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.

  • Analysis
  • Published:

The impact of nonsense-mediated mRNA decay on genetic disease, gene editing and cancer immunotherapy

Abstract

Premature termination codons (PTCs) can result in the production of truncated proteins or the degradation of messenger RNAs by nonsense-mediated mRNA decay (NMD). Which of these outcomes occurs can alter the effect of a mutation, with the engagement of NMD being dependent on a series of rules. Here, by applying these rules genome-wide to obtain a resource called NMDetective, we explore the impact of NMD on genetic disease and approaches to therapy. First, human genetic diseases differ in whether NMD typically aggravates or alleviates the effects of PTCs. Second, failure to trigger NMD is a cause of ineffective gene inactivation by CRISPR–Cas9 gene editing. Finally, NMD is a determinant of the efficacy of cancer immunotherapy, with only frameshifted transcripts that escape NMD predicting a response. These results demonstrate the importance of incorporating the rules of NMD into clinical decision-making. Moreover, they suggest that inhibiting NMD may be effective in enhancing cancer immunotherapy.

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: NMDetective catalogs the effects of all possible PTCs in the human genome.
Fig. 2: Disease phenotypes arising from germline PTCs are modulated by NMD.
Fig. 3: NMD rules determine the outcome of CRISPR–Cas9 gene editing.
Fig. 4: Efficacy of immunotherapy is predicted by the burden of NMD-evading frameshift indels but not other indels.

Similar content being viewed by others

Data availability

The NMD efficacy predictions have been made available through the Figshare repository at https://www.figshare.com/articles/NMDetective/7803398 and via a digital object identifier at https://doi.org/10.6084/m9.figshare.7803398.

References

  1. Lykke-Andersen, S. & Jensen, T. H. Nonsense-mediated mRNA decay: an intricate machinery that shapes transcriptomes. Nat. Rev. Mol. Cell Biol. 16, 665–677 (2015).

    CAS  PubMed  Google Scholar 

  2. Popp, M. W. & Maquat, L. E. Leveraging rules of nonsense-mediated mRNA decay for genome engineering and personalized medicine. Cell 165, 1319–1322 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Silva, A. L. & Romão, L. The mammalian nonsense-mediated mRNA decay pathway: to decay or not to decay! Which players make the decision? FEBS Lett. 583, 499–505 (2009).

    CAS  PubMed  Google Scholar 

  4. Nagy, E. & Maquat, L. E. A rule for termination-codon position within intron-containing genes: when nonsense affects RNA abundance. Trends Biochem. Sci. 23, 198–199 (1998).

    CAS  PubMed  Google Scholar 

  5. Le Hir, H., Gatfield, D., Izaurralde, E. & Moore, M. J. The exon–exon junction complex provides a binding platform for factors involved in mRNA export and nonsense-mediated mRNA decay. EMBO J. 20, 4987–4997 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kashima, I. et al. Binding of a novel SMG-1–Upf1–eRF1–eRF3 complex (SURF) to the exon junction complex triggers Upf1 phosphorylation and nonsense-mediated mRNA decay. Genes Dev. 20, 355–367 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Brogna, S. & Wen, J. Nonsense-mediated mRNA decay (NMD) mechanisms. Nat. Struct. Mol. Biol. 16, 107–113 (2009).

    CAS  PubMed  Google Scholar 

  8. Rivas, M. A. et al. Human genomics. Effect of predicted protein-truncating genetic variants on the human transcriptome. Science 348, 666–669 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Lindeboom, R. G., Supek, F. & Lehner, B. The rules and impact of nonsense-mediated mRNA decay in human cancers. Nat. Genet. 48, 1112–1118 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Hu, Z., Yau, C. & Ahmed, A. A. A pan-cancer genome-wide analysis reveals tumour dependencies by induction of nonsense-mediated decay. Nat. Commun. 8, 15943 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Hoek, T. A. et al. Single-molecule imaging uncovers rules governing nonsense-mediated mRNA decay. Mol. Cell 75, 324–339.e11 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Miller, J. N. & Pearce, D. A. Nonsense-mediated decay in genetic disease: friend or foe? Mutat. Res. Rev. Mutat. Res. 762, 52–64 (2014).

    CAS  PubMed  Google Scholar 

  13. Khajavi, M., Inoue, K. & Lupski, J. R. Nonsense-mediated mRNA decay modulates clinical outcome of genetic disease. Eur. J. Hum. Genet. 14, 1074–1081 (2006).

    CAS  PubMed  Google Scholar 

  14. Coban-Akdemir, Z. et al. Identifying genes whose mutant transcripts cause dominant disease traits by potential gain-of-function alleles. Am. J. Hum. Genet. 103, 171–187 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Mort, M., Ivanov, D., Cooper, D. N. & Chuzhanova, N. A. A meta-analysis of nonsense mutations causing human genetic disease. Hum. Mutat. 29, 1037–1047 (2008).

    CAS  PubMed  Google Scholar 

  16. Holbrook, J. A., Neu-Yilik, G., Hentze, M. W. & Kulozik, A. E. Nonsense-mediated decay approaches the clinic. Nat. Genet. 36, 801–808 (2004).

    CAS  PubMed  Google Scholar 

  17. Usuki, F. et al. Inhibition of SMG-8, a subunit of SMG-1 kinase, ameliorates nonsense-mediated mRNA decay-exacerbated mutant phenotypes without cytotoxicity. Proc. Natl Acad. Sci. USA 110, 15037–15042 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Bokhari, A. et al. Targeting nonsense-mediated mRNA decay in colorectal cancers with microsatellite instability. Oncogenesis 7, 70 (2018).

    PubMed  PubMed Central  Google Scholar 

  19. Huang, L. et al. Antisense suppression of the nonsense mediated decay factor Upf3b as a potential treatment for diseases caused by nonsense mutations. Genome Biol. 19, 4 (2018).

    PubMed  PubMed Central  Google Scholar 

  20. Lejeune, F. Triple effect of nonsense-mediated mRNA decay inhibition as a therapeutic approach for cancer. Single Cell Biol. 5, 1000136 (2016).

    Google Scholar 

  21. Gorlov, I. P., Gorlova, O. Y., Frazier, M. L., Spitz, M. R. & Amos, C. I. Evolutionary evidence of the effect of rare variants on disease etiology. Clin. Genet. 79, 199–206 (2011).

    CAS  PubMed  Google Scholar 

  22. Maher, M. C., Uricchio, L. H., Torgerson, D. G. & Hernandez, R. D. Population genetics of rare variants and complex diseases. Hum. Hered. 74, 118–128 (2012).

    PubMed  Google Scholar 

  23. Kerr, T. P., Sewry, C. A., Robb, S. A. & Roberts, R. G. Long mutant dystrophins and variable phenotypes: evasion of nonsense-mediated decay? Hum. Genet. 109, 402–407 (2001).

    CAS  PubMed  Google Scholar 

  24. Hall, G. W. & Thein, S. Nonsense codon mutations in the terminal exon of the β-globin gene are not associated with a reduction in β-mRNA accumulation: a mechanism for the phenotype of dominant β-thalassemia. Blood 83, 2031–2037 (1994).

    CAS  PubMed  Google Scholar 

  25. Spinner, N. B. et al. Jagged1 mutations in Alagille syndrome. Hum. Mutat. 17, 18–33 (2001).

    CAS  PubMed  Google Scholar 

  26. van Bon, B. W. et al. Intragenic deletion in DYRK1A leads to mental retardation and primary microcephaly. Clin. Genet. 79, 296–299 (2011).

    CAS  PubMed  Google Scholar 

  27. Brown, L. Y. et al. Holoprosencephaly due to mutations in ZIC2: alanine tract expansion mutations may be caused by parental somatic recombination. Hum. Mol. Genet. 10, 791–796 (2001).

    CAS  PubMed  Google Scholar 

  28. Banning, A., Schiff, M. & Tikkanen, R. Amlexanox provides a potential therapy for nonsense mutations in the lysosomal storage disorder Aspartylglucosaminuria. Biochim. Biophys. Acta Mol. Basis Dis. 1864, 668–675 (2018).

    CAS  PubMed  Google Scholar 

  29. Gonzalez-Hilarion, S. et al. Rescue of nonsense mutations by amlexanox in human cells. Orphanet J. Rare Dis. 7, 58 (2012).

    PubMed  PubMed Central  Google Scholar 

  30. Martin, L. et al. Identification and characterization of small molecules that inhibit nonsense-mediated RNA decay and suppress nonsense p53 mutations. Cancer Res. 74, 3104–3113 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, H., La Russa, M. & Qi, L. S. CRISPR/Cas9 in genome editing and beyond. Annu. Rev. Biochem. 85, 227–264 (2016).

    CAS  PubMed  Google Scholar 

  32. Wang, T. et al. Identification and characterization of essential genes in the human genome. Science 350, 1096–1101 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Meyers, R. M. et al. Computational correction of copy number effect improves specificity of CRISPR–Cas9 essentiality screens in cancer cells. Nat. Genet. 49, 1779–1784 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Wang, T. et al. Gene essentiality profiling reveals gene networks and synthetic lethal interactions with oncogenic Ras. Cell 168, 890–903.e15 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Findlay, G. M. et al. Accurate classification of BRCA1 variants with saturation genome editing. Nature 562, 217–222 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Doench, J. G. et al. Rational design of highly active sgRNAs for CRISPR–Cas9-mediated gene inactivation. Nat. Biotechnol. 32, 1262–1267 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  Google Scholar 

  38. Hargadon, K. M., Johnson, C. E. & Williams, C. J. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. Int. Immunopharmacol. 62, 29–39 (2018).

    CAS  PubMed  Google Scholar 

  39. Chalmers, Z. R. et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 9, 34 (2017).

    PubMed  PubMed Central  Google Scholar 

  40. Goodman, A. M. et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol. Cancer Ther. 16, 2598–2608 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Turajlic, S. et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol. 18, 1009–1021 (2017).

    CAS  PubMed  Google Scholar 

  42. Van Allen, E. M. et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350, 207–211 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Hugo, W. et al. Genomic and transcriptomic features of response to Anti-PD-1 therapy in metastatic melanoma. Cell 165, 35–44 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Miao, D. et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 359, 801–806 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Forde, P. M. et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N. Engl. J. Med. 378, 1976–1986 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Miao, D. et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat. Genet. 50, 1271–1281 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Durand, S. et al. Inhibition of nonsense-mediated mRNA decay (NMD) by a new chemical molecule reveals the dynamic of NMD factors in P-bodies. J. Cell Biol. 178, 1145–1160 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Keeling, K. M. et al. Attenuation of nonsense-mediated mRNA decay enhances in vivo nonsense suppression. PLoS ONE 8, e60478 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Pastor, F., Kolonias, D., Giangrande, P. H. & Gilboa, E. Induction of tumour immunity by targeted inhibition of nonsense-mediated mRNA decay. Nature 465, 227–230 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Gonzàlez-Porta, M., Frankish, A., Rung, J., Harrow, J. & Brazma, A. Transcriptome analysis of human tissues and cell lines reveals one dominant transcript per gene. Genome Biol. 14, R70 (2013).

    PubMed  PubMed Central  Google Scholar 

  51. Grossman, R. L. et al. Toward a shared vision for cancer genomic data. N. Engl. J. Med. 375, 1109–1112 (2016).

    PubMed  PubMed Central  Google Scholar 

  52. Friedel, C. C., Dölken, L., Ruzsics, Z., Koszinowski, U. H. & Zimmer, R. Conserved principles of mammalian transcriptional regulation revealed by RNA half-life. Nucleic Acids Res. 37, e115 (2009).

    PubMed  PubMed Central  Google Scholar 

  53. Haeussler, M. et al. The UCSC genome browser database: 2019 update. Nucleic Acids Res. 47, D853–D858 (2019).

    CAS  PubMed  Google Scholar 

  54. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Landrum, M. J. et al. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 46, D1062–D1067 (2018).

    CAS  PubMed  Google Scholar 

  56. Balasubramanian, S. et al. Using ALoFT to determine the impact of putative loss-of-function variants in protein-coding genes. Nat. Commun. 8, 382 (2017).

    PubMed  PubMed Central  Google Scholar 

  57. Karczewski, K. J. et al. Variation across 141,456 human exomes and genomes reveals the spectrum of loss-of-function intolerance across human protein-coding genes. Preprint at bioRxiv https://doi.org/10.1101/531210 (2019).

  58. Hart, T. & Moffat, J. BAGEL: a computational framework for identifying essential genes from pooled library screens. BMC Bioinformatics 17, 164 (2016).

    PubMed  PubMed Central  Google Scholar 

  59. Altschul, S. F., Gish, W., Miller, W., Myers, E. W. & Lipman, D. J. Basic local alignment search tool. J. Mol. Biol. 215, 403–410 (1990).

    CAS  PubMed  Google Scholar 

  60. Heigwer, F., Kerr, G. & Boutros, M. E-CRISP: fast CRISPR target site identification. Nat. Methods 11, 122–123 (2014).

    CAS  PubMed  Google Scholar 

  61. Thorsson, V. et al. The immune landscape of cancer. Immunity 48, 812–830.e14 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Eisenhauer, E. A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).

    CAS  PubMed  Google Scholar 

  63. Wolchok, J. D. et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin. Cancer Res. 15, 7412–7420 (2009).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was funded by the ERC Starting Grant HYPER-INSIGHT (757700, to F.S.) and the ERC Consolidator Grant IR-DC (616434, to B.L.). R.G.H.L. and M.V. are supported by the Oncode Institute, which is partly funded by the Dutch Cancer Society. M.V. also acknowledges support by the gravitation program Cancer GenomiCs.nl from the Netherlands Organization for Scientific Research. F.S. and B.L. are funded by the ICREA Research Professor programme. F.S. and B.L. acknowledge support of the Severo Ochoa Centres of Excellence programme to the IRB Barcelona and to the CRG, respectively. B.L. and F.S. were supported by the Spanish Ministry of Economy and Competitiveness (grant no. BFU2017-89488-P and BFU2017-89833-P, respectively). B.L. was further supported by the Bettencourt Schueller Foundation, Agencia de Gestio d’Ajuts Universitaris i de Recerca (no. 2017 SGR 1322) and the CERCA Program/Generalitat de Catalunya. B.L. also acknowledges the support of the Spanish Ministry of Economy, Industry and Competitiveness to the EMBL partnership.

Author information

Authors and Affiliations

Authors

Contributions

F.S., B.L. and R.G.H.L. conceptualized the study. F.S. and R.G.H.L. devised the methodology. R.G.H.L. carried out the formal analysis. R.G.H.L. and F.S. carried out the investigation. R.G.H.L. curated and validated the data. R.G.H.L. operated the software and was responsible for the data visualization. R.G.H.L., F.S. and B.L. wrote the original draft. B.L., R.G.H.L. and F.S. reviewed and edited the draft. M.V., B.L. and F.S. acquired the funding. F.S., B.L. and M.V. supervised the study.

Corresponding authors

Correspondence to Ben Lehner or Fran Supek.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 The distribution of genome-wide NMD efficacy scores and of NMD rules in all genes with more than 20 disease-associated PTC variants.

a, the distribution of NMDetective-A scores over all genes in hg38 reveals three global clusters of inefficient, intermediate-efficiency and efficient NMD. b, genes in which there is an excess of PTCs in NMD-evading regions (left barplot) and genes where there is a dearth of PTCs is NMD-evading regions (right barplot). The proportion of PTCs in different NMD-evading regions is shown as colored segments in the bar chart. The relative portion of the protein-coding mRNA sequence that is covered by the NMD rules is shown as a black vertical stripe. c, a schematic of a gene that illustrates how PTCs that trigger or evade NMD can lead to different outcomes in protein expression.

Extended Data Fig. 2 The sequence context of nonsense variants is not different between different types of NMD regions.

a, the trinucleotide spectrum of nonsense variants in human populations is consistent across gene regions that trigger or evade NMD, b. spectrum of variants shows high Pearson correlations between the different types of NMD regions. c, the baseline NMD-evasion rule coverage, obtained from nonsense variants simulated from the trinucleotide context of whole-genome population variants at different VAF ranges, exhibits a consistent distribution at different VAF ranges. Observed nonsense variants are increasingly enriched towards NMD- evading regions with an increasing VAF, compared to the simulated baseline at same VAFs. Odds ratios significant at P < 0.01 (Fisher’s exact test) are shown, comparing the distribution of simulated versus observed nonsense mutations.

Extended Data Fig. 3 Disease genes with a significant enrichment of PTC variants that do or do not trigger NMD, with and without normalization to local density of missense mutations.

a-b, significant enrichment of genes at FDR < 5% in a test that normalizes to the number of ClinVar missense variants observed in the same NMD regions. c-d, genes significant at an FDR < 25% are shown (see Fig. 2d–e for a list at FDR < 5%). Log2 odds ratios are for ClinVar frequencies of NMD-evading frameshift indel and nonsense variants versus NMD-detected frameshift indel and nonsense variants regions of a gene, normalized to the length of the NMD-evading versus NMD-detected regions. FDRs are by Fisher’s exact test, two-tailed, Benjamini-Hochberg adjusted. a-d, log2 odds ratios are shown separately for the four rules, for those rules which are significant in a particular gene.

Extended Data Fig. 4 Effect of NMD rules observed in CRISPR assays.

a, sgRNAs targeted to gene regions that evade NMD show a weaker enrichment in an experiment that selects for cells that do not express the targeted protein. Fold differences in sgRNA abundance are pooled per rule and shown for all proteins in a and broken down by protein in c. P values are by Mann-Whitney U test, two-sided. b, Models that classify essential from non-essential genes based on the fold-depletion of sgRNAs are more accurate for sgRNAs that target gene regions that trigger NMD than for sgRNAs targeted to different NMD-evading regions.

Extended Data Fig. 5 Relevance of NMD rules for CRISPR sgRNA design.

a, fitness loss upon targeting a non-essential gene (left) versus an essential gene (right) using a sgRNA directed at gene sections covered by various NMD-evasion rules. b-e, distribution of loci targeted by sgRNAs that are NMD-detected or NMD-evading (according to the individual NMD rules) for genome-wide CRISPR libraries (b, c) or for sgRNA design tools (d, e).

Extended Data Fig. 6 CRISPR screening data support canonical and non-canonical determinants of NMD efficacy.

a, the non-canonical long-exon NMD evasion rule has similar effects as the canonical last-exon NMD evasion rule, in terms of attenuated loss of fitness when targeting an essential gene (Methods). b-e, minor non-canonical NMD determinants, which are not included in the NMDetective-B model, but are included in the comprehensive NMDetective-A model. This includes: distance to downstream splice site in long exons (b), for the start-proximal rule, existence of a downstream in-frame AUG codon, presumably facilitating translation re-initiation (c), distance to the wild-type stop codon (d), and the effect of mRNA turnover on the observed NMD efficacy (e).

Extended Data Fig. 7 Tumor infiltration by immune cells is associated with a higher burden of NMD-evading frameshift indels.

a-b, Individual immune markers for the TCGA samples were estimated using gene expression data50. FDR is by two-sided Mann-Whitney U test, Benjamini-Hochberg adjusted. In panel b, only tests significant at FDR <25% are shown.

Extended Data Fig. 8 Evidence that NMD activity is a determinant of immune reactivity of tumors.

a, in the TCGA kidney cancer cohorts (KIRC, KICH and KIRP), a cancer type where indel burden is known to be strongly associated with immunogenicity41, higher relative burden of NMD-evading frameshifts was associated with longer survival (p = 0.011 for pooled data from both panels, by log-rank test) without application of immunotherapy. Patients were classified based on the number of frameshift indels that do not trigger NMD being higher than the number that trigger NMD (cyan) and those patients where the converse is true (red). b, in the TCGA UCEC cohort of uterine corpus endometrial carcinoma, where the key NMD gene UPF1 is commonly mutated, the predicted higher impact of UPF1 mutations is associated with multiple gene-expression based markers of lymphocyte, but not macrophage, infiltration. Patients with more than one UPF1 mutation were assigned to the group with the higher impact score. P values by Mann-Whitney U test.

Extended Data Fig. 9 NMD rules improve predictions of response to immunotherapy across multiple cancer types.

a, assigning NMD rules to frameshift mutations based on the location of the first downstream PTC in the new reading frame also shows that the burden of frameshifts that evade NMD is higher in patients that respond to immunotherapy. P values are by one-tailed Mann-Whitney U test. b, standardized regression coefficients and the 95% confidence interval in a logistic regression model that predicts responders versus nonresponders. c, pseudo-R2 for sequential addition of features to a joint model. The null model includes only the study (dataset) ID as a covariate. d, precision-recall curves for logistic regression models with three different sets of features: a tumor mutation burden (TMB) baseline, another baseline where TMB and all frameshifting indels are considered together, and the full model that considers TMB and NMD-evading and NMD-detected frameshifting indels separately. P values are by Chi-squared test. AUPRC, area under the precision-recall curve.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lindeboom, R.G.H., Vermeulen, M., Lehner, B. et al. The impact of nonsense-mediated mRNA decay on genetic disease, gene editing and cancer immunotherapy. Nat Genet 51, 1645–1651 (2019). https://doi.org/10.1038/s41588-019-0517-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-019-0517-5

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer