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.

  • Article
  • Published:

Human genetic diversity alters off-target outcomes of therapeutic gene editing

Abstract

CRISPR gene editing holds great promise to modify DNA sequences in somatic cells to treat disease. However, standard computational and biochemical methods to predict off-target potential focus on reference genomes. We developed an efficient tool called CRISPRme that considers single-nucleotide polymorphism (SNP) and indel genetic variants to nominate and prioritize off-target sites. We tested the software with a BCL11A enhancer targeting guide RNA (gRNA) showing promise in clinical trials for sickle cell disease and β-thalassemia and found that the top candidate off-target is produced by an allele common in African-ancestry populations (MAF 4.5%) that introduces a protospacer adjacent motif (PAM) sequence. We validated that SpCas9 generates strictly allele-specific indels and pericentric inversions in CD34+ hematopoietic stem and progenitor cells (HSPCs), although high-fidelity Cas9 mitigates this off-target. This report illustrates how genetic variants should be considered as modifiers of gene editing outcomes. We expect that variant-aware off-target assessment will become integral to therapeutic genome editing evaluation and provide a powerful approach for comprehensive off-target nomination.

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: CRISPRme provides web-based analysis of CRISPR-Cas gene editing off-target potential reflecting population genetic diversity.
Fig. 2: CRISPRme provides analysis of off-target potential of CRISPR-Cas gene editing reflecting population and private genetic diversity.
Fig. 3: Allele-specific off-target editing by a BCL11A enhancer targeting gRNA in clinical trials associated with a common variant in African-ancestry populations.
Fig. 4: Allele-specific pericentric inversion following BCL11A enhancer editing due to off-target cleavage.
Fig. 5: CRISPRme illustrates prevalent off-target potential due to genetic variation.

Similar content being viewed by others

Data availability

Sequencing data are deposited in the NCBI Sequence Read Archive database under accession number PRJNA733110. The data for 1000 G were downloaded from https://doi.org/10.12688/wellcomeopenres.15126.2. The data for the HGDP were downloaded from https://doi.org/10.1126/science.aay5012. The full CRISPRme results for NGG PAM gRNAs (including sg1617) are available at https://doi.org/10.5281/zenodo.7195706. Source data are provided with this paper.

Code availability

CRISPRme source code is available at https://github.com/pinellolab/crisprme and https://github.com/InfOmics/CRISPRme. The web app is available online at http://crisprme.di.univr.it. The versions of CRISPRme (1.8.8 and v1.7.7) used to generate the results presented in this manuscript have been deposited on Zenodo: https://doi.org/10.5281/zenodo.5047489. CRISPRitz v2.6.5 used to generate the data presented in this paper have been deposited to Zenodo: https://doi.org/10.5281/zenodo.7078220.

The scripts to generate the plots presented in the manuscript have been deposited to Zenodo: https://doi.org/10.5281/zenodo.7193131.

References

  1. 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  Google Scholar 

  2. Clement, K., Hsu, J. Y., Canver, M. C., Joung, J. K. & Pinello, L. Technologies and computational analysis strategies for CRISPR applications. Mol. Cell 79, 11–29 (2020).

    Article  CAS  Google Scholar 

  3. Bao, X. R., Pan, Y., Lee, C. M., Davis, T. H. & Bao, G. Tools for experimental and computational analyses of off-target editing by programmable nucleases. Nat. Protoc. 16, 10–26 (2021).

    Article  CAS  Google Scholar 

  4. Hsu, P. D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).

    Article  CAS  Google Scholar 

  5. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  Google Scholar 

  6. Chaudhari, H. G. et al. Evaluation of homology-independent CRISPR-Cas9 off-target assessment methods. CRISPR J. 3, 440–453 (2020).

    Article  CAS  Google Scholar 

  7. Lessard, S. et al. Human genetic variation alters CRISPR-Cas9 on- and off-targeting specificity at therapeutically implicated loci. Proc. Natl Acad. Sci. USA 114, E11257–E11266 (2017).

    Article  CAS  Google Scholar 

  8. Scott, D. A. & Zhang, F. Implications of human genetic variation in CRISPR-based therapeutic genome editing. Nat. Med. 23, 1095–1101 (2017).

    Article  CAS  Google Scholar 

  9. Concordet, J.-P. & Haeussler, M. CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens. Nucleic Acids Res. 46, W242–W245 (2018).

    Article  CAS  Google Scholar 

  10. Listgarten, J. et al. Prediction of off-target activities for the end-to-end design of CRISPR guide RNAs. Nat. Biomed. Eng. 2, 38–47 (2018).

    Article  CAS  Google Scholar 

  11. Labun, K. et al. CHOPCHOP v3: expanding the CRISPR web toolbox beyond genome editing. Nucleic Acids Res. 47, W171–W174 (2019).

    Article  CAS  Google Scholar 

  12. Park, J., Bae, S. & Kim, J.-S. Cas-Designer: a web-based tool for choice of CRISPR-Cas9 target sites. Bioinformatics 31, 4014–4016 (2015).

    CAS  Google Scholar 

  13. Cancellieri, S., Canver, M. C., Bombieri, N., Giugno, R. & Pinello, L. CRISPRitz: rapid, high-throughput and variant-aware in silico off-target site identification for CRISPR genome editing. Bioinformatics 36, 2001–2008 (2020).

    Article  CAS  Google Scholar 

  14. Lowy-Gallego, E. et al. Variant calling on the GRCh38 assembly with the data from phase three of the 1000 Genomes Project. Wellcome Open Res 4, 50 (2019).

    Article  Google Scholar 

  15. Bergström, A. et al. Insights into human genetic variation and population history from 929 diverse genomes. Science 367, eaay5012 (2020).

    Article  Google Scholar 

  16. Karczewski, K. J. et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581, 434–443 (2020).

    Article  CAS  Google Scholar 

  17. Frangoul, H. et al. CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. N. Engl. J. Med. 384, 252–260 (2021).

    Article  CAS  Google Scholar 

  18. Canver, M. C. et al. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature 527, 192–197 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  20. 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  Google Scholar 

  21. Fennell, T. et al. CALITAS: A CRISPR-Cas-aware ALigner for In silico off-TArget Search. CRISPR J. 4, 264–274 (2021).

    Article  CAS  Google Scholar 

  22. Frankish, A. et al. GENCODE reference annotation for the human and mouse genomes. Nucleic Acids Res. 47, D766–D773 (2019).

    Article  CAS  Google Scholar 

  23. ENCODE Project Consortium. et al. Expanded encyclopaedias of DNA elements in the human and mouse genomes. Nature 583, 699–710 (2020).

    Article  Google Scholar 

  24. Abadi, S., Yan, W. X., Amar, D. & Mayrose, I. A machine learning approach for predicting CRISPR-Cas9 cleavage efficiencies and patterns underlying its mechanism of action. PLoS Comput. Biol. 13, e1005807 (2017).

    Article  Google Scholar 

  25. Demirci, S. et al. Durable and robust fetal globin induction without Anemia in rhesus monkeys following autologous hematopoietic stem cell transplant with BCL11A erythroid enhancer editing.Blood 134, 4632 (2019).

    Article  Google Scholar 

  26. Schmid-Burgk, J. L. et al. Highly parallel profiling of Cas9 variant specificity. Mol. Cell 78, 794–800.e8 (2020).

    Article  CAS  Google Scholar 

  27. Vakulskas, C. A. et al. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells. Nat. Med. 24, 1216–1224 (2018).

    Article  CAS  Google Scholar 

  28. Xu, L. et al. CRISPR/Cas9-mediated CCR5 ablation in human hematopoietic stem/progenitor cells confers HIV-1 resistance in vivo. Mol. Ther. 25, 1782–1789 (2017).

    Article  CAS  Google Scholar 

  29. 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  Google Scholar 

  30. Stadtmauer, E. A. et al. CRISPR-engineered T cells in patients with refractory cancer. Science 367, eaba7365 (2020).

    Article  CAS  Google Scholar 

  31. Gillmore, J. D. et al. CRISPR-Cas9 in vivo gene editing for transthyretin amyloidosis. N. Engl. J. Med. 385, 493–502 (2021).

    Article  CAS  Google Scholar 

  32. 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  Google Scholar 

  33. Xu, S. et al. Editing aberrant splice sites efficiently restores β-globin expression in β-thalassemia. Blood 133, 2255–2262 (2019).

    Article  CAS  Google Scholar 

  34. Métais, J.-Y. et al. Genome editing of HBG1 and HBG2 to induce fetal hemoglobin. Blood Adv. 3, 3379–3392 (2019).

    Article  Google Scholar 

  35. Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187–197 (2015).

    Article  CAS  Google Scholar 

  36. Zeng, J. et al. Therapeutic base editing of human hematopoietic stem cells. Nat. Med. 26, 535–541 (2020).

    Article  CAS  Google Scholar 

  37. Musunuru, K. et al. In vivo CRISPR base editing of PCSK9 durably lowers cholesterol in primates. Nature 593, 429–434 (2021).

    Article  CAS  Google Scholar 

  38. Chu, S. H. et al. Rationally designed base editors for precise editing of the sickle cell disease mutation. CRISPR J. 4, 169–177 (2021).

    Article  CAS  Google Scholar 

  39. Newby, G. A. et al. Base editing of haematopoietic stem cells rescues sickle cell disease in mice. Nature 595, 295–302 (2021).

    Article  CAS  Google Scholar 

  40. 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).

    Article  CAS  Google Scholar 

  41. De Dreuzy, E. et al. EDIT-301: An experimental autologous cell therapy comprising Cas12a-RNP modified mPB-CD34+ cells for the potential treatment of SCD. Blood 134, 4636–4636 (2019).

    Article  Google Scholar 

  42. Zhao, M., Kim, P., Mitra, R., Zhao, J. & Zhao, Z. TSGene 2.0: an updated literature-based knowledgebase for tumor suppressor genes. Nucleic Acids Res. 44, D1023–D1031 (2016).

    Article  CAS  Google Scholar 

  43. Finkel, R. S. et al. Nusinersen versus sham control in infantile-onset spinal muscular atrophy. N. Engl. J. Med. 377, 1723–1732 (2017).

    Article  CAS  Google Scholar 

  44. Mercuri, E. et al. Nusinersen versus sham control in later-onset spinal muscular atrophy. N. Engl. J. Med. 378, 625–635 (2018).

    Article  CAS  Google Scholar 

  45. Raal, F. J. et al. Inclisiran for the treatment of heterozygous familial hypercholesterolemia. N. Engl. J. Med. 382, 1520–1530 (2020).

    Article  CAS  Google Scholar 

  46. Hickey, G. et al. Genotyping structural variants in pangenome graphs using the vg toolkit. Genome Biol. 21, 35 (2020).

    Article  Google Scholar 

  47. Ameur, A. Goodbye reference, hello genome graphs. Nat. Biotechnol. 37, 866–868 (2019).

    Article  CAS  Google Scholar 

  48. Center for Biologics Evaluation & Research. Human gene therapy products incorporating human genome editing. U.S. Food and Drug Administration https://www.fda.gov/regulatory-information/search-fda-guidance-documents/human-gene-therapy-products-incorporating-human-genome-editing (2022).

  49. Corces, M. R. et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Gen. 48, 1193–1203 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

L.P. received support from the National Institutes of Health (NIH) (R35 HG010717 and RM1 HG009490). D.E.B. was supported by the National Heart, Lung, and Blood Institute (OT2HL154984 and P01HL053749), Burroughs Wellcome Fund, Doris Duke Charitable Foundation and the St. Jude Children’s Research Hospital Collaborative Research Consortium. R.G received support from European Union’s ERA-NET JPCOFUND2 (JPND2019-466-037). We thank S. H. Orkin, G. Lettre, J. K. Joung, V. Pattanayak, K. Petri, A. H. Shen, E. Dirupo and F. Masillo for helpful input.

Author information

Authors and Affiliations

Authors

Contributions

S.C., L.Y.L., M.T., N.B., R.G. and L.P. created the software; J.Z., M.A.N., S.A.M., M.F.C., V.K., S.Q.T., M.A., S.A.W. and D.E.B. designed and conducted experiments; S.C., J.Z., L.Y.L., J.L., R.G., D.E.B. and L.P. performed data analysis; S.C., R.G., D.E.B. and L.P. conceived the work; S.C., J.Z., L.Y.L, R.G., D.E.B. and L.P. wrote the paper with input from all authors.

Corresponding authors

Correspondence to Rosalba Giugno, Daniel E. Bauer or Luca Pinello.

Ethics declarations

Competing interests

L.P. has financial interests in Edilytics, Excelsior Genomics and SeQure Dx. L.P.’s interests were reviewed and are managed by Massachusetts General Hospital and Partners HealthCare in accordance with their conflict of interest policies. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Krishanu Saha and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Top 100 predicted off-target sites for BCL11A-1617 spacer by CFD score.

CRISPRme search as in Fig. 1. Candidate off-target sites within coding regions based on GENCODE annotations and ATAC-seq peaks in HSCs based on user-provided annotations (data from ref. 49) are highlighted.

Extended Data Fig. 2 Plots with rank ordered correlation between CFD and CRISTA reported targets.

Scatter plots show from left to right, the correlation of ranked targets, extracted by selecting top 10000 targets ordered by CFD and CRISTA score, respectively. The left plot shows the rank correlation of targets with 0 bulges (Pearson’s correlation: 0.57, p < 1e-10, Spearman’s correlation: 0.55, p < 1e-10), the center plot shows rank correlation of targets with 1 bulge (Pearson’s correlation: −0.16, p < 1 e-10, Spearman’s correlation: −0.33, p < 1 e-10) and the right plot shows the rank correlation of targets with 2 bulges (Pearson’s correlation: −0.55, p < 1e-10, Spearman’s correlation: −0.80, p < 1e-10). The correlation values and p-values(two-sided) were calculated using standard functions from the Python scipy library. The colors represent the lowest count of bulges for each target, because the two scoring methods may prioritize different alignments and thus different number of mismatches and bulges of the same genomic target.

Extended Data Fig. 3 HGDP superpopulation distribution plots.

HGDP variant off-targets with CFD ≥ 0.2 and increase in CFD of ≥0.1. Individual samples from each of the seven superpopulations were shuffled 100 times to calculate the mean and 95% confidence interval. First panel shows distribution within all 54 discrete populations, colored by superpopulation. Additional seven panels show distribution of discrete populations within each listed superpopulation.

Extended Data Fig. 4 Candidate transcript off-targets introduced by common genetic variants for non-CRISPR sequence-based RNA-targeting therapeutic strategies.

a) A common SNP (in blue) introduces a candidate CDS off-target site with 2 mismatches for the FDA-approved antisense oligo Nusinersen. b) Top 1000 candidate transcript off-targets ranked by mismatches and bulges for Nusinersen from a search performed with the 1000 G and HGDP genetic variant datasets. c) A common insertion variant (in red) introduces a candidate 3’UTR off-target site with 4 mismatches + bulges for the FDA-approved RNAi therapy Inclisiran. d) Top 1000 candidate transcript off-targets ranked by mismatches and bulges for Inclisiran from a search performed with the 1000 G and HGDP genetic variant datasets.

Supplementary information

Supplementary Information

Supplementary Figures 1–13, Tables 1–5, Notes 1–9 and Data 1–4. Supplementary figures, tables and notesare included in the flat file. Supplementary data files are separate (see below).

Reporting Summary

Supplementary Data 1

Top candidate off-targets in CRISPRme search results for sg1617 using hg38, 1000 G and HGDP data with up to six mismatches and two bulges (including the integrated_results, all_results_with_alternative_alignments, and private_targets files).

Supplementary Data 2

Top candidate off-targets in CRISPRme search results for other example gRNAs with NGG PAMs.

Supplementary Data 3

Top candidate off-targets in CRISPRme search results for example gRNAs with non-NGG PAMs.

Supplementary Data 4

Top candidate off-targets in CRISPRme search results for example non-CRISPR-based, RNA-targeting strategies (antisense oligo and RNA interference).

Source data

Source Data Fig. 1

Gel image.

Source Data Fig. 2

ddPCR image files.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cancellieri, S., Zeng, J., Lin, L.Y. et al. Human genetic diversity alters off-target outcomes of therapeutic gene editing. Nat Genet 55, 34–43 (2023). https://doi.org/10.1038/s41588-022-01257-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01257-y

This article is cited by

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