1932

Abstract

Gene therapies are gaining momentum as promising early successes in clinical studies accumulate and examples of regulatory approval for licensing increase. Investigators are advancing with cautious optimism that effective, durable, and safe therapies will provide benefit to patients—not only those with single-gene disorders but those with complex acquired diseases as well. While the strategies being translated from the lab to the clinic are numerous, this review focuses on the clinical research that has forged the gene therapy field as it currently stands.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-012017-043332
2019-01-27
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/med/70/1/annurev-med-012017-043332.html?itemId=/content/journals/10.1146/annurev-med-012017-043332&mimeType=html&fmt=ahah

Literature Cited

  1. 1.  Ginn SL, Amaya AK, Alexander IE et al. 2018. Gene therapy clinical trials worldwide to 2017: an update. J. Gene Med. 20:e3015
    [Google Scholar]
  2. 2.  Komor AC, Kim YB, Packer MS et al. 2016. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533:420–24
    [Google Scholar]
  3. 3.  Naldini L 2011. Ex vivo gene transfer and correction for cell-based therapies. Nat. Rev. Genet. 12:301–15
    [Google Scholar]
  4. 4.  Mingozzi F, High KA 2011. Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nat. Rev. Genet. 12:341–55
    [Google Scholar]
  5. 5.  Hacein-Bey-Abina S, Garrigue A, Wang GP et al. 2008. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J. Clin. Investig. 118:3132–42
    [Google Scholar]
  6. 6.  Raper SE, Chirmule N, Lee FS et al. 2003. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol. Genet. Metab. 80:148–58
    [Google Scholar]
  7. 7.  Manno CS, Pierce GF, Arruda VR et al. 2006. Successful transduction of liver in hemophilia by AAV-factor IX and limitations imposed by the host immune response. Nat. Med. 12:342–47
    [Google Scholar]
  8. 8.  Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G et al. 2000. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 288:669–72
    [Google Scholar]
  9. 9.  Howe SJ, Mansour MR, Schwarzwaelder K et al. 2008. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J. Clin. Investig. 118:3143–50
    [Google Scholar]
  10. 10.  Aiuti A, Slavin S, Aker M et al. 2002. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science 296:2410–13
    [Google Scholar]
  11. 11.  Aiuti A, Cattaneo F, Galimberti S et al. 2009. Gene therapy for immunodeficiency due to adenosine deaminase deficiency. N. Engl. J. Med. 360:447–58
    [Google Scholar]
  12. 12.  Gaspar HB, Aiuti A, Porta F et al. 2009. How I treat ADA deficiency. Blood 114:3524–32
    [Google Scholar]
  13. 13.  Deichmann A, Hacein-Bey-Abina S, Schmidt M et al. 2007. Vector integration is nonrandom and clustered and influences the fate of lymphopoiesis in SCID-X1 gene therapy. J. Clin. Investig. 117:2225–32
    [Google Scholar]
  14. 14.  Schwarzwaelder K, Howe SJ, Schmidt M et al. 2007. Gammaretrovirus-mediated correction of SCID-X1 is associated with skewed vector integration site distribution in vivo. J. Clin. Investig. 117:2241–49
    [Google Scholar]
  15. 15.  Aiuti A, Cassani B, Andolfi G et al. 2007. Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy. J. Clin. Investig. 117:2233–40
    [Google Scholar]
  16. 16.  Wu C, Dunbar CE 2011. Stem cell gene therapy: the risks of insertional mutagenesis and approaches to minimize genotoxicity. Front. Med. 5:356–71
    [Google Scholar]
  17. 17.  Braun CJ, Boztug K, Paruzynski A et al. 2014. Gene therapy for Wiskott-Aldrich syndrome—long-term efficacy and genotoxicity. Sci. Transl. Med. 6:227ra33
    [Google Scholar]
  18. 18.  Stein S, Ott MG, Schultze-Strasser S et al. 2010. Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease. Nat. Med. 16:198–204
    [Google Scholar]
  19. 19.  Montini E, Cesana D, Schmidt M et al. 2006. Hematopoietic stem cell gene transfer in a tumor-prone mouse model uncovers low genotoxicity of lentiviral vector integration. Nat. Biotechnol. 24:687–96
    [Google Scholar]
  20. 20.  Wu X, Li Y, Crise B et al. 2003. Transcription start regions in the human genome are favored targets for MLV integration. Science 300:1749–51
    [Google Scholar]
  21. 21.  Cartier N, Hacein-Bey-Abina S, Bartholomae CC et al. 2009. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 326:818–23
    [Google Scholar]
  22. 22.  Biffi A, Montini E, Lorioli L et al. 2013. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science 341:1233158
    [Google Scholar]
  23. 23.  Rees DC, Williams TN, Gladwin MT 2010. Sickle-cell disease. Lancet 376:2018–31
    [Google Scholar]
  24. 24.  Cavazzana M, Antoniani C, Miccio A 2017. Gene therapy for beta-hemoglobinopathies. Mol. Ther. 25:1142–54
    [Google Scholar]
  25. 25.  Vichinsky EP, Neumayr LD, Earles AN et al. 2000. Causes and outcomes of the acute chest syndrome in sickle cell disease. National Acute Chest Syndrome Study Group. N. Engl. J. Med. 342:1855–65
    [Google Scholar]
  26. 26.  Cavazzana-Calvo M, Payen E, Negre O et al. 2010. Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature 467:318–22
    [Google Scholar]
  27. 27.  Thompson AA, Walters MC, Kwiatkowski J et al. 2018. Gene therapy in patients with transfusion-dependent beta-thalassemia. N. Engl. J. Med. 378:1479–93
    [Google Scholar]
  28. 28.  Ribeil JA, Hacein-Bey-Abina S, Payen E et al. 2017. Gene therapy in a patient with sickle cell disease. N. Engl. J. Med. 376:848–55
    [Google Scholar]
  29. 29.  Oldfield EH, Ram Z, Culver KW et al. 1993. Gene therapy for the treatment of brain tumors using intra-tumoral transduction with the thymidine kinase gene and intravenous ganciclovir. Hum. Gene Ther. 4:39–69
    [Google Scholar]
  30. 30.  Parato KA, Senger D, Forsyth PA et al. 2005. Recent progress in the battle between oncolytic viruses and tumours. Nat. Rev. Cancer 5:965–76
    [Google Scholar]
  31. 31.  Lawler SE, Speranza MC, Cho CF et al. 2017. Oncolytic viruses in cancer treatment: a review. JAMA Oncol 3:841–49
    [Google Scholar]
  32. 32.  Rehman H, Silk AW, Kane MP et al. 2016. Into the clinic: talimogene laherparepvec (T-VEC), a first-in-class intratumoral oncolytic viral therapy. J. Immunother. Cancer 4:53
    [Google Scholar]
  33. 33.  Kochenderfer JN, Wilson WH, Janik JE et al. 2010. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 116:4099–102
    [Google Scholar]
  34. 34.  Porter DL, Levine BL, Kalos M et al. 2011. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365:725–33
    [Google Scholar]
  35. 35.  Brentjens RJ, Riviere I, Park JH et al. 2011. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118:4817–28
    [Google Scholar]
  36. 36.  Schuster SJ, Svoboda J, Chong EA et al. 2017. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N. Engl. J. Med. 377:2545–54
    [Google Scholar]
  37. 37.  Neelapu SS, Locke FL, Bartlett NL et al. 2017. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 377:2531–44
    [Google Scholar]
  38. 38.  Maude SL, Laetsch TW, Buechner J et al. 2018. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 378:439–48
    [Google Scholar]
  39. 39.  Garfall AL, Maus MV, Hwang WT et al. 2015. Chimeric antigen receptor T cells against CD19 for multiple myeloma. N. Engl. J. Med. 373:1040–47
    [Google Scholar]
  40. 40.  Ormhoj M, Bedoya F, Frigault MJ et al. 2017. CARs in the lead against multiple myeloma. Curr. Hematol. Malig. Rep. 12:119–25
    [Google Scholar]
  41. 41.  Brown CE, Alizadeh D, Starr R et al. 2016. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N. Engl. J. Med. 375:2561–69
    [Google Scholar]
  42. 42.  Morgan RA, Dudley ME, Wunderlich JR et al. 2006. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314:126–29
    [Google Scholar]
  43. 43.  Johnson LA, June CH 2017. Driving gene-engineered T cell immunotherapy of cancer. Cell Res 27:38–58
    [Google Scholar]
  44. 44.  Rapoport AP, Stadtmauer EA, Binder-Scholl GK et al. 2015. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat. Med. 21:914–21
    [Google Scholar]
  45. 45.  Teachey DT, Lacey SF, Shaw PA et al. 2016. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov 6:664–79
    [Google Scholar]
  46. 46.  Cameron BJ, Gerry AB, Dukes J et al. 2013. Identification of a titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci. Transl. Med. 5:197ra03
    [Google Scholar]
  47. 47.  Morgan RA, Yang JC, Kitano M et al. 2010. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18:843–51
    [Google Scholar]
  48. 48.  Fry TJ, Shah NN, Orentas RJ et al. 2018. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat. Med. 24:20–28
    [Google Scholar]
  49. 49.  Lim WA, June CH 2017. The principles of engineering immune cells to treat cancer. Cell 168:724–40
    [Google Scholar]
  50. 50.  Maguire AM, Simonelli F, Pierce EA et al. 2008. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N. Engl. J. Med. 358:2240–48
    [Google Scholar]
  51. 51.  Cideciyan AV, Hauswirth WW, Aleman TS et al. 2009. Human RPE65 gene therapy for Leber congenital amaurosis: persistence of early visual improvements and safety at 1 year. Hum. Gene Ther. 20:999–1004
    [Google Scholar]
  52. 52.  Bainbridge JW, Smith AJ, Barker SS et al. 2008. Effect of gene therapy on visual function in Leber's congenital amaurosis. N. Engl. J. Med. 358:2231–39
    [Google Scholar]
  53. 53.  Bennett J, Wellman J, Marshall KA et al. 2016. Safety and durability of effect of contralateral-eye administration of AAV2 gene therapy in patients with childhood-onset blindness caused by RPE65 mutations: a follow-on phase 1 trial. Lancet 388:661–72
    [Google Scholar]
  54. 54.  Russell S, Bennett J, Wellman JA et al. 2017. Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial. Lancet 390:849–60
    [Google Scholar]
  55. 55.  Jacobson SG, Cideciyan AV, Roman AJ et al. 2015. Improvement and decline in vision with gene therapy in childhood blindness. N. Engl. J. Med. 372:1920–26
    [Google Scholar]
  56. 56.  Bainbridge JWB, Mehat MS, Sundaram V et al. 2015. Long-term effect of gene therapy on Leber's congenital amaurosis. N. Engl. J. Med. 372:1887–97
    [Google Scholar]
  57. 57.  Maguire AM, High KA, Auricchio A et al. 2009. Age-dependent effects of RPE65 gene therapy for Leber's congenital amaurosis: a phase 1 dose-escalation trial. Lancet 374:1597–605
    [Google Scholar]
  58. 58.  Bennett J, Ashtari M, Wellman J et al. 2012. AAV2 gene therapy readministration in three adults with congenital blindness. Sci. Transl. Med. 4:120ra15
    [Google Scholar]
  59. 59.  Mingozzi F, Maus MV, Hui DJ et al. 2007. CD8+ T-cell responses to adeno-associated virus capsid in humans. Nat. Med. 13:419–22
    [Google Scholar]
  60. 60.  Martino AT, Basner-Tschakarjan E, Markusic DM et al. 2013. Engineered AAV vector minimizes in vivo targeting of transduced hepatocytes by capsid-specific CD8+ T cells. Blood 121:2224–33
    [Google Scholar]
  61. 61.  Nathwani AC, Tuddenham EG, Rangarajan S et al. 2011. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N. Engl. J. Med. 365:2357–65
    [Google Scholar]
  62. 62.  Nathwani AC, Reiss UM, Tuddenham EG et al. 2014. Long-term safety and efficacy of factor IX gene therapy in hemophilia B. N. Engl. J. Med. 371:1994–2004
    [Google Scholar]
  63. 63.  George LA, Sullivan SK, Giermasz A et al. 2017. Hemophilia B gene therapy with a high-specific-activity factor IX variant. N. Engl. J. Med. 377:2215–27
    [Google Scholar]
  64. 64.  Wu Z, Yang H, Colosi P 2010. Effect of genome size on AAV vector packaging. Mol. Ther. 18:80–6
    [Google Scholar]
  65. 65.  Rangarajan S, Walsh L, Lester W et al. 2017. AAV5–factor VIII gene transfer in severe hemophilia A. N. Engl. J. Med. 377:2519–30
    [Google Scholar]
  66. 66.  George LA, Ragni MV, Samelson-Jones BJ et al. 2017. Spk-8011: preliminary results from a phase 1/2 dose escalation trial of an investigational AAV-mediated gene therapy for hemophilia A. Blood 130:604
    [Google Scholar]
  67. 67.  McPhee SW, Janson CG, Li C et al. 2006. Immune responses to AAV in a phase I study for Canavan disease. J. Gene Med. 8:577–88
    [Google Scholar]
  68. 68.  Kaplitt MG, Feigin A, Tang C et al. 2007. Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's disease: an open label, phase I trial. Lancet 369:2097–105
    [Google Scholar]
  69. 69.  Worgall S, Sondhi D, Hackett NR et al. 2008. Treatment of late infantile neuronal ceroid lipofuscinosis by CNS administration of a serotype 2 adeno-associated virus expressing CLN2 cDNA. Hum. Gene Ther. 19:463–74
    [Google Scholar]
  70. 70.  Davidson BL, Stein CS, Heth JA et al. 2000. Recombinant adeno-associated virus type 2, 4, and 5 vectors: transduction of variant cell types and regions in the mammalian central nervous system. PNAS 97:3428–32
    [Google Scholar]
  71. 71.  Foust KD, Nurre E, Montgomery CL et al. 2009. Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes. Nat. Biotechnol. 27:59–65
    [Google Scholar]
  72. 72.  Mendell JR, Al-Zaidy S, Shell R et al. 2017. Single-dose gene-replacement therapy for spinal muscular atrophy. N. Engl. J. Med. 377:1713–22
    [Google Scholar]
  73. 73.  Finkel RS, Chiriboga CA, Vajsar J et al. 2016. Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study. Lancet 388:3017–26
    [Google Scholar]
  74. 74.  Kim H, Kim JS 2014. A guide to genome engineering with programmable nucleases. Nat. Rev. Genet. 15:321–34
    [Google Scholar]
  75. 75.  Sharma R, Anguela XM, Doyon Y et al. 2015. In vivo genome editing of the albumin locus as a platform for protein replacement therapy. Blood 126:1777–84
    [Google Scholar]
  76. 76.  Tebas P, Stein D, Tang WW et al. 2014. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N. Engl. J. Med. 370:901–10
    [Google Scholar]
/content/journals/10.1146/annurev-med-012017-043332
Loading
/content/journals/10.1146/annurev-med-012017-043332
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