Skip to main content
Log in

Analysis of Off-target Effects and Risk Assessment Leading from Preclinical to Clinical Trials of Gene-edited Therapeutic Products

  • Original Research
  • Published:
Therapeutic Innovation & Regulatory Science Aims and scope Submit manuscript

Abstract

Objective

Concerns about off-target effects (OTEs) of genomic DNA cleavages by gene-editing enzymes have been raised, especially for OTEs that go undetected due to technical limitations. Since no explicit guidelines have been in place for risk assessment of OTEs, the regulatory authorities’ concept of an acceptable evaluation scheme for OTEs in the investigational drug application (IND) has not been clear. Here, we clarify the regulatory expectations by examining reports on OTE evaluations of leading gene-editing products that have achieved IND clearance.

Methods

We collected and analyzed the gene-editing products that have entered clinical trials by searching on ClinicalTrials.gov and EU-Clinical-Trial-Registries, and related reports for OTE evaluations from Google Scholar, PubMed, and the developers’ websites.

Results

We found a common two-step verification method used for different products at the preclinical stage. First, numerous potential off-target loci (POLs) are listed with state-of-the-art high-sensitivity detection methods and theoretical screens; Second, these OTEs are checked by amplicon sequencing of the POLs after treatment by enzymes in in vitro models close to clinical use conditions. Only the OTEs that can be detected and verified are addressed in the risk assessment in the translational phase from preclinical to clinical study.

Discussion

Here, we describe a clear scheme for risk assessment of OTEs at the key translational phase, based on the common features in protocols for gene-editing products that were cleared for use in clinical trials. This report will provide a guide for those newly attempting to conduct clinical development in this field.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Figure 1
Figure 2
Figure 3
Figure 4

Similar content being viewed by others

References

  1. Urnov FD, Miller JC, Lee Y-L, et al. Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature. 2005;435:646–51.

    Article  CAS  PubMed  Google Scholar 

  2. Cermak T, Doyle EL, Christian M, et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 2011;39:e82.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Jinek M, Chylinski K, Fonfara I, et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816–21.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  4. Zhang X-H, Tee LY, Wang X-G, et al. Off-target effects in CRISPR/Cas9-mediated genome engineering. Mol Ther Nucleic Acids. 2015;4:e264.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Kosicki M, Tomberg K, Bradley A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements. Nat Biotechnol. 2018;36:765–71.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. FDA: Long Term Follow-Up After Administration of Human Gene Therapy Products; Guidance for Industry. 37

  7. Bao XR, Pan Y, Lee CM, et al. Tools for experimental and computational analyses of off-target editing by programmable nucleases. Nat Protoc. 2021;16:10–26.

    Article  CAS  PubMed  Google Scholar 

  8. Tebas P, Stein D, Tang WW, et al. Gene editing of CCR5 in autologous CD4 T Cells of persons infected with HIV. N Engl J Med. 2014;370:901–10.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Qasim W, Zhan H, Samarasinghe S, et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci Transl Med. 2017. https://doi.org/10.1126/scitranslmed.aaj2013.

    Article  PubMed  Google Scholar 

  10. Frangoul H, Altshuler D, Cappellini MD, et al. CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. N Engl J Med. 2021;384:252–60.

    Article  CAS  PubMed  Google Scholar 

  11. Holmes MC, Reik A, Rebar EJ, et al. A potential therapy for beta-thalassemia (ST-400) and sickle cell disease (BIVV003). Biol Blood Marrow Transplant. 2018;24:S172.

    Article  Google Scholar 

  12. Bianconi E, Piovesan A, Facchin F, et al. An estimation of the number of cells in the human body. Ann Hum Biol. 2013;40:463–71.

    Article  PubMed  Google Scholar 

  13. EMA: Guideline on the quality, non-clinical and clinical aspects of gene therapy medicinal products.

  14. ClinicalTrials.gov. https://clinicaltrials.gov/. Accessed 17 July 2022.

  15. EU Clinical Trials Register - Update. https://www.clinicaltrialsregister.eu/.Accessed 17 July 2022.

  16. De Dreuzy E, Heath J, Zuris JA, et al. EDIT-301: an experimental autologous cell therapy comprising Cas12a-RNP modified mPB-CD34+ Cells for the potential treatment of SCD. Blood. 2019;134:4636–4636.

    Article  Google Scholar 

  17. Intellia Therapeutics. https://www.intelliatx.com/.Accessed 17 July 2022.

  18. Kanter J, DiPersio JF, Leavey P, et al. Cedar trial in progress: a first in human, phase 1/2 study of the correction of a single nucleotide mutation in autologous HSCs (GPH101) to convert HbS to HbA for treating severe SCD. Blood. 2021;138:1864.

    Article  Google Scholar 

  19. CRISPR. http://www.crisprtx.com/.Accessed 17 July 2022.

  20. Dar H, Henderson D, Padalia Z, et al. Preclinical development of CTX120, an allogeneic CAR-T cell targeting Bcma. Blood. 2018;132:1921.

    Article  Google Scholar 

  21. Dequeant M-L, Sagert J, Kalaitzidis D, et al. Abstract 1537: CD70 knockout: a novel approach to augment CAR-T cell function. Cancer Res. 2021;81:1537.

    Article  Google Scholar 

  22. Cossette D, Aiyer S, Kimball C, et al. Clinical-scale production and characterization of Ntla-5001—a novel approach to manufacturing CRISPR/Cas9 engineered T cell therapies. Blood. 2021;138:3881.

    Article  Google Scholar 

  23. Caribou Biosciences | Developing Sophisticated Allogeneic Cell Therapies. https://www.cariboubio.com/. Accessed 17 July 2022.

  24. Vor Bio. https://www.vorbio.com/. Accessed 17 July 2022.

  25. Maeder ML, Stefanidakis M, Wilson CJ, et al. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat Med. 2019;25:229–33.

    Article  CAS  PubMed  Google Scholar 

  26. Gillmore JD, Gane E, Taubel J, et al. CRISPR-Cas9 In vivo gene editing for transthyretin amyloidosis. N Engl J Med. 2021;385:493–502.

    Article  CAS  PubMed  Google Scholar 

  27. Seitzer J. NTLA-2002: CRISPR/Cas9-mediated gene knockout of KLKB1 to treat hereditary angioedema. J Allergy Clin Immunol. 2021. https://doi.org/10.1016/j.jaci.2020.12.531.

    Article  Google Scholar 

  28. Excision Bio. https://www.excision.bio. Accessed 17 July 2022.

  29. Perez EE, Wang J, Miller JC, et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol. 2008;26:808–16.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Sangamo Therapeutics, Inc. Sangamo. https://www.sangamo.com/programs/. aAccessed 17 July 2022.

  31. Harmatz P, Lau HA, Heldermon C, et al. Empowers: a phase 1/2 clinical trial of SB-318 ZFN-mediated in vivo human genome editing for treatment of MPS I (Hurler syndrome). Mol Genet Metab. 2019;126:S68.

    Google Scholar 

  32. Muenzer J, Prada CE, Burton B, et al. Champions: a phase 1/2 clinical trial with dose escalation of SB-913 ZFN-mediated in vivo human genome editing for treatment of MPS II (Hunter syndrome). Mol Genet Metab. 2019;126:S104.

    Google Scholar 

  33. Sangamo Therapeutics, Inc.. https://investor.sangamo.com/news-releases/news-release-details/sangamo-announces-treatment-first-patient-phase-12-clinical-0. Accessed 17 July 2022.

  34. DiGiusto DL, Cannon PM, Holmes MC, et al. Preclinical development and qualification of ZFN-mediated CCR5 disruption in human hematopoietic stem/progenitor cells. Mol Ther Method Clin Dev. 2016. https://doi.org/10.1038/mtm.2016.67.

    Article  Google Scholar 

  35. Lattanzi A, Camarena J, Lahiri P, et al. Development of β-globin gene correction in human hematopoietic stem cells as a potential durable treatment for sickle cell disease. Sci Transl Med. 2021. https://doi.org/10.1126/scitranslmed.abf2444.

    Article  PubMed Central  PubMed  Google Scholar 

  36. Bae S, Park J, Kim J-S. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinform Oxf Engl. 2014;30:1473–5.

    Article  CAS  Google Scholar 

  37. Cradick TJ, Qiu P, Lee CM, et al. COSMID: a web-based tool for identifying and validating CRISPR/Cas off-target sites. Mol Ther Nucleic Acids. 2014;3:e214.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  38. ClinicalTrials.gov. https://clinicaltrials.gov/ProvidedDocs/52/NCT01543152/Prot_000.pdf Accessed 17 July 2022.

  39. ClinicalTrials.gov. https://clinicaltrials.gov/ProvidedDocs/65/NCT02225665/Prot_001.pdf Accessed 17 July 2022.

  40. Clinical Trial Registration NCT01044654, Clinicaltrials.gov.https://clinicaltrials.gov/ct2/show/NCT01044654. Accessed 17 July 2022.

  41. Clinical Trial Registration NCT02500849, Clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT02500849. Accessed 17 July 2022.

  42. Gabriel R, Lombardo A, Arens A, et al. An unbiased genome-wide analysis of zinc-finger nuclease specificity. Nat Biotechnol. 2011;29:816–23. https://doi.org/10.1038/nbt.1948.

    Article  CAS  PubMed  Google Scholar 

  43. Kim D, Bae S, Park J, et al. Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-target effects in human cells. Nat Method. 2015;12:237–43.

    Article  CAS  Google Scholar 

  44. Cameron P, Fuller CK, Donohoue PD, et al. Mapping the genomic landscape of CRISPR–Cas9 cleavage. Nat Method. 2017;14:600–6.

    Article  CAS  Google Scholar 

  45. Tsai SQ, Nguyen NT, Malagon-Lopez J, et al. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets. Nat Method. 2017;14:607–14.

    Article  CAS  Google Scholar 

  46. Kim D, Kim J-S. DIG-seq: a genome-wide CRISPR off-target profiling method using chromatin DNA. Genome Res. 2018;28:1894–900.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  47. Clinical Trial Registration NCT03872479, Clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT03872479. Accessed 17 July 2022.

  48. Clinical Trial Registration NCT04601051, Clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT04601051.Accessed 17 July 2022.

  49. Clinical Trial Registration NCT04819841, Clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT04819841. Accessed 17 July 2022.

  50. Kuscu C, Arslan S, Singh R, et al. Genome-wide analysis reveals characteristics of off-target sites bound by the Cas9 endonuclease. Nat Biotechnol. 2014;32:677–83.

    Article  CAS  PubMed  Google Scholar 

  51. Tsai SQ, Zheng Z, Nguyen NT, et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat Biotechnol. 2015;33:187–97.

    Article  CAS  PubMed  Google Scholar 

  52. Frock RL, Hu J, Meyers RM, et al. Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases. Nat Biotechnol. 2015;33:179–86.

    Article  CAS  PubMed  Google Scholar 

  53. Wienert B, Wyman SK, Yeh CD, et al. CRISPR off-target detection with DISCOVER-seq. Nat Protoc. 2020;15:1775–99.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  54. Yan W, Mirzazadeh R, Garnerone S, et al. BLISS is a versatile and quantitative method for genome-wide profiling of DNA double-strand breaks. Nat Commun. 2017;8:15058.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

NY contributed to the study design, analysis, and interpretation, and he agrees to be accountable for all aspects of this work’s integrity and accuracy. AA contributed to the study design and interpretation, and he agrees to be accountable for all aspects of this work’s integrity and accuracy.

Corresponding author

Correspondence to Atsushi Aruga.

Ethics declarations

Conflict of interest

Naoki Yamada is an employee of Fujifilm Pharmaceuticals U.S.A. Inc. However, the views of the author in this paper do not reflect the views of the company. Atsushi Aruga has no conflicts of interest to disclose.

Additional information

Publisher's Note

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

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

Yamada, N., Aruga, A. Analysis of Off-target Effects and Risk Assessment Leading from Preclinical to Clinical Trials of Gene-edited Therapeutic Products. Ther Innov Regul Sci 57, 538–551 (2023). https://doi.org/10.1007/s43441-022-00481-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s43441-022-00481-2

Keywords

Navigation