SPECIAL ISSUE ARTICLE

1789

Diagnostics for the control and elimination of neglected tropical diseases ROSANNA W. PEELING* and DAVID MABEY London School of Hygiene and Tropical Medicine, Keppel Street, London SW1E 7HT, UK (Received 14 March 2014; revised 19 May 2014; accepted 19 May 2014; first published online 23 September 2014) SUMMARY

Practical diagnostic tools of sufficient sensitivity to detect levels of infection that can lead to transmission have been identified as a critical component of successful disease elimination programmes. In this review we describe the diagnostic tests currently available for six neglected tropical diseases that have been targeted for elimination; assess their performance in the light of the requirements for surveillance, certification of elimination and post-elimination surveillance; consider the unmet need for diagnostic tests for these diseases; and review recent technical developments that could meet these needs. Key words: Disease elimination, diagnostics, schistosomiasis, onchoceriasis, lymphatic filariasis, trachoma, African trypanosomiasis, visceral leishmaniasis.

INTRODUCTION

The London Declaration of 2012, endorsed by the World Health Organization (WHO), the Gates Foundation and 13 leading pharmaceutical companies, resolved to sustain, expand and extend programmes that ensure the necessary supply of drugs and other interventions to help eradicate Guinea worm disease, and help eliminate by 2020 lymphatic filariasis (LF), leprosy, human African trypanosomiasis (HAT) and blinding trachoma; and to help control by 2020 schistosomiasis, soil-transmitted helminths (STH), Chagas disease, visceral leishmaniasis (VL) and onchocerciasis [www.who.int/ntd]. This was to be done through partnerships and provision of funding to find next-generation treatments and interventions for neglected tropical diseases (NTDs). The role of diagnostics in the control and elimination of NTDs was not specifically addressed at the London meeting, but was raised as a high priority at a follow-up meeting held at the Institut Pasteur in Paris in April 2014. Experience with disease elimination and eradication programmes has shown that adequate tools for surveillance are critical determinants of success. The report of the 1997 Dahlem workshop on the eradication of infectious diseases identified ‘practical diagnostic tools of sufficient sensitivity and specificity to detect levels of infection that can lead to transmission’ as one of the three essential requirements for disease elimination or eradication (Dowdle, 1998). In the case of Guinea worm there is no need for a diagnostic test, since the clinical features are unmistakable (Al-Awadi et al. 2014). The other NTDs * Corresponding author: London School of Hygiene and Tropical Medicine, Keppel Street, London SW1E 7HT, UK. E-mail: [email protected]

mentioned in the declaration can be divided into those for which mass drug administration (MDA) is recommended, and those for which individual case management is required. The diagnostic needs for these two groups are clearly different. MDA is recommended for the control and elimination of LF, onchocerciasis, schistosomiasis, STH and trachoma. Traditionally, with the exception of trachoma which is diagnosed clinically, these diseases have been diagnosed microscopically, using day or night bloods, urine or stool samples or skin snips. Microscopy can be a sensitive technique when parasite burdens are high, but requires trained technicians and a laboratory with a functioning microscope; quality assurance is often lacking. As the prevalence and intensity of infection falls following repeated rounds of MDA, a more sensitive test is required. Microscopy, despite concentration techniques, may no longer be sufficiently sensitive to detect the remaining cases. As elimination targets are approached, large numbers of samples need to be tested to be sure that transmission has been interrupted, so highthroughput tests are needed. Moreover, as the prevalence of infection falls, so does the positive predictive value of a diagnostic test, and highly specific tests are required to certify elimination. Antigen detection assays either in the form of rapid immunochromatographic tests (ICTs) or enzyme linked immunoassays (ELISAs) offer alternative means of detecting infection. While ICTs offer ease of use and on-site results, ELISAs offer higher sensitivity and high throughput capability. As communities achieve interruption of transmission, it may be necessary to use molecular assays such as real-time PCR to detect the few remaining cases. However, for NTDs, these techniques remain laboratory-based, as they are expensive and require

Parasitology (2014), 141, 1789–1794. © Cambridge University Press 2014 doi:10.1017/S0031182014000973

Rosanna W. Peeling and David Mabey

highly trained personnel and sophisticated equipment. For monitoring interruption of transmission and for certifying elimination, the consensus is that detection of specific antibodies to a pathogen may be the most sensitive marker of exposure. Detection of antibodies to immunodominant epitopes of a pathogen in sentinel populations (e.g. young children) is the ideal tool for certifying elimination, and for postelimination surveillance. It may be necessary to use a combination of tests to achieve the required sensitivity and specificity. The role of diagnostics as a surveillance tool to support policy decisions related to treatment and control strategies for Schistosomiasis japonica towards elimination in China is an example of effective use of different types of diagnostics in disease control and elimination programmes (Zhou et al. 2011). In the case of HAT and VL, for which treatment is difficult to administer, toxic, or both, it is necessary to diagnose and treat individual cases, and the diagnostic needs are different. Point-of-care (POC) tests are needed for these diseases, which are often found in remote rural areas far from a laboratory (Mabey et al. 2004; Peeling and Mabey, 2010). In the following sections we describe the tools currently available for the diagnosis of the major NTDs mentioned in the London Declaration, the need for new diagnostics, and emerging diagnostic technologies that could meet these needs. Due to space constraints this is not a comprehensive review of diagnostics for NTDs; our aim was to focus on the key issues. CURRENT DIAGNOSTICS TOOLS AND UNMET NEEDS

Lymphatic filariasis Antigen detection tests adapted to ICT or ELISA formats are now used routinely by control programmes. These assays can be adapted to a bead or microfluidics-based assay to allow multiplexing. The ICT antigen detection test has been widely used for monitoring the impact of MDA programmes on the prevalence of Wuchereria bancrofti infection and for transmission assessment surveys. Tests for antigen are preferable to tests for antibody, which cannot distinguish between past, treated and current infection. Antibody detection assays using the BmR1 antigen have been used to monitor the impact of MDA on Brugia malayi infection. A recent multicentre study showed that transmission assessment surveys, in which children aged 6–7 years were tested using these two assays, were an effective and practical tool for deciding when to discontinue MDA (Chu et al. 2013). A backlog of disabled patients with lymphoedema caused by LF will remain after transmission of the parasite has been interrupted, and it will be important for elimination programmes to address the needs of these patients.

1790

Onchocerciasis In 2001 the WHO published criteria for the certification of interruption of transmission/elimination of human onchocerciasis (WHO, 2001). The two key criteria were absence, or near absence, of infective stage larvae in the vector population, and infection rates of 95% in Brazil and ranged from 90·8 to 98·0% in East Africa (Cunningham et al. 2012). The lower sensitivity may be due to antigenic differences between parasite strains (Bhattacharyya et al. 2013). Human African trypanosomiasis Because microscopic examination of a blood film is an insensitive test for the diagnosis of Trypanosoma brucei gambiense infection, a serological test (the card agglutination test, CATT) is used to screen at-risk populations. If low titres are considered positive this is a sensitive test, but lacks specificity (Inojosa et al. 2006). Treatment is not recommended unless the infection can be confirmed parasitologically on a blood film or lymph node aspirate. Various techniques have been developed to increase the sensitivity of microscopy, such as centrifugation of blood and examination of the buffy coat, and the mini anion exchange column technique, but these are not always available in the remote rural areas where most cases of HAT are found (Chappuis et al. 2005). The management of subjects who are CATT positive but parasitologically negative is problematic (Wastling et al. 2011). The recommendation is that they should be followed up and the tests repeated, but this is not always possible. Since the recommended treatment depends on whether the central nervous system (CNS) is involved, a lumbar puncture and

1791

cerebrospinal fluid (CSF) microscopy for staging the disease is also required. This is not always culturally acceptable, and the criteria for diagnosing stage 2 when parasites cannot be found in the CSF are not entirely clear. The presence of 5 or more white blood cells per mm3 of CSF is generally taken to indicate CNS involvement (Chappuis et al. 2005).

EMERGING DIAGNOSTIC TECHNOLOGI ES TO TRANSFORM DISEASE SURVEILLANCE

A new generation of immunoassays and molecular technologies has been developed that can be combined with readers or mobile phones to provide realtime NTD surveillance data with improved accuracy to inform treatment and disease-control strategies and monitor progress towards elimination. These technology platforms can be used to detect multiple targets from a single specimen. Many can yield quantitative results to give estimates of transmission intensity linked to precise geographical locations using GPS.

Bead-based immunoassays Bead-based technologies are more versatile and potentially offer increased sensitivity over ELISAs. Instead of using enzymatic amplification of a colorimetric substrate, bead-based assays can use a range of labels including laser or fluorescence to detect the binding of the secondary antibody to the antigen or antibody target. The beads are in solution offering more sites for ligand binding compared with solid phase support used for ELISAs. Fluorescent intensities can be quantitative. Protein arrays offer the potential of quantification of the antigen target as a surrogate measure of parasite load remaining in the community to inform treatment strategies. Detection of multiple targets from the same pathogen can lead to increased sensitivity over single-target assays. Detection of targets from multiple pathogens using a single specimen can offer substantial cost and sample savings over traditional ELISA measurements.

Microfluidic immunoassays Immunoassays developed on a microfluidic platform that miniaturizes but reproduces all the steps of an ELISA have been developed (Chin et al. 2011). The microfluidic disc can fit into the palm of a hand and is rapid and inexpensive to manufacture. These platforms allow multiplexing but they work best for antibody detection in blood samples. Antigen detection from urine or stool samples will require multistep specimen processing and/or purification and concentration before initiating the antigen-antibody binding and detection steps within the microfluidic channels.

Rosanna W. Peeling and David Mabey

Molecular assays Bead-based molecular assays offer a promising approach to increase the performance of diagnostics beyond limit of detection of immunoassays. There are a number of exciting lab-on-a-chip developments in the pipeline with nanowires and quantum dot barcodes using microfluidics and magnetism (Giri et al. 2011). These are simple to use and give answers in less than 30 min. The convergence of these nanotechnologies with rapidly evolving smart phone technology will lead to simple, rapid, low-cost multiplex DNA hybridization assays that can also transmit data to a central database, yielding real-time surveillance data from the field (Gao et al. 2013). These lab-on-a-chip technologies offer an integrated approach combining pathogen detection and speciation with genetic analysis such as detection of single nucleotide polymorphisms. Hence they can be used for high-throughput screening of drug mutation targets. Advocacy to apply these novel technologies to NTDs is an urgent priority (Jani and Peter 2013).

POC MOLECULAR ASSAYS

A number of nucleic acid amplification tests (NAATs) have been developed in the last 20 years and have been used as the gold standard to which the performance of other diagnostic tests is compared. They have largely remained laboratory based but, in recent years, a number of POC NAATs have been developed. The first sample in–answer out real-time PCR platform that can be used wherever there is a source of electricity has been introduced for the diagnosis of tuberculosis and detection of rifampicin resistance (Boehme et al. 2011). The GeneXpert realtime PCR platform allows random access so that different tests can be initiated as required. It requires minimal onsite expertise and has remote quality control. Isothermal amplification platforms such as helicase-dependent amplification (HDA), cross-priming amplification (CPA), recombinase polymerase amplification (RPA) and loop-mediated amplification (LAMP) assays can be made into POC NAATs as there is no requirement for sophisticated equipment to perform thermal cycling. The details of these technologies are described in an excellent review article (Niemz et al. 2011). POC NAATs based on these technologies have largely been applied to highburden diseases such as HIV and tuberculosis, but LAMP assays have been developed and evaluated for the diagnosis of VL and HAT. In the case of VL, the LAMP assay for Leishmania donovani is highly sensitive and specific for the diagnosis of VL (using a blood sample), and for the diagnosis of post kalaazar dermal leishmaniasis (PKDL) using a skin biopsy (Verma et al. 2013). Diagnosis of PKDL will be important for the elimination agenda, since

1792

patients with this condition are an important source of infection. In the case of HAT, the LAMP assay was shown to have a sensitivity of 87·3% (95% CI 80·9–91·8%, and a specificity of 92·8% (95% CI 86·4–96·3%) against a PCR gold standard (Mitashi et al. 2013). Advocacy and investments are needed to apply these technologies to the control and elimination of NTDs.

M O V I N G FO R WA R D

The development of appropriate diagnostics starts with defining target product profiles. At a WHOsponsored meeting held at the London School of Hygiene & Tropical Medicine in 2010, the first steps towards defining TPPs for diagnostic tools for measuring the impact of MDA, and for postelimination surveillance for these diseases were taken (Solomon et al. 2012). Further consultations were organized by the Task Force for Global Health with key stakeholders to define target product profiles for diagnostic tools needed in two phases of disease control: pre-elimination and post-elimination. An important issue that needs consensus is the sampling strategy as the throughput needs to be matched to appropriate technologies. The sampling strategy depends on a number of factors, including the prevalence threshold above which MDA is implemented, the inherent spatial heterogeneity of the individual infections, and how this varies for different NTDs and different stages of control; cost of conducting surveys in the field and cost and accuracy of diagnostic tests in classifying populations, which could be termed efficiency. This will require modelling the cost-effectiveness of using different tools for different phases of control and needs to take into account spatial characteristics for the distribution of infections and differing diagnostic accuracies and costs. The models can be used to investigate the required sensitivity and specificity within given control programme contexts (Drain et al. 2013). Consideration should be given the use of multiplex testing for more than one NTD, which could prove costeffective for post-MDA surveillance in some settings. Once a test is developed and standardized, its performance and utility need to be validated. A bank of well-characterized specimens is critical to the evaluation of the analytical performance of these tests. POC tests will also need to be validated in settings of intended use, covering the full spectrum of populations in which the test will be used (Banoo et al. 2006). Standardized protocols for laboratory and field evaluation and a network of competent evaluation sites that can perform evaluations and pilot studies to implement new technologies are needed to accelerate the adoption and introduction of new diagnostic tools for NTDs.

Diagnostics for the control and elimination of NTDs CONCLUSIONS

Appropriate diagnostics to monitor disease trends and assess the impact of interventions are essential for guiding treatment strategies for NTDs at different thresholds of control, interruption of transmission, elimination and post-elimination surveillance. While there remains a need for standardization and evaluation of new tests in a variety of settings before they are widely adopted, it is clear that recent technical advances have led to improved diagnostic tests for NTDs, which are sensitive, specific, able to diagnose multiple infections using a single specimen, and can be used at the point of care. Since there is extensive geographical overlap between different NTDs targeted by MDA, multiplex surveillance platforms are likely to prove cost-effective. However, barriers remain to the rapid deployment of these new technologies for the control and elimination of NTDs. The importance of better diagnostics for NTD control and elimination must be brought to the attention of the policymakers and funding agencies.

ACKNOWLEDGEMENT

This paper is based on a plenary lecture delivered by Professor Peeling at a meeting of the British Society of Parasitology in Ness Gardens, Liverpool in 2013.

REFERENCES Al-Awadi, A. R., Al-Kuhlani, A., Breman, J. G., Doumbo, O., Eberhard, M. L., Guiguemde, R. T., Magnussen, P., Molyneux, D. H. and Nadim, A. (2014). Guinea worm (Dracunculiasis) eradication: update on progress and endgame challenges. Transactions of the Royal Society of Tropical Medicine and Hygiene 108, 249–251. Banoo, S., Bell, D., Bossuyt, P., Herring, A., Mabey, D., Poole, F., Smith, P. G., Sriram, N., Wongsrichanalai, C., Linke, R., O’Brien, R., Perkins, M., Cunningham, J., Matsoso, P., Nathanson, C. M., Olliaro, P., Peeling, R. W., Ramsay, A. and TDR Diagnostics Evaluation Expert Panel (2006). Evaluation of diagnostic tests for infectious diseases: general principles. Nature Reviews Microbiology 4 (Suppl.), S21–S31. Bhattacharyya, T., Boelaert, M. and Miles, M. A. (2013). Comparison of visceral leishmaniasis diagnostic antigens in African and Asian Leishmania donovani reveals extensive diversity and region-specific polymorphisms. PLOS Neglected Tropical Diseases 7, e2057. Boehme, C. C., Nicol, M. P., Nabeta, P., Michael, J. S., Gotuzzo, E., Tahirli, R., Gler, M. T., Blakemore, R., Worodria, W., Gray, C., Huang, L., Caceres, T., Mehdiyev, R., Raymond, L., Whitelaw, A., Sagadevan, K., Alexander, H., Albert, H., Cobelens, F., Cox, H., Alland, D., Perkins, M. D. (2011). Feasibility, diagnostic accuracy, and effectiveness of decentralised use of the Xpert MTB/RIF test for diagnosis of tuberculosis and multidrug resistance: a multicentre implementation study. Lancet 377, 1495–1505. Chappuis, F., Loutan, L., Simarro, P., Lejon, V. and Büscher, P. (2005). Options for field diagnosis of human African trypanosomiasis. Clinical Microbiology Reviews 18, 133–146. Chappuis, F., Sundar, S., Hailu, A., Ghalib, H., Rijal, S., Peeling, R. W., Alvar, J. and Boelaert, M. (2010). Visceral leishmaniasis: what are the needs for diagnosis, treatment and control? Nature Reviews Microbiology 5, 873–882. Chin, C. D., Lakasanasopin, L., Cheung, Y. K., Steinmiller, D., Linder, V., Parsa, H., Wang, J., Moore, H., Rouse, R., Umviligihozo, G., Karita, E., Mwambarangwe, L., Braunstein, S. L., van de Wijgert, J., Sahabo, R., Justman, J. E., ElSadr, W. and Sia, S. K. (2011). Microfluidics-based diagnostics of infectious diseases in the developing world. Nature Medicine 17, 1015–1019.

1793 Chu, B. K., Deming, M., Biritwum, N-K., Bougma, W. R., Dorkenoo, A. M., El-Setouhy, M., Fischer, P. U., Gass, K., Gonzalez de Peña, M., Mercado-Hernandez, L., Kyelem, D., Lammie, P. J., Flueckiger, R. M., Mwingira, U. J., Noordin, R., Offei Owusu, I., Ottesen, E. A., Pavluck, A., Pilotte, N., Rao, R. U., Samarasekera, D., Schmaedick, M. A., Settinayake, S., Simonsen, P. E., Supali, T., Taleo, F., Torres, M., Weil, G. J. and Won, K. Y. (2013). Transmission Assessment Surveys (TAS) to define endpoints for lymphatic filariasis mass drug administration: a multicenter evaluation. PLOS Neglected Tropical Diseases 7, e2584. Cunningham, J., Hasker, E., Das, P., El Safi, S., Goto, H., Mondal, D., Mbuchi, M., Mukhtar, M., Rabello, A., Rijal, S., Sundar, S., Wasunna, M., Adams, E., Menten, J., Peeling, R., Boelaert, M. (2012). A global comparative evaluation of commercial immunochromatographic rapid diagnostic tests for visceral leishmaniasis. Clinical Infectious Diseases 55, 1312–1319. Dowdle, W. R. (1998). The principles of disease elimination and eradication. Bulletin of the World Health Organization 76 (Suppl. 2), 22–25. Drain, P. K., Hyle, E. P., Noubary, F., Freedberg, K. A., Wilson, D., Bishai, W. R., Rodriguez, W. and Bassett, I. V. (2014). Diagnostic pointof-care tests in resource-limited settings. Lancet Infectious Diseases 14, 239–249. Fung, M. S., Xiao, N., Wang, S. and Carlton, E. J. (2012). Field evaluation of a PCR test for Schistosoma japonicum egg detection in lowprevalence regions of China. American Journal of Tropical Medicine and Hygiene 87, 1053–1058. Gao, Y., Lam, A. W. Y. and Chan, W. C. W. (2013). Automating quantum dot barcode assays using microfluidics and magnetism for the development of a point-of-care device. ACS Applied Materials and Interfaces 5, 2853–2860. Giri, S., Sykes, E. A., Jennings, T. L. and Chan, W. C. W. (2011). Rapid screening of genetic biomarkers of infectious agents using quantum dot barcodes. ACS Nano 5, 1580–1587. Goodhew, E. B., Priest, J. W., Moss, D. M., Zhong, G., Munoz, B., Mkocha, H., Martin, D. L., West, S. K., Gaydos, C. and Lammie, P. J. (2012). CT694 and pgp3 as serological tools for monitoring trachoma programs. PLOS Neglected Tropical Diseases 6, e1873. Gopal, H., Hassan, H. K., Rodrıguez-Perez, M. A., Toe, L. D., Lustigman, S. and Unnasch, T. R. (2012). Oligonucleotide based magnetic bead capture of Onchocerca volvulus DNA for PCR pool screening of vector black flies. PLOS Neglected Tropical Diseases 6, e1712. Harding-Esch, E. M., Sillah, A., Edwards, T., Burr, S. E., Hart, J. D., Joof, H., Laye, M., Makalo, P., Manjang, A., Molina, S., SarrSissoho, I., Quinn, T. C., Lietman, T., Holland, M. J., Mabey, D., West, S. K. and Bailey, R., on behalf of the Partnership for Rapid Elimination of Trachoma (PRET) study group (2013). Mass treatment with azithromycin for trachoma: when is one round enough? Results from the PRET trial in The Gambia. PLOS Neglected Tropical Diseases 7, e2115. Inojosa, W. O., Augusto, I., Bisoffi, Z., Josenado, T., Abel, P. M., Stich, A. and Whitty, C. J. (2006). Diagnosing human African trypanosomiasis in Angola using a card agglutination test: observational study of active and passive case finding strategies. British Medical Journal 332, 1479. Jani, I. V. and Peter, T. F. (2013). How point-of-care testing could drive innovation in global health. New England Journal of Medicine 368, 2319–2324. Mabey, D., Peeling, R. W., Ustianowski, A. and Perkins, M. D. (2004). Diagnostics for the developing world. Nature Reviews Microbiology 2, 231–240. Mitashi, P., Hasker, E., Ngoyi, D. M., Pyana, P. P., Lejon, V., Van der Veken, W., Lutumba, P., Büscher, P., Boelaert, M. and Deborggraeve, S. (2013). Diagnostic accuracy of loopamp Trypanosoma brucei detection kit for diagnosis of human African trypanosomiasis in clinical samples. PLOS Neglected Tropical Diseases 7, e2504. Niemz, A., Ferguson, T. M. and Boyle, D. S. (2011). Point-of-care nucleic acid testing for infectious diseases. Trends in Biotechnology 29, 240–250. Oguttu, D., Byamukama, E., Katholi, C. R., Habomugisha, P., Nahabwe, C., Ngabirano, M., Hassan, H. K., Lakwo, T., Katabarwa, M., Richards, F. O. and Unnasch, T. R. (2014). Serosurveillance to monitor onchocerciasis elimination: the Ugandan experience. American Journal of Tropical Medicine and Hygiene 90, 339–345. Peeling, R. W. and Mabey, D. (2010). Point-of-care tests for diagnosing infections in the developing world. Microbiology and Infection 16, 1062– 1069. Roberts, C. H., Last, A., Molina-Gonzalez, S., Cassama, E., Butcher, R., Nabicassa, M., McCarthy, E., Burr, S. E., Mabey, D. C.,

Rosanna W. Peeling and David Mabey Bailey, R. L. and Holland, M. J. (2013). Development and evaluation of a next-generation digital PCR diagnostic assay for ocular Chlamydia trachomatis infections. Journal of Clinical Microbiology 51, 2195–2203. Solomon, A. W., Holland, M. J., Burton, M. J., West, S. K., Alexander, N. D. E., Aguirre, A., Massae, P. A., Mkocha, H., Munoz, B., Johnson, G. J., Peeling, R. W., Bailey, R. L., Foster, A. and Mabey, D. C. W. (2003). Strategies for control of trachoma: observational study with quantitative PCR. Lancet 362, 198–204. Solomon, A. W., Foster, A. and Mabey, D. C. W. (2006). Clinical examination versus Chlamydia trachomatis assays to guide antibiotic use in trachoma control programmes. Lancet Infectious Diseases 6, 5–6. Solomon, A. W., Engels, D., Bailey, R. L., Blake, I. M., Brooker, S., Chen, J-X., Chen, J.-H., Churcher, T. S., Drakeley, C. J., Edwards, T., Fenwick, A., French, M., Gabrielli, A. F., Grassly, N. C., HardingEsch, E. M., Holland, M. J., Koukounari, A., Lammie, P. J., Leslie, J., Mabey, D. C., Rhajaoui, M., Secor, W. E., Stothard, J. R., Wei, H., Willingham, A. L., Zhou, X-N. and Peeling, R. W. (2012). A diagnostics platform for the integrated mapping, monitoring, and surveillance of neglected tropical diseases: rationale and target product profiles. PLOS Neglected Tropical Diseases 6, e1746. Sousa-Figueiredo, J. C., Betson, M., Kabatereine, N. B. and Stothard, J. R. (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.

1794 Tsang, V. C. and Wilkins, P. P. (1991). Immunodiagnosis of schistosomiasis: screen with FAST-ELISA and confirm with immunoblot. Clinics in Laboratory Medicine 11, 1029–1039. Verma, S., Avishek, K., Sharma, V., Negi, N. S., Ramesh, V. and Salotra, P. (2013). Application of loop-mediated isothermal amplification assay for the sensitive and rapid diagnosis of visceral leishmaniasis and post-kala-azar dermal leishmaniasis. Diagnostic Microbiology and Infectious Disease 75, 390–395. Wastling, S. L., Picozzi, K., Wamboga, C., von Wissmann, B., Amongi-Accup, C., Wardrop, N. A., Stothard, J. R., Kakembo, A. and Welburn, S. C. (2011). Latent Trypanosoma brucei gambiense foci in Uganda: a silent epidemic in children and adults? Parasitology 138, 1480– 1487. World Health Organization. (2001). Criteria for certification of interruption of transmission/elimination of human onchocerciasis. WHO/CDS/CPE/ CEE/2001.18a. World Health Organization, Geneva. Xu, J., Feng, T., Lin, D. D., Wang, Q. Z., Tang, L., Wu, X. H., Guo, J. G., Peeling, R. W. and Zhou, X. N. (2011). Performance of a dipstick dye immunoassay for rapid screening of Schistosoma japonicum infection in areas of low endemicity. Parasites and Vectors 4, 87. Zhou, X-N., Xu, J., Chen, H-G., Wang, T-P., Huang, X-B., Lin, D-D., Wang, Q-Z., Tang, L., Guo, J-G., Wu, X-H., Feng, T., Chen, J-X., Guo, J., Chen, S-H., Li, H., Wu, Z-D. and Peeling, R. W. (2011). Tools to support policy decisions related to treatment strategies and surveillance of schistosomiasis japonica towards elimination. PLOS Neglected Tropical Diseases 5, e1408.

Diagnostics for the control and elimination of neglected tropical diseases.

Practical diagnostic tools of sufficient sensitivity to detect levels of infection that can lead to transmission have been identified as a critical co...
89KB Sizes 5 Downloads 9 Views