Skip to main content
  • Brief Report
  • Open access
  • Published:

Comparison of multi-parallel quantitative real-time PCRs targeting different DNA regions and detecting soil-transmitted helminths in stool

Abstract

Background

Soil-transmitted helminths infect an estimated 18% of the world’s population, causing a significant health burden. Microscopy has been the primary tool for diagnosing eggs from fecal samples, but its sensitivity drops in low-prevalence settings. Quantitative real-time polymerase chain reaction (qPCR) is slowly increasing in research and clinical settings. However, there is still no consensus on preferred qPCR targets.

Methods

We aimed to compare soil-transmitted helminth (STH) DNA detection methods by testing naïve stool samples spiked with known quantities of STH eggs and larvae. DNA extracts from spiked samples were tested using independent quantitative realtime PCR (qPCR) assays targeting ribosomal or putative non-protein coding satellite sequences.

Results

For Trichuris trichiura, there was a strong correlation between egg/larvae counts and qPCR results using either qPCR method (0.86 and 0.87, respectively). Strong correlations also existed for A. lumbricoides (0.60 and 0.63, respectively), but weaker correlations were found for Ancylostoma duodenale (0.41 for both assays) and Strongyloides stercoralis (0.48 and 0.65, respectively). No correlation for Necator americanus was observed when testing with either qPCR assay. Both assays had fair-to-moderate agreement across targets when using field-collected stool samples (0.28–0.45, for all STHs), except for S. stercoralis (0.12) with slight agreement.

Conclusions

There is a strong correlation between qPCR results and egg/larvae counts. Our study confirms that qPCR is an effective diagnostic tool, even with low-intensity infections, regardless of the DNA-based diagnostic marker used. However, the moderate agreement between the two different qPCR assays when testing field samples highlights the need to understand the role of these targets in the genome so that the parasite burden can be quantified more accurately and consistently by qPCR.

Graphical abstract

Background

Soil-transmitted helminths (STHs) (Ascaris lumbricoides, Trichuris trichiura, Necator americanus, Ancylostoma duodenale) and Strongyloides stercoralis infect more than 1.4 billion people worldwide [1], resulting in years of disability and extensive morbidity. The most widely used techniques for diagnosing STH infections are microscopy-based, despite the repeated demonstration of their shortcomings [2]. Microscopy is still recommended by the World Health Organization (WHO) for use in epidemiological interventions and monitoring progress in deworming programs [3]. Advances in molecular testing have spurred interest in developing new tools for monitoring STH infections, such as assays utilizing real-time polymerase chain reaction (qPCR) [4,5,6]. There is a need for more sensitive tools to complement WHO efforts to monitor the elimination of STHs [7]. Intervention success and impact assessment are highly reliant upon sensitive and accurate diagnostic tools for STH detection. However, attempts to evaluate molecular methods remain incomplete, including assessing one or more microscopic techniques against molecular methods [8,9,10]. Different real-time PCR assays targeting various DNA regions [ribosomal internal transcribed spacer sequences (ITS), ribosomal subunit sequences, or mitochondrial genes] have been developed to detect STHs [11]. Assays targeting mitochondrial and ribosomal sequences leverage their relatively high copy numbers, providing moderate-to-high sensitivity for real-time PCR. Ribosomal assays have been validated in numerous studies and clinical settings for STH detection [6, 8, 12, 13]. However, ribosomal targets tend to be conserved between species and present in lower copy numbers than other genome repeats. Given their conservation, they are frequently less specific than targets designed from other repeat types. Other nuclear tandemly arranged repeats can reach up to 37% of the parasite’s genome in certain species of STHs [14]. The development of new bioinformatics tools [15] and the assembly of improved genomes for the various STHs [16] has facilitated the exploration of the function of these target sequences and the genetic variation. Novel, optimized assays targeting highly repetitive elements in a parasite’s genome have the potential to reduce time and cost through high throughput automation [14]. Such targets have enhanced the sensitivity and specificity of qPCR assays, allowing for the differentiation of closely related species and facilitating target detection at copy numbers below those found within a single egg [5, 17].

This study assesses the agreement when two different molecular assays utilize different target sequences and compares results across a panel of samples spiked with known quantities of STH eggs or larvae. Similar testing was also performed on a panel of field-collected samples to assess the transferability of results.

Methods

Spiked sample preparation

Known numbers of parasitic eggs (1, 2, 5, 10, 15, 20, 40 egg or larvae) were used to spike 10 mg samples of naïve stool at Baylor College of Medicine (BCM), followed by DNA extraction using the FastDNA Spin Kit for Soil (MP Biomedicals, Santa Ana, CA) and a high-speed homogenizer (FastPrep-24, MP Biomedicals). In total, 19 samples containing A. lumbricoides eggs were created, as were 20 containing T. trichiura eggs, 24 containing S. stercoralis larvae, and 10 containing hookworm eggs. Details on the number of replicates per egg/larvae quantity used for spiking can be found in Supplementary Table S1. Aliquots of the same DNA extracts were shipped from the BCM to the Natural History Museum (NHM) for testing.

qPCR testing

Two independent laboratories, BCM and NHM, tested aliquots of the same DNA extracts. The NHM assay was initially developed at Smith College in Northampton, MA, USA [5], and the BCM assay was initially developed at the National Institutes of Health (NIH) in Bethesda, MD, USA [6]. The assays used for testing target repetitive genomic elements, except for the A. lumbricoides assay, which targets the internal transcribed spacer 1 (ITS1) region. The assays used at BCM all target ribosomal genes (ITS1 for A. lumbricoides, ITS1 for T. trichiura, 18S for S. stercoralis, ITS2 for N. americanus, and ITS2 for A. duodenale).

Field sample testing

A panel of 130 samples was collected as part of ongoing field studies in Orán, Argentina (approved by the bioethics committee of Colegio de Médicos de la Provincia de Salta and the IRBs of BCM; protocol number H-34926). All samples underwent direct smear stool microscopy and were frozen, without preservatives, until 50 mg of each sample was subjected to DNA extraction. Aliquots were sent to BCM and NHM for qPCR analysis using the above assays.

Statistical analysis

The correlation between target concentration (fg/µl or copies/µl, for the BCM assays and the NHM assays, respectively) and spiked egg numbers was assessed by the Kendall rank correlation test [18]. Correlations were visualized in R v.4.2.2; N. americanus was excluded from the graphs due to too few data points, but the correlation values are still presented in Table 1. P-values < 0.05 were considered statistically significant. Comparisons between the two qPCRs were depicted using unweighted Cohen’s kappa agreement [19]. Fleiss kappa was calculated to evaluate the agreement between microscopy and the two qPCR tests (three raters), treating results as categorical values (presence/absence).

Table 1 Kendall correlation, as Tau-b value and respective P-values, between the number of larvae/eggs and the quantitative qPCR method for each soil-transmitted helminth for NHM and BCM assays

Results

Concordance between qPCR and egg/larvae counts

The Kendall Tau-b values for the NHM and BCM assays were 0.86 and 0.87 for T. trichiura and 0.60 and 0.63 for A. lumbricoides, indicating strong concordance between DNA quantity measured using qPCR and egg numbers as determined by microscopy for both STHs. Using both assays, the Tau-b values for A. duodenale (0.41 for both, but not significant) and S. stercoralis (0.48 and 0.65, respectively) were less strong but still significant. With 0 and −0.816 Tau-b values for the NHM and BCM assays, respectively, results for N. americanus are probably due to ineffective extraction or insufficient eggs/larvae, making it difficult to draw any conclusions (Table 1). The graphs in Additional File 1: Fig. S1. also show the linearity and correlation between the qPCR quantitative method and eggs or larvae spiked (N. americanus graph not shown).

Two- and three-rater agreement on field samples

We calculated the overall percentage agreement (total number of agreed positives or negatives in a given sample set). Cohen’s kappa for agreement on a sample-by-sample case, for both sets of qPCR assays, treats data as categorical values (presence/absence) since the qPCR output interpretation (i.e., quantitation) still does not correspond directly to worm burden or worm intensity [9, 20]. Fleiss kappa was calculated to show the greater discordance between microscopy and both qPCR assays; the results are presented in Table 2. Between the two sets of qPCR assays, there was a moderate agreement for A. lumbricoides (kappa = 0.43) and fair agreement for N. americanus (kappa = 0.33), T. trichiura (kappa value = 0.366), and A. duodenale (kappa value = 0.28). However, both assays showed a slight agreement for S. stercoralis (kappa value = 0.121). As expected, Fleiss kappa showed weak agreement between microscopy (ranging from 0.06 to 0.22) and qPCR assays (Table 2).

Table 2 Performance comparison between (i) NHM and BMC qPCR methods and (ii) between the two qPCRs and microscopy in field-collected stool samples

Discussion

We present a comparative study evaluating two independent qPCR assay platforms for four STH species and S. stercoralis using laboratory-spiked and field-collected samples. We showed concordance and moderate-to-strong correlation between the presence of helminth eggs or larvae and the amount of parasite DNA. A strong correlation between spiked eggs and qPCR output has been demonstrated previously in similar settings [21]. Greater discordance between both qPCR platforms and microscopy illustrates the superior sensitivity (true positives) and specificity (true negatives) of the molecular methods compared with coprological tests. This comparative study demonstrates the benefits of qPCR when STH prevalence and intensity are low in a population. Molecular assays can be the foundation for reliable diagnostic results, irrespective of the target used, and opportunities for technological transfer, even to resource-limited areas, are expanding [22, 23]. We acknowledge that the number of spiked samples (eggs and larvae) used in this study was limited, requiring further scaling. This could partially explain the poor correlation observed for A. duodenale. The poor concordance between larval counts and qPCR for S. stercoralis could be explained by the low sensitivity of qPCR for Strongyloides [24]. We focused solely on spiking stool samples with small numbers of eggs/larvae, as the correlation between larger egg counts and qPCR in field-collected stool samples has already been demonstrated [7, 9, 25]. Another limitation was that an extraction control to check the efficacy of the DNA extraction method was not available at the time of this study. Suboptimal extraction of the samples spiked with N. americanus could explain the 0 and negative correlations for both N. americanus-targeting assays. Although DNA extraction products were sent to each institution, false negatives will impact both assays. One downside of using repeat-based assays is that the copy number of highly repetitive sequences can vary significantly between individual organisms and even within different stages of the same organism’s life cycle. However, larger monomer repeats might be more consistent within species [14, 26]. Further and future genomic work will highlight structural and geographical differences in repeat-based diagnostics. Currently, the full function of these nuclear repeats is mainly unknown. As a result, we are unsure how random mutations affect these areas and whether this variability can lead to bias in the quantification of STHs. These mutations make it challenging to compare results between samples accurately. Another limitation was the lack of egg/larvae counts from stool microscopy. However, previous studies have shown that the amount of egg burden directly correlates with detecting parasite DNA with qPCR in field-collected stool samples [6, 9].

Conclusions

In summary, we present further evidence that qPCR is a valid alternative to fecal microscopy for the detection of STH, and our study supports current attempts [7] toward replacing coprological tools to assess low-intensity STH infections using qPCR. Results highlight assay-specific strengths and weaknesses. This study represents the first comparison of two distinct qPCR platforms with unique molecular targets. The study involved samples spiked with known quantities of STH eggs or larvae. The findings provide a framework for better understanding what constitutes a reliable diagnostic target. Developing standardized, accurate, and quality-controlled measures is crucial to successfully testing for STHs in fecal samples using qPCR. Standardization can be achieved through an External Quality Assessment Scheme (EQAS) program [27], which ensures consistency and diagnostic precision.

Availability of data and materials

Data availability declaration in the manuscript.

References

  1. Jourdan PM, Lamberton PHL, Fenwick A, Addiss DG. Soil-transmitted helminth infections. Lancet. 2018;391:252–65.

    Article  PubMed  Google Scholar 

  2. Nikolay B, Brooker SJ, Pullan RL. Sensitivity of diagnostic tests for human soil-transmitted helminth infections: a meta-analysis in the absence of a true gold standard. Int J Parasitol. 2014;44:765–74.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Montresor A, Porta N, Albonico M, Gabrielli AF, Jankovic D, Fitzpatrick C, Vercruysse J, Levecke B. Soil-transmitted helminthiasis: the relationship between prevalence and classes of intensity of infection. Trans R Soc Trop Med Hyg. 2015;109:262–7.

    Article  PubMed  Google Scholar 

  4. Autier B, Gangneux J-P, Robert-Gangneux F. Evaluation of the AllplexTM GI-Helminth(I) Assay, the first marketed multiplex PCR for helminth diagnosis. Parasite. 2021;28:33.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Pilotte N, Papaiakovou M, Grant JR, Bierwert LA, Llewellyn S, McCarthy JS, Williams SA. Improved PCR-based detection of soil transmitted helminth infections using a Next-Generation Sequencing approach to assay design. PLoS Negl Trop Dis. 2016;10:e0004578.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Mejia R, Vicuña Y, Broncano N, Sandoval C, Vaca M, Chico M, Cooper PJ, Nutman TB. A novel, multi-parallel, real-time polymerase chain reaction approach for eight gastrointestinal parasites provides improved diagnostic capabilities to resource-limited at-risk populations. Am J Trop Med Hyg. 2013;88:1041–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Papaiakovou M, Littlewood DTJ, Gasser RB, Anderson RM. How qPCR complements the WHO roadmap (2021–2030) for soil-transmitted helminths. Trends Parasitol. 2021;37:698–708.

    Article  CAS  PubMed  Google Scholar 

  8. Cimino RO, Jeun R, Juarez M, Cajal PS, Vargas P, Echazú A, Bryan PE, Nasser J, Krolewiecki A, Mejia R. Identification of human intestinal parasites affecting an asymptomatic peri-urban Argentinian population using multi-parallel quantitative real-time polymerase chain reaction. Parasit Vectors. 2015;8:380.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Dunn JC, Papaiakovou M, Han KT, Chooneea D, Bettis AA, Wyine NY, Lwin AMM, Maung NS, Misra R, Littlewood DTJ, Anderson RM. The increased sensitivity of qPCR in comparison to Kato-Katz is required for the accurate assessment of the prevalence of soil-transmitted helminth infection in settings that have received multiple rounds of mass drug administration. Parasit Vectors. 2020;13:324.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Levecke B, Cools P, Albonico M, Ame S, Angebault C, Ayana M, et al. Identifying thresholds for classifying moderate-to-heavy soil-transmitted helminth intensity infections for FECPAKG2, McMaster, Mini-FLOTAC and qPCR. PLoS Negl Trop Dis. 2020;14:e0008296.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. O’Connell EM, Nutman TB. Molecular diagnostics for soil-transmitted helminths. Am J Trop Med Hyg. 2016;95:508–13.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Kaisar MMM, Brienen EAT, Djuardi Y, Sartono E, Yazdanbakhsh M, Verweij JJ, Supali T, van Lieshout L. Improved diagnosis of Trichuris trichiura by using a bead-beating procedure on ethanol preserved stool samples prior to DNA isolation and the performance of multiplex real-time PCR for intestinal parasites. Parasitology. 2017;144:965–74.

    Article  CAS  PubMed  Google Scholar 

  13. Verweij JJ. Application of PCR-based methods for diagnosis of intestinal parasitic infections in the clinical laboratory. Parasitology. 2014;141:1863–72.

    Article  PubMed  Google Scholar 

  14. Grant JR, Pilotte N, Williams SA. A case for using genomics and a bioinformatics pipeline to develop sensitive and species-specific PCR-based diagnostics for soil-transmitted helminths. Front Genet. 2019;10:883.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Novák P, Neumann P, Macas J. Global analysis of repetitive DNA from unassembled sequence reads using RepeatExplorer2. Nat Protoc. 2020;15:3745–76.

    Article  PubMed  Google Scholar 

  16. Doyle SR. Improving helminth genome resources in the post-genomic era. Trends Parasitol. 2022;38:831–40.

    Article  CAS  PubMed  Google Scholar 

  17. Pilotte N, Maasch JRMA, Easton AV, Dahlstrom E, Nutman TB, Williams SA. Targeting a highly repeated germline DNA sequence for improved real-time PCR-based detection of Ascaris infection in human stool. PLoS Negl Trop Dis. 2019;13:e0007593.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ng SK, Holden L, Sun J. Identifying comorbidity patterns of health conditions via cluster analysis of pairwise concordance statistics. Stat Med. 2012;31:3393–405.

    Article  PubMed  Google Scholar 

  19. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics. 1977;33:159–74.

    Article  CAS  PubMed  Google Scholar 

  20. Papaiakovou M, Gasser RB, Littlewood DTJ. Quantitative PCR-based diagnosis of soil-transmitted helminth infections: Faecal or Fickle? Trends Parasitol. 2019;35:491–500.

    Article  CAS  PubMed  Google Scholar 

  21. Zendejas-Heredia PA, Colella V, Hii SF, Traub RJ. Comparison of the egg recovery rates and limit of detection for soil-transmitted helminths using the Kato-Katz thick smear, faecal flotation and quantitative real-time PCR in human stool. PLoS Negl Trop Dis. 2021;15:e0009395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Carter JY. External quality assessment in resource-limited countries. Biochem Med. 2017;27:97–109.

    Article  Google Scholar 

  23. Cantera JL, White HN, Forrest MS, Stringer OW, Belizario VY Jr, Storey HL, de Hostos EL, de los Santos T. Sensitive and semiquantitative detection of soil-transmitted helminth infection in stool using a recombinase polymerase amplification-based assay. PLoS Negl Trop Dis. 2021;15:e0009782.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Knopp S, Salim N, Schindler T, Karagiannis Voules DA, Rothen J, Lweno O, Mohammed AS, Singo R, Benninghoff M, Nsojo AA, Genton B, Daubenberger C. Diagnostic accuracy of Kato-Katz, FLOTAC, Baermann, and PCR methods for the detection of light-intensity hookworm and Strongyloides stercoralis infections in Tanzania. Am J Trop Med Hyg. 2014;90:535–45.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Easton AV, Oliveira RG, O’Connell EM, Kepha S, Mwandawiro CS, Njenga SM, Kihara JH, Mwatele C, Odiere MR, Brooker SJ, Webster JP, Anderson RM, Nutman TB. Multi-parallel qPCR provides increased sensitivity and diagnostic breadth for gastrointestinal parasites of humans: field-based inferences on the impact of mass deworming. Parasit Vectors. 2016;9:38.

    Article  PubMed  PubMed Central  Google Scholar 

  26. López-Flores I, Garrido-Ramos MA. The repetitive DNA content of eukaryotic genomes. Genome Dyn. 2012;7:1–28.

    Article  PubMed  Google Scholar 

  27. Schutte AHJ, Koelewijn R, Ajjampur SSR, Levecke B, McCarthy JS, Mejia R, Williams SA, Verweij JJ, van Lieshout L, van Hellemond JJ. Detection of soil-transmitted helminths and Schistosoma spp. by nucleic acid amplification test: Results of the first 5 years of the only international external quality assessment scheme. PLoS Negl Trop Dis. 2024;18:e0012404.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

MP would like to thank Professor Cinzia Cantacessi and Dr. Stephen R Doyle for their continuous support. MP, AJK and RM would like to thank Nicolas R Caro, Marisa Juarez and Paola A. Vargas, for their help with egg collection and logistical support in Orán, Argentina.

Funding

This work received financial support from the Coalition for Operational Research on Neglected Tropical Diseases (COR-NTD), which is funded by The Task Force for Global Health primarily by the Bill & Melinda Gates Foundation (OPP1190754), by UK aid from the UK Government, and by the United States Agency for International Development through its Neglected Tropical Diseases Program. M.P. was partly funded by an NHM SIF award (2018) for laboratory work. Under the grant conditions of the Foundation, a Creative Commons Attribution 4.0 Generic License has already been assigned to the Author Accepted Manuscript version that might arise from this submission. M.P. is a grateful recipient of a Harding Distinguished Postgraduate Scholarship from the University of Cambridge and Cambridge Commonwealth Trust.

Author information

Authors and Affiliations

Authors

Contributions

M.P., N.P., S.A.W., R.J., A.K., and R.O. conceived the concept of the pilot study. M.P. conducted the analysis and drafted the original draft of the manuscript. N.P., J.C.D., D.T.J.L., R.O.C., A.K., R.J., and S.A.W. provided valuable feedback on the analysis and draft. All authors approved the final manuscript.

Corresponding authors

Correspondence to Marina Papaiakovou or Rojelio Mejia.

Ethics declarations

Ethical approval and consent to participate

Not applicable.

Competing Interests

M.P. was a member of the World Health Organization Diagnostics Technical Advisory Group (WHO DTAG) for Neglected Tropical Diseases (NTDs) between March 2021 and December 2023. The WHO played no role in producing the contents of this short report. All other authors declare no conflict of interest.

Consent for publication

Not applicable.

Additional information

Publisher’s Note

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

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Papaiakovou, M., Cimino, R.O., Pilotte, N. et al. Comparison of multi-parallel quantitative real-time PCRs targeting different DNA regions and detecting soil-transmitted helminths in stool. Parasites Vectors 17, 390 (2024). https://doi.org/10.1186/s13071-024-06464-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13071-024-06464-6