Laboratory methods supporting measles surveillance in Queensland, Australia, 2010–2017 Open Access

Abstract

Australia was officially recognised as having eliminated endemic measles transmission in 2014. Maintaining laboratory support for surveillance of vaccine-preventable diseases, such as measles, is an essential component of reaching and maintaining transmission-free status.

Real-time and conventional PCR-based tools were used to detect, differentiate from measles vaccine virus (MeVV), and sequence fragments of measles viruses (MeV) identified from specimens collected in Queensland. Specimens were mostly from travellers who had visited or returned to Queensland from international or interstate sites or been in contact with a case from either group.

Between 2010 and 2017, 13 678 specimens were tested in our laboratory using real-time RT-PCR (RT-rPCR), identifying 533 positives. Most specimens were swabs (70.98 %) and urines (25.56 %). A MeVV RT-rPCR was used on request and identified 154 instances of MeVV. MeV-positive extracts were genotyped as required. Genotypes identified among sequenced specimens included B3, D4, D8, D9, G3, and H1 as well as members of clade A as expected from the detection of MeV among virus introductions due to global travel and vaccination.

We describe the workflow employed and results from our laboratory between 2010 and 2017 for the sensitive detection of MeV infection, supporting high-quality surveillance to ensure the maintenance of Australia’s measles-free status.

Loading

Article metrics loading...

/content/journal/acmi/10.1099/acmi.0.000093
2020-01-27
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/acmi/2/3/acmi000093.html?itemId=/content/journal/acmi/10.1099/acmi.0.000093&mimeType=html&fmt=ahah

References

  1. Wang L-F, Collins PLF RAM, Kurath G, Lamb RA, Randall RE et al. Paramyxoviridae: International Committee on taxonomy of viruses (ICTV).
  2. Benamar T, Tajounte L, Alla A, Khebba F, Ahmed H et al. Real-Time PCR for measles virus detection on clinical specimens with negative IgM result in Morocco. PLoS One 2016; 11:e0147154 [View Article][PubMed]
    [Google Scholar]
  3. Thomas B, Beard S, Jin L, Brown KE, Brown DWG. Development and evaluation of a real-time PCR assay for rapid identification and semi-quantitation of measles virus. J Med Virol 2007; 79:1587–1592 [View Article][PubMed]
    [Google Scholar]
  4. Sanz JC, Mosquera M, Ramos B, Ramírez R, de Ory F et al. Assessment of RNA amplification by multiplex RT-PCR and IgM detection by indirect and capture ELISAs for the diagnosis of measles and rubella. APMIS 2010; 118:203–209 [View Article][PubMed]
    [Google Scholar]
  5. Michel Y, Saloum K, Tournier C, Quinet B, Lassel L et al. Rapid molecular diagnosis of measles virus infection in an epidemic setting. J Med Virol 2013; 85:723–730 [View Article][PubMed]
    [Google Scholar]
  6. Lo NC, Hotez PJ. Public health and economic consequences of vaccine Hesitancy for measles in the United States. JAMA Pediatr 2017; 171:887–892 [View Article][PubMed]
    [Google Scholar]
  7. Gidding HF, Martin NV, Stambos V, Tran T, Dey A et al. Verification of measles elimination in Australia: application of World Health organization regional guidelines. J Epidemiol Glob Health 2016; 6:197–209 [View Article][PubMed]
    [Google Scholar]
  8. Moss WJ, Measles MWJ. Measles. Lancet 2017; 390:2490–2502 [View Article][PubMed]
    [Google Scholar]
  9. Rota PA, Brown K, Mankertz A, Santibanez S, Shulga S et al. Global distribution of measles genotypes and measles molecular epidemiology. J Infect Dis 2011; 204:S514–S523 [View Article][PubMed]
    [Google Scholar]
  10. World Health Organization The role of extended and whole genome sequencing for tracking transmission of measles and rubella viruses: report from the global measles and rubella laboratory network meeting, 2017. Wkly Epidemiol Rec 2018; 93:55–59[PubMed]
    [Google Scholar]
  11. World Health Organization Measles virus nomenclature update: 2012. Wkly Epidemiol Rec 2012; 87:73–81[PubMed]
    [Google Scholar]
  12. Rota PA, Featherstone D, Bellini DJ. Molecular epidemiology of measles virus. in: Griffin de, Oldstone MM, editors. measles pathogenesis and control introduction. Curr Top Microbiol Immunol 2009; 330:
    [Google Scholar]
  13. Muñoz-Alía Miguel Ángel, Muller CP, Russell SJ. Antigenic drift defines a new D4 subgenotype of measles virus. J Virol 2017; 91: [View Article][PubMed]
    [Google Scholar]
  14. Brown KE, Rota PA, Goodson JL, Williams D, Abernathy E et al. Genetic characterization of measles and rubella viruses detected through global measles and rubella elimination surveillance, 2016-2018. MMWR Morb Mortal Wkly Rep 2019; 68:587–591 [View Article][PubMed]
    [Google Scholar]
  15. McMahon J, Mackay IM, Lambert IM. Measles vaccine virus RNA in children more than 100 days after vaccination. Viruses 2019; 11:636 [View Article][PubMed]
    [Google Scholar]
  16. Finger M, Lyon M, Northill J, Mackay IM. Measles virus TaqMan RT-PCR (F gene)2019.
  17. Finger M, Lyon M, Northill J et al. Measles vaccine virus Taqman-MGB2019.
  18. Smith G, Smith I, Harrower B, Warrilow D, Bletchly C. A simple method for preparing synthetic controls for conventional and real-time PCR for the identification of endemic and exotic disease agents. J Virol Methods 2006; 135:229–234 [View Article][PubMed]
    [Google Scholar]
  19. Mackay IM, Northil J. Binary synthetic template oligonucleotide positive control for in-house diagnostic real-time RT-PCR 2019. https://www.protocols.io/view/binary-synthetic-template-oligonucleotide-positive-yxjfxkn
  20. Chibo D, Birch CJ, Rota PA, Catton MG. Molecular characterization of measles viruses isolated in Victoria, Australia, between 1973 and 1998. J Gen Virol 2000; 81:2511–2518 [View Article][PubMed]
    [Google Scholar]
  21. Kearse M, Moir R, Wilson A, Stones-Havas S, Cheung M et al. Geneious basic: an integrated and extendable desktop software platform for the organization and analysis of sequence data. Bioinformatics 2012; 28:1647–1649 [View Article][PubMed]
    [Google Scholar]
  22. Kumar S, Stecher G, Tamura K. MEGA7: molecular evolutionary genetics analysis version 7.0 for bigger datasets. Mol Biol Evol 2016; 33:1870–1874 [View Article][PubMed]
    [Google Scholar]
  23. Woudenberg T, van Binnendijk RS, Sanders EAM, Wallinga J, de Melker HE et al. Large measles epidemic in the Netherlands, may 2013 to March 2014: changing epidemiology. Euro Surveill 2017; 22:30443 [View Article][PubMed]
    [Google Scholar]
  24. World Health Organization Third Annual Meeting of the Regional Verification Commission for Measles Elimination in the Western Pacific, Seoul, Republic of Korea, 18-21 March 2014: report2014 14/11/2018. http://iris.wpro.who.int/handle/10665.1/10675
  25. Saitou N, Nei M. The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol Biol Evol 1987; 4:406–425 [View Article][PubMed]
    [Google Scholar]
  26. Tamura K, Nei M, Kumar S. Prospects for inferring very large phylogenies by using the neighbor-joining method. Proc Natl Acad Sci U S A 2004; 101:11030–11035 [View Article][PubMed]
    [Google Scholar]
  27. Kumar S, Tamura K, Nei M. MEGA3: integrated software for molecular evolutionary genetics analysis and sequence alignment. Brief Bioinform 2004; 5:150–163 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/acmi/10.1099/acmi.0.000093
Loading
/content/journal/acmi/10.1099/acmi.0.000093
Loading

Data & Media loading...

Most cited Most Cited RSS feed