Hostname: page-component-8448b6f56d-t5pn6 Total loading time: 0 Render date: 2024-04-19T22:33:46.275Z Has data issue: false hasContentIssue false

An update on non-invasive urine diagnostics for human-infecting parasitic helminths: what more could be done and how?

Published online by Cambridge University Press:  13 December 2019

John Archer*
Affiliation:
Wolfson Wellcome Biomedical Laboratories, Department of Zoology, Natural History Museum, Cromwell Road, LondonSW7 5BD, UK Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, LiverpoolL3 5QA, UK.
James E. LaCourse
Affiliation:
Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, LiverpoolL3 5QA, UK.
Bonnie L. Webster
Affiliation:
Wolfson Wellcome Biomedical Laboratories, Department of Zoology, Natural History Museum, Cromwell Road, LondonSW7 5BD, UK
J. Russell. Stothard
Affiliation:
Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Pembroke Place, LiverpoolL3 5QA, UK.
*
Author for correspondence: John Archer, E-mail: j.archer@nhm.ac.uk

Abstract

Reliable diagnosis of human helminth infection(s) is essential for ongoing disease surveillance and disease elimination. Current WHO-recommended diagnostic assays are unreliable in low-endemic near-elimination settings and typically involve the invasive, onerous and potentially hazardous sampling of bodily fluids such as stool and blood, as well as tissue via biopsy. In contrast, diagnosis by use of non-invasive urine sampling is generally painless, more convenient and low risk. It negates the need for specialist staff, can usually be obtained immediately upon request and is better accepted by patients. In some instances, urine-based diagnostic assays have also been shown to provide a more reliable diagnosis of infection when compared to traditional methods that require alternative and more invasive bodily samples, particularly in low-endemicity settings. Given these relative benefits, we identify and review current research literature to evaluate whether non-invasive urine sampling is currently exploited to its full potential in the development of diagnostic tools for human helminthiases. Though further development, assessment and validation are needed before their routine use in control programmes, low-cost, rapid and reliable assays capable of detecting transrenal helminth-derived antigens and cell-free DNA show excellent promise for future use at the point-of-care in high-, medium- and even low-endemicity elimination settings.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © Cambridge University Press 2019

Introduction

Parasitic worms, often referred to as helminths, form the most common human infectious parasites in low- and middle-income countries (LMICs), causing a global burden of disease exceeding that of both malaria and tuberculosis (Hotez et al., Reference Hotez, Brindley, Bethony, King, Pearce and Jacobson2008; Lustigman et al., Reference Lustigman, Prichard, Gazzinelli, Grant, Boatin, McCarthy and Basáñez2012). The rapid, straightforward and reliable diagnosis of helminthiases is essential for ongoing disease surveillance and successful disease control, particularly as control programmes advance towards disease elimination within endemic areas (Fig. 1), (Bergquist et al., Reference Bergquist, Johansen and Utzinger2009; Gordon et al., Reference Gordon, Gray, Gobert and McManus2011; McCarthy et al., Reference McCarthy, Lustigman, Yang, Barakat, García, Sripa, Willingham, Prichard and Basáñez2012; Rollinson et al., Reference Rollinson, Knopp, Levitz, Stothard, Tchuem Tchuenté, Garba, Mohammed, Schur, Person, Colley and Utzinger2013; Werkman et al., Reference Werkman, Wright, Truscott, Easton, Oliveira, Toor, Ower, Ásbjörnsdóttir, Means, Farrell, Walson and Anderson2018).

Fig. 1. Schematic outlining changes in diagnostic priorities as control programmes progress (adapted from Bergquist et al., Reference Bergquist, Johansen and Utzinger2009).

Current ‘gold standard’ diagnostic assays for the majority of these diseases typically involve the invasive and cumbersome sampling of bodily fluids such as stool and blood, as well as tissue via biopsy (Table 1), (WHO, 2012). Not only are these procedures often painful, onerous and carry a risk of infection (with, for example, HIV), but they also require specific equipment and specialist health workers seldom available in endemic areas. A reliable assessment of disease prevalence within a given community can therefore often prove challenging as a result of patient aversions to being assessed, as well as through a lack of resources (Itoh et al., Reference Itoh, Weerasooriya, Yahathugoda, Takagi, Samarawickrema, Nagaoka and Kimura2011). Although widely considered low-cost, when taking into consideration the cumulative costs of equipment, number of personnel needed and remuneration of specialist staff, the true costs of gold standard assays are also being realized now and may likely be far more expensive than previously assumed (Turner et al., Reference Turner, Bettis, Dunn, Whitton, Hollingsworth, Fleming and Anderson2017). In addition, whilst these techniques may be sufficiently sensitive to confirm or refute individual infection status in areas of high disease endemicity or when assessing patients burdened with a high degree of infection, in areas of low-endemicity, for example during control programme near-elimination settings, sensitivity of these techniques can seriously wane (Appendix Fig. A1), (Bergquist et al., Reference Bergquist, Johansen and Utzinger2009; Klepac et al., Reference Klepac, Metcalf, McLean and Hampson2013; Hawkins et al., Reference Hawkins, Cantera, Storey, Leader and de los Santos2016).

Table 1. WHO-recommended diagnostic techniques for major human helminth infections and how technique invasiveness compares to that of urine sampling.

*Positive/negative symbols denote degree of increase in sample invasiveness when compared to urine sampling where: ‘±’ indicates relative comparable invasiveness; ‘+’ indicates a moderate increase in sample invasiveness; ‘++’ indicates a considerable increase in sample invasiveness and; ‘+++’ indicates a major increase in sample invasiveness.

In contrast, diagnosis by use of non-invasive urine sampling is generally painless, more convenient, less expensive and low risk. It negates the need for specialist staff, can usually be obtained immediately upon request and is better accepted by patients (Castillo et al., Reference Castillo, Rodriguez, García, Brandt, Van Hul, Silva, Rodriguez-Hidalgo, Portocarrero, Melendez, Gonzalez, Gilman and Dorny2009). Further to these clear practical advantages, some urine-based diagnostic assays have also been shown to provide a more sensitive diagnosis of infection when compared to traditional methods that require alternative and more invasive bodily samples, particularly in low-endemicity settings (Sousa-Figueiredo et al., Reference Sousa-Figueiredo, Betson, Kabatereine and Stothard2013; Adriko et al., Reference Adriko, Standley, Tinkitina, Tukahebwa, Fenwick, Fleming, Sousa-Figueiredo, Stothard and Kabatereine2014). Given these relative benefits in ease of collection, greater patient acceptability and possible improved diagnostic performance, the following review aims to evaluate whether urine is currently being exploited to its full potential with regards to the diagnosis of the major human helminth infections and highlight future research needed to further advance helminth urine-diagnostics.

Literature search strategy

A systematic online literature search was conducted, beginning in October of 2018 and ending in October of 2019. The PubMed, Cochrane Library, Google Scholar and Web of Science databases were used, following stipulated database guidelines, to search for any literature published between 1919 and 2019 within peer-reviewed journals relevant to inputted search terms (National Center for Biotechnology Information., 2019; Cochrane Library., 2019; Google Scholar., 2019; Web of Science., 2019).

Three focal search terms, ‘diagnosis’, ‘diagnostic’ and ‘detection’, were used in conjunction with either disease name(s) (e.g. ‘schistosomiasis’, ‘Bilharzia’ and ‘snail fever’ or ‘lymphatic filariasis’ and ‘elephantiasis’), or pathogen species (e.g. ‘Schistosoma haematobium’ or ‘Wuchereria bancrofti’) and ‘urine’ or ‘transrenal’. Following this initial search, additional terms were included, such as diagnostic marker (e.g. ‘antigen’) and/or assay method (e.g. ‘enzyme-linked immunosorbent assay’), to potentially uncover additional literature. The abstracts of all publication hits were read and assessed for their relevance to review. Irrelevant articles were not included in the review, whereas all relevant articles were read in full. Publications deemed relevant were those that highlighted any primary research concerning the detection of any human-infecting parasitic helminth outlined in Table 1, or closely related non-human animal-infecting species, within urine samples taken from humans or non-human animals. Any secondary research, for example, systematic reviews or meta-analyses that met these criteria were also included. All literature cited within relevant articles was also screened, again to potentially uncover additional literature not provided by initial database searches.

Macroscopic changes to urine as a means of diagnosing urogenital schistosomiasis

Visible haematuria is often indicative of active urogenital schistosomiasis, caused by infection with Schistosoma haematobium (Colley et al., Reference Colley, Bustinduy, Secor and King2014). As such, cost-effective questionnaires involving either the self-reporting of blood in the urine by patients or the observation of blood in the urine by health workers have been used in an attempt to rapidly identify infected individuals and disease prevalence within endemic areas (Lengeler et al., Reference Lengeler, Utzinger and Tanner2002a,Reference Lengeler, Utzinger and Tannerb; Okeke and Ubachukwu, Reference Okeke and Ubachukwu2014; Atalabi et al., Reference Atalabi, Adubi and Lawal2017).

The sensitivity of self-reporting the presence of blood in the urine for diagnosis of S. haematobium infection has been extensively assessed (Bogoch et al., Reference Bogoch, Andrews, Dadzie Ephraim and Utzinger2012; Bassiouny et al., Reference Bassiouny, Hasab, El-Nimr, Al-Shibani and Al-Waleedi2014). Comparing patient questionnaire responses to the diagnostic gold standard (identification of ova in concentrated urine samples via microscopy), it has been concluded that despite the method's practical advantages and relatively low cost, self-reported macrohaematuria alone is unreliable at the individual level primarily because visible haematuria typically only presents in individuals burdened with particularly heavy infections (Bogoch et al., Reference Bogoch, Andrews, Dadzie Ephraim and Utzinger2012). In addition, macrohaematuria is also often a symptom of common urinary tract infections and bladder stones (Appendix Fig. A1), (Le and Hsieh, Reference Le and Hsieh2017). It has also been highlighted that the self-reporting of blood in the urine by school-aged children, the demographic customarily selected for helminth surveillance within a given community, can be unreliable due to either a young girl's reluctance to admit the onset of menses, or a young boy's eagerness to proclaim his ‘coming of age’ as a result of gross haematuria often being considered a natural sign of the onset of puberty (Montresor et al., Reference Montresor, Crompton, Gyorkos, Savioli and Organization2002; Colley et al., Reference Colley, Bustinduy, Secor and King2014).

For these reasons, the diagnostic reliability of having trained and experienced personnel to identify the presence of macroscopic blood in the urine has also been assessed, again, comparing the method to urine-egg detection by microscopy (Okeke and Ubachukwu, Reference Okeke and Ubachukwu2014). Once more it was concluded that, unless used in conjunction with more taxing and costly methods, macrohematuria does not provide adequate sensitivity when compared to egg microscopy, and in using only this method low-, or even moderate-intensity, infections would likely be missed.

It is generally accepted that although a useful and easily implemented tool in initial baseline observations to confirm S. haematobium presence in highly-endemic populations, in areas of low-endemicity, or when evaluating programmatic intervention success in reducing disease prevalence and transmission, alternative and more accurate diagnostic approaches should be used (Utzinger et al., Reference Utzinger, Becker, van Lieshout, van Dam and Knopp2015; Mutapi et al., Reference Mutapi, Maizels, Fenwick and Woolhouse2017).

Microscopic changes to urine as a means of diagnosing urogenital schistosomiasis

The current gold standard of S. haematobium diagnosis involves the filtering, staining and observation of morphologically distinct eggs excreted in urine (Le and Hsieh, Reference Le and Hsieh2017). Using a syringe and polycarbonate filters with a pore size of 8–30 μm, eggs from 10 mL of a well-shaken urine sample can be isolated, stained and examined under a microscope (Peters et al., Reference Peters, Warren and Mahmoud1976; Colley et al., Reference Colley, Bustinduy, Secor and King2014; Utzinger et al., Reference Utzinger, Becker, van Lieshout, van Dam and Knopp2015). This has long been the preferred method of S. haematobium diagnosis as it allows for a straightforward and reasonably inexpensive means of confirming infection within an individual or presence within a community (through sample pooling), using relatively unsophisticated and somewhat field-appropriate equipment. Additionally, and importantly, eggs can be quantified; providing a moderately accurate assessment of infection intensity within an individual that can then be used to estimate the degree of clinical morbidity (Colley et al., Reference Colley, Bustinduy, Secor and King2014; Utzinger et al., Reference Utzinger, Becker, van Lieshout, van Dam and Knopp2015; Corstjens et al., Reference Corstjens, Hoekstra, de Dood and van Dam2017).

The many shortcomings of urine-egg microscopy, however, are well understood (Braun-Munzinger and Southgate, Reference Braun-Munzinger and Southgate1992; Le and Hsieh, Reference Le and Hsieh2017; Ajibola et al., Reference Ajibola, Gulumbe, Eze and Obishakin2018). Owing to heterogeneities in egg output occurring between different periods of the same day, between different days and even between different seasons, accurate diagnosis and morbidity assessment of any given individual using just one urine sample is unlikely (Braun-Munzinger and Southgate, Reference Braun-Munzinger and Southgate1992; Le and Hsieh, Reference Le and Hsieh2017; Christensen et al., Reference Christensen, Taylor, Zulu, Lillebo, Gundersen, Holmen, Kleppa, Vennervald, Ndhlovu and Kjetland2018). To mitigate this, multiple urine samples from the same individual can be taken over consecutive days, ideally between the hours of 10:00am and 2:00pm to coincide with optimum egg passage (Le and Hsieh, Reference Le and Hsieh2017). Repeated bouts of urine filtration and microscopy is, however, taxing work; a reasonable balance between diagnostic accuracy, time spent and financial cost must be met and even then, overt improvements in diagnostic sensitivity are rarely seen (Stothard et al., Reference Stothard, Stanton, Bustinduy, Sousa-Figueiredo, Van Dam, Betson, Waterhouse, Ward, Allan, Hassan, Al-Helal, Memish and Rollinson2014). Differences in diagnostic sensitivity between more and lesser-experienced technicians are also often found, further complicating matters when large quantities of urine samples require assessment (Knopp et al., Reference Knopp, Corstjens, Koukounari, Cercamondi, Ame, Ali, de Dood, Mohammed, Utzinger, Rollinson and van Dam2015).

Of more urgent concern is urine-egg microscopy's poor sensitivity when used in areas of low- or even moderate-prevalence settings (WHO, 2013; Le and Hsieh, Reference Le and Hsieh2017). As egg output declines, the sensitivity of urine-egg microscopy is significantly reduced resulting in a variety of challenges beyond just reliably identifying individuals burdened with low-intensity infections. Some of these challenges include accurately estimating clinical morbidity, evaluating the impact of programmatic interventions, diagnosing pre-school aged children and assessing new diagnostic tools (Stete et al., Reference Stete, Krauth, Coulibaly, Knopp, Hattendorf, Müller, Lohourignon, Kern, N'Goran and Utzinger2012; Knopp et al., Reference Knopp, Becker, Ingram, Keiser and Utzinger2013, Reference Knopp, Ame, Hattendorf, Ali, Khamis, Bakar, Khamis, Person, Kabole and Rollinson2018; Le and Hsieh, Reference Le and Hsieh2017). Recent concern has also been raised about urine-egg microscopy's poor sensitivity when attempting to detect ‘ultra-light’ infections, regarded as those that result in the expulsion of between only one and five eggs per 10 mL of urine (Knopp et al., Reference Knopp, Ame, Hattendorf, Ali, Khamis, Bakar, Khamis, Person, Kabole and Rollinson2018). Given the reproductive biology of schistosomes, just one infected individual excreting such minute numbers of eggs that may go on to infect and asexually reproduce within the appropriate intermediate freshwater snail host, potentially producing hundreds of cercariae per day, can cause the re-infection of an entire community (Colley et al., Reference Colley, Bustinduy, Secor and King2014). As such, in elimination settings or where treatment is targeted only to infected individuals that may be tracked, reassessed and retreated, any infected individuals must be quickly identified to ensure prompt treatment and total interruption of transmission; highlighting the urgent need for rapid, simple-to-use diagnostic tools deployable at the point-of-care (POC) and able to detect ultra-light infections (Hawkins et al., Reference Hawkins, Cantera, Storey, Leader and de los Santos2016; Knopp et al., Reference Knopp, Ame, Hattendorf, Ali, Khamis, Bakar, Khamis, Person, Kabole and Rollinson2018).

Although macrohematuria is typically present only in those harbouring heavy S. haematobium infections, microhaematuria, i.e. trace amounts of blood in the urine not visible to the naked-eye, can occur even in moderate- and low-intensity infections and can be detected using rapid, simple-to-use and relatively inexpensive reagent-strips that can be used at the point-of-care (Ochodo et al., Reference Ochodo, Gopalakrishna, Spek, Reitsma, van Lieshout, Polman, Lamberton, Bossuyt and Leeflang2015; Le and Hsieh, Reference Le and Hsieh2017; Knopp et al., Reference Knopp, Ame, Hattendorf, Ali, Khamis, Bakar, Khamis, Person, Kabole and Rollinson2018).

The accuracy of urine-heme reagent-strips, or ‘dipsticks’ for the indirect diagnosis of urogenital schistosomiasis have also been extensively assessed (Robinson et al., Reference Robinson, Picon, Sturrock, Sabasio, Lado, Kolaczinski and Brooker2009; Krauth et al., Reference Krauth, Greter, Stete, Coulibaly, Traoré, Ngandolo, Achi, Zinsstag, N'Goran and Utzinger2015; Hassan et al., Reference Hassan, Mohammed, Opaluwa, Adamu, Nataala, Garba, Bello and Bunza2018; Musa and Dadah, Reference Musa and Dadah2018; Knopp et al., Reference Knopp, Ame, Hattendorf, Ali, Khamis, Bakar, Khamis, Person, Kabole and Rollinson2018). Recent reviews and meta-analyses have been undertaken to evaluate their diagnostic accuracy with a specific focus on high-, medium- and low-prevalence settings and in populations that have previously undergone repeated mass drug administration (MDA) treatment with praziquantel (King and Bertsch, Reference King and Bertsch2013; Ochodo et al., Reference Ochodo, Gopalakrishna, Spek, Reitsma, van Lieshout, Polman, Lamberton, Bossuyt and Leeflang2015). In most cases, it has been concluded that although the diagnostic performance of urine-heme dipsticks is reduced in low-transmission areas and despite a range of possible confounding reasons for the presence of blood in the urine (such as urinary tract infections, bladder stones and menstrual blood), at the population level, urine-heme dipsticks should be considered more accurate than urine-egg microscopy (Ochodo et al., Reference Ochodo, Gopalakrishna, Spek, Reitsma, van Lieshout, Polman, Lamberton, Bossuyt and Leeflang2015). In addition, urine-heme dipsticks do not require specially trained microscopists, are less influenced by daily fluctuations in egg passage and take far less time to carry out (Krauth et al., Reference Krauth, Greter, Stete, Coulibaly, Traoré, Ngandolo, Achi, Zinsstag, N'Goran and Utzinger2015). It has also been concluded, however, that whilst urine-heme dipsticks should continue to be used to monitor the early-stage population-level impact of schistosomiasis control programmes (i.e. when assessing the initial baseline prevalence or when evaluating changes in overall prevalence after early intervention implementation), in elimination settings, or again when treatment is targeted only to infected individuals, neither the urine-heme dipstick or urine-egg microscopy can reliably identify individuals burdened with low- or ultra-light-intensity infections still capable of maintaining disease transmission (King and Bertsch, Reference King and Bertsch2013; Ochodo et al., Reference Ochodo, Gopalakrishna, Spek, Reitsma, van Lieshout, Polman, Lamberton, Bossuyt and Leeflang2015; Knopp et al., Reference Knopp, Ame, Hattendorf, Ali, Khamis, Bakar, Khamis, Person, Kabole and Rollinson2018). Further assessment of urine-heme dipstick diagnostic performance using more sophisticated and sensitive methods such as Schistosoma antigen or DNA detection, rather than egg microscopy, has been encouraged (King and Bertsch, Reference King and Bertsch2013).

As well as microhaematuria, leukocyturia (the abnormal presence of white blood cells in the urine) and proteinuria (the abnormal presence of proteins in the urine) may also be used as proxy to diagnose urogenital schistosomiasis, though both methods been found to be significantly less sensitive and specific than urine-heme dipsticks (Ochodo et al., Reference Ochodo, Gopalakrishna, Spek, Reitsma, van Lieshout, Polman, Lamberton, Bossuyt and Leeflang2015). It has been suggested, however, that the use of urine-heme dipsticks in conjunction with low-cost and field-deployable assays capable of detecting albuminuria (urine-albumin concentrations of >40 mg L−1), may provide a reliable diagnosis of infection in high-endemicity settings whilst also allowing assessment of kidney and urinary tract morbidity associated with chronic disease (Rollinson et al., Reference Rollinson, Klinger, Mgeni, Khamis and Stothard2005; Sousa-Figueiredo et al., Reference Sousa-Figueiredo, Basáñez, Khamis, Garba, Rollinson and Stothard2009).

Like macroscopic changes, microscopic changes to urine are also considered now insufficiently sensitive to detect S. haematobium infection in low-prevalence settings or within individuals harbouring low-level infections. In addition, these changes only occur as a result of infection with S. haematobium. In endemic areas, co-infection with multiple helminth species is commonplace, highlighting the need for multiplex assays capable of reliably detecting multiple helminth species using just one bodily sample.

Detection of anti-helminth urine-antibodies

Immunodiagnostic assays for the detection of blood-circulating anti-helminth antibodies have been used to diagnose infection with many human-infecting helminthiases (Rebollo and Bockarie, Reference Rebollo and Bockarie2014; Kemal et al., Reference Kemal, Alemu, Yimer and Terefe2015; Vlaminck et al., Reference Vlaminck, Fischer and Weil2015; Akue et al., Reference Akue, Eyang-Assengone and Dieki2018). Of all antibody-targeting immunological assays, the most frequently employed is some form of the enzyme-linked immunosorbent assay (ELISA), the diagnostic functionality of which relies on the highly specific antigen-antibody binding that occurs during the body's immune response to invading foreign pathogens (Lazcka et al., Reference Lazcka, Del Campo and Muñoz2007).

Due to ease of sample procurement relative to blood sampling, the diagnostic potential of targeting anti-helminth antibodies expelled within the urine using immunodiagnostic assays has also been assessed; targeting and successfully detecting urine-based antibodies formulated against a range of helminth species (Table 2). Of those studies comparing the diagnostic accuracy between targeting urine- and serum-based antibodies, all reported good association in diagnostic performance whilst no additional effort in urine-sample preparation was required, presenting a compelling argument for moving beyond invasive blood-based diagnostics (Elhag et al., Reference Elhag, Abdelkareem, Yousif, Frah and Mohamed2011; Nagaoka et al., Reference Nagaoka, Itoh, Samad, Takagi, Weerasooriya, Yahathugoda, Hossain, Moji and Kimura2013; Eamudomkarn et al., Reference Eamudomkarn, Sithithaworn, Kamamia, Yakovleva, Sithithaworn, Kaewkes, Techasen, Loilome, Yongvanit, Wangboon, Saichua, Itoh and Bethony2018).

Table 2. Anti-helminth antibodies detected within urine and immunodiagnostic assay used.

Although highly specific even in low-endemicity settings, antibody detection using the ELISA requires sophisticated equipment, specially trained health workers and expensive reagents that require cold chain typically unavailable to those in disease-endemic regions, particularly when hoping to obtain a quantitative diagnosis that indicates degree of infection within an individual (Bergquist et al., Reference Bergquist, Johansen and Utzinger2009; Tchuem Tchuenté, Reference Tchuem Tchuenté2011). As such, regardless of the bodily sample taken, these requirements make it difficult to envisage the future scale-up and field-deployment of the ELISA at the point-of-care, where the simple-to-use, rapid and sensitive diagnosis is needed. It is for this reason that much attention has been given to the development of simple-to-use point-of-care rapid diagnostic test (POC-RDT) devices capable of rapidly detecting blood-circulating anti-helminth antibodies (Weil et al., Reference Weil, Steel, Liftis, Li, Mearns, Lobos and Nutman2000; Coulibaly et al., Reference Coulibaly, N'Goran, Utzinger, Doenhoff and Dawson2013; Steel et al., Reference Steel, Golden, Kubofcik, LaRue, de los Santos, Domingo and Nutman2013). Further development of these for use with urine samples, however, is lacking. Two novel transrenal antibody-detecting RDTs that have been developed and assessed involve the use of antigen-coated coloured latex beads for the detection of filaria-specific IgG4 (Nagaoka et al., Reference Nagaoka, Itoh, Samad, Takagi, Weerasooriya, Yahathugoda, Hossain, Moji and Kimura2013), and the filtering of urine to isolate human IgG bound to S. haemotobium ova, both requiring significantly less equipment, reagents and technical expertise than conventional immunodiagnostic assays (Sheele et al., Reference Sheele, Kihara, Baddorf, Byrne and Ravi2013). Although promising, further evaluation for reliability, field-applicability, upscale and deployment is needed.

Another principal concern when targeting antibodies to determine infection status is the inability to distinguish between active and past infections owing to high antibody titres remaining within the body long after treatment success and infection clearance (Rollinson et al., Reference Rollinson, Knopp, Levitz, Stothard, Tchuem Tchuenté, Garba, Mohammed, Schur, Person, Colley and Utzinger2013; Utzinger et al., Reference Utzinger, Becker, van Lieshout, van Dam and Knopp2015). This becomes particularly problematic when attempting to evaluate the impact of programmatic control strategies in areas that have undergone control intervention. As an example, individuals within areas having undergone mass administration with albendazole for the treatment of ascariasis may have indeed cleared any infection, however, any diagnostic assay targeting anti-Ascaris antibodies used to assess these individuals may remain positive (Jourdan et al., Reference Jourdan, Lamberton, Fenwick and Addiss2018). In areas where disease elimination is sought, it has been suggested that antibody-targeting assays may be appropriate for use with young children who have not yet received treatment as a means of assessing whether the transmission is still taking place (Jourdan et al., Reference Jourdan, Lamberton, Fenwick and Addiss2018; Takagi et al., Reference Takagi, Yahathugoda, Tojo, Rathnapala, Nagaoka, Weerasooriya and Itoh2019). In doing so, seroconversion rate, typically somewhere between at least 4 and 8 weeks after initial exposure, must be taken into consideration (van Grootveld et al., Reference van Grootveld, van Dam, de Dood, de Vries, Visser, Corstjens and van Lieshout2018; Vlaminck et al., Reference Vlaminck, Lagatie, Verheyen, Dana, Van Dorst, Mekonnen, Levecke and Stuyver2019).

Persistent post-infection circulating antibodies also cause difficulty when attempting to evaluate the true accuracy of antibody-targeting diagnostic assays; typically performed via comparison to gold standard assays that may themselves have poor-sensitivity. In doing this, antibody assays will consistently appear highly-sensitive with likely concurrent low positive predictive values (PPV), (Appendix Figure A1), whereas individuals testing negative by gold standard methods but positive by antibody-detecting methods may plausibly be harbouring active but low-level infections, or may indeed be currently uninfected after having cleared a previous infection (Doenhoff et al., Reference Doenhoff, Chiodini and Hamilton2004).

Cross-reactivity of antibodies between different helminth genera is also an issue (Genta, Reference Genta1988; Lammie et al., Reference Lammie, Weil, Noordin, Kaliraj, Steel, Goodman, Lakshmikanthan and Ottesen2004; Weerakoon et al., Reference Weerakoon, Gobert, Cai and McManus2015; Lamberton and Jourdan, Reference Lamberton and Jourdan2015; Garcia et al., Reference Garcia, Castillo, Gonzales, Bustos, Saavedra, Jacob, Del Brutto, Wilkins, Gonzalez and Gilman2018; Song et al., Reference Song, Kim, Jin, Lee, Jeoung, Lee, Saeed and Hong2018). In some cases, genera-, or even species-specific identification of infecting helminths is essential for safe treatment strategies, for example when providing ivermectin to treat onchocerciasis in loiasis-endemic areas (Gardon et al., Reference Gardon, Gardon-Wendel, Demanga-Ngangue, Kamgno, Chippaux and Boussinesq1997), or for diagnosis of species-specific pathologies such as female and male genital schistosomiasis (Itoh et al., Reference Itoh, Weerasooriya, Yahathugoda, Takagi, Samarawickrema, Nagaoka and Kimura2011; Vlaminck et al., Reference Vlaminck, Supali, Geldhof, Hokke, Fischer and Weil2016; Kayuni et al., Reference Kayuni, Lampiao, Makaula, Juziwelo, Lacourse, Reinhard-Rupp, Leutscher and Stothard2019; Kukula et al., Reference Kukula, MacPherson, Tsey, Stothard, Theobald and Gyapong2019). In circumstances such as these, diagnostic assays with a higher degree of specificity than that of antibody-targeting assays are needed.

Because of the technical, financial and logistical challenges presented by anti-helminth antibody detection and when considering the very limited resources available for the development and validation of novel diagnostic assays, perhaps focus is best placed elsewhere, on more user-friendly, cost-effective and reliable methods.

Detection of helminth-derived urine-antigens

Targeting urine-antigens has multiple advantages over targeting transrenal antibodies; detection of antigens indicates active infection; diagnostic assays that target antigens can, therefore, be used to evaluate disease intervention strategies such as MDA and vector control; invading parasites may be detected soon after infection and antigen levels generally correlate well with parasite load (Corstjens et al., Reference Corstjens, De Dood, Kornelis, Fat, Wilson, Kariuki, Nyakundi, Loverde, Abrams, Tanke, Van Lieshout, Deelder and Van Dam2014; Worasith et al., Reference Worasith, Kamamia, Yakovleva, Duenngai, Wangboon, Sithithaworn, Watwiengkam, Namwat, Techasen, Loilome, Yongvanit, Loukas, Sithithaworn and Bethony2015; Ochodo et al., Reference Ochodo, Gopalakrishna, Spek, Reitsma, van Lieshout, Polman, Lamberton, Bossuyt and Leeflang2015; Kamel et al., Reference Kamel, Salah, Demerdash, Maher, Atta, Badr, Afifi and El Baz2019; Sousa et al., Reference Sousa, van Dam, Pinheiro, de Dood, Peralta, Peralta, de Daher, Corstjens and Bezerra2019). As with antibody detection, good association between urine- and serum-based antigen detection has been found in high-, medium- and low-endemicity settings, further strengthening the argument for moving towards non-invasive urine sampling (van Dam et al., Reference van Dam, Wichers, Ferreira, Ghati, van Amerongen and Deelder2004; Kamel et al., Reference Kamel, Salah, Demerdash, Maher, Atta, Badr, Afifi and El Baz2019; Sousa et al., Reference Sousa, van Dam, Pinheiro, de Dood, Peralta, Peralta, de Daher, Corstjens and Bezerra2019).

Again, most immunodiagnostic assays used to detect helminth-derived urine-antigens, such as conventional ELISAs, are currently unsuited for point-of-care use (Table 3). At present, efforts to develop simple-to-use POC-RDTs for the detection of helminth urine-antigens have focused primarily on test devices capable of diagnosing urogenital and intestinal schistosomiasis, though a point-of-care lateral-flow dipstick to detect O. volvulus-derived urine-antigens has also been developed (Ayong et al., Reference Ayong, Tume, Wembe, Simo, Asonganyi, Lando and Ngu2005).

Table 3. Helminth-derived antigens detected within the urine and immunodiagnostic assay used.

The reliability of urine-antigen POC-RDTs when used in low-endemicity settings or when assessing individuals with low-intensity infections that may give unclear ‘trace’ results is, however, disputed (Coelho et al., Reference Coelho, Siqueira, Grenfell, Almeida, Katz, Almeida, Carneiro and Oliveira2016; Peralta and Cavalcanti, Reference Peralta and Cavalcanti2018). Recent meta-analyses suggest that, although more rapid and sensitive than stool-microscopy, under these circumstances' targeting the schistosome urine circulating cathodic antigen (CCA) by use of the CCA-POC-RDT is not sufficiently sensitive to reliably detect S. mansoni infection at the individual level (Danso-Appiah et al., Reference Danso-Appiah, Minton, Boamah, Otchere, Asmah, Rodgers, Bosompem, Eusebi and De Vlas2016). It has been concluded that because of their low-cost, ease of use and patient-compliance, the CCA-POC-RDT may serve as a useful tool for disease-prevalence mapping and monitoring of control programmes relevant to S. mansoni in high- and medium-endemicity settings. In low-endemicity settings, however, when highly sensitive diagnostics capable of detecting low-intensity infections with a range of helminth species at the individual level are required, alternative and more sensitive assays are needed.

Revisions in assay protocols can improve the sensitivity and specificity of RDT's capable of detecting schistosome-urine-antigens beyond that of even lab-based ELISA assays, even in light-infections (Coelho et al., Reference Coelho, Siqueira, Grenfell, Almeida, Katz, Almeida, Carneiro and Oliveira2016; Kamel et al., Reference Kamel, Salah, Demerdash, Maher, Atta, Badr, Afifi and El Baz2019). Recently, the development of an up-converting phosphor lateral-flow (UCP-LF) assay targeting transrenal circulating anodic antigens (CAA) has shown that diagnosis of ultra-light schistosome infections through urine-CAA detection is possible (Corstjens et al., Reference Corstjens, De Dood, Kornelis, Fat, Wilson, Kariuki, Nyakundi, Loverde, Abrams, Tanke, Van Lieshout, Deelder and Van Dam2014; van Dam et al., Reference van Dam, Xu, Bergquist, de Dood, Utzinger, Qin, Guan, Feng, Yu, Zhou, Zheng, Zhou and Corstjens2015a,Reference van Dam, Odermatt, Acosta, Bergquist, de Dood, Kornelis, Muth, Utzinger and Corstjensb). The genus-specific assay has shown extremely high sensitivity for the detection of S. haemotobium, S. mansoni, S. japonicum and S. mekongi urine-CAA, even in low-endemicity settings (Corstjens et al., Reference Corstjens, De Dood, Kornelis, Fat, Wilson, Kariuki, Nyakundi, Loverde, Abrams, Tanke, Van Lieshout, Deelder and Van Dam2014; van Dam et al., Reference van Dam, Xu, Bergquist, de Dood, Utzinger, Qin, Guan, Feng, Yu, Zhou, Zheng, Zhou and Corstjens2015a,Reference van Dam, Odermatt, Acosta, Bergquist, de Dood, Kornelis, Muth, Utzinger and Corstjensb; Knopp et al., Reference Knopp, Corstjens, Koukounari, Cercamondi, Ame, Ali, de Dood, Mohammed, Utzinger, Rollinson and van Dam2015; de Dood et al., Reference de Dood, Hoekstra, Mngara, Kalluvya, van Dam, Downs and Corstjens2018; Sousa et al., Reference Sousa, van Dam, Pinheiro, de Dood, Peralta, Peralta, de Daher, Corstjens and Bezerra2019). Further to its high-specificity, the UCP-LF CAA assay offers additional advantages over urine- and stool- microscopy in that the UPC-LF CAA is much higher-throughput and that only urine sampling is required to diagnose both urogenital and intestinal schistosomiasis (Knopp et al., Reference Knopp, Corstjens, Koukounari, Cercamondi, Ame, Ali, de Dood, Mohammed, Utzinger, Rollinson and van Dam2015; Corstjens et al., Reference Corstjens, De Dood, Kornelis, Fat, Wilson, Kariuki, Nyakundi, Loverde, Abrams, Tanke, Van Lieshout, Deelder and Van Dam2014). Though treatment of both forms of schistosomiasis is identical, (40 mg praziquantel per kg body weight), if using this assay in areas co-endemic for both S. haemotobium and S. mansoni, additional steps would be required to diagnose species-specific infection, associated pathologies, cure rates and drug efficacies.

Although not yet fully suited for point-of-care use, the UCP-LF CAA assay requires only a reliable source of electricity, simple centrifugation facilities and pipetting capacities; offering a high-throughput and highly-sensitive means of diagnosing schistosomiasis through urine sampling whilst requiring lesser-equipped laboratory infrastructure than conventional immunodiagnostic assays (Knopp et al., Reference Knopp, Corstjens, Koukounari, Cercamondi, Ame, Ali, de Dood, Mohammed, Utzinger, Rollinson and van Dam2015; Sousa et al., Reference Sousa, van Dam, Pinheiro, de Dood, Peralta, Peralta, de Daher, Corstjens and Bezerra2019). As the assay is also currently too expensive for commercial and routine use in schistosomiasis control programmes, efforts to develop a less expensive, rapid and simple-to-use CAA-POC-RDT that retains the UCP-LF CAA's high-sensitivity have begun (Knopp et al., Reference Knopp, Corstjens, Koukounari, Cercamondi, Ame, Ali, de Dood, Mohammed, Utzinger, Rollinson and van Dam2015). Until then, it has been suggested that the existing assay could be used as a robust means of confirming or refuting indecisive test results at the individual level given by alternative, less sensitive but more field-appropriate, methods (de Dood et al., Reference de Dood, Hoekstra, Mngara, Kalluvya, van Dam, Downs and Corstjens2018).

Additional advancements in antigen-detecting immunodiagnostic POC-RDTs include the development of a lateral flow immunochromatographic test strip capable of detecting circulating S. mansoni antigen (CSA) within the urine using colloidal gold and mesoporous silica nanoparticles (Kamel et al., Reference Kamel, Salah, Demerdash, Maher, Atta, Badr, Afifi and El Baz2019). Though currently adapted only for diagnosis of infection with S. mansoni, these rapid and field-applicable test strips showed extremely high sensitivity when used to assess patients burdened with light infections and were even found to provide a more sensitive diagnosis than the conventional lab-based sandwich ELISA. Further assessment and validation of these RDT test strips, as well as adaptation for detection of other helminth-species urine-antigens, is encouraged.

It should be noted that the diagnostic potential of targeting any helminth-derived antigen through urine sampling will greatly depend on whether or not that antigen is expelled in the urine. Blood-circulating antigens with a high molecular mass may be too large to pass from the glomerular capillaries into the glomerular capsule and onto the bladder, and even of those that do, some will undoubtedly degrade into smaller products not recognised by monoclonal antibodies prior to diagnosis (Chanteau et al., Reference Chanteau, Moulia-Pelat, Glaziou, Nguyen, Luquiaud, Plichart, Martin and Cartel1994). Moreover, some helminth antigens may not be expelled in the urine because of that helminth species' bodily habitat. The adult form of Ascaris lumbricoides and various species of cestode, for example, reside within the gastrointestinal lumen and so do not directly interact with circulating blood (Lamberton and Jourdan, Reference Lamberton and Jourdan2015).

Additionally, as with antibodies, any transrenal antigens targeted for diagnostic purposes will require assessment as to whether or not and to the degree with which they may cross-react with other antigens and/or other proteins expelled in the urine. Helminth-derived blood-circulating antigens from various genera of filarial nematode, for example, are known to cross-react in co-endemic areas; severely hampering the diagnostic efficacy of assays needed to identify infected individuals and provide safe treatment (Hertz et al., Reference Hertz, Nana-Djeunga, Kamgno, Jelil Njouendou, Chawa Chunda, Wanji, Rush, Fischer, Weil and Budge2018). In addition, the Schistosoma CCA assay has been found to cross-react with antigens from other parasites, general inflammatory biomarkers and even metabolites expelled in the urine of pregnant women; also hampering diagnostic efficacy (van Dam et al., Reference van Dam, Claas, Yazdanbakhsh, Kruize, van Keulen, Ferreira, Rotmans and Deelder1996; Utzinger et al., Reference Utzinger, Greter, Ngandolo, Krauth, Alfaroukh and Zinsstag2016).

Detection of helminth-derived transrenal nucleic acid

Diagnosis via detection of helminth DNA expelled in the urine has many advantages beyond ease of sample procurement; it can be highly sensitive (trace levels of DNA can be detected), highly specific, parasite load can be quantified, assays can be high through-put and multiple species of parasitic helminth can be identified within one multiplex assay (Gordon et al., Reference Gordon, Gray, Gobert and McManus2011; Phuphisut et al., Reference Phuphisut, Yoonuan, Sanguankiat, Chaisiri, Maipanich, Pubampen, Komalamisra and Adisakwattana2014; Melchers et al., Reference Melchers, van Dam, Shaproski, Kahama, Brienen, Vennervald and van Lieshout2014). Possible further benefits include the early detection of anthelminthic drug resistance development, the ability to monitor helminth population genetic variation over time, relatively less arduous sample preparation when compared to blood, stool or tissue samples and the ability to detect pre-patent infections (Enk et al., Reference Enk, Oliveira e Silva and Rodrigues2010, Reference Enk, Oliveira e Silva and Rodrigues2012; Lamberton and Jourdan, Reference Lamberton and Jourdan2015; Minetti et al., Reference Minetti, Lacourse, Reimer and Stothard2016).

Cell-Free DNA (cfDNA) has been defined as extracellular fragments of DNA found in bodily fluids or tissues, including the urine (Weerakoon et al., Reference Weerakoon, Gobert, Cai and McManus2015, Reference Weerakoon, Gordon and McManus2018; Weerakoon and McManus, Reference Weerakoon and McManus2016). It can be detected through use of nucleic acid amplification tests (NAATs), the more common of which include conventional polymerase chain reaction (PCR), nested PCR (nPCR) and quantitative or real-time PCR (qPCR/rtPCR) (Gordon et al., Reference Gordon, Gray, Gobert and McManus2011; Verweij and Stensvold, Reference Verweij and Stensvold2014). Praised for their high sensitivity and specificity, NAATs are now becoming recognised as a more reliable means of helminth diagnosis than current gold standard and immunodiagnostic assays, particularly in low-intensity infections and even when targeting cfDNA expelled in the urine (Enk et al., Reference Enk, Oliveira e Silva and Rodrigues2012; Ibironke et al., Reference Ibironke, Koukounari, Asaolu, Moustaki and Shiff2012; Melchers et al., Reference Melchers, van Dam, Shaproski, Kahama, Brienen, Vennervald and van Lieshout2014; Lodh et al., Reference Lodh, Caro, Sofer, Scott, Krolewiecki and Shiff2016; Krolewiecki et al., Reference Krolewiecki, Koukounari, Romano, Caro, Scott, Fleitas, Cimino and Shiff2018).

To date, using NAATs, numerous studies have evaluated the diagnostic efficacy of targeting transrenal cfDNA from helminths known to reside within a range of bodily habitats, all of which have reported higher sensitivity when compared to gold standard techniques (Table 4). Although the detectible presence of transrenal cfDNA has not yet been confirmed for all human-infecting parasitic helminths, as validated assays do currently exist for the detection of many helminth species' cfDNA in other bodily samples, adaptation of these to assess presence and diagnostic efficacy of helminth-derived cfDNA within the urine should be straightforward (Gordon et al., Reference Gordon, Gray, Gobert and McManus2011; Minetti et al., Reference Minetti, Lacourse, Reimer and Stothard2016; Weerakoon and McManus, Reference Weerakoon and McManus2016). Of particular interest would be to determine the presence of transrenal Onchocerca volvulus and Loa loa cfDNA, given their subcutaneous and deep tissue habitats and understandable patient aversions to invasive skin-snip biopsies currently used to confirm onchocerciasis infection (Knopp et al., Reference Knopp, Steinmann, Hatz, Keiser and Utzinger2012). Of additional interest would be to determine the presence of transrenal cfDNA from Trichuris trichiura and hookworm parasites as, despite sharing their gastrointestinal tract habitat with Ascaris lumbricoides and cestodes, adult forms do interact with circulating blood (Jourdan et al., Reference Jourdan, Lamberton, Fenwick and Addiss2018). As with A. lumbricoides and cestode urine-antigen detection, detection of transrenal cfDNA from these helminths may be unlikely. Cell-free DNA from Strongyloides stercoralis, another gut-dwelling helminth, has been successfully detected in urine, though it is speculated this is due to tissue dissemination during larval-form autoinfection (Lodh et al., Reference Lodh, Caro, Sofer, Scott, Krolewiecki and Shiff2016).

Table 4. Helminth cfDNA detected within the urine and nucleic acid amplification test (NAAT) used.

Although clearly a highly sensitive method of confirming or refuting infection, many financial, logistical and methodological challenges must be overcome if NAATs are to replace current diagnostic standards, regardless of the bodily sample taken. Perhaps of primary concern are the high costs associated with NAATs, such as PCR and qPCR, when compared to current, less costly, gold standard assays. Expensive reagents, sophisticated equipment and remuneration of specialist technical staff all contribute to overall expenditure, again resulting in a diagnostic assay likely unaffordable to most health workers in resource-poor settings (Minetti et al., Reference Minetti, Lacourse, Reimer and Stothard2016). Another major challenge is the upscale and field-applicability of assays targeting cfDNA. In programmatic elimination settings where rapid and reliable detection of few infected individuals is crucial for disease elimination, diagnostic assays must be deployable at the point-of-care. Not only are conventional NAATs themselves currently unsuited for point-of-care use, but essential sample preparation steps, such as DNA extraction, that also require specific laboratory equipment and reagents, prevent the use of conventional NAATs anywhere lacking sophisticated laboratory infrastructure. In addition, to what extent helminth-derived cfDNA continues to be expelled in the urine after infection clearance is largely unknown and likely varies between parasite species' bodily habitat and degree of the previous infection.

The recently developed loop-mediated isothermal amplification (LAMP) assay shows promise for future point-of-care use; DNA fragments are amplified under isothermal conditions, negating the need for thermocycling equipment essential for PCR-based assays; the assay is rapid; results can be seen with the naked-eye; multiple pathogens can be targeted and detected using one assay run; assays can be carried out by non-specialist staff; reagents can be lyophilised and initial DNA extraction steps may be less laborious (Gordon et al., Reference Gordon, Gray, Gobert and McManus2011; Zhang et al., Reference Zhang, Lowe and Gooding2014; Weerakoon and McManus, Reference Weerakoon and McManus2016; Bayoumi et al., Reference Bayoumi, Al-Refai and Badr2016; Deng et al., Reference Deng, Zhong, Kamolnetr, Limpanont and Lv2019). To date, LAMP has been used to successfully detect and amplify helminth DNA from other bodily samples; S. mansoni-derived cfDNA in urine samples taken from experimentally infected mouse models; cfDNA of Strongyloides venezuelensis (a rodent-infecting species) in urine samples taken from experimentally infected rat models and S. haematobium DNA in human urine samples (Takagi et al., Reference Takagi, Itoh, Kasai, Yahathugoda, Weerasooriya and Kimura2011; Fernández-Soto et al., Reference Fernández-Soto, Gandasegui Arahuetes, Sánchez Hernández, López Abán, Vicente Santiago and Muro2014, Reference Fernández-Soto, Sánchez-Hernández, Gandasegui, Bajo Santos, López-Abán, Saugar, Rodríguez, Vicente and Muro2016, Reference Fernández-Soto, Gandasegui, Carranza Rodríguez, Pérez-Arellano, Crego-Vicente, García-Bernalt Diego, López-Abán, Vicente and Muro2019; Shiraho et al., Reference Shiraho, Eric, Mwangi, Maina, Kinuthia, Mutuku, Mugambi, Mwandi and Mkoji2016; Bayoumi et al., Reference Bayoumi, Al-Refai and Badr2016; Lagatie et al., Reference Lagatie, Merino, Batsa Debrah, Debrah and Stuyver2016). Despite these advantages, however, the LAMP assay does still require heat-blocks or waterbaths to heat reactions for long periods; often up to 2 hours. A reliable source of electricity is therefore still essential. Furthermore, unlike qPCR/rtPCR, LAMP assays are only semi-quantitative, meaning estimations of infection intensity are subjective and may vary between personnel; individual assays are low-throughput; amplified fragments cannot be sequenced, preventing the monitoring of genetic variation in populations over time and ambiguity exists around LAMP sensitivity when compared to alternative PCR-based approaches (Verweij and Stensvold, Reference Verweij and Stensvold2014; Zhang et al., Reference Zhang, Lowe and Gooding2014; Minetti et al., Reference Minetti, Lacourse, Reimer and Stothard2016).

To overcome many of the logistical and methodological challenges presented by PCR-based diagnostics, the point-of-care recombinase DNA-polymerase amplification (RPA) assay has also recently been developed and has been used to successfully detect and amplify S. japonicum and F. hepatica cfDNA in human stool samples, as well as S. haematobium DNA within human urine samples (Piepenburg et al., Reference Piepenburg, Williams, Stemple and Armes2006; Sun et al., Reference Sun, Xing, Yu, Fu, Wang, Zou, Luo and Xu2016; Xing et al., Reference Xing, Yu, Feng, Sun, Fu, Wang, Zou, Xia, Luo, He, Li and Xu2017; Cabada et al., Reference Cabada, Malaga, Castellanos-Gonzalez, Bagwell, Naeger, Rogers, Maharsi, Mbaka and White2017; Li et al., Reference Li, Macdonald and von Stetten2019; Rostron et al., Reference Rostron, Pennance, Bakar, Rollinson, Knopp, Allan, Kabole, Ali, Ame and Webster2019). Assays to detect S. mansoni DNA by the use of the RPA have also recently been developed, though these have not yet been tested on clinical samples (Poulton and Webster, Reference Poulton and Webster2018).

Capable of detecting even trace levels of DNA, the RPA provides a promising means of reliably detecting ultra-light levels of infection in low-endemic areas (Rosser et al., Reference Rosser, Rollinson, Forrest and Webster2015; Lai et al., Reference Lai, Ooi and Lau2017; Poulton and Webster, Reference Poulton and Webster2018; Rostron et al., Reference Rostron, Pennance, Bakar, Rollinson, Knopp, Allan, Kabole, Ali, Ame and Webster2019). In addition, the assay itself offers many advantages over PCR and qPCR in terms of its methodology and potential use at the point-of-care (Aryeetey et al., Reference Aryeetey, Essien-Baidoo, Larbi, Ahmed, Amoah, Obeng, van Lieshout, Yazdanbakhsh, Boakye and Verweij2013; Lodh et al., Reference Lodh, Naples, Bosompem, Quartey and Shiff2014; Sady et al., Reference Sady, Al-Mekhlafi, Ngui, Atroosh, Al-Delaimy, Nasr, Dawaki, Abdulsalam, Ithoi, Lim, Chua and Surin2015; Minetti et al., Reference Minetti, Lacourse, Reimer and Stothard2016).

Firstly, assay reactions take place within a robust, hand-held, easily programmed, portable and battery-powered device, omitting the need for specialist technical personnel and sophisticated laboratory infrastructure. Moreover, assay results can be easily interpreted using the same device or, alternatively, via simple-to-use and low-cost lateral-flow immunoassay strips; both omitting the need for sophisticated and delicate readout equipment (Rosser et al., Reference Rosser, Rollinson, Forrest and Webster2015; Xing et al., Reference Xing, Yu, Feng, Sun, Fu, Wang, Zou, Xia, Luo, He, Li and Xu2017; Poulton and Webster, Reference Poulton and Webster2018; Rostron et al., Reference Rostron, Pennance, Bakar, Rollinson, Knopp, Allan, Kabole, Ali, Ame and Webster2019).

Assay reactions are also isothermal; optimal amplification occurs within 25°C – 42°C with use of the device's battery-powered heater, though testing can take place at ambient temperature in some endemic areas, or even at body temperature (Crannell et al., Reference Crannell, Rohrman and Richards-Kortum2014; Kersting et al., Reference Kersting, Rausch, Bier and von Nickisch-Rosenegk2014; Minetti et al., Reference Minetti, Lacourse, Reimer and Stothard2016). Reaction time at reduced temperatures is, however, prolonged. Isothermal reactions not only negate the need for thermocycling equipment that may only amplify specific DNA strands based on cycle conditions within one cycle run but also allow for the detection and amplification of DNA from multiple helminth species or even other pathogens, e.g. malaria or intestinal protozoa, using only different primer combinations, within the same assay run (Crannell et al., Reference Crannell, Castellanos-Gonzalez, Nair, Mejia, White and Richards-Kortum2016). Additionally, and unlike LAMP amplicons, RPA amplicons can be sequenced, allowing the monitoring of genetic variation in populations over time (Oyola et al., Reference Oyola, Otto, Gu, Maslen, Manske, Campino, Turner, MacInnis, Kwiatkowski, Swerdlow and Quail2012).

Another advantage of RPA over PCR-based techniques is assay runtime; results can often be seen within 30 min of urine sample procurement as purification of total DNA from urine is not required (Kersting et al., Reference Kersting, Rausch, Bier and von Nickisch-Rosenegk2014; Krõlov et al., Reference Krõlov, Frolova, Tudoran, Suhorutsenko, Lehto, Sibul, Mäger, Laanpere, Tulp and Langel2014; Rosser et al., Reference Rosser, Rollinson, Forrest and Webster2015; Rostron et al., Reference Rostron, Pennance, Bakar, Rollinson, Knopp, Allan, Kabole, Ali, Ame and Webster2019). Therefore, sample preparation is also less laborious and more field-applicable than these alternative DNA amplification methods as only crude preparations are needed. In addition, assay reagents can be lyophilised for easy transportation and can be stored without refrigeration even in tropical ambient temperatures for several weeks (Crannell et al., Reference Crannell, Rohrman and Richards-Kortum2014; Oriero et al., Reference Oriero, Jacobs, Van Geertruyden, Nwakanma and D'Alessandro2015).

The RPA assay is, however, much higher in cost when compared to alternative DNA amplification approaches. Estimated to cost between 4 USD$ and 5 USD$ per sample, the assay is currently too expensive for routine use in population-level control programmes within endemic areas (Minetti et al., Reference Minetti, Lacourse, Reimer and Stothard2016). With further development, adaptation and uptake of the assay, however, the cost per assay sample is very likely to decrease in the near future (Rosser et al., Reference Rosser, Rollinson, Forrest and Webster2015).

Further drawbacks to RPA include, like LAMP, the assay's low throughput when compared to alternative PCR-based methods, though this can be resolved through manufacture of larger-capacity devices. In addition, again like LAMP results, RPA results are only semi-quantitative, making estimations of infection intensity less reliable. As such, the assay in its current form may be best suited for small sample sizes and individual test-and-treat scenarios in low-endemicity settings.

Despite these disadvantages, the RPA is an extremely promising means of rapid, straightforward and sensitive diagnosis at the point-of-care in low-endemicity settings. Further development and validation of the RPA assay for use in diagnosing helminthiases using non-invasive urine sampling, is therefore recommended.

Novel biomarkers

Proteomic and metabolomic technologies can be used to screen bodily samples, including urine, to identify novel biomarkers that may potentially be used for diagnostic purposes. Using liquid chromatography and mass spectrometry, for example, it was recently reported that as many as 31 Schistosoma-derived proteins were differently abundant within the urine of patients infected with S. haematobium when compared to an uninfected control group and so may be used to detect active infection (Onile et al., Reference Onile, Calder, Soares, Anumudu and Blackburn2017). Here, it was also suggested that the presence and abundance of some transrenal host-derived proteins such as human growth/differentiation factor 15 (GDF15), upregulated in response to organ damage, may even provide a reliable means of determining disease severity and infection intensity, and so should be further evaluated.

Eosinophil cationic protein (ECP), involved in the body's immune response to foreign pathogens, has also been found to be significantly elevated in the urine of individuals infected with a variety of helminth species including S. haematobium, S. mansoni, O. volvulus, W. bancrofti, and hookworm (Tischendorf et al., Reference Tischendorf, Brattig, Burchard, Kubica, Kreuzpainer and Lintzel1999; Tischendorf et al., Reference Tischendorf, Brattig, Lintzel, Buttner, Burchard, Bork and Muller2000; Klion and Nutman, Reference Klion and Nutman2004; Fayez et al., Reference Fayez, Zaki, Elawady and El-Gebaly2010; Asuming-Brempong et al., Reference Asuming-Brempong, Gyan, Amoah, van der Puije, Bimi, Boakye and Ayi2015). In addition to GDF15, when assessing the use of transrenal ECP as a biomarker for infection with S. haematobium, a positive correlation between expelled ECP and urine egg count was found, suggesting urinal ECP may too increase with infection intensity and may therefore potentially be used to assess disease severity and worm burden (Leutscher et al., Reference Leutscher, van Dam, Reimert, Ramarakoto, Deelder and Ørnbjerg2008; Leutscher et al., Reference Leutscher, Reimert, Vennervald, Ravaoalimalala, Ramarokoto, Serieye, Raobelison, Rasendramino, Christensen and Esterre2000). These findings have since been replicated not only in S. haematobium, but also in S. mansoni infections (Asuming-Brempong et al., Reference Asuming-Brempong, Gyan, Amoah, van der Puije, Bimi, Boakye and Ayi2015). More recently, ECP levels in serum samples taken from individuals infected with hookworm have also been shown to positively correlate with infection intensity (Amoani et al., Reference Amoani, Adu, Frempong, Sarkodie-Addo, Nuvor, Wilson and Gyan2019). Repeated assessment using lesser-invasive urine samples was recommended.

Liquid chromatography has also been used in conjunction with infrared spectrophotometry to screen urine for expelled metabolites associated with helminth infections. Using this approach, it was reported that two metabolites, 2-methyl-butyramide and 2-methyl-valeramide, can be detected within the urine of individuals infected with Ascaris (Hall and Romanova., Reference Hall and Romanova1990). These findings have, however, recently been contested after neither of metabolite was detected in Indonesian individuals harbouring active Ascaris infections (Lagatie et al., Reference Lagatie, Njumbe Ediage, Pikkemaat, Djuardi and Stuyver2017). In addition to liquid chromatography, nuclear magnetic resonance (NMR) spectroscopy has been used to screen urine samples taken from mice experimentally infected with S. mansoni for expelled metabolites (Wang et al., Reference Wang, Holmes, Nicholson, Cloarec, Chollet, Tanner, Singer and Utzinger2004). A range of transrenal metabolites was associated with active infection and so warrant further exploration in human urine samples and in other helminth species infections.

Newly discovered transrenal biomarkers with the potential to indicate active infection, parasite burden and morbidity status may help to inform and shape future point-of-care diagnostic tools. The continued use of proteomic and metabolomic technologies for biomarker discovery is therefore strongly encouraged.

Discussion

Rapid, simple-to-use and low-cost diagnostic tools, deployable at the point-of-care and reliable in low-endemicity near-elimination settings, are urgently needed to help facilitate the elimination of debilitating parasitic helminth diseases. Current WHO-recommended gold standard assays do not meet these criteria and typically require invasive and potentially hazardous bodily samples. Many of these criteria are, however, met by non-invasive urine-based diagnostic assays capable of detecting a range of parasitic helminth species.

Macroscopic and microscopic changes to urine are not adequately sensitive to detect urogenital schistosomiasis in light-infections, preventing their use in near-elimination settings. In addition, although anti-helminth antibodies from a range of helminth species can be detected within the urine with high sensitivity, technical, financial and logistical challenges impede the reliability and routine use of urine-antibody diagnostic assays in helminth control programmes.

POC-RDT devices capable of detecting transrenal helminth-derived antigens may offer a simple, rapid, sensitive and low-cost diagnostic format at the point-of-care in high- and medium-prevalence settings. Although not currently adequately sensitive in low-endemicity settings or at the individual-level in patients burdened with light-infections, technological advancements and protocol revisions show promise for future improvements in POC-RDT diagnostic sensitivity that may facilitate their use in low-endemicity near-elimination settings.

Targeting transrenal helminth cfDNA is extremely sensitive and specific even in low-endemicity settings. The majority of assays capable of detecting urine-cfDNA are, however, both unsuited for point-of-care use and unaffordable to most control-programmes in disease-endemic areas. The recently developed LAMP and RPA assays may offer a promising, reliable and field-deployable means of detecting helminth-derived urine-cfDNA and, through further research, development and validation is needed before their routine application in disease-endemic areas, these assays have the potential for reliable test-and-treat use in low-endemicity near-elimination settings to rapidly identify lightly infected individuals capable of maintaining disease transmission.

The majority of current literature concerned with diagnosing helminthiases through urine sampling focuses primarily on the diagnosis of urogenital and intestinal schistosomiasis. As outlined here, many other helminth species, from a range of boldily habitats, can be detected through non-invasive urine sampling, particularly via targeting transrenal helminth-derived antigens and cell-free DNA. As such, the following research priorities are proposed:

  • To ascertain the detectable presence of transrenal antigens and cfDNA from all of the major human-infecting parasitic helminth species.

  • To determine any potential cross-reactivity of transrenal helminth-derived antigens with other antigens and/or proteins expelled in the urine and decipher how long helminth-derived cfDNA continues to be expelled within the urine after infection clearance.

  • Further development and validation of rapid diagnostic tests and field-deployable assays suitable for point-of-care use and able to reliably detect trace levels of helminth-derived urine-antigens and cfDNA known to be expelled in the urine. Assay assessment should not use traditional and unreliable gold standard techniques as reference, but rather more sensitive and specific assays, such as qPCR, as reference.

Despite the high financial costs associated with developing, validating and implementing novel diagnostic tools, the programmatic and economic benefits, as well as the health benefits to those in disease-endemic areas, gained from improved diagnostics capable of detecting even trace infections at the point-of-care will very likely outweigh any initial expenditure (Turner et al., Reference Turner, Bettis, Dunn, Whitton, Hollingsworth, Fleming and Anderson2017). The continued investment in and development of reliable, low-cost and non-invasive urine-based diagnostic assays deployable at the point-of-care is therefore highly encouraged.

Concluding remarks

Sensitive and specific diagnosis of many major parasitic helminthiases at the point-of-care is likely possible through non-invasive urine sampling. Though further development, assessment and validation are needed before their routine use in control programmes, low-cost and rapid assays capable of detecting transrenal helminth-derived antigens and cell-free DNA show promise for future use at the point-of-care in high-, medium- and even low-endemicity elimination settings. Ultimately, however, until these techniques are more affordable and easily implemented, less-reliable assays that require more invasive bodily samples will remain the diagnostic standard.

Acknowledgements

We wish to thank Mr Tom Pennance for his support in reading the manuscript as well as Alison Derbyshire of the Liverpool School of Tropical Medicine (LSTM) Library for essential training and extensive, valuable advice on systematic approaches to literature searching.

Financial support

None.

Conflict of interest

The authors declare that they have no competing interests.

Ethical standards

Not applicable.

Appendix

Fig. A1. Defining sensitivity, specificity and predictive values (Akobeng, Reference Akobeng2007; Bergquist et al., Reference Bergquist, Johansen and Utzinger2009).

References

Adriko, M, Standley, CJ, Tinkitina, B, Tukahebwa, EM, Fenwick, A, Fleming, FM, Sousa-Figueiredo, JC, Stothard, JR and Kabatereine, NB (2014) Evaluation of circulating cathodic antigen (CCA) urine-cassette assay as a survey tool for Schistosoma mansoni in different transmission settings within Bugiri district, Uganda. Acta Tropica 136, 5057.CrossRefGoogle ScholarPubMed
Ajibola, O, Gulumbe, BH, Eze, AA and Obishakin, E (2018) Tools for detection of Schistosomiasis in resource limited settings. Medical Sciences 6, 112. doi: 10.3390/medsci6020039.CrossRefGoogle ScholarPubMed
Akobeng, AK (2007) Understanding diagnostic tests 1: sensitivity, specificity and predictive values. Acta. Paediatricia 96, 333341. doi. 10.1111/j.1651-2227.2006.00180.Google ScholarPubMed
Akue, JP, Eyang-Assengone, E-R and Dieki, R (2018) Loa loa infection detection using biomarkers: current perspectives. Research and Reports in Tropical Medicine 9, 4348.CrossRefGoogle ScholarPubMed
Amoani, B, Adu, B, Frempong, MT, Sarkodie-Addo, T, Nuvor, SV, Wilson, MD and Gyan, B (2019) Levels of serum eosinophil cationic protein are associated with hookworm infection and intensity in endemic communities in Ghana. PLoS ONE 14, e0222382.CrossRefGoogle ScholarPubMed
Aryeetey, YA, Essien-Baidoo, S, Larbi, IA, Ahmed, K, Amoah, AS, Obeng, BB, van Lieshout, L, Yazdanbakhsh, M, Boakye, DA and Verweij, JJ (2013) Molecular diagnosis of Schistosoma infections in urine samples of school children in Ghana. The American Journal of Tropical Medicine and Hygiene 88, 10281031.CrossRefGoogle ScholarPubMed
Asuming-Brempong, E, Gyan, B, Amoah, A, van der Puije, W, Bimi, L, Boakye, D and Ayi, I (2015) Relationship between eosinophil cationic protein and infection intensity in a schistosomiasis endemic community in Ghana. Research and Reports in Tropical Medicine 1, 110. doi: 10.2147/RRTM.S51713.Google Scholar
Atalabi, TE, Adubi, TO and Lawal, U (2017) Rapid mapping of urinary schistosomiasis: an appraisal of the diagnostic efficacy of some questionnaire-based indices among high school students in Katsina state, northwestern Nigeria. PLOS Neglected Tropical Diseases 11, e0005518.CrossRefGoogle ScholarPubMed
Ayong, LS, Tume, CB, Wembe, FE, Simo, G, Asonganyi, T, Lando, G and Ngu, JL (2005) Development and evaluation of an antigen detection dipstick assay for the diagnosis of human onchocerciasis. Tropical Medicine & International Health: TM & IH 10, 228233.CrossRefGoogle Scholar
Bassiouny, HK, Hasab, AA, El-Nimr, NA, Al-Shibani, LA and Al-Waleedi, AA (2014) Rapid diagnosis of schistosomiasis in Yemen using a simple questionnaire and urine reagent strips. Eastern Mediterranean Health Journal = La Revue De Sante De La Mediterranee Orientale = Al-Majallah Al-Sihhiyah Li-Sharq Al-Mutawassit 20, 242249.Google ScholarPubMed
Bayoumi, A, Al-Refai, SA and Badr, MS (2016) Loop-Mediated isothermal amplification (LAMP): sensitive and rapid detection of Schistosoma Haematobium DNA in urine samples of Egyptian suspected cases. Journal of the Egyptian Society of Parasitology 46, 299308.Google ScholarPubMed
Bergquist, R, Johansen, MV and Utzinger, J (2009) Diagnostic dilemmas in helminthology: what tools to use and when? Trends in Parasitology 25, 151156.CrossRefGoogle ScholarPubMed
Bogoch, II, Andrews, JR, Dadzie Ephraim, RK and Utzinger, J (2012) Simple questionnaire and urine reagent strips compared to microscopy for the diagnosis of Schistosoma haematobium in a community in northern Ghana. Tropical Medicine & International Health: TM & IH 17, 12171221.CrossRefGoogle Scholar
Braun-Munzinger, RA and Southgate, BA (1992) Repeatability and reproducibility of egg counts of Schistosoma haematobium in urine. Tropical medicine and parasitology: official organ of Deutsche Tropenmedizinische Gesellschaft and of Deutsche Gesellschaft fur Technische Zusammenarbeit (GTZ) 43, 149154.Google ScholarPubMed
Cabada, MM, Malaga, JL, Castellanos-Gonzalez, A, Bagwell, KA, Naeger, PA, Rogers, HK, Maharsi, S, Mbaka, M and White, AC (2017) Recombinase polymerase amplification compared to real-time polymerase chain reaction test for the detection of Fasciola hepatica in human stool. The American Journal of Tropical Medicine and Hygiene 96, 341346.CrossRefGoogle ScholarPubMed
Castillo, Y, Rodriguez, S, García, HH, Brandt, J, Van Hul, A, Silva, M, Rodriguez-Hidalgo, R, Portocarrero, M, Melendez, DP, Gonzalez, AE, Gilman, RH and Dorny, P and Cysticercosis Working Group in Perú (2009). Urine antigen detection for the diagnosis of human neurocysticercosis. The American Journal of Tropical Medicine and Hygiene 80, 379383.CrossRefGoogle Scholar
Chanteau, S, Moulia-Pelat, JP, Glaziou, P, Nguyen, NL, Luquiaud, P, Plichart, C, Martin, PM and Cartel, JL (1994) Og4c3 circulating antigen: a marker of infection and adult worm burden in Wuchereria bancrofti filariasis. The Journal of Infectious Diseases 170, 247250.CrossRefGoogle ScholarPubMed
Chenthamarakshan, V, Padigel, UM, Ramaprasad, P, Reddy, MVR and Harinath, BC (1993) Diagnostic utility of fractionated urinary filarial antigen. Journal of Biosciences 18, 319326.CrossRefGoogle Scholar
Chenthamarakshan, V, Reddy, MVR and Harinath, BC (1996) Detection of filarial antigen by inhibition enzyme linked immunosorbent assay using fractionated Brugia malayi microfilarial excretory secretory antigen. Journal of Biosciences 21, 2734.CrossRefGoogle Scholar
Christensen, EE, Taylor, M, Zulu, SG, Lillebo, K, Gundersen, SG, Holmen, S, Kleppa, E, Vennervald, BJ, Ndhlovu, PD and Kjetland, EF (2018) Seasonal variations in Schistosoma haematobium egg excretion in school-age girls in rural KwaZulu-Natal Province, South Africa. South African Medical Journal = Suid-Afrikaanse Tydskrif Vir Geneeskunde 108, 352355.Google ScholarPubMed
Cochrane Library. (2019). How to use the Cochrane Library. St Albans House, 57–59. Haymarket, St. James's, London, SW1Y 4QX. Website: https://www.cochranelibrary.com/help/how-to-use/ (Accessed 1 September 2019).Google Scholar
Coelho, PMZ, Siqueira, LMV, Grenfell, RFQ, Almeida, NBF, Katz, N, Almeida, Á, Carneiro, NFdF and Oliveira, E (2016) Improvement of POC-CCA interpretation by using lyophilization of urine from patients with Schistosoma mansoni Low worm burden: towards an elimination of doubts about the concept of trace. PLoS Neglected Tropical Diseases 10, e0004778.CrossRefGoogle ScholarPubMed
Colley, DG, Bustinduy, AL, Secor, WE and King, CH (2014) Human schistosomiasis. The Lancet 383, 22532264.CrossRefGoogle ScholarPubMed
Corstjens, PLAM, De Dood, CJ, Kornelis, D, Fat, EMTK, Wilson, RA, Kariuki, TM, Nyakundi, RK, Loverde, PT, Abrams, WR, Tanke, HJ, Van Lieshout, L, Deelder, AM and Van Dam, GJ (2014) Tools for diagnosis, monitoring and screening of Schistosoma infections utilizing lateral-flow based assays and upconverting phosphor labels. Parasitology 141, 18411855.CrossRefGoogle ScholarPubMed
Corstjens, PLAM, Hoekstra, PT, de Dood, CJ and van Dam, GJ (2017) Corstjens, P. L. A. M., hoekstra, P. T., de dood, C. J. and van Dam, G. J. (2017). utilizing the ultrasensitive Schistosoma up-converting phosphor lateral flow circulating anodic antigen (UCP-LF CAA) assay for sample pooling-strategies. Infectious Diseases of Poverty 6, 155.CrossRefGoogle Scholar
Coulibaly, JT, N'Goran, EK, Utzinger, J, Doenhoff, MJ and Dawson, EM (2013) A new rapid diagnostic test for detection of anti-Schistosoma mansoni and anti-Schistosoma haematobium antibodies. Parasites & Vectors 6, 29.CrossRefGoogle ScholarPubMed
Crannell, ZA, Rohrman, B and Richards-Kortum, R (2014) Equipment-free incubation of recombinase polymerase amplification reactions using body heat. PLoS ONE 9, e112146.CrossRefGoogle ScholarPubMed
Crannell, Z, Castellanos-Gonzalez, A, Nair, G, Mejia, R, White, AC and Richards-Kortum, R (2016) Multiplexed recombinase polymerase amplification assay to detect intestinal protozoa. Analytical Chemistry 88, 16101616.CrossRefGoogle ScholarPubMed
Danso-Appiah, A, Minton, J, Boamah, D, Otchere, J, Asmah, RH, Rodgers, M, Bosompem, KM, Eusebi, P and De Vlas, SJ (2016) Accuracy of point-of-care testing for circulatory cathodic antigen in the detection of schistosome infection: systematic review and meta-analysis. Bulletin of the World Health Organization 94, 522533A.CrossRefGoogle ScholarPubMed
de Dood, CJ, Hoekstra, PT, Mngara, J, Kalluvya, SE, van Dam, GJ, Downs, JA and Corstjens, PLAM (2018) Refining diagnosis of Schistosoma haematobium infections: antigen and antibody detection in urine. Frontiers in Immunology 9, 19. doi: 10.3389/fimmu.2018.02635.CrossRefGoogle ScholarPubMed
Deng, M-H, Zhong, L-Y, Kamolnetr, O, Limpanont, Y and Lv, Z-Y (2019) Detection of helminths by loop-mediated isothermal amplification assay: a review of updated technology and future outlook. Infectious Diseases of Poverty 8, 20.CrossRefGoogle ScholarPubMed
Doenhoff, MJ, Chiodini, PL and Hamilton, JV (2004) Specific and sensitive diagnosis of schistosome infection: can it be done with antibodies? Trends in Parasitology 20, 3539.CrossRefGoogle ScholarPubMed
Drame, PM, Meng, Z, Bennuru, S, Herrick, JA, Veenstra, TD and Nutman, TB (2016) Identification and validation of Loa Loa microfilaria-specific biomarkers: a rational design approach using proteomics and novel immunoassays. mBio 7, e02132–15, /mbio/7/1/e02132-15.atom.CrossRefGoogle ScholarPubMed
Eamudomkarn, C, Sithithaworn, P, Kamamia, C, Yakovleva, A, Sithithaworn, J, Kaewkes, S, Techasen, A, Loilome, W, Yongvanit, P, Wangboon, C, Saichua, P, Itoh, M and Bethony, MJ (2018) Diagnostic performance of urinary IgG antibody detection: a novel approach for population screening of strongyloidiasis. PLoS ONE 13, e0192598.CrossRefGoogle ScholarPubMed
Elhag, SM, Abdelkareem, EA, Yousif, AS, Frah, EA and Mohamed, AB (2011) Detection of schistosomiasis antibodies in urine patients as a promising diagnostic maker. Asian Pacific Journal of Tropical Medicine 4, 773777.CrossRefGoogle ScholarPubMed
Enk, MJ, Oliveira e Silva, G and Rodrigues, NB (2010) A salting out and resin procedure for extracting Schistosoma mansoni DNA from human urine samples. BMC Research Notes 3, 115.CrossRefGoogle ScholarPubMed
Enk, MJ, Oliveira e Silva, G and Rodrigues, NB (2012) Diagnostic accuracy and applicability of a PCR system for the detection of Schistosoma mansoni DNA in human urine samples from an endemic area. PLoS ONE 7, e38947.CrossRefGoogle ScholarPubMed
Fayez, S, Zaki, MM, Elawady, AA and El-Gebaly, NSM (2010) Assessment of the Role of Serum and Urine Eosinophil Cationic Protein in Diagnosis of Wuchereria bancrofti Infection. 9.Google Scholar
Fernández-Soto, P, Gandasegui Arahuetes, J, Sánchez Hernández, A, López Abán, J, Vicente Santiago, B and Muro, A (2014) A loop-mediated isothermal amplification (LAMP) assay for early detection of Schistosoma mansoni in stool samples: a diagnostic approach in a murine model. PLoS Neglected Tropical Diseases 8, e3126.CrossRefGoogle ScholarPubMed
Fernández-Soto, P, Sánchez-Hernández, A, Gandasegui, J, Bajo Santos, C, López-Abán, J, Saugar, JM, Rodríguez, E, Vicente, B and Muro, A (2016) Strong-LAMP: a LAMP assay for Strongyloides spp. Detection in stool and urine samples. Towards the diagnosis of human strongyloidiasis starting from a rodent model. PLoS Neglected Tropical Diseases 10, e0004836.CrossRefGoogle Scholar
Fernández-Soto, P, Gandasegui, J, Carranza Rodríguez, C, Pérez-Arellano, JL, Crego-Vicente, B, García-Bernalt Diego, J, López-Abán, J, Vicente, B and Muro, A (2019) Detection of Schistosoma mansoni-derived DNA in human urine samples by loop-mediated isothermal amplification (LAMP). PLoS ONE 14, e0214125.CrossRefGoogle Scholar
Garcia, HH, Castillo, Y, Gonzales, I, Bustos, JA, Saavedra, H, Jacob, L, Del Brutto, OH, Wilkins, PP, Gonzalez, AE and Gilman, RH and Cysticercosis Working Group in Peru (2018). Low sensitivity and frequent cross-reactions in commercially available antibody detection ELISA assays for taenia solium cysticercosis. Tropical Medicine & International Health: TM & IH 23, 101105.CrossRefGoogle ScholarPubMed
Gardon, J, Gardon-Wendel, N, Demanga-Ngangue, N, Kamgno, J, Chippaux, JP and Boussinesq, M (1997) Serious reactions after mass treatment of onchocerciasis with ivermectin in an area endemic for Loa loa infection. Lancet (London, England) 350, 1822.CrossRefGoogle Scholar
Genta, RM (1988) Predictive value of an enzyme-linked immunosorbent assay (ELISA) for the serodiagnosis of strongyloidiasis. American Journal of Clinical Pathology 89, 391394.CrossRefGoogle ScholarPubMed
Google Scholar (2019) 1600 Amphitheatre Parkway, California, USA: Mountain View. Website: https://scholar.google.com (Accessed 1 September 2019).Google Scholar
Gordon, CA, Gray, DJ, Gobert, GN and McManus, DP (2011) DNA Amplification approaches for the diagnosis of key parasitic helminth infections of humans. Molecular and Cellular Probes 25, 143152.CrossRefGoogle Scholar
Hall, A and Romanova, T (1990) Ascaris lumbricoides: detecting its metabolites in the urine of infected people using gas-liquid chromatography. Exp Parasitol. 70, 2542.CrossRefGoogle ScholarPubMed
Hamburger, J, Turetski, T, Kapeller, I and Deresiewicz, R (1991) Highly repeated short DNA sequences in the genome of Schistosoma mansoni recognised by a species-specific probe. Molecular and Biochemical Parasitology 44, 7380. doi:10.1016/0166-6851(91)90222-R.CrossRefGoogle Scholar
Hamburger, J, Abbasi, I, Kariuki, C, Wanjala, A, Mzungu, E, Mungai, P, Muchiri, E and King, CH (2013) Evaluation of loop-mediated isothermal amplification suitable for molecular monitoring of schistosome-infected snails in field laboratories. The American Journal of Tropical Medicine and Hygiene 88, 344351. doi: 10.4269/ajtmh.2012.12-0208.CrossRefGoogle ScholarPubMed
Hassan, J, Mohammed, K, Opaluwa, S, Adamu, T, Nataala, S, Garba, M, Bello, M and Bunza, N (2018) Diagnostic potentials of haematuria and proteinuria in urinary schistosomiasis among school-Age children in aliero local government area, kebbi state, north-western Nigeria. Asian Journal of Research in Medical and Pharmaceutical Sciences 2, 19.CrossRefGoogle Scholar
Hawkins, KR, Cantera, JL, Storey, HL, Leader, BT and de los Santos, T (2016) Diagnostic tests to support late-stage control programs for schistosomiasis and soil-transmitted helminthiases. PLoS Neglected Tropical Diseases 10, 115. doi: 10.1371/journal.pntd.0004985.CrossRefGoogle ScholarPubMed
Henry, D, Dessaint, JP, Wandji, K and Centre, AC (1987) Lymphatic filariasis: detection of circulating and urinary antigen and differences in antibody isotypes complexed with circulating antigen between symptomatic and asymptomatic subjects. Clinical Experimental Immunology 71, 253260, PMC 3280185.Google Scholar
Hertz, MI, Nana-Djeunga, H, Kamgno, J, Jelil Njouendou, A, Chawa Chunda, V, Wanji, S, Rush, A, Fischer, PU, Weil, GJ and Budge, PJ (2018) Identification and characterization of Loa loa antigens responsible for cross-reactivity with rapid diagnostic tests for lymphatic filariasis. PLoS Neglected Tropical Diseases 12, e0006963.CrossRefGoogle ScholarPubMed
Hotez, PJ, Brindley, PJ, Bethony, JM, King, CH, Pearce, EJ and Jacobson, J (2008) Helminth infections: the great neglected tropical diseases. The Journal of Clinical Investigation 118, 13111321.CrossRefGoogle ScholarPubMed
Huijun, Z, Zhenghou, T, Reddy, MVR, Harinath, BC and Piessens, WF (1987) Parasite antigens in Sera and urine of patients with Bancroftian and Brugian Filariasis detected by sandwich Elisa with monoclonal antibodies. The American Journal of Tropical Medicine and Hygiene 36, 554560.CrossRefGoogle Scholar
Ibironke, O, Koukounari, A, Asaolu, S, Moustaki, I and Shiff, C (2012) Validation of a new test for Schistosoma haematobium based on detection of Dra1 DNA fragments in urine: evaluation through latent class analysis. PLoS Neglected Tropical Diseases 6, e1464.CrossRefGoogle ScholarPubMed
Itoh, M, Kimura, E, Fujimaki, Y, Weerasooriya, MV, Islam, MZ and Qiu, X-G (2003a) Prevalence and levels of filaria-specific urinary IgG4 among children less than five years of age and the association of antibody positivity between children and their mothers. The American Journal of Tropical Medicine and Hygiene 68, 465468.Google Scholar
Itoh, M, Ohta, N, Kanazawa, T, Nakajima, Y, Sho, M, Minai, M, Daren, Z, Chen, Y, He, H, He, Y-K and Zhong, Z (2003b) Sensitive enzyme-linked immunosorbent assay with urine samples: a tool for surveillance of schistosomiasis japonica. The Southeast Asian Journal of Tropical Medicine and Public Health 34, 469472.Google Scholar
Itoh, M, Wu, W, Sun, D, Yao, L, Li, Z, Islam, MZ, Chen, R, Zhang, K, Wang, F, Zhu, S and Kimura, E (2007) Confirmation of elimination of lymphatic filariasis by an IgG4 enzyme-linked immunosorbent assay with urine samples in Yongjia, Zhejiang province and Gaoan, Jiangxi province, People's Republic of China. The American Journal of Tropical Medicine and Hygiene 77, 330333.CrossRefGoogle ScholarPubMed
Itoh, M, Weerasooriya, MV, Yahathugoda, TC, Takagi, H, Samarawickrema, WA, Nagaoka, F and Kimura, E (2011) Effects of 5 rounds of mass drug administration with diethylcarbamazine and albendazole on filaria-specific IgG4 titers in urine: 6-year follow-up study in Sri Lanka. Parasitology International 60, 393397.CrossRefGoogle ScholarPubMed
Jourdan, PM, Lamberton, PHL, Fenwick, A and Addiss, DG (2018) Soil-transmitted helminth infections. The Lancet 391, 252265.CrossRefGoogle ScholarPubMed
Kamel, M, Salah, F, Demerdash, Z, Maher, S, Atta, S, Badr, A, Afifi, A and El Baz, H (2019) Development of new lateral-flow immunochromatographic strip using colloidal gold and mesoporous silica nanoparticles for rapid diagnosis of active schistosomiasis. Asian Pacific Journal of Tropical Biomedicine 9, 315.Google Scholar
Kanjanavas, P, Tan-ariya, P, Khawsak, P, Pakpitcharoen, A, Phantana, S and Chansiri, K (2005) Detection of lymphatic Wuchereria bancrofti in carriers and long-term storage blood samples using semi-nested PCR. Molecular and Cellular Probes 19(3), 169172. doi: 10.1016/j.mcp.2004.11.003.CrossRefGoogle ScholarPubMed
Kato-Hayashi, N, Leonardo, LR, Arevalo, NL, Tagum, MNB, Apin, J, Agsolid, LM, Chua, JC, Villacorte, EA, Kirinoki, M, Kikuchi, M, Ohmae, H, Haruki, K and Chigusa, Y (2015) Detection of active schistosome infection by cell-free circulating DNA of Schistosoma japonicum in highly endemic areas in Sorsogon Province, the Philippines. Acta Tropica 141, 178183.CrossRefGoogle ScholarPubMed
Kayuni, S, Lampiao, F, Makaula, P, Juziwelo, L, Lacourse, EJ, Reinhard-Rupp, J, Leutscher, PDC and Stothard, JR (2019) A systematic review with epidemiological update of male genital schistosomiasis (MGS): a call for integrated case management across the health system in sub-Saharan Africa. Parasite Epidemiology and Control 4, e00077.CrossRefGoogle Scholar
Kemal, J, Alemu, S, Yimer, M and Terefe, G (2015) Immunological and molecular diagnostic tests for Cestodes and metacestodes: review. World Applied Sciences Journal 33, 18671879.Google Scholar
Kersting, S, Rausch, V, Bier, FF and von Nickisch-Rosenegk, M (2014) Rapid detection of Plasmodium falciparum with isothermal recombinase polymerase amplification and lateral flow analysis. Malaria Journal 13, 99.CrossRefGoogle ScholarPubMed
King, CH and Bertsch, D (2013) Meta-analysis of urine heme dipstick diagnosis of Schistosoma haematobium infection, including low-prevalence and previously-treated populations. PLoS Neglected Tropical Diseases 7, e2431.CrossRefGoogle ScholarPubMed
Klepac, P, Metcalf, CJE, McLean, AR and Hampson, K (2013) Towards the endgame and beyond: complexities and challenges for the elimination of infectious diseases. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences 368, 20120137.CrossRefGoogle ScholarPubMed
Klion, AD and Nutman, TB (2004) The role of eosinophils in host defense against helminth parasites. Journal of Allergy and Clinical Immunology 113, 3037.CrossRefGoogle ScholarPubMed
Knopp, S, Steinmann, P, Hatz, C, Keiser, J and Utzinger, J (2012) Nematode infections: filariases. Infectious Disease Clinics of North America 26, 359381.CrossRefGoogle ScholarPubMed
Knopp, S, Becker, SL, Ingram, KJ, Keiser, J and Utzinger, J (2013) Diagnosis and treatment of schistosomiasis in children in the era of intensified control. Expert Review of Anti-Infective Therapy 11, 12371258.CrossRefGoogle ScholarPubMed
Knopp, S, Corstjens, PLAM, Koukounari, A, Cercamondi, CI, Ame, SM, Ali, SM, de Dood, CJ, Mohammed, KA, Utzinger, J, Rollinson, D and van Dam, GJ (2015) Sensitivity and specificity of a urine circulating anodic antigen test for the diagnosis of Schistosoma haematobium in low endemic settings. PLOS Neglected Tropical Diseases 9, e0003752.CrossRefGoogle Scholar
Knopp, S, Ame, SM, Hattendorf, J, Ali, SM, Khamis, IS, Bakar, F, Khamis, MA, Person, B, Kabole, F and Rollinson, D (2018) Urogenital schistosomiasis elimination in Zanzibar: accuracy of urine filtration and haematuria reagent strips for diagnosing light intensity Schistosoma haematobium infections. Parasites & Vectors 11, 552.CrossRefGoogle ScholarPubMed
Krauth, SJ, Greter, H, Stete, K, Coulibaly, JT, Traoré, SI, Ngandolo, BNR, Achi, LY, Zinsstag, J, N'Goran, EK and Utzinger, J (2015) All that is blood is not schistosomiasis: experiences with reagent strip testing for urogenital schistosomiasis with special consideration to very-low prevalence settings. Parasites & Vectors 8, 584.CrossRefGoogle Scholar
Krolewiecki, AJ, Koukounari, A, Romano, M, Caro, RN, Scott, AL, Fleitas, P, Cimino, R and Shiff, CJ (2018) Transrenal DNA-based diagnosis of Strongyloides stercoralis (grassi, 1879) infection: bayesian latent class modeling of test accuracy. PLOS Neglected Tropical Diseases 12, e0006550.CrossRefGoogle ScholarPubMed
Krõlov, K, Frolova, J, Tudoran, O, Suhorutsenko, J, Lehto, T, Sibul, H, Mäger, I, Laanpere, M, Tulp, I and Langel, Ü (2014) Sensitive and rapid detection of Chlamydia trachomatis by recombinase polymerase amplification directly from urine samples. The Journal of Molecular Diagnostics 16, 127135.CrossRefGoogle ScholarPubMed
Kukula, VA, MacPherson, EE, Tsey, IH, Stothard, JR, Theobald, S and Gyapong, M (2019) A major hurdle in the elimination of urogenital schistosomiasis revealed: identifying key gaps in knowledge and understanding of female genital schistosomiasis within communities and local health workers. PLOS Neglected Tropical Diseases 13, e0007207.CrossRefGoogle Scholar
Lagatie, O, Merino, M, Batsa Debrah, L, Debrah, AY and Stuyver, LJ (2016) An isothermal DNA amplification method for detection of Onchocerca volvulus infection in skin biopsies. Parasites & Vectors 9, 624.CrossRefGoogle ScholarPubMed
Lagatie, O, Njumbe Ediage, E, Pikkemaat, JA, Djuardi, Y and Stuyver, LJ (2017) 2-methyl Butyramide, a previously identified urine biomarker for Ascaris lumbricoides, is not present in infected Indonesian individuals. Parasites & Vectors 10, 629.CrossRefGoogle Scholar
Lai, M-Y, Ooi, C-H and Lau, Y-L (2017) Rapid detection of Plasmodium knowlesi by isothermal recombinase polymerase amplification assay. The American Journal of Tropical Medicine and Hygiene 97, 15971599.CrossRefGoogle ScholarPubMed
Lamberton, PHL and Jourdan, PM (2015) Human ascariasis: diagnostics update. Current Tropical Medicine Reports 2, 189200.CrossRefGoogle ScholarPubMed
Lammie, PJ, Weil, G, Noordin, R, Kaliraj, P, Steel, C, Goodman, D, Lakshmikanthan, VB and Ottesen, E (2004) Recombinant antigen-based antibody assays for the diagnosis and surveillance of lymphatic filariasis–a multicenter trial. Filaria Journal 3, 9.CrossRefGoogle ScholarPubMed
Lazcka, O, Del Campo, FJ and Muñoz, FX (2007) Pathogen detection: a perspective of traditional methods and biosensors. Biosensors & Bioelectronics 22, 12051217.CrossRefGoogle ScholarPubMed
Le, L and Hsieh, MH (2017) Diagnosing urogenital schistosomiasis: dealing with diminishing returns. Trends in Parasitology 33, 378387.CrossRefGoogle ScholarPubMed
Lengeler, C, Utzinger, J and Tanner, M (2002a) Questionnaires for rapid screening of schistosomiasis in sub-Saharan Africa. Bulletin of the World Health Organization 80, 235242.Google Scholar
Lengeler, C, Utzinger, J and Tanner, M (2002b) Screening for schistosomiasis with questionnaires. Trends in Parasitology 18, 375377.CrossRefGoogle Scholar
Leutscher, PDC, Reimert, CM, Vennervald, BJ, Ravaoalimalala, VE, Ramarokoto, CE, Serieye, J, Raobelison, A, Rasendramino, M, Christensen, NO and Esterre, P (2000) Morbidity assessment in urinary schistosomiasis infection through ultrasonography and measurement of eosinophil cationic protein (ECP) in urine. Tropical Medicine and International Health 5, 8893.CrossRefGoogle ScholarPubMed
Leutscher, PDC, van Dam, GTJ, Reimert, CM, Ramarakoto, CE, Deelder, AM and Ørnbjerg, N (2008) Eosinophil Cationic Protein, Soluble Egg Antigen, Circulating Anodic Antigen, and Egg Excretion in Male Urogenital Schistosomiasis. The American Journal of Tropical Medicine and Hygiene 79, 422426. doi: 10.4269/ajtmh.2008.79.422.CrossRefGoogle ScholarPubMed
Li, J, Macdonald, J and von Stetten, F (2019) Review: a comprehensive summary of a decade development of the recombinase polymerase amplification. The Analyst 144, 3167.CrossRefGoogle Scholar
Lodh, N, Mwansa, JCL, Mutengo, MM and Shiff, CJ (2013) Diagnosis of Schistosoma mansoni without the stool: comparison of three diagnostic tests to detect Schistosoma [corrected] mansoni infection from filtered urine in Zambia. The American Journal of Tropical Medicine and Hygiene 89, 4650.CrossRefGoogle ScholarPubMed
Lodh, N, Naples, JM, Bosompem, KM, Quartey, J and Shiff, CJ (2014) Detection of parasite-specific DNA in urine sediment obtained by filtration differentiates between single and mixed infections of Schistosoma mansoni and S. haematobium from endemic areas in Ghana. PLoS ONE 9, e91144.CrossRefGoogle ScholarPubMed
Lodh, N, Caro, R, Sofer, S, Scott, A, Krolewiecki, A and Shiff, C (2016) Diagnosis of Strongyloides stercoralis: detection of parasite-derived DNA in urine. Acta Tropica 163, 913.CrossRefGoogle ScholarPubMed
Lodh, N, Mikita, K, Bosompem, KM, Anyan, WK, Quartey, JK, Otchere, J and Shiff, CJ (2017) Point of care diagnosis of multiple schistosome parasites: species-specific DNA detection in urine by loop-mediated isothermal amplification (LAMP). Acta Tropica 173, 125129.CrossRefGoogle Scholar
Lucena, WA, Dhalia, R, Abath, FG, Nicolas, L, Regis, LN and Furtado, AF (1998) Diagnosis of Wuchereria bancrofti infection by the polymerase chain reaction using urine and day blood samples from amicrofilaraemic patients. Transactions of the Royal Society of Tropical Medicine and Hygiene 92, 290293.CrossRefGoogle ScholarPubMed
Lustigman, S, Prichard, RK, Gazzinelli, A, Grant, WN, Boatin, BA, McCarthy, JS and Basáñez, M-G (2012) A research agenda for helminth diseases of humans: the problem of helminthiases. PLOS Neglected Tropical Diseases 6, e1582.CrossRefGoogle ScholarPubMed
McCarthy, JS, Lustigman, S, Yang, G-J, Barakat, RM, García, HH, Sripa, B, Willingham, AL, Prichard, RK and Basáñez, M-G (2012) A research agenda for helminth diseases of humans: diagnostics for control and elimination programmes. PLoS Neglected Tropical Diseases 6, e1601.CrossRefGoogle ScholarPubMed
Melchers, NVSV, van Dam, GJ, Shaproski, D, Kahama, AI, Brienen, EAT, Vennervald, BJ and van Lieshout, L (2014) Diagnostic performance of Schistosoma real-time PCR in urine samples from Kenyan children infected with Schistosoma haematobium: day-to-day variation and follow-up after praziquantel treatment. PLOS Neglected Tropical Diseases 8, e2807.CrossRefGoogle Scholar
Minetti, C, Lacourse, EJ, Reimer, L and Stothard, JR (2016) Focusing nucleic acid-based molecular diagnostics and xenomonitoring approaches for human helminthiases amenable to preventive chemotherapy. Parasitology Open 2, e16.CrossRefGoogle Scholar
Montresor, A, Crompton, DWT, Gyorkos, TW, Savioli, L and Organization, WH (2002) Helminth Control in School-Age Children: A Guide for Managers of Control Programmes. Second edition. Geneva, Switzerland: World Health Organization.Google Scholar
Musa, NY and Dadah, AJ (2018) Comparing the sensitivity of microscopy to diagnostic strip in the survey for urinary schistosomiasis. Bioprocess Engineering 2, 14.CrossRefGoogle Scholar
Mutapi, F, Maizels, R, Fenwick, A and Woolhouse, M (2017) Human schistosomiasis in the post mass drug administration era. The Lancet Infectious Diseases 17, e42e48.CrossRefGoogle ScholarPubMed
Mwape, KE, Praet, N, Benitez-Ortiz, W, Muma, JB, Zulu, G, Celi-Erazo, M, Phiri, IK, Rodriguez-Hidalgo, R, Dorny, P and Gabriël, S (2011) Field evaluation of urine antigen detection for diagnosis of Taenia solium cysticercosis. Transactions of the Royal Society of Tropical Medicine and Hygiene 105, 10. 574578. doi: 10.1016/j.trstmh.2011.05.010.CrossRefGoogle ScholarPubMed
Nagaoka, F, Itoh, M, Samad, MS, Takagi, H, Weerasooriya, MV, Yahathugoda, TC, Hossain, M, Moji, K and Kimura, E (2013) Visual detection of filaria-specific IgG4 in urine using red-colored high density latex beads. Parasitology International 62, 3235.CrossRefGoogle ScholarPubMed
National Center for Biotechnology Information (2019) Using PubMed. U.S. National Library of Medicine. 8600 Rockville Pike, Bethesda MD, 20894. USA. Website: https://www.ncbi.nlm.nih.gov/pubmed/ (Accessed 1 September 2019).Google Scholar
Ochodo, EA, Gopalakrishna, G, Spek, B, Reitsma, JB, van Lieshout, L, Polman, K, Lamberton, P, Bossuyt, PMM and Leeflang, MMG (2015) Circulating antigen tests and urine reagent strips for diagnosis of active schistosomiasis in endemic areas. The Cochrane Database of Systematic Reviews 3, CD009579. doi: 10.1002/14651858.CD009579.pub2.Google Scholar
Okeke, OC and Ubachukwu, PO (2014) Performance of three rapid screening methods in the detection of Schistosoma haematobium infection in school-age children in southeastern Nigeria. Pathogens and Global Health 108, 111117.CrossRefGoogle ScholarPubMed
Onile, OS, Calder, B, Soares, NC, Anumudu, CI and Blackburn, JM (2017) Quantitative label-free proteomic analysis of human urine to identify novel candidate protein biomarkers for schistosomiasis. PLoS Neglected Tropical Diseases 11, e0006045.CrossRefGoogle ScholarPubMed
Oriero, EC, Jacobs, J, Van Geertruyden, J-P, Nwakanma, D and D'Alessandro, U (2015) Molecular-based isothermal tests for field diagnosis of malaria and their potential contribution to malaria elimination. Journal of Antimicrobial Chemotherapy 70, 213.CrossRefGoogle ScholarPubMed
Oyola, SO, Otto, TD, Gu, Y, Maslen, G, Manske, M, Campino, S, Turner, DJ, MacInnis, B, Kwiatkowski, DP, Swerdlow, HP and Quail, MA (2012) Optimizing illumina next-generation sequencing library preparation for extremely at-biased genomes. BMC Genomics 13, 1.CrossRefGoogle ScholarPubMed
Paredes, A, Sáenz, P, Marzal, MW, Orrego, MA, Castillo, Y, Rivera, A, Mahanty, S, Guerra-Giraldez, C, García, HH and Nash, TE (2016) Anti-Taenia solium monoclonal antibodies for the detection of parasite antigens in body fluids from patients with neurocysticercosis. Experimental Parasitology 166, 3743. doi: 10.1016/j.exppara.2016.03.025.CrossRefGoogle ScholarPubMed
Peralta, JM and Cavalcanti, MG (2018) Is POC-CCA a truly reliable test for schistosomiasis diagnosis in low endemic areas? The trace results controversy. PLoS Neglected Tropical Diseases 12, e0006813.CrossRefGoogle ScholarPubMed
Peters, PA, Warren, KS and Mahmoud, AA (1976) Rapid, accurate quantification of schistosome eggs Via Nuclepore filters. The Journal of Parasitology 62, 154155.CrossRefGoogle ScholarPubMed
Phuphisut, O, Yoonuan, T, Sanguankiat, S, Chaisiri, K, Maipanich, W, Pubampen, S, Komalamisra, C and Adisakwattana, P (2014) Ascaris lumbricoides, trichuris trichiura, necator americanus, in fecal samples. Southeast Asian Journal of Tropical Medicine and Public Health 45, 9.Google ScholarPubMed
Piepenburg, O, Williams, CH, Stemple, DL and Armes, NA (2006) DNA Detection using recombination proteins. PLoS Biology 4, e204.CrossRefGoogle ScholarPubMed
Poulton, K and Webster, B (2018) Development of a lateral flow recombinase polymerase assay for the diagnosis of Schistosoma mansoni infections. Analytical Biochemistry 546, 6571.CrossRefGoogle Scholar
Ramaprasad, P, Prasad, GB and Harinath, BC (1988) Microfilaraemia, filarial antibody, antigen and immune complex levels in human filariasis before, during and after DEC therapy. A two-year follow-up. Acta Tropica 45, 245255.Google ScholarPubMed
Rattanaxay, P, Gunawardena, NK, Itoh, M, Fujimaki, Y, Anantaphruti, MT, Weerasooriya, MV, Kimura, E, Tesana, S and Qiu, G (2001) Sensitive and specific enzyme-linked immunosorbent assay for the diagnosis of Wuchereria bancrofti infection in urine samples. The American Journal of Tropical Medicine and Hygiene 65, 362365.Google Scholar
Rebollo, MP and Bockarie, MJ (2014) Shrinking the lymphatic filariasis map: update on diagnostic tools for mapping and transmission monitoring. Parasitology 141, 19121917.CrossRefGoogle ScholarPubMed
Robinson, E, Picon, D, Sturrock, HJ, Sabasio, A, Lado, M, Kolaczinski, J and Brooker, S (2009) The performance of haematuria reagent strips for the rapid mapping of urinary schistosomiasis: field experience from southern Sudan. Tropical Medicine & International Health 14, 14841487.CrossRefGoogle ScholarPubMed
Rollinson, D, Klinger, EV, Mgeni, AF, Khamis, IS and Stothard, JR (2005) Urinary schistosomiasis on Zanzibar: application of two novel assays for the detection of excreted albumin and haemoglobin in urine. Journal of Helminthology 79, 199206.CrossRefGoogle ScholarPubMed
Rollinson, D, Knopp, S, Levitz, S, Stothard, JR, Tchuem Tchuenté, L-A, Garba, A, Mohammed, KA, Schur, N, Person, B, Colley, DG and Utzinger, J (2013) Time to set the agenda for schistosomiasis elimination. Acta Tropica 128, 423440.CrossRefGoogle ScholarPubMed
Rosser, A, Rollinson, D, Forrest, M and Webster, BL (2015) Isothermal recombinase polymerase amplification (RPA) of Schistosoma haematobium DNA and oligochromatographic lateral flow detection. Parasites & Vectors 8, 446.CrossRefGoogle ScholarPubMed
Rostron, P, Pennance, T, Bakar, F, Rollinson, D, Knopp, S, Allan, F, Kabole, F, Ali, SM, Ame, SM and Webster, BL (2019) Development of a recombinase polymerase amplification (RPA) fluorescence assay for the detection of Schistosoma Haematobium. Parasites & Vectors 12, 17. doi: 10.1186/s13071-019-3755-6.CrossRefGoogle ScholarPubMed
Sady, H, Al-Mekhlafi, HM, Ngui, R, Atroosh, WM, Al-Delaimy, AK, Nasr, NA, Dawaki, S, Abdulsalam, AM, Ithoi, I, Lim, YAL, Chua, KH and Surin, J (2015) Detection of Schistosoma mansoni and Schistosoma haematobium by real-time PCR with high resolution melting analysis. International Journal of Molecular Sciences 16, 1608516103.CrossRefGoogle ScholarPubMed
Samad, MS, Itoh, M, Moji, K, Hossain, M, Mondal, D, Alam, MS and Kimura, E (2013) Enzyme-linked immunosorbent assay for the diagnosis of Wuchereria bancrofti infection using urine samples and its application in Bangladesh. Parasitology International 62, 564567.CrossRefGoogle Scholar
Sandoval, N, Siles-Lucas, M, Pérez-Arellano, JL, Carranza, C, Puente, S, López-Abán, J and Muro, A (2006) A new PCR-based approach for the specific amplification of DNA from different Schistosoma species applicable to human urine samples. Parasitology 133, 581587.CrossRefGoogle ScholarPubMed
Sawangsoda, P, Sithithaworn, J, Tesana, S, Pinlaor, S, Boonmars, T, Mairiang, E, Yongvanit, P, Duenngai, K and Sithithaworn, P (2012) Diagnostic values of parasite-specific antibody detections in saliva and urine in comparison with serum in opisthorchiasis. Parasitology International 61, 196202.CrossRefGoogle ScholarPubMed
Sheele, JM, Kihara, JH, Baddorf, S, Byrne, J and Ravi, B (2013) Evaluation of a novel rapid diagnostic test for Schistosoma haematobium based on the detection of human immunoglobulins bound to filtered Schistosoma haematobium eggs. Tropical Medicine & International Health: TM & IH 18, 477484.CrossRefGoogle ScholarPubMed
Shiff, C, Garba, A, Phillips, AE, Lamine, SM and Ibironke, OA (2011) Diagnosis of Schistosoma haematobium by detection of specific DNA fragments from filtered urine samples. The American Journal of Tropical Medicine and Hygiene 84, 9981001.Google Scholar
Shiraho, EA, Eric, AL, Mwangi, IN, Maina, GM, Kinuthia, JM, Mutuku, MW, Mugambi, RM, Mwandi, JM and Mkoji, GM (2016) Development of a loop mediated isothermal amplification for diagnosis of Ascaris lumbricoides in fecal samples. Journal of Parasitology Research 2016, 17.CrossRefGoogle ScholarPubMed
Song, HB, Kim, J, Jin, Y, Lee, JS, Jeoung, HG, Lee, YH, Saeed, AAW and Hong, S-T (2018) Comparison of ELISA and urine microscopy for diagnosis of Schistosoma haematobium infection. Journal of Korean Medical Science 33, 19. doi: 10.3346/jkms.2018.33.e238.CrossRefGoogle ScholarPubMed
Sousa, MS, van Dam, GJ, Pinheiro, MCC, de Dood, CJ, Peralta, JM, Peralta, RHS, de Daher, EF, Corstjens, PLAM and Bezerra, FSM (2019) Performance of an ultra-sensitive assay targeting the circulating anodic antigen (CAA) for detection of Schistosoma mansoni infection in a Low endemic area in Brazil. Frontiers in Immunology 10, 682.CrossRefGoogle Scholar
Sousa-Figueiredo, JC, Basáñez, M-G, Khamis, IS, Garba, A, Rollinson, D and Stothard, JR (2009) Measuring morbidity associated with urinary schistosomiasis: assessing levels of excreted urine albumin and urinary tract pathologies. PLoS Neglected Tropical Diseases 3, e526.CrossRefGoogle ScholarPubMed
Sousa-Figueiredo, JC, Betson, M, Kabatereine, NB and Stothard, JR (2013) The urine circulating cathodic antigen (CCA) dipstick: a valid substitute for microscopy for mapping and point-Of-care diagnosis of intestinal schistosomiasis. PLoS Neglected Tropical Diseases 7, e2008.CrossRefGoogle ScholarPubMed
Steel, C, Golden, A, Kubofcik, J, LaRue, N, de los Santos, T, Domingo, GJ and Nutman, TB (2013) Rapid Wuchereria bancrofti-specific antigen Wb123-based IgG4 immunoassays as tools for surveillance following mass drug administration programs on lymphatic filariasis. Clinical and Vaccine Immunology 20, 11551161.CrossRefGoogle ScholarPubMed
Stete, K, Krauth, SJ, Coulibaly, JT, Knopp, S, Hattendorf, J, Müller, I, Lohourignon, LK, Kern, WV, N'Goran, EK and Utzinger, J (2012) Dynamics of Schistosoma haematobium egg output and associated infection parameters following treatment with praziquantel in school-aged children. Parasites & Vectors 5, 298.CrossRefGoogle ScholarPubMed
Stothard, JR, Kabatereine, NB, Tukahebwa, EM, Kazibwe, F, Rollinson, D, Mathieson, W, Webster, JP and Fenwick, A (2006) Use of circulating cathodic antigen (CCA) dipsticks for detection of intestinal and urinary schistosomiasis. Acta Tropica 97, 219228.CrossRefGoogle ScholarPubMed
Stothard, JR, Stanton, MC, Bustinduy, AL, Sousa-Figueiredo, JC, Van Dam, GJ, Betson, M, Waterhouse, D, Ward, S, Allan, F, Hassan, AA, Al-Helal, MA, Memish, ZA and Rollinson, D (2014) Diagnostics for schistosomiasis in Africa and Arabia: a review of present options in control and future needs for elimination. Parasitology 141, 19471961.CrossRefGoogle ScholarPubMed
Sun, K, Xing, W, Yu, X, Fu, W, Wang, Y, Zou, M, Luo, Z and Xu, D (2016) Recombinase polymerase amplification combined with a lateral flow dipstick for rapid and visual detection of Schistosoma japonicum. Parasites & Vectors 9, 476.CrossRefGoogle ScholarPubMed
Takagi, H, Itoh, M, Kasai, S, Yahathugoda, TC, Weerasooriya, MV and Kimura, E (2011) Development of loop-mediated isothermal amplification method for detecting Wuchereria bancrofti DNA in human blood and vector mosquitoes. Parasitology International 60, 493497.CrossRefGoogle ScholarPubMed
Takagi, H, Yahathugoda, TC, Tojo, B, Rathnapala, UL, Nagaoka, F, Weerasooriya, MV and Itoh, M (2019) Surveillance of Wuchereria bancrofti infection by anti-filarial IgG4 in urine among schoolchildren and molecular xenomonitoring in Sri Lanka: a post mass drug administration study. Tropical Medicine and Health 47, 39.CrossRefGoogle ScholarPubMed
Tchuem Tchuenté, LA (2011) Control of soil-transmitted helminths in sub-saharan Africa: diagnosis, drug efficacy concerns and challenges. Acta Tropica 120, S4S11.CrossRefGoogle ScholarPubMed
Tesana, S, Srisawangwong, T, Sithithaworn, P, Itoh, M and Phumchaiyothin, R (2007) The ELISA-based detection of anti-Opisthorchis viverrini IgG and IgG4 in samples of human urine and serum from an endemic area of north-eastern Thailand. Annals of Tropical Medicine and Parasitology 101, 585591.CrossRefGoogle ScholarPubMed
Tischendorf, FW, Brattig, NW, Burchard, GD, Kubica, T, Kreuzpainer, G and Lintzel, M (1999) Eosinophils, eosinophil cationic protein and eosinophil-derived neurotoxin in serum and urine of patients with onchocerciasis coinfected with intestinal nematodes and in urinary schistosomiasis. Acta Tropica 72, 157173.CrossRefGoogle ScholarPubMed
Tischendorf, FW, Brattig, NW, Lintzel, M, Buttner, DW, Burchard, GD, Bork, K and Muller, M (2000) Eosinophil granule proteins in serum and urine of patients with helminth infections and atopic dermatitis. Tropical Medicine and International Health 5, 898905.CrossRefGoogle ScholarPubMed
Toribio, L., Romano, M., Scott, A. L., Gonzales, I., Saavedra, H., Garcia, H. H. and Shiff, C. and Peru, for the C. W. G. (2019). Detection of Taenia solium DNA in the urine of neurocysticercosis patients. The American Journal of Tropical Medicine and Hygiene 100, 327329.CrossRefGoogle ScholarPubMed
Turner, HC, Bettis, AA, Dunn, JC, Whitton, JM, Hollingsworth, TD, Fleming, FM and Anderson, RM (2017) Economic considerations for moving beyond the kato-katz technique for diagnosing intestinal parasites as we move towards elimination. Trends in Parasitology 33, 435443.CrossRefGoogle ScholarPubMed
Utzinger, J, Becker, SL, van Lieshout, L, van Dam, GJ and Knopp, S (2015) New diagnostic tools in schistosomiasis. Clinical Microbiology and Infection 21, 529542.CrossRefGoogle ScholarPubMed
Utzinger, J, Greter, H, Ngandolo, BNR, Krauth, SJ, Alfaroukh, IO and Zinsstag, J (2016) Validation of a point-of-care circulating cathodic antigen urine cassette test for Schistosoma mansoni diagnosis in the sahel, and potential cross-reaction in pregnancy. The American Journal of Tropical Medicine and Hygiene 94, 361364.Google Scholar
van Dam, GJ, Claas, FH, Yazdanbakhsh, M, Kruize, YC, van Keulen, AC, Ferreira, ST, Rotmans, JP and Deelder, AM (1996) Schistosoma mansoni excretory circulating cathodic antigen shares Lewis-x epitopes with a human granulocyte surface antigen and evokes host antibodies mediating complement-dependent lysis of granulocytes. Blood 88, 42464251.CrossRefGoogle ScholarPubMed
van Dam, GJ, Wichers, JH, Ferreira, TMF, Ghati, D, van Amerongen, A and Deelder, AM (2004) Diagnosis of schistosomiasis by reagent strip test for detection of circulating cathodic antigen. Journal of Clinical Microbiology 42, 54585461.CrossRefGoogle ScholarPubMed
van Dam, GJ, Xu, J, Bergquist, R, de Dood, CJ, Utzinger, J, Qin, Z-Q, Guan, W, Feng, T, Yu, X-L, Zhou, J, Zheng, M, Zhou, X-N and Corstjens, PLAM (2015a) An ultra-sensitive assay targeting the circulating anodic antigen for the diagnosis of Schistosoma japonicum in a low-endemic area, People's Republic of China. Acta Tropica 141, 190197.CrossRefGoogle Scholar
van Dam, GJ, Odermatt, P, Acosta, L, Bergquist, R, de Dood, CJ, Kornelis, D, Muth, S, Utzinger, J and Corstjens, PLAM (2015b) Evaluation of banked urine samples for the detection of circulating anodic and cathodic antigens in Schistosoma mekongi and S. japonicum infections: a proof-of-concept study. Acta Tropica 141, 198203.CrossRefGoogle Scholar
van Grootveld, R, van Dam, GJ, de Dood, C, de Vries, JJC, Visser, LG, Corstjens, PLAM and van Lieshout, L (2018) Improved diagnosis of active Schistosoma infection in travellers and migrants using the ultra-sensitive in-house lateral flow test for detection of circulating anodic antigen (CAA) in serum. European Journal of Clinical Microbiology & Infectious Diseases: Official Publication of the European Society of Clinical Microbiology 37, 17091716.CrossRefGoogle ScholarPubMed
Verweij, JJ and Stensvold, CR (2014) Molecular testing for clinical diagnosis and epidemiological investigations of intestinal parasitic infections. Clinical Microbiology Reviews 27, 371418.CrossRefGoogle ScholarPubMed
Vlaminck, J, Fischer, PU and Weil, GJ (2015) Diagnostic tools for onchocerciasis elimination programs. Trends in Parasitology 31, 571582.CrossRefGoogle ScholarPubMed
Vlaminck, J, Supali, T, Geldhof, P, Hokke, CH, Fischer, PU and Weil, GJ (2016) Community rates of IgG4 antibodies to Ascaris haemoglobin reflect changes in community Egg loads following mass drug administration. PLoS Neglected Tropical Diseases 10, e0004532.CrossRefGoogle ScholarPubMed
Vlaminck, J, Lagatie, O, Verheyen, A, Dana, D, Van Dorst, B, Mekonnen, Z, Levecke, B and Stuyver, LJ (2019) Patent infections with soil-transmitted helminths and Schistosoma mansoni are not associated with increased prevalence of antibodies to the Onchocerca volvulus peptide epitopes OvMP-1 and OvMP-23. Parasites & Vectors 12, 63.CrossRefGoogle Scholar
Vonghachack, Y, Sayasone, S, Khieu, V, Bergquist, R, van Dam, GJ, Hoekstra, PT, Corstjens, PLAM, Nickel, B, Marti, H, Utzinger, J, Muth, S and Odermatt, P (2017) Comparison of novel and standard diagnostic tools for the detection of Schistosoma mekongi infection in Lao people's democratic republic and Cambodia. Infectious Diseases of Poverty 6, 127.CrossRefGoogle ScholarPubMed
Wang, Y, Holmes, E, Nicholson, JK, Cloarec, O, Chollet, J, Tanner, M, Singer, BH and Utzinger, J (2004) Metabonomic investigations in mice infected with Schistosoma mansoni: an approach for biomarker identification. Proceedings of the National Academy of Sciences 101, 1267612681.CrossRefGoogle ScholarPubMed
Web of Science (2019) Friars House, Blackfriars Road, London. SE1 8EZ. Website: https://apps.webofknowledge.com (Accessed 1 September 2019).Google Scholar
Weerakoon, KG and McManus, DP (2016) Cell-Free DNA as a diagnostic tool for human parasitic infections. Trends in Parasitology 32, 378391.CrossRefGoogle ScholarPubMed
Weerakoon, KGAD, Gobert, GN, Cai, P and McManus, DP (2015) Advances in the diagnosis of human schistosomiasis. Clinical Microbiology Reviews 28, 939967.CrossRefGoogle Scholar
Weerakoon, K, Gordon, C and McManus, D (2018) DNA Diagnostics for schistosomiasis control. Tropical Medicine and Infectious Disease 3, 81.CrossRefGoogle ScholarPubMed
Weerasooriya, M, Itoh, M, Islam, M, Aoki, Y, Samarawickrema, W and Kimura, E (2008) Presence and gradual disappearance of filaria-specific urinary IgG4 in babies born to antibody-positive mothers: a 2-year follow-up study. Parasitology International 57, 386389.CrossRefGoogle ScholarPubMed
Weil, GJ, Kumar, H, Santhanam, S, Sethumadhavan, KVP and Jain, DC (1986) Detection of circulating parasite antigen in Bancroftian Filariasis by counterimmunoelectrophoresis. The American Journal of Tropical Medicine and Hygiene 35, 565570.CrossRefGoogle ScholarPubMed
Weil, GJ, Steel, C, Liftis, F, Li, BW, Mearns, G, Lobos, E and Nutman, TB (2000) A rapid-format antibody card test for diagnosis of onchocerciasis. The Journal of Infectious Diseases 182, 17961799.CrossRefGoogle ScholarPubMed
Werkman, M, Wright, JE, Truscott, JE, Easton, AV, Oliveira, RG, Toor, J, Ower, A, Ásbjörnsdóttir, KH, Means, AR, Farrell, SH, Walson, JL and Anderson, RM (2018) Testing for soil-transmitted helminth transmission elimination: analysing the impact of the sensitivity of different diagnostic tools. PLoS Neglected Tropical Diseases 12, e0006114.CrossRefGoogle ScholarPubMed
Worasith, C, Kamamia, C, Yakovleva, A, Duenngai, K, Wangboon, C, Sithithaworn, J, Watwiengkam, N, Namwat, N, Techasen, A, Loilome, W, Yongvanit, P, Loukas, A, Sithithaworn, P and Bethony, JM (2015) Advances in the diagnosis of human opisthorchiasis: development of Opisthorchis viverrini antigen detection in urine. PLoS Neglected Tropical Diseases 9, e0004157.CrossRefGoogle Scholar
World Health Organisation (2012). Research Priorities for Helminth Infections. WHO. Technical Report Series No. 972. Geneva, Switzerland: World Health Organization.Google Scholar
World Health Organization (2013). Schistosomiasis: Progress Report 2001–2011 and. Strategic Plan 2012–2020. Geneva, Switzerland: World Health Organisation.Google Scholar
Ximenes, C, Brandão, E, Oliveira, P, Rocha, A, Rego, T, Medeiros, R, Aguiar-Santos, A, Ferraz, J, Reis, C, Araujo, P, Carvalho, L and Melo, FL (2014) Detection of Wuchereria bancrofti DNA in paired serum and urine samples using polymerase chain reaction-based systems. Memórias do Instituto Oswaldo Cruz 109, 978983.CrossRefGoogle ScholarPubMed
Xing, W, Yu, X, Feng, J, Sun, K, Fu, W, Wang, Y, Zou, M, Xia, W, Luo, Z, He, H, Li, Y and Xu, D (2017) Field evaluation of a recombinase polymerase amplification assay for the diagnosis of Schistosoma japonicum infection in hunan province of China. BMC Infectious Diseases 17, 164.CrossRefGoogle Scholar
Zhang, X, Lowe, SB and Gooding, JJ (2014) Brief review of monitoring methods for loop-mediated isothermal amplification (LAMP). Biosensors and Bioelectronics 61, 491499.CrossRefGoogle Scholar
Figure 0

Fig. 1. Schematic outlining changes in diagnostic priorities as control programmes progress (adapted from Bergquist et al., 2009).

Figure 1

Table 1. WHO-recommended diagnostic techniques for major human helminth infections and how technique invasiveness compares to that of urine sampling.

Figure 2

Table 2. Anti-helminth antibodies detected within urine and immunodiagnostic assay used.

Figure 3

Table 3. Helminth-derived antigens detected within the urine and immunodiagnostic assay used.

Figure 4

Table 4. Helminth cfDNA detected within the urine and nucleic acid amplification test (NAAT) used.

Figure 5

Fig. A1. Defining sensitivity, specificity and predictive values (Akobeng, 2007; Bergquist et al., 2009).