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:

Curated variation benchmarks for challenging medically relevant autosomal genes

Abstract

The repetitive nature and complexity of some medically relevant genes poses a challenge for their accurate analysis in a clinical setting. The Genome in a Bottle Consortium has provided variant benchmark sets, but these exclude nearly 400 medically relevant genes due to their repetitiveness or polymorphic complexity. Here, we characterize 273 of these 395 challenging autosomal genes using a haplotype-resolved whole-genome assembly. This curated benchmark reports over 17,000 single-nucleotide variations, 3,600 insertions and deletions and 200 structural variations each for human genome reference GRCh37 and GRCh38 across HG002. We show that false duplications in either GRCh37 or GRCh38 result in reference-specific, missed variants for short- and long-read technologies in medically relevant genes, including CBS, CRYAA and KCNE1. When masking these false duplications, variant recall can improve from 8% to 100%. Forming benchmarks from a haplotype-resolved whole-genome assembly may become a prototype for future benchmarks covering the whole genome.

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: GIAB developed a process to create new phased small variant and SV benchmarks for 273 CMRGs.
Fig. 2: The new CMRG benchmark contains more challenging variants and regions than previous benchmarks.
Fig. 3: The new benchmark covers the gene SMN1, which was previously excluded due to mapping challenges for all technologies in the highly identical segmental duplication.
Fig. 4: The benchmark resolves the gene CBS, which has a highly homologous gene (CBSL) due to a false duplication in GRCh38 that is not in HG002 or GRCh37.
Fig. 5: The new CMRG small variant benchmark includes more challenging variants and identifies more false negatives in a standard short-read callset (Illumina–BWA-MEM–GATK) than the previous v4.2.1 benchmark in these challenging genes.

Similar content being viewed by others

Data availability

The PacBio HiFi reads used to generate the hifiasm assembly for the benchmark are in the NCBI Sequence Read Archive with accession numbers SRR10382245, SRR10382244, SRR10382249, SRR10382248, SRR10382247 and SRR10382246. The v1.00 benchmark VCF and BED files, as well as Liftoff gene annotations, assembly–assembly alignments and variant calls, are available at https://trace.ncbi.nlm.nih.gov/ReferenceSamples/giab/release/AshkenazimTrio/HG002_NA24385_son/CMRG_v1.00/, and as a DOI at https://doi.org/10.18434/mds2-2475. This is released as a separate benchmark from v4.2.1, because it includes a small fraction of the genome, it has different characteristics from the mapping-based v4.2.1 and v4.2.1 only includes small variants. Using v4.2.1 and the CMRG benchmarks as two separate benchmarks enables users to obtain broader performance metrics for most of the genome and for a small set of particularly challenging genes, respectively. The masked GRCh38 reference, recently updated to v2 with additional false duplications from the Telomere-to-Telomere Consortium, is under https://trace.ncbi.nlm.nih.gov/ReferenceSamples/giab/release/references. We recommend using v3.0 GA4GH/GIAB stratification bed files intended for use with hap.py when benchmarking, which are available at https://trace.ncbi.nlm.nih.gov/ReferenceSamples/giab/release/genome-stratifications/. These stratifications include bed files corresponding to false duplications and collapsed duplications in GRCh38. All data have no restrictions, as the HG002 sample has an open consent from the Personal Genome Project.

Code availability

Scripts used to develop the CMRG benchmark and generate figures and tables for the manuscript are available at https://github.com/usnistgov/cmrg-benchmarkset-manuscript. The previously developed assembly, which was used as the basis of this benchmark, was from hifiasm v0.11.

A variety of open source software was used for variant calling for the evaluations of the benchmark, including NextDenovo2.2-beta.0, DRAGEN 3.6.3, NeuSomatic’s submission for the PrecisionFDA truth challenge v2 (ref. 12) (BWA-MEM50 version 0.7.17-r1188 (https://github.com/lh3/bwa) and GATK version gatk-4.1.4.1 (https://gatk.broadinstitute.org/hc/en-us)), Parabricks_DeepVariant (Parabricks Pipelines DeepVariant v3.0.0_2 (https://developer.nvidia.com/clara-parabricks)), Sentieon (DNAscope) version sentieon_release_201911 (https://www.sentieon.com/products/#dnaseq), BWA-MEM and Strelka2 (BWA-MEM version 0.7.17-r1188 (https://github.com/lh3/bwa) and Strelka2 version 2.9.10 (https://github.com/Illumina/strelka)), BWA-MEM50(v0.7.8), Picard tools (https://broadinstitute.github.io/picard/) (ver. 1.83), GATK52 (v3.4-0), GATK (v3.5), BWA-MEM v0.7.15-r1140, SAMtools53 v1.3, Picard v2.10.10, GATK v3.8, DELLY54 v0.8.5, GRIDSS55 v2.9.4, LUMPY56 v0.3.1, Manta57 v1.6.0, Wham58 v1.7.0, NanoPlot60 v1.27.0, Filtlong v0.2.0, minimap2 (refs. 40,60) v2.17-r941, cuteSV v1.0.8, Sniffles61 v1.0.12, SURVIVOR59 v1.0.7, BWA v0.7.15, GATK v3.6, Java v1.8.0_74 (OpenJDK), Picard Tools v2.6.0, Sambamba63 v0.6.7, Samblaster64 v0.1.24, Samtools v1.9, DeepVariant v1.0 and Liftoff32 v1.4.0.

References

  1. Wenger, A. M. et al. Accurate circular consensus long-read sequencing improves variant detection and assembly of a human genome. Nat. Biotechnol. 37, 1155–1162 (2019).

    Article  CAS  Google Scholar 

  2. Cheng, H., Concepcion, G. T., Feng, X., Zhang, H. & Li, H. Haplotype-resolved de novo assembly using phased assembly graphs with hifiasm. Nat. Methods 18, 170–175 (2021).

    Article  CAS  Google Scholar 

  3. Nurk, S. et al. HiCanu: accurate assembly of segmental duplications, satellites, and allelic variants from high-fidelity long reads. Genome Res. 30, 1291–1305 (2020).

    Article  CAS  Google Scholar 

  4. Shafin, K. et al. Nanopore sequencing and the Shasta toolkit enable efficient de novo assembly of eleven human genomes. Nat. Biotechnol. 38, 1044–1053 (2020).

    Article  CAS  Google Scholar 

  5. Mahmoud, M. et al. Structural variant calling: the long and the short of it. Genome Biol. 20, 246 (2019).

    Article  Google Scholar 

  6. De Coster, W., Weissensteiner, M. H. & Sedlazeck, F. J. Towards population-scale long-read sequencing. Nat. Rev. Genet. 22, 572–587 (2021).

    Article  Google Scholar 

  7. Mandelker, D. et al. Navigating highly homologous genes in a molecular diagnostic setting: a resource for clinical next-generation sequencing. Genet. Med. 18, 1282–1289 (2016).

    Article  CAS  Google Scholar 

  8. Ebbert, M. T. W. et al. Systematic analysis of dark and camouflaged genes reveals disease-relevant genes hiding in plain sight. Genome Biol. 20, 1–23 (2019).

    Article  Google Scholar 

  9. Lincoln, S. E. et al. One in seven pathogenic variants can be challenging to detect by NGS: an analysis of 450,000 patients with implications for clinical sensitivity and genetic test implementation. Genet. Med. 23, 1673–1680 (2021).

  10. Zook, J. M. et al. An open resource for accurately benchmarking small variant and reference calls. Nat. Biotechnol. 37, 561–566 (2019).

    Article  CAS  Google Scholar 

  11. Zook, J. M. et al. A robust benchmark for detection of germline large deletions and insertions. Nat. Biotechnol. 38, 1347–1355 (2020) ; erratum 38, 1357 (2020).

    Article  CAS  Google Scholar 

  12. Olson, N. D. et al. precisionFDA Truth Challenge V2: calling variants from short- and long-reads in difficult-to-map regions. Preprint at bioRxiv https://doi.org/10.1101/2020.11.13.380741 (2020).

  13. Wagner, J. et al. Benchmarking challenging small variants with linked and long reads. Preprint at bioRxiv https://doi.org/10.1101/2020.07.24.212712 (2020).

  14. Chin, C.-S. et al. A diploid assembly-based benchmark for variants in the major histocompatibility complex. Nat. Commun. 11, 4794 (2020).

    Article  CAS  Google Scholar 

  15. Goldfeder, R. L. et al. Medical implications of technical accuracy in genome sequencing. Genome Med. 8, 24 (2016).

    Article  Google Scholar 

  16. Ball, M. P. et al. A public resource facilitating clinical use of genomes. Proc. Natl Acad. Sci. USA 109, 11920–11927 (2012).

    Article  CAS  Google Scholar 

  17. Tate, J. G. et al. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 47, D941–D947 (2019).

    Article  CAS  Google Scholar 

  18. Ross, M. G. et al. Characterizing and measuring bias in sequence data. Genome Biol. 14, R51 (2013).

    Article  Google Scholar 

  19. Prior, T. W., Leach, M. E. & Finanger, E. Spinal muscular atrophy. In GeneReviews [Internet] (University of Washington, 2020).

  20. Biros, I. & Forrest, S. Spinal muscular atrophy: untangling the knot? J. Med. Genet. 36, 1–8 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Leiding, J. W. & Holland, S. M. Chronic granulomatous disease. In GeneReviews [Internet] (University of Washington, 2016).

  22. Innan, H. A two-locus gene conversion model with selection and its application to the human RHCE and RHD genes. Proc. Natl. Acad. Sci. USA 100, 8793–8798 (2003).

    Article  CAS  Google Scholar 

  23. Hayakawa, T. et al. Coevolution of Siglec-11 and Siglec-16 via gene conversion in primates. BMC Evol. Biol. 17, 228 (2017).

    Article  Google Scholar 

  24. Garg, P. et al. Pervasive cis effects of variation in copy number of large tandem repeats on local DNA methylation and gene expression. Am. J. Hum. Genet. https://doi.org/10.1016/j.ajhg.2021.03.016 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Lennerz, J. K. et al. Addition of H19 ‘loss of methylation testing’ for Beckwith-Wiedemann syndrome (BWS) increases the diagnostic yield. J. Mol. Diagn. 12, 576–588 (2010).

    Article  CAS  Google Scholar 

  26. Nurk, S. et al. The complete sequence of a human genome. Preprint at bioRxiv https://doi.org/10.1101/2021.05.26.445798 (2021).

  27. Aganezov, S. et al. A complete reference genome improves analysis of human genetic variation. Preprint at bioRxiv https://doi.org/10.1101/2021.07.12.452063 (2021).

  28. Boisson, B. et al. Rescue of recurrent deep intronic mutation underlying cell type–dependent quantitative NEMO deficiency. J. Clin. Invest. 129, 583–597 (2018).

    Article  Google Scholar 

  29. 1000 Genomes Project Consortium et al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Article  Google Scholar 

  30. Schmidt, K., Noureen, A., Kronenberg, F. & Utermann, G. Structure, function, and genetics of lipoprotein (a). J. Lipid Res. 57, 1339–1359 (2016).

    Article  CAS  Google Scholar 

  31. Li, H., Feng, X. & Chu, C. The design and construction of reference pangenome graphs with minigraph. Genome Biol. 21, 265 (2020).

    Article  Google Scholar 

  32. Shumate, A. & Salzberg, S. L. Liftoff: accurate mapping of gene annotations. Bioinform. 37, 1639–1643 (2020).

  33. Theunissen, F. et al. Structural variants may be a source of missing heritability in sALS. Front. Neurosci. 14, 47 (2020).

    Article  Google Scholar 

  34. Guo, Y. et al. Improvements and impacts of GRCh38 human reference on high throughput sequencing data analysis.Genomics 109, 83–90 (2017).

    Article  CAS  Google Scholar 

  35. Pan, B. et al. Similarities and differences between variants called with human reference genome HG19 or HG38. BMC Bioinform. 20, 101 (2019).

  36. Miller, C. A. et al. Failure to detect mutations in U2AF1 due to changes in the GRCh38 reference sequence. Preprint at bioRxiv https://doi.org/10.1101/2021.05.07.442430 (2021).

  37. Li, H. et al. Exome variant discrepancies due to reference-genome differences. Am. J. Hum. Genet. 108, 1239–1250 (2021).

    Article  CAS  Google Scholar 

  38. Collins, R. L. et al. A structural variation reference for medical and population genetics. Nature 590, E55 (2021).

    Article  CAS  Google Scholar 

  39. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinform. 26, 841–842 (2010).

  40. Li, H. Minimap2: pairwise alignment for nucleotide sequences. Bioinform. 34, 3094–3100 (2018).

  41. Krusche, P. et al. Best practices for benchmarking germline small-variant calls in human genomes. Nat. Biotechnol. 37, 555–560 (2019).

    Article  CAS  Google Scholar 

  42. Van der Auwera, G. A. & O’Connor, B. D. Genomics in the Cloud: Using Docker, GATK, and WDL in Terra (O’Reilly Media, 2020).

  43. Farek, J. et al. xAtlas: scalable small variant calling across heterogeneous next-generation sequencing experiments. Preprint at bioRxiv https://doi.org/10.1101/295071 (2018).

  44. Edge, P. & Bansal, V. Longshot enables accurate variant calling in diploid genomes from single-molecule long read sequencing. Nat. Commun. 10, 4660 (2019).

    Article  Google Scholar 

  45. Shafin, K. et al. Haplotype-aware variant calling with PEPPER-Margin-DeepVariant enables high accuracy in nanopore long-reads. Nat. Meth. 18, 1322–1332 (2021).

  46. Sahraeian, S. M. E. et al. Deep convolutional neural networks for accurate somatic mutation detection. Nat. Commun. 10, 1041 (2019).

    Article  Google Scholar 

  47. Walker, B. J. et al. Pilon: an integrated tool for comprehensive microbial variant detection and genome assembly improvement. PLoS One 9, e112963 (2014).

    Article  Google Scholar 

  48. Patterson, M. et al. WhatsHap: weighted haplotype assembly for future-generation sequencing reads. J. Comput. Biol. 6, 498–509 (2015).

  49. Zook, J. M. et al. Extensive sequencing of seven human genomes to characterize benchmark reference materials. Sci. Data 3, 160025 (2016).

    Article  CAS  Google Scholar 

  50. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at https://arxiv.org/abs/1303.3997 (2013).

  51. Regier, A. A. et al. Functional equivalence of genome sequencing analysis pipelines enables harmonized variant calling across human genetics projects.Nat. Commun. 9, 4038 (2018).

    Article  Google Scholar 

  52. Poplin, R. et al. Scaling accurate genetic variant discovery to tens of thousands of samples. Preprint at bioRxiv https://doi.org/10.1101/201178 (2018).

  53. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinform. 25, 2078–2079 (2009).

  54. Rausch, T. et al. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinform. 28, 333–339 (2012).

  55. Cameron, D. L. et al. GRIDSS: sensitive and specific genomic rearrangement detection using positional de Bruijn graph assembly. Genome Res. 27, 2050–2060 (2017).

    Article  CAS  Google Scholar 

  56. Layer, R. M., Chiang, C., Quinlan, A. R. & Hall, I. M. LUMPY: a probabilistic framework for structural variant discovery. Genome Biol. 15, R84 (2014).

    Article  Google Scholar 

  57. Chen, X. et al. Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications. Bioinform. 32, 1220–1222 (2016).

  58. Kronenberg, Z. N. et al. Wham: identifying structural variants of biological consequence. PLoS Comput. Biol. 11, e1004572 (2015).

    Article  Google Scholar 

  59. Jeffares, D. C. et al. Transient structural variations have strong effects on quantitative traits and reproductive isolation in fission yeast. Nat. Commun. 8, 14061 (2017).

    Article  CAS  Google Scholar 

  60. De Coster, W., D’Hert, S., Schultz, D. T., Cruts, M. & Van Broeckhoven, C. NanoPack: visualizing and processing long-read sequencing data. Bioinform. 34, 2666–2669 (2018).

  61. Sedlazeck, F. J. et al. Accurate detection of complex structural variations using single-molecule sequencing. Nat. Methods 15, 461–468 (2018).

    Article  CAS  Google Scholar 

  62. Jiang, T. et al. Long-read-based human genomic structural variation detection with cuteSV. Genome Biol. 21, 189 (2020).

    Article  CAS  Google Scholar 

  63. Tarasov, A., Vilella, A. J., Cuppen, E., Nijman, I. J. & Prins, P. Sambamba: fast processing of NGS alignment formats. Bioinform. 31, 2032–2034 (2015).

  64. Faust, G. G. & Hall, I. M. SAMBLASTER: fast duplicate marking and structural variant read extraction. Bioinform. 30, 2503–2505 (2014).

  65. Poplin, R. et al. A universal SNP and small-indel variant caller using deep neural networks. Nat. Biotechnol. 36, 983–987 (2018).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the Genome Reference Consortium for their curation efforts of GRCh37 and GRCh38 (https://www.genomereference.org), especially V.A. Schneider and P.A. Kitts from the National Institutes of Health (NIH)/NCBI for developing the falsely duplicated regions that should be masked in GRCh38. We thank S. Miller at NIST for helping make available benchmark sets and READMEs. Certain commercial equipment, instruments or materials are identified to adequately specify experimental conditions or reported results. Such identification does not imply recommendation or endorsement by NIST, nor does it imply that the equipment, instruments or materials identified are necessarily the best available for the purpose. C.F. was funded by Instituto de Salud Carlos III (PI20/00876) and Ministerio de Ciencia e Innovación (RTC-2017-6471-1; AEI/FEDER, UE), cofinanced by the European Regional Development Fund ‘A Way of Making Europe’ from the European Union, and Cabildo Insular de Tenerife (CGIEU0000219140). J.M.L.-S. was funded by Consejería de Educación-Gobierno de Canarias and Cabildo Insular de Tenerife (BOC 163, 24/08/2017). F.J.S. and M.M. was supported by the NIH (UM1 HG008898). C.X. was supported by the Intramural Research Program of the National Library of Medicine, NIH. K.H.M. was supported by the NIH/National Human Genome Research Institute (R01 1R01HG011274-01 and U01 1U01HG010971). H.L. was supported by the NIH (R01 HG010040 and U01 HG010961). C.E.M. thanks funding from the WorldQuant Foundation, NASA (NNX14AH50G), the National Institutes of Health (R01MH117406, R01CA249054, R01AI151059, P01CA214274) and the Leukemia and Lymphoma Society (LLS) (MCL7001-18, LLS 9238-16, LLS-MCL7001-18).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: J.W., N.D.O., A.F., K.H.M., S.E.L., M.T.W.E., H.L., C.-S.C., J.M.Z. and F.J.S. Data curation: J.W., N.D.O. and J.M. Formal analysis – benchmark: J.W., N.D.O., J.M. and J.M.Z. Formal analysis – assembly: H.C., A.S., H.L. and C.-S.C. Methodology: J.W., H.C., H.L., C.-S.C., J.M.Z. and F.J.S. Project administration: J.W., J.M.Z. and F.J.S. Resources: C.X. Software: J.W. and N.D.O. Supervision: C.-S.C., J.M.Z. and F.J.S. Validation: J.W., N.D.O., L.H., J.M., H.C., A.F., Y.-C.H., R.G., A.M.W., W.J.R., Z.M.K., J.F., Y.Z., A.P., M.M., C.X., B.Y., S.M.E.S., D.J., J.M.L.-S., A.M.-B., L.A.R.-R., C.F., G.N., U.S.E., S.E.C., J.L., H.L., C.-S.C., J.M.Z. and F.J.S. Visualization: J.W., N.D.O., H.C., H.L. and C.-S.C. Writing – original draft: J.W., L.H., C.-S.C., J.M.Z. and F.J.S. Writing – review and editing: J.W., N.D.O., D.E.M., J.L., C.E.M., S.E.L., M.T.W.E., C.-S.C., J.M.Z. and F.J.S.

Corresponding authors

Correspondence to Chen-Shan Chin, Justin M. Zook or Fritz J. Sedlazeck.

Ethics declarations

Competing interests

A.M.W. and W.J.R. are employees and shareholders of Pacific Biosciences. A.F., Y.-C.H, R.G., and C.-S.C. are employees and shareholders of DNAnexus. S.M.E.S. is an employee of Roche. J.L. is a former employee and shareholder of Bionano Genomics. S.E.L. was an employee of Invitae. F.J.S. has sponsored travel from Pacific Biosciences and Oxford Nanopore Technologies. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Adam Ameur, Christian Marshall 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.

Supplementary information

Supplementary Information

Supplementary Figures 1–17, Notes 1–5 and Table 1.

Reporting Summary.

Supplementary Data 1

Additional characteristics of high-priority clinical genes.

Supplementary Data 2

Overlaps of the 5,038 genes on GRCh38 primary assembly between both HG002 GRCh38 v4.2.1 and HG002 hifiasm v0.11.

Supplementary Data 3

Benchmarking of the hifiasm v0.11 assembly-based variants called with dipcall against the GIAB v4.2.1 benchmark for HG002.

Supplementary Data 4

Benchmarking statistics against CMRG benchmark and evaluation callsets.

Supplementary Data 5

Manual curation results for evaluation and common errors in v0.02.03 small variant benchmark.

Supplementary Data 6

Primer designs and reaction conditions for Long-Range PCR and Sanger confirmation.

Supplementary Data 7

Genes excluded from the CMRG benchmarks, with likely reasons for exclusion annotated for GRCh38 in the last column.

Supplementary Data 8

Commands for BWA-GATK variant calling on normal GRCh38 reference.

Supplementary Data 9

Commands for BWA-GATK variant calling on v1 masked GRCh38 reference.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wagner, J., Olson, N.D., Harris, L. et al. Curated variation benchmarks for challenging medically relevant autosomal genes. Nat Biotechnol 40, 672–680 (2022). https://doi.org/10.1038/s41587-021-01158-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-021-01158-1

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