
  35. 35.  Xu J, Bauer DE, Kerenyi MA et al. 2013. Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A. PNAS 110:166518–23
    [Google Scholar]
  36. 36.  Reed Moody R, Lo MC, Meagher JL et al. 2018. Probing the interaction between the histone methyltransferase/deacetylase subunit RBBP4/7 and the transcription factor BCL11A in epigenetic complexes. J. Biol. Chem. 293:62125–36
    [Google Scholar]
  37. 37.  Bowen N, Fujita N, Kajita M et al. 2004. Mi-2/NuRD: multiple complexes for many purposes. Biochim. Biophys. Acta 1677:52–57
    [Google Scholar]
  38. 38.  Amaya M, Desai M, Gnanapragasam MN et al. 2013. Mi2β-mediated silencing of the fetal γ-globin gene in adult erythroid cells. Blood 121:173493–501
    [Google Scholar]
  39. 39.  Desai MA, Webb HD, Sinanan LM et al. 2015. An intrinsically disordered region of methyl-CpG binding domain protein 2 (MBD2) recruits the histone deacetylase core of the NuRD complex. Nucleic Acids Res 43:63100–13
    [Google Scholar]
  40. 40.  Liu N, Hargreaves VV, Zhu Q et al. 2018. Direct promoter repression by BCL11A controls the fetal to adult hemoglobin switch. Cell 173:2430–42.e1740, 42. These studies identify γ-globin promoter mutations in HPFH as sites of BCL11A and LRF binding.
    [Google Scholar]
  41. 41.  Sher F, Schoonenberg V, Cole MA et al. 2017. High-resolution CRISPR functional genomics reveals critical vulnerabilities of the NuRD complex for fetal hemoglobin control. Blood 130:286 (Abstr.)
    [Google Scholar]
  42. 42.  Martyn GE, Wienert B, Yang L et al. 2018. Natural regulatory mutations elevate fetal globin via disruption of BCL11A or ZBTB7A binding. Nat. Genet. 50:498–50340, 42. These studies identify γ-globin promoter mutations in HPFH as sites of BCL11A and LRF binding.
    [Google Scholar]
  43. 43.  Skene PJ, Henikoff S 2017. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6:e21856
    [Google Scholar]
  44. 44.  Wienert B, Funnell APW, Norton LJ et al. 2015. Editing the genome to introduce a beneficial naturally occurring mutation associated with increased fetal globin. Nat. Commun. 6:May7085
    [Google Scholar]
  45. 45.  Wienert B, Martyn GE, Kurita R et al. 2017. KLF1 drives the expression of fetal hemoglobin in British HPFH. Blood 130:6803–8
    [Google Scholar]
  46. 46.  Fitzhugh CD, Hsieh MM, Bolan CD et al. 2009. Granulocyte colony-stimulating factor (G-CSF) administration in individuals with sickle cell disease: time for a moratorium?. Cytotherapy 11:4464–71
    [Google Scholar]
  47. 47.  Kanter J, Walters MC, Hsieh MM et al. 2016. Interim results from a phase 1/2 clinical study of lentiglobin gene therapy for severe sickle cell disease. Blood 128:221176
    [Google Scholar]
  48. 48.  Boulad F, Shore T, van Besien K et al. 2018. Safety and efficacy of plerixafor dose escalation for the mobilization of CD34+ hematopoietic progenitor cells in patients with sickle cell disease: interim results. Haematologica 103:770–77
    [Google Scholar]
  49. 49.  Lagresle-Peyrou C, Lefrère F, Magrin E et al. 2018. Plerixafor enables the safe, rapid, efficient mobilization of haematopoietic stem cells in sickle cell disease patients after exchange transfusion. Haematologica 103:778–86
    [Google Scholar]
  50. 50.  Tisdale JF, Pierciey FJ, Kamble R et al. 2017. Successful plerixafor-mediated mobilization, apheresis, and lentiviral vector transduction of hematopoietic stem cells in patients with severe sickle cell disease. Blood 130:Suppl. 1990
    [Google Scholar]
  51. 50a.  Esrick EB, Manis JP, Daley H et al. 2018. Successful hematopoietic stem cell mobilization and apheresis collection using plerixafor alone in sickle cell patients. Blood Adv. 2:2505–12
    [Google Scholar]
  52. 51.  Ribeil J-A, Hacein-Bey-Abina S, Payen E et al. 2017. Gene therapy in a patient with sickle cell disease. N. Engl. J. Med. 376:9848–55Reports clinical outcomes for globin gene addition therapy in SCD and β-thalassemia (also Reference 56).
    [Google Scholar]
  53. 52.  Dunbar CE 2018. Gene therapy comes of age. Science 359:6372eaan7672
    [Google Scholar]
  54. 53.  May C, Rivella S, Callegari J et al. 2000. Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin. Nature 406:82–86
    [Google Scholar]
  55. 54.  Pawliuk R, Westerman KA, Fabry ME et al. 2001. Correction of sickle cell disease in transgenic mouse models by gene therapy. Science 294:55502368–71
    [Google Scholar]
  56. 55.  Cavazzana-Calvo M, Payen E, Negre O et al. 2010. Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia. Nature 467:7313318–22
    [Google Scholar]
  57. 56.  Thompson A, Walters M, Kwiatkowski J et al. 2018. Gene therapy in patients with transfusion-dependent beta-thalassemia. N. Engl. J. Med. 378:161479–93Reports clinical outcomes for globin gene addition therapy in SCD and β-thalassemia (also Reference 51).
    [Google Scholar]
  58. 57.  Rouet P, Smih F, Jasin M 1994. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol. Cell. Biol. 14:128096–106
    [Google Scholar]
  59. 58.  Hoban MD, Bauer DE 2016. A genome editing primer for the hematologist. Blood 127:2525–35
    [Google Scholar]
  60. 59.  Komor AC, Kim YB, Packer MS et al. 2016. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533:7603420–24
    [Google Scholar]
  61. 60.  Gaudelli NM, Komor AC, Rees HA et al. 2017. Programmable base editing of A • T to G • C in genomic DNA without DNA cleavage. Nature 551:464–71
    [Google Scholar]
  62. 61.  Hoban MD, Cost GJ, Mendel MC et al. 2015. Correction of the sickle-cell disease mutation in human hematopoietic stem/progenitor cells. Blood 125:172597–604
    [Google Scholar]
  63. 62.  DeWitt MA, Magis W, Bray NL et al. 2016. Selection-free genome editing of the sickle mutation in human adult hematopoietic stem/progenitor cells. Sci. Transl. Med. 8:360360ra134
    [Google Scholar]
  64. 63.  Mohrin M, Bourke E, Alexander D et al. 2010. Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. Cell Stem Cell 7:2174–85
    [Google Scholar]
  65. 64.  Genovese P, Schiroli G, Escobar G et al. 2014. Targeted genome editing in human repopulating haematopoietic stem cells. Nature 510:7504235–40
    [Google Scholar]
  66. 65.  Ricciardi AS, Quijano E, Putman R et al. 2018. Peptide nucleic acids as a tool for site-specific gene editing. Molecules 23:632
    [Google Scholar]
  67. 66.  Bahal R, Ali McNeer N, Quijano E et al. 2016. In vivo correction of anaemia in β-thalassemic mice by γPNA-mediated gene editing with nanoparticle delivery. Nat. Commun. 7:13304
    [Google Scholar]
  68. 67.  Canver MC, Bauer DE, Dass A et al. 2014. Characterization of genomic deletion efficiency mediated by CRISPR/Cas9 in mammalian cells. J. Biol. Chem. 289:3121312–24
    [Google Scholar]
  69. 68.  Ye L, Wang J, Tan Y et al. 2016. Genome editing using CRISPR-Cas9 to create the HPFH genotype in HSPCs: an approach for treating sickle cell disease and β-thalassemia. PNAS 113:3810661–65
    [Google Scholar]
  70. 69.  Antoniani C, Meneghini V, Lattanzi A et al. 2018. Induction of fetal hemoglobin synthesis by CRISPR/Cas9-mediated editing of the human β-globin locus. Blood 131:171960–73
    [Google Scholar]
  71. 70.  Traxler EA, Yao Y, Wang YD et al. 2016. A genome-editing strategy to treat β-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition. Nat. Med. 22:9987–90
    [Google Scholar]
  72. 71.  Vierstra J, Reik A, Chang K-H et al. 2015. Functional footprinting of regulatory DNA. Nat. Methods 12:10927–30Elucidates a therapeutic genome editing strategy employing NHEJ to disrupt the BCL11A enhancer (also Reference 26).
    [Google Scholar]
  73. 72.  Chang K-H, Smith SE, Sullivan T et al. 2017. Long-term engraftment and fetal globin induction upon BCL11A gene editing in bone-marrow-derived CD34+ hematopoietic stem and progenitor cells. Mol. Ther. Methods Clin. Dev. 4:Mar.137–48
    [Google Scholar]
  74. 73.  Deng W, Rupon JW, Krivega I et al. 2014. Reactivation of developmentally silenced globin genes by forced chromatin looping. Cell 158:4849–60
    [Google Scholar]
  75. 74.  Fitzhugh CD, Cordes S, Taylor T et al. 2017. At least 20% donor myeloid chimerism is necessary to reverse the sickle phenotype after allogeneic HSCT. Blood 130:171946–48
    [Google Scholar]
  76. 75.  Altrock PM, Brendel C, Renella R et al. 2016. Mathematical modeling of erythrocyte chimerism informs genetic intervention strategies for sickle cell disease. Am. J. Hematol. 91:9931–37
    [Google Scholar]
  77. 76.  Palchaudhuri R, Saez B, Hoggatt J et al. 2016. Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin. Nat. Biotechnol. 34:7738–47
    [Google Scholar]
  78. 77.  Orkin SH, Reilly P 2016. Paying for future success in gene therapy. Science 352:62891059–61
    [Google Scholar]
/content/journals/10.1146/annurev-med-041817-125507
Loading
/content/journals/10.1146/annurev-med-041817-125507
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error