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:

Prime editing for precise and highly versatile genome manipulation

Abstract

Programmable gene-editing tools have transformed the life sciences and have shown potential for the treatment of genetic disease. Among the CRISPR–Cas technologies that can currently make targeted DNA changes in mammalian cells, prime editors offer an unusual combination of versatility, specificity and precision. Prime editors do not require double-strand DNA breaks and can make virtually any substitution, small insertion and small deletion within the DNA of living cells. Prime editing minimally requires a programmable nickase fused to a polymerase enzyme, and an extended guide RNA that both specifies the target site and templates the desired genome edit. In this Review, we summarize prime editing strategies to generate programmed genomic changes, highlight their limitations and recent developments that circumvent some of these bottlenecks, and discuss applications and future directions.

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

Fig. 1: Precision genome editing in mammalian cells.
Fig. 2: Original prime editing systems.
Fig. 3: Advancements in prime editing systems.
Fig. 4: Prime editing variants.

Similar content being viewed by others

References

  1. Barrangou, R. et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709–1712 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Marraffini, L. A. & Sontheimer, E. J. CRISPR interference limits horizontal gene transfer in Staphylococci by targeting DNA. Science 322, 1843–1845 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Deltcheva, E. et al. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 471, 602–607 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jinek, M. et al. A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816 (2012). This paper reports the use of SpCas9 nuclease and nickases and the development of single-guide RNAs for programmable DNA cutting.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Anzalone, A. V., Koblan, L. W. & Liu, D. R. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 38, 824–844 (2020).

    Article  CAS  PubMed  Google Scholar 

  7. Lieber, M. R. The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu. Rev. Biochem. 79, 181–211 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. van Overbeek, M. et al. DNA repair profiling reveals nonrandom outcomes at Cas9-mediated breaks. Mol. Cell 63, 633–646 (2016).

    Article  PubMed  Google Scholar 

  9. Shen, M. W. et al. Predictable and precise template-free CRISPR editing of pathogenic variants. Nature 563, 646–651 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Allen, F. et al. Predicting the mutations generated by repair of Cas9-induced double-strand breaks. Nat. Biotechnol. 37, 64–72 (2019).

    Article  CAS  Google Scholar 

  11. Chen, W. et al. Massively parallel profiling and predictive modeling of the outcomes of CRISPR/Cas9-mediated double-strand break repair. Nucleic Acids Res. 47, 7989–8003 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Iyer, S. et al. Precise therapeutic gene correction by a simple nuclease-induced double-stranded break. Nature 568, 561–565 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Rouet, P., Smih, F. & Jasin, M. Expression of a site-specific endonuclease stimulates homologous recombination in mammalian cells. Proc. Natl Acad. Sci. USA 91, 6064–6068 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Rouet, P., Smih, F. & Jasin, M. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol. Cell. Biol. 14, 8096–8106 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Heyer, W.-D., Ehmsen, K. T. & Liu, J. Regulation of homologous recombination in eukaryotes. Annu. Rev. Genet. 44, 113–139 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Komor, A. C., Badran, A. H. & Liu, D. R. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell 168, 20–36 (2017).

    Article  CAS  PubMed  Google Scholar 

  17. Paquet, D. et al. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature 533, 125–129 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Suzuki, K. et al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature 540, 144–149 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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 

  20. Cullot, G. et al. CRISPR-Cas9 genome editing induces megabase-scale chromosomal truncations. Nat. Commun. 10, 1136 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Alanis-Lobato, G. et al. Frequent loss of heterozygosity in CRISPR-Cas9 edited early human embryos. Proc. Natl Acad. Sci. 118, e2004832117 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Leibowitz, M. L. et al. Chromothripsis as an on-target consequence of CRISPR–Cas9 genome editing. Nat. Genet. 53, 895–905 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tao, J., Wang, Q., Mendez-Dorantes, C., Burns, K. H. & Chiarle, R. Frequency and mechanisms of LINE-1 retrotransposon insertions at CRISPR/Cas9 sites. Nat. Commun. https://doi.org/10.1038/s41467-022-31322-3 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  24. 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 

  25. 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 

  26. Enache, O. M. et al. Cas9 activates the p53 pathway and selects for p53-inactivating mutations. Nat. Genet. 52, 662–668 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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). This paper details the development of the first cytosine base editor.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nishida, K. et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353, aaf8729 (2016).

    Article  PubMed  Google Scholar 

  29. 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). This article describes the engineering and evolution of the first adenine base editor.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Mok, B. Y. et al. A bacterial cytidine deaminase toxin enables CRISPR-free mitochondrial base editing. Nature 583, 631–637 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Cho, S.-I. et al. Targeted A-to-G base editing in human mitochondrial DNA with programmable deaminases. Cell 185, 1764–1776.e1712 (2022).

    Article  CAS  PubMed  Google Scholar 

  32. Yang, L. et al. Engineering and optimising deaminase fusions for genome editing. Nat. Commun. 7, 13330 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kurt, I. C. et al. CRISPR C-to-G base editors for inducing targeted DNA transversions in human cells. Nat. Biotechnol. 39, 41–46 (2021).

    Article  CAS  PubMed  Google Scholar 

  34. Zhao, D. et al. Glycosylase base editors enable C-to-A and C-to-G base changes. Nat. Biotechnol. 39, 35–40 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Chen, L. et al. Programmable C:G to G:C genome editing with CRISPR-Cas9-directed base excision repair proteins. Nat. Commun. 12, 1384 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Koblan, L. W. et al. Efficient C•G-to-G•C base editors developed using CRISPRi screens, target-library analysis, and machine learning. Nat. Biotechnol. 39, 1414–1425 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ferrari, S. et al. Efficient gene editing of human long-term hematopoietic stem cells validated by clonal tracking. Nat. Biotechnol. 38, 1298–1308 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Song, Y. et al. Large-fragment deletions induced by Cas9 cleavage while not in the BEs system. Mol. Ther. Nucleic Acids 21, 523–526 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 19, 770–788 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Jin, S. et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science 364, 292–295 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  43. Gehrke, J. M. et al. An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nat. Biotechnol. 36, 977–982 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kim, Y. B. et al. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat. Biotechnol. 35, 371–376 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nishimasu, H. et al. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science 361, 1259–1262 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Huang, T. P. et al. Circularly permuted and PAM-modified Cas9 variants broaden the targeting scope of base editors. Nat. Biotechnol. 37, 626–631 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Walton, R. T., Christie, K. A., Whittaker, M. N. & Kleinstiver, B. P. Unconstrained genome targeting with near-PAMless engineered CRISPR-Cas9 variants. Science 368, 290–296 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Miller, S. M. et al. Continuous evolution of SpCas9 variants compatible with non-G PAMs. Nat. Biotechnol. 38, 471–481 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Doman, J. L., Raguram, A., Newby, G. A. & Liu, D. R. Evaluation and minimization of Cas9-independent off-target DNA editing by cytosine base editors. Nat. Biotechnol. 38, 620–628 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Rees, H. A., Wilson, C., Doman, J. L. & Liu, D. R. Analysis and minimization of cellular RNA editing by DNA adenine base editors. Sci. Adv. 5, eaax5717 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Grünewald, J. et al. CRISPR DNA base editors with reduced RNA off-target and self-editing activities. Nat. Biotechnol. 37, 1041–1048 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019). This article describes the original development of prime editing, which includes the PE1, PE2 and PE3 prime editing systems.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Chen, P. J. et al. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 184, 5635–5652.e5629 (2021). This article uncovers the inhibitory effect of mismatch repair on prime editing and details the development of the PE4 and PE5 systems, as well as the PEmax architecture.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Nelson, J. W. et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. 40, 402–410 (2022). This paper identifies pegRNA degradation as a mechanism that reduces prime editing efficiency and reports the development of epegRNAs with 3ʹ structural motifs that can reduce pegRNA degradation.

    Article  CAS  PubMed  Google Scholar 

  55. Choi, J. et al. Precise genomic deletions using paired prime editing. Nat. Biotechnol. 40, 218–226 (2022). This article describes the PRIME-Del strategy, which uses two pegRNAs to mediate large, targeted deletions.

    Article  CAS  PubMed  Google Scholar 

  56. Anzalone, A. V. et al. Programmable deletion, replacement, integration and inversion of large DNA sequences with twin prime editing. Nat. Biotechnol. 40, 731–740 (2022). This article describes the creation of twin prime editing and its use with serine recombinases to enable deletion, replacement, inversion and integration of large sequences.

    Article  CAS  PubMed  Google Scholar 

  57. Jiang, T., Zhang, X.-O., Weng, Z. & Xue, W. Deletion and replacement of long genomic sequences using prime editing. Nat. Biotechnol. 40, 227–234 (2022).

    Article  CAS  PubMed  Google Scholar 

  58. Ioannidi, E. I. et al. Drag-and-drop genome insertion without DNA cleavage with CRISPR-directed integrases. Preprint at bioRxiv https://doi.org/10.1101/2021.11.01.466786 (2021).

    Article  Google Scholar 

  59. Ferreira Da Silva, J. et al. Prime editing efficiency and fidelity are enhanced in the absence of mismatch repair. Nat. Commun. https://doi.org/10.1038/s41467-022-28442-1 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Gao, R. et al. Genomic and transcriptomic analyses of prime editing guide RNA–independent off-target effects by prime editors. CRISPR J. 5, 276–293 (2022).

    Article  CAS  PubMed  Google Scholar 

  61. Schene, I. F. et al. Prime editing for functional repair in patient-derived disease models. Nat. Commun. https://doi.org/10.1038/s41467-020-19136-7 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Geurts, M. H. et al. Evaluating CRISPR-based prime editing for cancer modeling and CFTR repair in organoids. Life Sci. Alliance 4, e202000940 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Park, S.-J. et al. Targeted mutagenesis in mouse cells and embryos using an enhanced prime editor. Genome Biol. https://doi.org/10.1186/s13059-021-02389-w (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Liu, Y. et al. Efficient generation of mouse models with the prime editing system. Cell Discov. https://doi.org/10.1038/s41421-020-0165-z (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Gao, P. et al. Prime editing in mice reveals the essentiality of a single base in driving tissue-specific gene expression. Genome Biol. https://doi.org/10.1186/s13059-021-02304-3 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Lin, J. et al. Modeling a cataract disorder in mice with prime editing. Mol. Ther. Nucleic Acids 25, 494–501 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Jin, S. et al. Genome-wide specificity of prime editors in plants. Nat. Biotechnol. 39, 1292–1299 (2021).

    Article  CAS  PubMed  Google Scholar 

  68. Liu, P. et al. Improved prime editors enable pathogenic allele correction and cancer modelling in adult mice. Nat. Commun. https://doi.org/10.1038/s41467-021-22295-w (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Spencer, J. M. & Zhang, X. Deep mutational scanning of S. pyogenes Cas9 reveals important functional domains. Sci. Rep. https://doi.org/10.1038/s41598-017-17081-y (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Velimirovic, M. et al. Peptide fusion improves prime editing efficiency. Nat. Commun. https://doi.org/10.1038/s41467-022-31270-y (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Song, M. et al. Generation of a more efficient prime editor 2 by addition of the Rad51 DNA-binding domain. Nat. Commun. https://doi.org/10.1038/s41467-021-25928-2 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Zong, Y. et al. An engineered prime editor with enhanced editing efficiency in plants. Nat. Biotechnol. 40, 1394–1402 (2022).

    Article  CAS  PubMed  Google Scholar 

  73. Xu, W. et al. A design optimized prime editor with expanded scope and capability in plants. Nat. Plants 8, 45–52 (2022).

    Article  CAS  PubMed  Google Scholar 

  74. Zhang, G. et al. Enhancement of prime editing via xrRNA motif-joined pegRNA. Nat. Commun. https://doi.org/10.1038/s41467-022-29507-x (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Li, X. et al. Enhancing prime editing efficiency by modified pegRNA with RNA G-quadruplexes. J. Mol. Cell Biol. https://doi.org/10.1093/jmcb/mjac022 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Feng, Y. et al. Enhancing prime editing efficiency and flexibility with tethered and split pegRNAs. Protein Cell https://doi.org/10.1093/procel/pwac014 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Liu, Y. et al. Enhancing prime editing by Csy4-mediated processing of pegRNA. Cell Res. 31, 1134–1136 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Petri, K. et al. CRISPR prime editing with ribonucleoprotein complexes in zebrafish and primary human cells. Nat. Biotechnol. 40, 189–193 (2022).

    Article  CAS  PubMed  Google Scholar 

  79. Huang, S. et al. Broadening prime editing toolkits using RNA-Pol-II-driven engineered pegRNA. Mol. Ther. 30, 2923–2932 (2022).

    Article  CAS  PubMed  Google Scholar 

  80. Li, X. et al. Highly efficient prime editing by introducing same-sense mutations in pegRNA or stabilizing its structure. Nat. Commun. https://doi.org/10.1038/s41467-022-29339-9 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Liu, B. et al. A split prime editor with untethered reverse transcriptase and circular RNA template. Nat. Biotechnol. 40, 1388–1393 (2022). This paper describes the split prime editor system and petRNAs, in which the prime editor and pegRNA are each separated into two modular components.

    Article  CAS  PubMed  Google Scholar 

  82. Habib, O., Habib, G., Hwang, G.-H. & Bae, S. Comprehensive analysis of prime editing outcomes in human embryonic stem cells. Nucleic Acids Res. 50, 1187–1197 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Bothmer, A. et al. Characterization of the interplay between DNA repair and CRISPR/Cas9-induced DNA lesions at an endogenous locus. Nat. Commun. 8, 13905 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Liu, N. et al. HDAC inhibitors improve CRISPR/Cas9 mediated prime editing and base editing. Mol. Ther. Nucleic Acids 29, 36–46 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Kweon, J. et al. Engineered prime editors with PAM flexibility. Mol. Ther. 29, 2001–2007 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Böck, D. et al. In vivo prime editing of a metabolic liver disease in mice. Sci. Transl. Med. 14, eabl9238 (2022). This article demonstrates the phenotypic rescue of a phenylketonuria mouse model through viral delivery of prime editors.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Oh, Y. et al. Expansion of the prime editing modality with Cas9 from Francisella novicida. Genome Biol. https://doi.org/10.1186/s13059-022-02644-8 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Zheng, C. et al. A flexible split prime editor using truncated reverse transcriptase improves dual-AAV delivery in mouse liver. Mol. Ther. 30, 1343–1351 (2022).

    Article  CAS  PubMed  Google Scholar 

  89. Gao, Z. et al. A truncated reverse transcriptase enhances prime editing by split AAV vectors. Mol. Ther. 30, 2942–2951 (2022).

    Article  CAS  PubMed  Google Scholar 

  90. Lin, Q. et al. Prime genome editing in rice and wheat. Nat. Biotechnol. 38, 582–585 (2020).

    Article  CAS  PubMed  Google Scholar 

  91. Doman, J. L., Sousa, A. A., Randolph, P. B., Chen, P. J. & Liu, D. R. Designing and executing prime editing experiments in mammalian cells. Nat. Protoc. https://doi.org/10.1038/s41596-022-00724-4 (2022).

  92. Bhagwat, A. M. et al. Multicrispr: gRNA design for prime editing and parallel targeting of thousands of targets. Life Sci. Alliance 3, e202000757 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  93. Chow, R. D., Chen, J. S., Shen, J. & Chen, S. A web tool for the design of prime-editing guide RNAs. Nat. Biomed. Eng. 5, 190–194 (2021).

    Article  CAS  PubMed  Google Scholar 

  94. Hwang, G.-H. et al. PE-Designer and PE-Analyzer: web-based design and analysis tools for CRISPR prime editing. Nucleic Acids Res. 49, W499–W504 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Li, Y., Chen, J., Tsai, S. Q. & Cheng, Y. Easy-prime: a machine learning–based prime editor design tool. Genome Biol. https://doi.org/10.1186/s13059-021-02458-0 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  96. Standage-Beier, K., Tekel, S. J., Brafman, D. A. & Wang, X. Prime editing guide RNA design automation using PINE-CONE. ACS Synth. Biol. 10, 422–427 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Siegner, S. M., Karasu, M. E., Schröder, M. S., Kontarakis, Z. & Corn, J. E. PnB Designer: a web application to design prime and base editor guide RNAs for animals and plants. BMC Bioinform. https://doi.org/10.1186/s12859-021-04034-6 (2021).

    Article  Google Scholar 

  98. Hsu, J. Y. et al. PrimeDesign software for rapid and simplified design of prime editing guide RNAs. Nat. Commun. 12, 1034 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Lin, Q. et al. High-efficiency prime editing with optimized, paired pegRNAs in plants. Nat. Biotechnol. 39, 923–927 (2021). This paper reports the use of dual pegRNAs to increase the efficiency of substitutions, small insertions and small deletions with prime editing.

    Article  CAS  PubMed  Google Scholar 

  100. Kim, H. K. et al. Predicting the efficiency of prime editing guide RNAs in human cells. Nat. Biotechnol. 39, 198–206 (2021).

    Article  CAS  PubMed  Google Scholar 

  101. Zhuang, Y. et al. Increasing the efficiency and precision of prime editing with guide RNA pairs. Nat. Chem. Biol. 18, 29–37 (2021).

    Article  PubMed  Google Scholar 

  102. Wang, J. et al. Efficient targeted insertion of large DNA fragments without DNA donors. Nat. Methods 19, 331–340 (2022).

    Article  CAS  PubMed  Google Scholar 

  103. Tao, R. et al. Bi-PE: bi-directional priming improves CRISPR/Cas9 prime editing in mammalian cells. Nucleic Acids Res. 50, 6423–6434 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Tao, R. et al. WT-PE: prime editing with nuclease wild-type Cas9 enables versatile large-scale genome editing. Signal Transduct. Target. Ther. https://doi.org/10.1038/s41392-022-00936-w (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  105. Kweon, J. et al. Targeted genomic translocations and inversions generated using a paired prime editing strategy. Mol. Ther. https://doi.org/10.1016/j.ymthe.2022.09.008 (2022).

    Article  PubMed  Google Scholar 

  106. Adikusuma, F. et al. Optimized nickase- and nuclease-based prime editing in human and mouse cells. Nucleic Acids Res. 49, 10785–10795 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Peterka, M. et al. Harnessing DSB repair to promote efficient homology-dependent and -independent prime editing. Nat. Commun. https://doi.org/10.1038/s41467-022-28771-1 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Jang, H. et al. Application of prime editing to the correction of mutations and phenotypes in adult mice with liver and eye diseases. Nat. Biomed. Eng. 6, 181–194 (2022).

    Article  CAS  PubMed  Google Scholar 

  109. Eggenschwiler, R. et al. A selectable all-in-one CRISPR prime editing piggyBac transposon allows for highly efficient gene editing in human cell lines. Sci. Rep. https://doi.org/10.1038/s41598-021-01689-2 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  110. Wolff, J. H., Haldrup, J., Thomsen, E. A., Andersen, S. & Mikkelsen, J. G. piggyPrime: high-efficacy prime editing in human cells using piggyBac-based DNA transposition. Front. Genome Ed. https://doi.org/10.3389/fgeed.2021.786893 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Yuan, Q. & Gao, X. Multiplex base- and prime-editing with drive-and-process CRISPR arrays. Nat. Commun. https://doi.org/10.1038/s41467-022-30514-1 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  112. Bharucha, N., Ataam, J. A., Gavidia, A. A. & Karakikes, I. Generation of AAVS1 integrated doxycycline-inducible CRISPR-Prime Editor human induced pluripotent stem cell line. Stem Cell Res. 57, 102610 (2021).

    Article  CAS  PubMed  Google Scholar 

  113. Li, H. et al. Highly efficient generation of isogenic pluripotent stem cell models using prime editing. eLife https://doi.org/10.7554/elife.79208 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Thomas, C. E., Ehrhardt, A. & Kay, M. A. Progress and problems with the use of viral vectors for gene therapy. Nat. Rev. Genet. 4, 346–358 (2003).

    Article  CAS  PubMed  Google Scholar 

  115. Zhi, S. et al. Dual-AAV delivering split prime editor system for in vivo genome editing. Mol. Ther. 30, 283–294 (2022).

    Article  CAS  PubMed  Google Scholar 

  116. Wang, Q. et al. Broadening the reach and investigating the potential of prime editors through fully viral gene-deleted adenoviral vector delivery. Nucleic Acids Res. 49, 11986–12001 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Aulicino, F. et al. Highly efficient CRISPR-mediated large DNA docking and multiplexed prime editing using a single baculovirus. Nucleic Acids Res. 50, 7783–7799 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Sürün, D. et al. Efficient generation and correction of mutations in human iPS cells utilizing mRNAs of CRISPR base editors and prime editors. Genes 11, 511 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Aida, T. et al. Prime editing primarily induces undesired outcomes in mice. Preprint at bioRxiv https://doi.org/10.1101/2020.08.06.239723 (2020).

    Article  Google Scholar 

  120. Hou, X., Zaks, T., Langer, R. & Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 6, 1078–1094 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Landrum, M. J. et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 44, D862–D868 (2016).

    Article  CAS  PubMed  Google Scholar 

  122. Chemello, F. et al. Precise correction of Duchenne muscular dystrophy exon deletion mutations by base and prime editing. Sci. Adv. 7, eabg4910 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Zhou, M. et al. Targeted-deletion of a tiny sequence via prime editing to restore SMN expression. Int. J. Mol. Sci. 23, 7941 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Torkamani, A., Wineinger, N. E. & Topol, E. J. The personal and clinical utility of polygenic risk scores. Nat. Rev. Genet. 19, 581–590 (2018).

    Article  CAS  PubMed  Google Scholar 

  125. Li, H. et al. Multiplex precision gene editing by a surrogate prime editor in rice. Mol. Plant. 15, 1077–1080 (2022).

    Article  PubMed  Google Scholar 

  126. Qian, Y. et al. Efficient and precise generation of Tay–Sachs disease model in rabbit by prime editing system. Cell Discov. https://doi.org/10.1038/s41421-021-00276-z (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  127. Kim, D. E. et al. Prime editor-mediated correction of a pathogenic mutation in purebred dogs. Sci. Rep. https://doi.org/10.1038/s41598-022-17200-4 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  128. Bosch, J. A., Birchak, G. & Perrimon, N. Precise genome engineering in Drosophila using prime editing. Proc. Natl Acad. Sci. USA 118, e2021996118 (2021).

    Article  CAS  PubMed  Google Scholar 

  129. Doench, J. G. Am I ready for CRISPR? A user’s guide to genetic screens. Nat. Rev. Genet. 19, 67–80 (2018).

    Article  CAS  PubMed  Google Scholar 

  130. Bock, C. et al. High-content CRISPR screening. Nat. Rev. Methods Prim. https://doi.org/10.1038/s43586-021-00093-4 (2022).

    Article  Google Scholar 

  131. Hanna, R. E. et al. Massively parallel assessment of human variants with base editor screens. Cell 184, 1064–1080.e1020 (2021).

    Article  CAS  PubMed  Google Scholar 

  132. Kim, Y. et al. High-throughput functional evaluation of human cancer-associated mutations using base editors. Nat. Biotechnol. 40, 874–884 (2022).

    Article  CAS  PubMed  Google Scholar 

  133. Cuella-Martin, R. et al. Functional interrogation of DNA damage response variants with base editing screens. Cell 184, 1081–1097.e1019 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Erwood, S. et al. Saturation variant interpretation using CRISPR prime editing. Nat. Biotechnol. 40, 885–895 (2022). This paper performs genetic screens and classifies variants of unknown significance using prime editing.

    Article  CAS  PubMed  Google Scholar 

  135. Xu, R., Liu, X., Li, J., Qin, R. & Wei, P. Identification of herbicide resistance OsACC1 mutations via in planta prime-editing-library screening in rice. Nat. Plants 7, 888–892 (2021).

    Article  CAS  PubMed  Google Scholar 

  136. Jiao, Y. et al. Random-PE: an efficient integration of random sequences into mammalian genome by prime editing. Mol. Biomed. 2, 36 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  137. Oh-hashi, K., Furuta, E., Fujimura, K. & Hirata, Y. Application of a novel HiBiT peptide tag for monitoring ATF4 protein expression in Neuro2a cells. Biochem. Biophys. Rep. 12, 40–45 (2017).

    PubMed  PubMed Central  Google Scholar 

  138. Leonetti, M. D., Sekine, S., Kamiyama, D., Weissman, J. S. & Huang, B. A scalable strategy for high-throughput GFP tagging of endogenous human proteins. Proc. Natl Acad. Sci. USA 113, E3501–E3508 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Choi, J. et al. A time-resolved, multi-symbol molecular recorder via sequential genome editing. Nature 608, 98–107 (2022). This article explains the development of the DNA typewriter lineage tracing system, which uses prime editing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Bortesi, L. & Fischer, R. The CRISPR/Cas9 system for plant genome editing and beyond. Biotechnol. Adv. 33, 41–52 (2015).

    Article  CAS  PubMed  Google Scholar 

  141. Yin, K., Gao, C. & Qiu, J.-L. Progress and prospects in plant genome editing. Nat. Plants 3, 17107 (2017).

    Article  CAS  PubMed  Google Scholar 

  142. Tang, X. et al. Plant prime editors enable precise gene editing in rice cells. Mol. Plant 13, 667–670 (2020).

    Article  CAS  PubMed  Google Scholar 

  143. Xu, R. et al. Development of plant prime-editing systems for precise genome editing. Plant Commun. 1, 100043 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  144. Li, J. et al. Development of a highly efficient prime editor 2 system in plants. Genome Biol. https://doi.org/10.1186/s13059-022-02730-x (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  145. Jiang, Y. et al. Optimized prime editing efficiently generates glyphosate-resistant rice plants carrying homozygous TAP-IVS mutation in EPSPS. Mol. Plant. https://doi.org/10.1016/j.molp.2022.09.006 (2022).

    Article  PubMed  Google Scholar 

  146. Lu, Y. et al. Precise genome modification in tomato using an improved prime editing system. Plant Biotechnol. J. 19, 415–417 (2021).

    Article  PubMed  Google Scholar 

  147. Perroud, P.-F. et al. Prime editing in the model plant Physcomitrium patens and its potential in the tetraploid potato. Plant Sci. 316, 111162 (2022).

    Article  CAS  PubMed  Google Scholar 

  148. Biswas, S., Bridgeland, A., Irum, S., Thomson, M. J. & Septiningsih, E. M. Optimization of prime editing in rice, peanut, chickpea, and cowpea protoplasts by restoration of GFP activity. Int. J. Mol. Sci. 23, 9809 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

D.R.L. acknowledges support from US National Institutes of Health awards U01 AI142756, RM1 HG009490 and R35 GM118062, from the Bill and Melinda Gates Foundation, and from the Howard Hughes Medical Institute. P.J.C. acknowledges support from a US National Science Foundation (NSF) graduate research fellowship. The authors thank A. Anzalone for helpful comments.

Author information

Authors and Affiliations

Authors

Contributions

P.J.C. and D.R.L. contributed to all aspects of the manuscript.

Corresponding author

Correspondence to David R. Liu.

Ethics declarations

Competing interests

The authors have filed patent applications on gene-editing technologies through the Broad Institute of MIT and Harvard. P.J.C. is currently an employee of Prime Medicine. D.R.L. is a consultant and equity owner of Beam Therapeutics, Pairwise Plants, Prime Medicine, Chroma Medicine, and Nvelop Therapeutics, companies that use or deliver genome-editing or genome-engineering technologies.

Additional information

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

Glossary

Chromothripsis

A process in which tens to thousands of chromosomal rearrangements occur in a single event.

Heteroduplex

Double-stranded DNA in which the sequences of the strands are not perfectly complementary.

Mosaicism

A condition in which an animal contains multiple cell lineages with different genotypes.

Nickase Cas9

(nCas9). Cas9 that has either its HNH or RuvC nuclease domain catalytically inactivated, resulting in a Cas9 enzyme that can only cut one strand of targeted double-stranded DNA.

Polygenic diseases

Diseases that are mediated by numerous genetic variants that each individually contribute small effects.

R loop

A three-stranded nucleic acid structure that contains a DNA:RNA hybrid and a displaced strand of DNA.

Single-guide RNA

(sgRNA). A single-guide RNA molecule, composed of a CRISPR RNA (crRNA) fused to its corresponding trans-activating CRISPR RNA (tracrRNA) scaffold sequence, that directs the binding and nuclease activity of Cas9 enzymes.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, P.J., Liu, D.R. Prime editing for precise and highly versatile genome manipulation. Nat Rev Genet 24, 161–177 (2023). https://doi.org/10.1038/s41576-022-00541-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-022-00541-1

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research