1887

Abstract

A rapid and accurate diagnostic assay represents an important means to detect , identify drug-resistant strains and ensure treatment success. Currently employed techniques to diagnose drug-resistant tuberculosis include slow phenotypic tests or more rapid molecular assays that evaluate a limited range of drugs. Whole-genome-sequencing-based approaches can detect known drug-resistance-conferring mutations and novel variations; however, the dependence on growing samples in culture, and the associated delays in achieving results, represents a significant limitation. As an alternative, targeted sequencing strategies can be directly performed on clinical samples at high throughput. This study proposes a targeted sequencing assay to rapidly detect drug-resistant strains of using the Nanopore MinION sequencing platform. We designed a single-tube assay that targets nine genes associated with drug resistance to seven drugs and two phylogenetic-determining regions to determine strain lineage and tested it in nine clinical isolates and six sputa. The study’s main aim is to calibrate MinNION variant calling to detect drug-resistance-associated mutations with different frequencies to match the accuracy of Illumina (the current gold-standard sequencing technology) from both culture and sputum samples. After calibrating Nanopore MinION variant calling, we demonstrated 100% agreement between Illumina WGS and our MinION set up to detect known drug resistance and phylogenetic variants in our dataset. Importantly, other variants in the amplicons are also detected, decreasing the recall. We identify minority variants and insertions/deletions as crucial bioinformatics challenges to fully reproduce Illumina WGS results.

Funding
This study was supported by the:
  • Conselleria d'Educació, Investigació, Cultura i Esport (Award PROMETEO/2020/012)
    • Principle Award Recipient: ComasIñaki
  • Ministerio de Economía, Indústria y Competividad (Award PID2019-104477RB-I00)
    • Principle Award Recipient: ComasIñaki
  • H2020 European Research Council (Award 101001038)
    • Principle Award Recipient: ComasIñaki
  • H2020 European Research Council (Award 638553)
    • Principle Award Recipient: IñakiComas
  • This is an open-access article distributed under the terms of the Creative Commons Attribution NonCommercial License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/mgen/10.1099/mgen.0.000740
2021-12-17
2024-04-20
Loading full text...

Full text loading...

/deliver/fulltext/mgen/7/12/mgen000740.html?itemId=/content/journal/mgen/10.1099/mgen.0.000740&mimeType=html&fmt=ahah

References

  1. World Health Organization Global Tuberculosis Report 2019; 2019
  2. Hoang TTT, Nguyen NV, Dinh SN, Nguyen HB, Cobelens F et al. Challenges in detection and treatment of multidrug resistant tuberculosis patients in Vietnam. BMC Public Health 2015; 15:1–10 [View Article]
    [Google Scholar]
  3. World Health Organization Global tuberculosis control: a short update to the 2009 report. World Health Organization; 2009. Report No.: WHO/HTM/TB/2009.426; 2009
  4. Lee A, Xie YL, Barry CE, Chen RY. Current and future treatments for tuberculosis. BMJ 2020m216 [View Article]
    [Google Scholar]
  5. Marks SM, Hirsch-Moverman Y, Salcedo K, Graviss EA, Oh P et al. Characteristics and costs of multidrug-resistant tuberculosis in-patient care in the United States, 2005–2007. Int J Tuberc Lung Dis 2016; 20:435–441 [View Article]
    [Google Scholar]
  6. World Health Organization Rapid сommunication: key changes to treatment of multidrug-and rifampicin-resistant tuberculosis (MDR/RR-TB) [Internet]. World Health Organization; 2019 https://www.who.int/tb/publications/2019/WHO_RapidCommunicationMDR_TB2019.pdf
  7. Roelens M, Battista Migliori G, Rozanova L, Estill J, Campbell JR et al. Evidence-based definition for extensively drug-resistant tuberculosis. Am J Respir Crit Care Med 2021; 204:713–722 [View Article]
    [Google Scholar]
  8. Mirzayev F, Viney K, Linh NN, Gonzalez-Angulo L, Gegia M et al. World Health Organization recommendations on the treatment of drug-resistant tuberculosis, 2020 update. Eur Respir J 2021; 57:2003300 [View Article]
    [Google Scholar]
  9. Kim SJ. Drug-susceptibility testing in tuberculosis: methods and reliability of results. European Respiratory Journal 2005; 25:564–569 [View Article]
    [Google Scholar]
  10. Falkinham JO. The Mycobacterium avium complex and slowly growing mycobacteria. In Molecular Medical Microbiology Academic Press; 2015 pp 1669–1678 [View Article]
    [Google Scholar]
  11. Cancino-Muñoz I, Moreno-Molina M, Furió V, Goig GA, Torres-Puente M et al. Cryptic resistance mutations associated with misdiagnoses of multidrug-resistant tuberculosis. J Infect Dis 2019; 220:316–320 [View Article] [PubMed]
    [Google Scholar]
  12. World Health Organization The use of molecular line probe assay for the detection of resistance to isoniazid and rifampicin: policy update. World Health Organization 2016
    [Google Scholar]
  13. Mäkinen J, Marttila HJ, Marjamäki M, Viljanen MK, Soini H. Comparison of two commercially available DNA line probe assays for detection of multidrug-resistant Mycobacterium tuberculosis. J Clin Microbiol 2006; 44:350–352 [View Article] [PubMed]
    [Google Scholar]
  14. Bai Y, Wang Y, Shao C, Hao Y, Jin Y et al. GenoType MTBDRplus assay for rapid detection of multidrug resistance in Mycobacterium tuberculosis: a meta-analysis. PLoS ONE 2016; 11:e0150321 [View Article]
    [Google Scholar]
  15. Ninan MM, Gowri M, Christopher DJ, Rupali P, Michael JS. The diagnostic utility of line probe assays for multidrug-resistant tuberculosis. Pathog Glob Health 2016; 110:194–199 [View Article] [PubMed]
    [Google Scholar]
  16. Osei Sekyere J, Maphalala N, Malinga LA, Mbelle NM, Maningi NE. A comparative evaluation of the new genexpert MTB/RIF ultra and other rapid diagnostic assays for detecting tuberculosis in pulmonary and extra pulmonary specimens. Sci Rep 2019; 9:16587. [View Article] [PubMed]
    [Google Scholar]
  17. Bunsow E, Ruiz-Serrano MJ, López Roa P, Kestler M, Viedma DG et al. Evaluation of GeneXpert MTB/RIF for the detection of Mycobacterium tuberculosis and resistance to rifampin in clinical specimens. J Infect 2014; 68:338–343 [View Article] [PubMed]
    [Google Scholar]
  18. Sanchez-Padilla E, Merker M, Beckert P, Jochims F, Dlamini T et al. Detection of drug-resistant tuberculosis by Xpert MTB/RIF in Swaziland. N Engl J Med 2015; 372:1181–1182 [View Article] [PubMed]
    [Google Scholar]
  19. Goig GA, Cancino-Muñoz I, Torres-Puente M, Villamayor LM, Navarro D et al. Whole-genome sequencing of Mycobacterium tuberculosis directly from clinical samples for high-resolution genomic epidemiology and drug resistance surveillance: an observational study. The Lancet Microbe 2020; 1:e175–e183 [View Article]
    [Google Scholar]
  20. Jouet A, Gaudin C, Badalato N, Allix-Béguec C, Duthoy S et al. Deep amplicon sequencing for culture-free prediction of susceptibility or resistance to 13 anti-tuberculous drugs. Eur Respir J 2021; 57:2002338 [View Article]
    [Google Scholar]
  21. López B, Aguilar D, Orozco H, Burger M, Espitia C et al. A marked difference in pathogenesis and immune response induced by different Mycobacterium tuberculosis genotypes. Clin Exp Immunol 2003; 133:30–37 [View Article]
    [Google Scholar]
  22. Ford CB, Shah RR, Maeda MK, Gagneux S, Murray MB et al. Mycobacterium tuberculosis mutation rate estimates from different lineages predict substantial differences in the emergence of drug-resistant tuberculosis. Nat Genet 2013; 45:784–790 [View Article] [PubMed]
    [Google Scholar]
  23. Colman RE, Schupp JM, Hicks ND, Smith DE, Buchhagen JL et al. Detection of low-level mixed-population drug resistance in Mycobacterium tuberculosis using high fidelity amplicon sequencing. PLoS One 2015; 10:e0126626 [View Article] [PubMed]
    [Google Scholar]
  24. Rinder H, Mieskes KT, Löscher T. Heteroresistance in Mycobacterium tuberculosis. Int J Tuberc Lung Dis 2001; 5:339–345 [PubMed]
    [Google Scholar]
  25. Magi A, Semeraro R, Mingrino A, Giusti B, D’Aurizio R. Nanopore sequencing data analysis: state of the art, applications and challenges. Brief Bioinform 2018; 19:1256–1272 [View Article] [PubMed]
    [Google Scholar]
  26. Xu Y, Lewandowski K, Lumley S, Pullan S, Vipond R et al. Detection of viral pathogens with multiplex nanopore MinION sequencing: be careful with cross-talk. Front Microbiol 2018; 9:2225. [View Article] [PubMed]
    [Google Scholar]
  27. Schmidt K, Mwaigwisya S, Crossman LC, Doumith M, Munroe D et al. Identification of bacterial pathogens and antimicrobial resistance directly from clinical urines by nanopore-based metagenomic sequencing. J Antimicrob Chemother 2017; 72:104–114 [View Article] [PubMed]
    [Google Scholar]
  28. Bainomugisa A, Duarte T, Lavu E, Pandey S, Coulter C et al. A complete high-quality MinION nanopore assembly of an extensively drug-resistant Mycobacterium tuberculosis Beijing lineage strain identifies novel variation in repetitive PE/PPE gene regions. Microbial Genomics 2018; 4: [View Article]
    [Google Scholar]
  29. Public health teams in Madagascar pioneer use of portable, real time DNA sequencing to fight drug-resistant tuberculosis (TB) [Internet]; 2018 http://nanoporetech.com/about-us/news/public-health-teams-madagascar-pioneer-use-portable-real-time-dna-sequencing-fight
  30. Tyler AD, Mataseje L, Urfano CJ, Schmidt L, Antonation KS et al. Evaluation of Oxford Nanopore’s MinION sequencing device for microbial whole genome sequencing applications. Sci Rep 2018; 8:10931. [View Article] [PubMed]
    [Google Scholar]
  31. Votintseva AA, Bradley P, Pankhurst L, Del Ojo Elias C, Loose M et al. Same-day diagnostic and surveillance data for tuberculosis via whole-genome sequencing of direct respiratory samples. J Clin Microbiol 2017; 55:1285–1298 [View Article] [PubMed]
    [Google Scholar]
  32. Somerville W, Thibert L, Schwartzman K, Behr MA. Extraction of Mycobacterium tuberculosis DNA: a question of containment. J Clin Microbiol 2005; 43:2996–2997 [View Article] [PubMed]
    [Google Scholar]
  33. Cancino-Muñoz I, Gil-Brusola A, Torres-Puente M, Mariner-Llicer C, Dogba J et al. Development and application of affordable SNP typing approaches to genotype Mycobacterium tuberculosis complex strains in low and high burden countries. Sci Rep 2019; 9:15343. [View Article] [PubMed]
    [Google Scholar]
  34. Wick RR, Judd LM, Holt KE. Performance of neural network basecalling tools for Oxford Nanopore sequencing. Genome Biol 2019; 20:129. [View Article] [PubMed]
    [Google Scholar]
  35. Li H. Minimap2: pairwise alignment for nucleotide sequences. Bioinformatics 2018; 34:3094–3100 [View Article] [PubMed]
    [Google Scholar]
  36. Comas I, Chakravartti J, Small PM, Galagan J, Niemann S et al. Human T cell epitopes of Mycobacterium tuberculosis are evolutionarily hyperconserved. Nat Genet 2010; 42:498–503 [View Article] [PubMed]
    [Google Scholar]
  37. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 2009; 25:2078–2079 [View Article] [PubMed]
    [Google Scholar]
  38. Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res 2012; 22:568–576 [View Article] [PubMed]
    [Google Scholar]
  39. Wood DE, Salzberg SL. Kraken: ultrafast metagenomic sequence classification using exact alignments. Genome Biol 2014; 15:R46. [View Article] [PubMed]
    [Google Scholar]
  40. Comas I, Chakravartti J, Small PM, Galagan J, Niemann S et al. Human T cell epitopes of Mycobacterium tuberculosis are evolutionarily hyperconserved. Nat Genet 2010; 42:498–503 [View Article] [PubMed]
    [Google Scholar]
  41. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 2009; 25:1754–1760 [View Article] [PubMed]
    [Google Scholar]
  42. Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res 2012; 22:568–576 [View Article] [PubMed]
    [Google Scholar]
  43. Feuerriegel S, Schleusener V, Beckert P, Kohl TA, Miotto P et al. PhyResSE: a web tool delineating Mycobacterium tuberculosis antibiotic resistance and lineage from whole-genome sequencing data. J Clin Microbiol 2015; 53:1908–1914 [View Article] [PubMed]
    [Google Scholar]
  44. Minervini CF, Cumbo C, Orsini P, Anelli L, Zagaria A et al. Nanopore sequencing in blood diseases: a wide range of opportunities. Front Genet 2020; 11: [View Article]
    [Google Scholar]
  45. Orsini P, Minervini CF, Cumbo C, Anelli L, Zagaria A et al. Design and MinION testing of a nanopore targeted gene sequencing panel for chronic lymphocytic leukemia. Sci Rep 2018; 8:11798. [View Article] [PubMed]
    [Google Scholar]
  46. Ammar R, Paton TA, Torti D, Shlien A, Bader GD. Long read nanopore sequencing for detection of HLA and CYP2D6 variants and haplotypes. F1000Res 2015; 4:17 [View Article] [PubMed]
    [Google Scholar]
  47. Tafess K, Ng TTL, Lao HY, Leung KSS, KKG T et al. Targeted sequencing workflows for comprehensive drug resistance profiling of Mycobacterium tuberculosis cultures using Illumina MiSeq and Nanopore MinION: Comparison of analytical and diagnostic performance, turnaround time and cost [Internet]. bioRxiv 2019760462
    [Google Scholar]
  48. World Health Organization Meeting report of the WHO expert consultation on the definition of extensively drug-resistant tuberculosis, 27-29 October 2020, Geneva; 2021 https://apps.who.int/iris/bitstream/handle/10665/338776/9789240018662-eng.pdf
  49. Greig DR, Jenkins C, Gharbia S, Dallman TJ. Comparison of single-nucleotide variants identified by Illumina and Oxford Nanopore technologies in the context of a potential outbreak of Shiga toxin–producing Escherichia coli. GigaScience 2019; 8: [View Article]
    [Google Scholar]
  50. Tafess K, Ng TTL, Lao HY, Leung KSS, KKG T et al. Targeted-sequencing workflows for comprehensive drug resistance profiling of Mycobacterium tuberculosis cultures using two commercial sequencing platforms: comparison of analytical and diagnostic performance, turnaround time, and cost. Clin Chem 2020; 66:809–820
    [Google Scholar]
  51. Cabibbe AM, Spitaleri A, Battaglia S, Colman RE, Suresh A et al. Application of targeted next-generation sequencing assay on a portable sequencing platform for culture-free detection of drug-resistant tuberculosis from clinical samples. J Clin Microbiol 2020; 58: [View Article]
    [Google Scholar]
  52. Chan WS, Au CH, Chung Y, Leung HCM, Ho DN et al. Rapid and economical drug resistance profiling with Nanopore MinION for clinical specimens with low bacillary burden of Mycobacterium tuberculosis. BMC Res Notes 2020; 13:444. [View Article] [PubMed]
    [Google Scholar]
  53. Vargas R, Freschi L, Marin M, Epperson LE, Smith M et al. In-host population dynamics of complex during active disease. Elife [Internet] 2021; 10:
    [Google Scholar]
  54. Rigouts L, Miotto P, Schats M, Lempens P, Cabibbe AM et al. Fluoroquinolone heteroresistance in Mycobacterium tuberculosis: detection by genotypic and phenotypic assays in experimentally mixed populations. Sci Rep 2019; 9:11760 [View Article]
    [Google Scholar]
  55. Karst SM, Ziels RM, Kirkegaard RH, Sørensen EA, McDonald D et al. High-accuracy long-read amplicon sequences using unique molecular identifiers with Nanopore or PacBio sequencing. Nat Methods 2021; 18:165–169 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/mgen/10.1099/mgen.0.000740
Loading
/content/journal/mgen/10.1099/mgen.0.000740
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error