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
  • Published:

The promise and challenge of therapeutic genome editing

Abstract

Genome editing, which involves the precise manipulation of cellular DNA sequences to alter cell fates and organism traits, has the potential to both improve our understanding of human genetics and cure genetic disease. Here I discuss the scientific, technical and ethical aspects of using CRISPR (clustered regularly interspaced short palindromic repeats) technology for therapeutic applications in humans, focusing on specific examples that highlight both opportunities and challenges. Genome editing is—or will soon be—in the clinic for several diseases, with more applications under development. The rapid pace of the field demands active efforts to ensure that this breakthrough technology is used responsibly to treat, cure and prevent genetic disease.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Ex vivo and in vivo genome editing to treat human disease.
Fig. 2: The genome editing toolbox.
Fig. 3: Emerging tools.
Fig. 4: Editing the human germline.

Similar content being viewed by others

References

  1. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012). This study demonstrates dual RNA-programmed DNA cutting by CRISPR–Cas9 and establishes a sgRNA format to direct Cas9 applications, providing a road map for genome editing in human, animal and plant cells.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jinek, M. et al. RNA-programmed genome editing in human cells. eLife 2, e00471 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Cho, S. W., Kim, S., Kim, J. M. & Kim, J.-S. Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat. Biotechnol. 31, 230–232 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Knott, G. J. & Doudna, J. A. CRISPR–Cas guides the future of genetic engineering. Science 361, 866–869 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hidalgo-Cantabrana, C., Goh, Y. J. & Barrangou, R. Characterization and repurposing of type I and type II CRISPR–Cas systems in bacteria. J. Mol. Biol. 431, 21–33 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Bao, A. et al. The CRISPR/Cas9 system and its applications in crop genome editing. Crit. Rev. Biotechnol. 39, 321–336 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Terns, M. P. CRISPR-based technologies: impact of RNA-targeting systems. Mol. Cell 72, 404–412 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. High, K. A. & Roncarolo, M. G. Gene therapy. N. Engl. J. Med. 381, 455–464 (2019).

    Article  PubMed  CAS  Google Scholar 

  11. Pauling, L. et al. Sickle cell anemia, a molecular disease. Science 110, 543–548 (1949).

    Article  ADS  CAS  PubMed  Google Scholar 

  12. Ingram, V. M. Gene mutations in human haemoglobin: the chemical difference between normal and sickle cell haemoglobin. Nature 180, 326–328 (1957).

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Shieh, P. B. Emerging strategies in the treatment of Duchenne muscular dystrophy. Neurotherapeutics 15, 840–848 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Min, Y.-L., Bassel-Duby, R. & Olson, E. N. CRISPR correction of Duchenne muscular dystrophy. Annu. Rev. Med. 70, 239–255 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Long, C. et al. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA. Science 345, 1184–1188 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Miller, J. C. et al. A TALE nuclease architecture for efficient genome editing. Nat. Biotechnol. 29, 143–148 (2011).

    Article  CAS  PubMed  Google Scholar 

  17. Hoban, M. D. et al. Zinc finger nucleases targeting the β-globin locus drive efficient correction of the sickle mutation in CD34+ cells. Blood 122, 2904 (2013).

    Article  Google Scholar 

  18. Chang, K.-H. et al. 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, 137–148 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Qasim, W. et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci. Transl. Med. 9, eaaj2013 (2017).

    Article  PubMed  Google Scholar 

  20. Gasiunas, G., Barrangou, R., Horvath, P. & Siksnys, V. Cas9–crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc. Natl Acad. Sci. USA 109, E2579–E2586 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Szostak, J. W., Orr-Weaver, T. L., Rothstein, R. J. & Stahl, F. W. The double-strand-break repair model for recombination. Cell 33, 25–35 (1983). The authors proposed a surprising but ultimately correct cellular DNA repair mechanism in which double-stranded breaks are enlarged to double-stranded gaps to initiate genetic recombination, forming the basis for genome editing mediated by DNA repair.

    Article  CAS  PubMed  Google Scholar 

  22. Stark, J. M., Pierce, A. J., Oh, J., Pastink, A. & Jasin, M. Genetic steps of mammalian homologous repair with distinct mutagenic consequences. Mol. Cell. Biol. 24, 9305–9316 (2004). Double-stranded DNA breaks in mammalian cells trigger DNA repair that can introduce site-specific changes in the genome sequence.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liu, J.-J. et al. CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature 566, 218–223 (2019).

    Google Scholar 

  24. Yan, W. X. et al. Cas13d is a compact RNA-targeting type VI CRISPR effector positively modulated by a WYL-domain-containing accessory protein. Mol. Cell 70, 327–339 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016). A DNA-nicking version of CRISPR–Cas9 was fused to a DNA-editing enzyme that enables targeted nucleotide changes to be introduced at Cas9-directed genome locations.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nishida, K. et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353, aaf8729 (2016). CRISPR–Cas9 was fused to a DNA-editing enzyme that enables targeted nucleotide editing at genome locations recognized by Cas9, while avoiding double-stranded DNA breaks.

    Article  PubMed  CAS  Google Scholar 

  27. Sharon, E. et al. Functional genetic variants revealed by massively parallel precise genome editing. Cell 175, 544–557 (2018). This study showed that an RNA template can be used together with a Cas9–reverse transcriptase fusion protein to introduce small targeted changes in cellular genomes without involving double-stranded DNA break repair.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019). A CRISPR–Cas9-reverse transcriptase fusion protein was used together with extended guide-RNA templates to introduce small sequence changes within approximately 50 base pairs of the location of Cas9 binding.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013). This study demonstrated the use of a catalytically deactivated form of CRISPR–Cas9 for transcriptional control in cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gilbert, L. A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pickar-Oliver, A. & Gersbach, C. A. The next generation of CRISPR–Cas technologies and applications. Nat. Rev. Mol. Cell Biol. 20, 490–507 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Xu, X. & Qi, L. S. A CRISPR–dCas toolbox for genetic engineering and synthetic biology. J. Mol. Biol. 431, 34–47 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. Grünewald, J. et al. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 569, 433–437 (2019).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  34. Zhou, C. et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature 571, 275–278 (2019).

    Article  CAS  PubMed  Google Scholar 

  35. Antoniani, C. et al. Induction of fetal hemoglobin synthesis by CRISPR/Cas9-mediated editing of the human β-globin locus. Blood 131, 1960–1973 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Chung, J. E. et al. CRISPR–Cas9 interrogation of a putative fetal globin repressor in human erythroid cells. PLoS ONE 14, e0208237 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bjurström, C. F. et al. Reactivating fetal hemoglobin expression in human adult erythroblasts through BCL11A knockdown using targeted endonucleases. Mol. Ther. Nucleic Acids 5, e351 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Liu, N. et al. Direct promoter repression by BCL11A controls the fetal to adult hemoglobin switch. Cell 173, 430–442 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Shariati, L. et al. Genetic disruption of the KLF1 gene to overexpress the γ-globin gene using the CRISPR/Cas9 system. J. Gene Med. 18, 294–301 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Martyn, G. E. et al. Natural regulatory mutations elevate the fetal globin gene via disruption of BCL11A or ZBTB7A binding. Nat. Genet. 50, 498–503 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. Grevet, J. D. et al. Domain-focused CRISPR screen identifies HRI as a fetal hemoglobin regulator in human erythroid cells. Science 361, 285–290 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Martyn, G. E. et al. A natural regulatory mutation in the proximal promoter elevates fetal globin expression by creating a de novo GATA1 site. Blood 133, 852–856 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Lomova, A. et al. Improving gene editing outcomes in human hematopoietic stem and progenitor cells by temporal control of DNA repair. Stem Cells 37, 284–294 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Schumann, K. et al. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc. Natl Acad. Sci. USA 112, 10437–10442 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Dever, D. P. et al. CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells. Nature 539, 384–389 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. DeWitt, M. A. et al. Selection-free genome editing of the sickle mutation in human adult hematopoietic stem/progenitor cells. Sci. Transl. Med. 8, 360ra134 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Wu, Y. et al. Highly efficient therapeutic gene editing of human hematopoietic stem cells. Nat. Med. 25, 776–783 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Amoasii, L. et al. Single-cut genome editing restores dystrophin expression in a new mouse model of muscular dystrophy. Sci. Transl. Med. 9, eaan8081 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551, 464–471 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  50. Amoasii, L. et al. Gene editing restores dystrophin expression in a canine model of Duchenne muscular dystrophy. Science 362, 86–91 (2018). This article presents evidence that CRISPR–Cas9 can induce corrective genome edits in sufficient cell numbers in vivo to provide therapeutic benefit in a dog model of DMD.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zuo, E. et al. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 364, 289–292 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  52. Sharma, R. et al. In vivo genome editing of the albumin locus as a platform for protein replacement therapy. Blood 126, 1777–1784 (2015). A potential therapeutic strategy in which blood cells are edited to enable the high-level expression of a desired protein is described.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Laoharawee, K. et al. Dose-dependent prevention of metabolic and neurologic disease in murine MPS II by ZFN-mediated in vivo genome editing. Mol. Ther. 26, 1127–1136 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Maeder, M. L. et al. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat. Med. 25, 229–233 (2019). This study explored a method for treating inherited retinal disease using a genome-editing approach that uses an AAV5 vector to deliver the S. aureus Cas9 and sgRNAs to photoreceptor cells by subretinal injection.

    Article  CAS  PubMed  Google Scholar 

  55. Finn, J. D. et al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing. Cell Rep. 22, 2227–2235 (2018). Lipid nanoparticle-based delivery of mRNA-encoded Cas9 and sgRNAs provided therapeutically relevant levels of genome editing in the liver in mice.

    Article  CAS  PubMed  Google Scholar 

  56. Hultquist, J. F. et al. A Cas9 ribonucleoprotein platform for functional genetic studies of HIV–host interactions in primary human T cells. Cell Rep. 17, 1438–1452 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wang, J.-Z., Wu, P., Shi, Z.-M., Xu, Y.-L. & Liu, Z.-J. The AAV-mediated and RNA-guided CRISPR/Cas9 system for gene therapy of DMD and BMD. Brain Dev. 39, 547–556 (2017).

    Article  PubMed  Google Scholar 

  58. Mendell, J. R. et al. Single-dose gene-replacement therapy for spinal muscular atrophy. N. Engl. J. Med. 377, 1713–1722 (2017).

    Article  CAS  PubMed  Google Scholar 

  59. Yang, Y. et al. A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice. Nat. Biotechnol. 34, 334–338 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Lau, C.-H. & Suh, Y. In vivo genome editing in animals using AAV–CRISPR system: applications to translational research of human disease. F1000Res. 6, 2153 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Bengtsson, N. E. et al. Muscle-specific CRISPR/Cas9 dystrophin gene editing ameliorates pathophysiology in a mouse model for Duchenne muscular dystrophy. Nat. Commun. 8, 14454 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  62. Nelson, C. E. et al. In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy. Science 351, 403–407 (2016). In this study, the feasibility of achieving therapeutically meaningful levels of genome editing in affected tissues in a mouse model of muscular dystrophy was demonstrated.

    Article  ADS  CAS  PubMed  Google Scholar 

  63. Tabebordbar, M. et al. In vivo gene editing in dystrophic mouse muscle and muscle stem cells. Science 351, 407–411 (2016). The feasibility of achieving therapeutically meaningful levels of genome editing in affected tissues in a mouse model of muscular dystrophy was demonstrated.

    Article  ADS  CAS  PubMed  Google Scholar 

  64. Li, H. et al. Inhibition of HBV expression in HBV transgenic mice using AAV-delivered CRISPR–SaCas9. Front. Immunol. 9, 2080 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Kim, E. et al. In vivo genome editing with a small Cas9 orthologue derived from Campylobacter jejuni. Nat. Commun. 8, 14500 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  66. Charlesworth, C. T. et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nat. Med. 25, 249–254 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Simhadri, V. L. et al. Prevalence of pre-existing antibodies to CRISPR-associated nuclease Cas9 in the USA population. Mol. Ther. Methods Clin. Dev. 10, 105–112 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Sandoval, I. M., Kuhn, N. M. & Manfredsson, F. P. Multimodal production of adeno-associated virus. Methods Mol. Biol. 1937, 101–124 (2019).

    Article  CAS  PubMed  Google Scholar 

  69. Sandro, Q., Relizani, K. & Benchaouir, R. AAV production using baculovirus expression vector system. Methods Mol. Biol. 1937, 91–99 (2019).

    Article  CAS  PubMed  Google Scholar 

  70. Strobel, B. et al. Standardized, scalable, and timely flexible adeno-associated virus vector production using frozen high-density HEK-293 cell stocks and CELLdiscs. Hum. Gene Ther. Methods 30, 23–33 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Miller, J. B. et al. Non-viral CRISPR/Cas gene editing in vitro and in vivo enabled by synthetic nanoparticle co-delivery of Cas9 mRNA and sgRNA. Angew. Chem. Int. Edn 56, 1059–1063 (2017).

    Article  CAS  Google Scholar 

  72. Wang, M. et al. Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles. Proc. Natl Acad. Sci. USA 113, 2868–2873 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  73. Yeh, W.-H., Chiang, H., Rees, H. A., Edge, A. S. B. & Liu, D. R. In vivo base editing of post-mitotic sensory cells. Nat. Commun. 9, 2184 (2018).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  74. Gao, X. et al. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature 553, 217–221 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  75. Zuris, J. A. et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat. Biotechnol. 33, 73–80 (2015).

    Article  CAS  PubMed  Google Scholar 

  76. Staahl, B. T. et al. Efficient genome editing in the mouse brain by local delivery of engineered Cas9 ribonucleoprotein complexes. Nat. Biotechnol. 35, 431–434 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Ding, Y. et al. Gold nanoparticles for nucleic acid delivery. Mol. Ther. 22, 1075–1083 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Glass, Z., Li, Y. & Xu, Q. Nanoparticles for CRISPR–Cas9 delivery. Nat. Biomed. Eng. 1, 854–855 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Lee, K. et al. Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair. Nat. Biomed. Eng. 1, 889–901 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Sago, C. D. et al. High-throughput in vivo screen of functional mRNA delivery identifies nanoparticles for endothelial cell gene editing. Proc. Natl Acad. Sci. USA 115, E9944–E9952 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Qin, W. & Wang, H. Delivery of CRISPR–Cas9 into mouse zygotes by electroporation. Methods Mol. Biol. 1874, 179–190 (2019).

    Article  CAS  PubMed  Google Scholar 

  82. Tanihara, F. et al. Generation of a TP53-modified porcine cancer model by CRISPR/Cas9-mediated gene modification in porcine zygotes via electroporation. PLoS ONE 13, e0206360 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Xu, L. et al. CRISPR-mediated genome editing restores dystrophin expression and function in mdx mice. Mol. Ther. 24, 564–569 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Kim, S., Kim, D., Cho, S. W., Kim, J. & Kim, J.-S. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. 24, 1012–1019 (2014). This study demonstrated the use of purified protein–guide RNA complexes for genome editing in human cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Lin, S., Staahl, B. T., Alla, R. K. & Doudna, J. A. Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery. eLife 3, e04766 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Gaj, T. et al. Targeted gene knock-in by homology-directed genome editing using Cas9 ribonucleoprotein and AAV donor delivery. Nucleic Acids Res. 45, e98 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  87. Rouet, R. et al. Receptor-mediated delivery of CRISPR–Cas9 endonuclease for cell-type-specific gene editing. J. Am. Chem. Soc. 140, 6596–6603 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Yin, J. et al. Potent protein delivery into mammalian cells via a supercharged polypeptide. J. Am. Chem. Soc. 140, 17234–17240 (2018).

    Article  CAS  PubMed  Google Scholar 

  89. Riley, R. S., June, C. H., Langer, R. & Mitchell, M. J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 18, 175–196 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Sun, Y. et al. Advances of blood cell-based drug delivery systems. Eur. J. Pharm. Sci. 96, 115–128 (2017).

    Article  CAS  PubMed  Google Scholar 

  91. Sharma, P. et al. Efficient intracellular delivery of biomacromolecules employing clusters of zinc oxide nanowires. Nanoscale 9, 15371–15378 (2017).

    Article  CAS  PubMed  Google Scholar 

  92. Kim, D., Luk, K., Wolfe, S. A. & Kim, J.-S. Evaluating and enhancing target specificity of gene-editing nucleases and deaminases. Annu. Rev. Biochem. 88, 191–220 (2019).

    Article  CAS  PubMed  Google Scholar 

  93. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 36, 765–771 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Poirot, L. et al. Multiplex genome-edited T-cell manufacturing platform for “off-the-shelf” adoptive T-cell immunotherapies. Cancer Res. 75, 3853–3864 (2015).

    Article  CAS  PubMed  Google Scholar 

  95. Bak, R. O. et al. Multiplexed genetic engineering of human hematopoietic stem and progenitor cells using CRISPR/Cas9 and AAV6. eLife 6, e27873 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  96. Tichy, E. D. et al. Mouse embryonic stem cells, but not somatic cells, predominantly use homologous recombination to repair double-strand DNA breaks. Stem Cells Dev. 19, 1699–1711 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Buechele, C. et al. MLL leukemia induction by genome editing of human CD34+ hematopoietic cells. Blood 126, 1683–1694 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Maddalo, D. et al. In vivo engineering of oncogenic chromosomal rearrangements with the CRISPR/Cas9 system. Nature 516, 423–427 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  99. NIST. NIST Genome Editing Consortium https://www.nist.gov/programs-projects/nist-genome-editing-consortium (NIST, 2017).

  100. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Liu, X. S. et al. Editing DNA methylation in the mammalian genome. Cell 167, 233–247 (2016). CRISPR–Cas9 fusion proteins were used to introduce targeted epigenetic changes into cellular genomes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Wilson, R. C. & Carroll, D. The daunting economics of therapeutic genome editing. CRISPR J. 2, 280–284 (2019).

    Article  PubMed  Google Scholar 

  103. Wagner, D. L. et al. High prevalence of Streptococcus pyogenes Cas9-reactive T cells within the adult human population. Nat. Med. 25, 242–248 (2019).

    Article  CAS  PubMed  Google Scholar 

  104. Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. & Taipale, J. CRISPR–Cas9 genome editing induces a p53-mediated DNA damage response. Nat. Med. 24, 927–930 (2018).

    Article  CAS  PubMed  Google Scholar 

  105. Ihry, R. J. et al. p53 inhibits CRISPR–Cas9 engineering in human pluripotent stem cells. Nat. Med. 24, 939–946 (2018).

    Article  CAS  PubMed  Google Scholar 

  106. Schiroli, G. et al. Precise gene editing preserves hematopoietic stem cell function following transient p53-mediated DNA damage response. Cell Stem Cell 24, 551–565.e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Xu, L. et al. CRISPR-edited stem cells in a patient with HIV and acute lymphocytic leukemia. N. Engl. J. Med. 381, 1240–1247 (2019).

    Article  CAS  PubMed  Google Scholar 

  108. Porteus, M. H. A new class of medicines through DNA editing. N. Engl. J. Med. 380, 947–959 (2019).

    Article  CAS  PubMed  Google Scholar 

  109. Liang, P. et al. CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes. Protein Cell 6, 363–372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Fogarty, N. M. E. et al. Genome editing reveals a role for OCT4 in human embryogenesis. Nature 550, 67–73 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  111. Tang, L. et al. CRISPR/Cas9-mediated gene editing in human zygotes using Cas9 protein. Mol. Genet. Genomics 292, 525–533 (2017).

    Article  CAS  PubMed  Google Scholar 

  112. Ma, H. et al. Correction of a pathogenic gene mutation in human embryos. Nature 548, 413–419 (2017).

    Article  ADS  CAS  PubMed  Google Scholar 

  113. Kaul, S., Heitner, S. B. & Mitalipov, S. Sarcomere gene mutation correction. Eur. Heart J. 39, 1506–1507 (2018).

    Article  CAS  PubMed  Google Scholar 

  114. Egli, D. et al. Inter-homologue repair in fertilized human eggs? Nature 560, E5–E7 (2018).

    Article  CAS  PubMed  Google Scholar 

  115. Ormond, K. E. et al. Human germline genome editing. Am. J. Hum. Genet. 101, 167–176 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

J.A.D. thanks M. Hochstrasser and M. Triplet for extensive expert assistance with manuscript editing, formatting, referencing and illustrations. D. Carroll, F. Urnov and R. Wilson provided comments on the manuscript. T. Tolpa created the artwork with input from M. Hochstrasser and E. Stahl, and with support from the Innovative Genomics Institute. J.A.D. is an investigator of the Howard Hughes Medical Institute (HHMI) and a Paul Allen Distinguished Investigator. In addition to funding from HHMI and the Paul Allen Frontiers Group, research in the Doudna laboratory is supported by the Defense Advanced Research Projects Agency (DARPA) (award HR0011-17-2-0043), the William M. Keck Foundation, a Collaborative MS Research Center Award from the National Multiple Sclerosis Society, the Centers for Excellence in Genomic Science of the National Institutes of Health under award number RM1HG009490, the Somatic Cell Genome Editing Program of the Common Fund of the National Institutes of Health under award number U01AI142817-02 and the National Science Foundation under award number 1817593.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jennifer A. Doudna.

Ethics declarations

Competing interests

J.A.D. is a co-founder of Caribou Biosciences, Editas Medicine, Intellia Therapeutics, Scribe Therapeutics and Mammoth Biosciences; a scientific adviser to Caribou Biosciences, Intellia Therapeutics, Scribe Therapeutics, Synthego, Inari and eFFECTOR Therapeutics; and a director of Johnson & Johnson. The Regents of the University of California have patents issued and pending for CRISPR-related technologies on which J.A.D. is an inventor.

Additional information

Peer review information Nature thanks R. Alta Charo and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Doudna, J.A. The promise and challenge of therapeutic genome editing. Nature 578, 229–236 (2020). https://doi.org/10.1038/s41586-020-1978-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-1978-5

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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