Performance of a High-Sensitivity Rapid Diagnostic Test for Plasmodium falciparum Malaria in Asymptomatic Individuals from Uganda and Myanmar and Naive Human Challenge Infections

Smita Das, Ihn Kyung Jang, Becky Barney, Roger Peck, John C Rek, Emmanuel Arinaitwe, Harriet Adrama, Maxwell Murphy, Mallika Imwong, Clare L Ling, Stephane Proux, Warat Haohankhunnatham, Melissa Rist, Annette M Seilie, Amelia Hanron, Glenda Daza, Ming Chang, Tomoka Nakamura, Michael Kalnoky, Paul Labarre, Sean C Murphy, James S McCarthy, Francois Nosten, Bryan Greenhouse, Sophie Allauzen, Gonzalo J Domingo, Smita Das, Ihn Kyung Jang, Becky Barney, Roger Peck, John C Rek, Emmanuel Arinaitwe, Harriet Adrama, Maxwell Murphy, Mallika Imwong, Clare L Ling, Stephane Proux, Warat Haohankhunnatham, Melissa Rist, Annette M Seilie, Amelia Hanron, Glenda Daza, Ming Chang, Tomoka Nakamura, Michael Kalnoky, Paul Labarre, Sean C Murphy, James S McCarthy, Francois Nosten, Bryan Greenhouse, Sophie Allauzen, Gonzalo J Domingo

Abstract

Sensitive field-deployable diagnostic tests can assist malaria programs in achieving elimination. The performance of a new Alere™ Malaria Ag P.f Ultra Sensitive rapid diagnostic test (uRDT) was compared with the currently available SD Bioline Malaria Ag P.f RDT in blood specimens from asymptomatic individuals in Nagongera, Uganda, and in a Karen Village, Myanmar, representative of high- and low-transmission areas, respectively, as well as in pretreatment specimens from study participants from four Plasmodium falciparum-induced blood-stage malaria (IBSM) studies. A quantitative reverse transcription PCR (qRT-PCR) and a highly sensitive enzyme-linked immunosorbent assay (ELISA) test for histidine-rich protein II (HRP2) were used as reference assays. The uRDT showed a greater than 10-fold lower limit of detection for HRP2 compared with the RDT. The sensitivity of the uRDT was 84% and 44% against qRT-PCR in Uganda and Myanmar, respectively, and that of the RDT was 62% and 0% for the same two sites. The specificities of the uRDT were 92% and 99.8% against qRT-PCR for Uganda and Myanmar, respectively, and 99% and 99.8% against the HRP2 reference ELISA. The RDT had specificities of 95% and 100% against qRT-PCR for Uganda and Myanmar, respectively, and 96% and 100% against the HRP2 reference ELISA. The uRDT detected new infections in IBSM study participants 1.5 days sooner than the RDT. The uRDT has the same workflow as currently available RDTs, but improved performance characteristics to identify asymptomatic malaria infections. The uRDT may be a useful tool for malaria elimination strategies.

Figures

Figure 1.
Figure 1.
Relationship between parasite density and histidine-rich protein II (HRP2) concentration in IBSM studies. (A) HRP2 concentration and parasite density profiles over days after infection with parasitized red blood cells. Dashed line and empty squares represent parasite density; solid line and empty circles represent HRP2 concentration. The means of results from 16 subjects are shown. (B) Relationship between parasite density and HRP2 concentration in the 0–5 parasites per µL range pre-treatment infection.
Figure 2.
Figure 2.
Distribution of specimens by parasite density and histidine-rich protein II (HRP2) concentration in induced blood-stage malaria challenge studies (A, D); Myanmar study (B, E); and Uganda study (C, F). (AC). The outer clear bars represent the specimens that were positive by quantitative reverse transcription polymerase chain reaction; overlaid are the number of positive test results by ultra sensitive rapid diagnostic test (uRDT) (checkered bars) and by RDT (gray bars). (DF). The outer clear bars represent the specimens that were positive by quantitative HRP2 assay; overlaid are the number of positive test results by uRDT (checkered bars) and by RDT (gray bars).
Figure 3.
Figure 3.
Relationship between parasite density and histidine-rich protein II (HRP2) concentration shown as box plots for ranges of parasite densities. The data shown are for the Uganda specimens. The upper and lower bounds of the boxes represent the third and first quartiles, respectively. The mean is indicated by a solid line and the whiskers indicate the 95th and 5th percentiles. The leftmost bin for 0 parasites per µL (p/µL) corresponds to HRP2-positive but quantitative reverse transcription polymerase chain reaction-negative specimens. The descriptive statistics are provided in Table 3.
Figure 4.
Figure 4.
Histogram of Uganda asymptomatic clinical specimens arranged by histidine-rich protein II (HRP2) concentration ranges and broken out into confirmed parasitemic (gray shaded bars) and non-parasitemic (spotted bars) specimens based on quantitative reverse transcription polymerase chain reaction data.
Figure 5.
Figure 5.
Cumulative proportion of the confirmed parasitemic asymptomatic specimens in the Uganda study that are uRDT (solid line) or RDT (dashed line) positive with (A) decreasing parasite density as determined by quantitative reverse transcription polymerase chain reaction (left to right) and (B) decreasing histidine-rich protein II (HRP2) concentration as determined by the HRP2 Q-plex enzyme-linked immunosorbent assay (left to right).

References

    1. WHO, 2016. World Malaria Report 2016.Geneva, Switzerland: WHO
    1. Okell LCGA, Lyons E, Drakeley CJ, 2009. Submicroscopic infection in Plasmodium falciparum-endemic populations: a systematic review and meta-analysis. J Infect Dis 200: 1509–1517.
    1. Hemingway J, Shretta R, Wells TNC, Bell D, Djimdé AA, Achee N, Qi G, 2016. Tools and strategies for malaria control and elimination: what do we need to achieve a grand convergence in malaria? PLoS Biol 14: e1002380.
    1. Slater HC, Ross A, Ouedraogo AL, White LJ, Nguon C, Walker PGT, Ngor P, Aguas R, Silal SP, Dondorp AM, La Barre P, Burton R, Sauerwein RW, Drakeley C, Smith TA, Bousema T, Ghani AC, 2015. Assessing the impact of next-generation rapid diagnostic tests on Plasmodium falciparum malaria elimination strategies. Nature 528: S94–S101.
    1. Lo E, Zhou G, Oo W, Afrane Y, Githeko A, Yan G, 2015. Low parasitemia in submicroscopic infections significantly impacts malaria diagnostic sensitivity in the highlands of western Kenya. PLoS One 10: e0121763.
    1. Daubersies PS-SS, Magne S, Trape JF, Contamin H, Fandeur T, Rogier C, Mercereau-Puijalon O, Druilhe P, 1996. Rapid turnover of Plasmodium falciparum populations in asymptomatic individuals living in a high transmission area. Am J Trop Med Hyg 54: 18–26.
    1. Gaye A, Bousema T, Libasse G, Ndiath MO, Konate L, Jawara M, Faye O, Sokhna C, 2015. Infectiousness of the human population to Anopheles arabiensis by direct skin feeding in an area hypoendemic for malaria in Senegal. Am J Trop Med Hyg 92: 648–652.
    1. Tadesse FG, van den Hoogen L, Lanke K, Schildkraut J, Tetteh K, Aseffa A, Mamo H, Sauerwein R, Felger I, Drakeley C, Gadissa E, Bousema T, 2017. The shape of the iceberg: quantification of submicroscopic Plasmodium falciparum and Plasmodium vivax parasitaemia and gametocytaemia in five low endemic settings in Ethiopia. Malar J 16: 99.
    1. Harris I, Sharrock WW, Bain LM, Gray K-A, Bobogare A, Boaz L, Lilley K, Krause D, Vallely A, Johnson M-L, Gatton ML, Shanks GD, Cheng Q, 2010. A large proportion of asymptomatic Plasmodium infections with low and sub-microscopic parasite densities in the low transmission setting of Temotu Province, Solomon Islands: challenges for malaria diagnostics in an elimination setting. Malar J 9: 254.
    1. Hawkins KBR, Labarre P, 2014. Diagnostic to Support Malaria Elimination: Choosing an Appropriate Biomarker to Target the Subclinical Plasmodium falciparum Reservoir. IEEE 2014 Global Humanitarian Technology Conference, San Jose, CA, 561–568.
    1. Bousema JT, Gouagna LC, Drakeley CJ, Meutstege AM, Okech BA, Akim IN, Beier JC, Githure JI, Sauerwein RW, 2004. Plasmodium falciparum gametocyte carriage in asymptomatic children in western Kenya. Malar J 3: 18.
    1. Bousema T, Okell L, Felger I, Drakeley C, 2014. Asymptomatic malaria infections: detectability, transmissibility and public health relevance. Nat Rev Microbiol 12: 833–840.
    1. Laishram DD, Sutton PL, Nanda N, Sharma VL, Sobti RC, Carlton JM, Joshi H, 2012. The complexities of malaria disease manifestations with a focus on asymptomatic malaria. Malar J 11: 29.
    1. Laban NM, Kobayashi T, Hamapumbu H, Sullivan D, Mharakurwa S, Thuma PE, Shiff CJ, Moss WJ;Southern Africa International Centers of Excellence for Malaria Research, 2015. Comparison of a PfHRP2-based rapid diagnostic test and PCR for malaria in a low prevalence setting in rural southern Zambia: implications for elimination. Malar J 14: 25.
    1. Roper MHTR, Goicochea CG, Andersen EM, Guarda JS, Calampa C, Hightower AW, Magill AJ, 2000. The epidemiology of malaria in an epidemic area of the Peruvian Amazon. Am J Trop Med Hyg 62: 247–256.
    1. Alves FPGL, Marrelli MT, Ribolla PE, Camargo EP, Da Silva LH, 2005. Asymptomatic carriers of Plasmodium spp. as infection source for malaria vector mosquitoes in the Brazilian Amazon. J Med Entomol 42: 777–779.
    1. Roshanravan BKE, Gilman RH, Cabrera L, Lee E, Metcalfe J, Calderon M, Lescano AG, Montenegro SH, Calampa C, Vinetz JM, 2003. Endemic malaria in the Peruvian Amazon region of Iquitos. Am J Trop Med Hyg 69: 45–52.
    1. Cerutti CJ, Boulos M, Coutinho AF, Hatab MC, Falqueto A, Rezende HR, Duarte AM, Collins W, Malafronte RS, 2007. Epidemiologic aspects of the malaria transmission cycle in an area of very low incidence in Brazil. Malar J 6: 33.
    1. Cucunubá ZM, Guerra ÁP, Rahirant SJ, Rivera JA, Cortés LJ, Nicholls RS, 2008. Asymptomatic Plasmodium spp. infection in Tierralta, Colombia. Mem Inst Oswaldo Cruz 103: 668–673.
    1. Imwong M, Nguyen TN, Tripura R, Peto TJ, Lee SJ, Lwin KM, Suangkanarat P, Jeeyapant A, Vihokhern B, Wongsaen K, Van Hue D, Dong LT, Nguyen T-U, Lubell Y, von Seidlein L, Dhorda M, Promnarate C, Snounou G, Malleret B, Rénia L, Keereecharoen L, Singhasivanon P, Sirithiranont P, Chalk J, Nguon C, Hien TT, Day N, White NJ, Dondorp A, Nosten F, 2015. The epidemiology of subclinical malaria infections in South-East Asia: findings from cross-sectional surveys in Thailand–Myanmar border areas, Cambodia, and Vietnam. Malar J 14: 381.
    1. Okell LC, Bousema T, Griffin JT, Ouédraogo AL, Ghani AC, Drakeley CJ, 2012. Factors determining the occurrence of submicroscopic malaria infections and their relevance for control. Nat Commun 3: 1237.
    1. Kifude CM, Rajasekariah HG, Sullivan DJ, Stewart VA, Angov E, Martin SK, Diggs CL, Waitumbi JN, 2008. Enzyme-linked immunosorbent assay for detection of Plasmodium falciparum histidine-rich protein 2 in blood, plasma, and serum. Clin Vaccine Immunol 15: 1012–1018.
    1. WHO, 1999. New Perspectives: Malaria Diagnosis. Geneva, Switzerland: Report of joint WHO/USAID informal consultation.
    1. Marquart L, Butterworth A, McCarthy JS, Gatton ML, 2012. Modelling the dynamics of Plasmodium falciparum histidine-rich protein 2 in human malaria to better understand malaria rapid diagnostic test performance. Malar J 11: 74.
    1. Jimenez A, Rees-Channer RR, Perera R, Gamboa D, Chiodini PL, Gonzalez IJ, Mayor A, Ding XC, 2017. Analytical sensitivity of current best-in-class malaria rapid diagnostic tests. Malar J 16: 128.
    1. Spring MPM, Ockenhouse C, 2014. Controlled human malaria infection. J Infect Dis 209: S40–S45.
    1. Andrews LAR, Webster D, Dunachie S, Walther RM, Bejon P, Hunt-Cooke A, Bergson G, Sanderson F, Hill AV, Gilbert SC, 2005. Quantitative real-time polymerase chain reaction for malaria diagnosis and its use in malaria vaccine clinical trials. Am J Trop Med Hyg 73: 191–198.
    1. Kamya MR, Arinaitwe E, Wanzira H, Katureebe A, Barusya C, Kigozi SP, Kilama M, Tatem AJ, Rosenthal PJ, Drakeley C, Lindsay SW, Staedke SG, Smith DL, Greenhouse B, Dorsey G, 2015. Malaria transmission, infection, and disease at three sites with varied transmission intensity in Uganda: implications for malaria control. Am J Trop Med Hyg 92: 903–912.
    1. Hodgson SH, Douglas AD, Edwards NJ, Kimani D, Elias SC, Chang M, Daza G, Seilie AM, Magiri C, Muia A, Juma EA, Cole AO, Rampling TW, Anagnostou NA, Gilbert SC, Hoffman SL, Draper SJ, Bejon P, Ogutu B, Marsh K, Hill AVS, Murphy SC, 2015. Increased sample volume and use of quantitative reverse-transcription PCR can improve prediction of liver-to-blood inoculum size in controlled human malaria infection studies. Malar J 14: 33.
    1. Murphy SC, Daza G, Chang M, Coombs R, 2012. Laser cutting eliminates nucleic acid cross-contamination in dried-blood-spot processing. J Clin Microbiol 50: 4128–4130.
    1. Billman ZP, Seilie AM, Murphy SC, 2016. Purification of Plasmodium Sporozoites Enhances Parasite-Specific CD8+ T Cell Responses. Infect Immun 84: 2233–2242.
    1. Imwong M, Hanchana S, Malleret B, Renia L, Day NP, Dondorp A, Nosten F, Snounou G, White NJ, 2014. High-throughput ultrasensitive molecular techniques for quantifying low-density malaria parasitemias. J Clin Microbiol 52: 3303–3309.
    1. Imwong M, Nguyen TN, Tripura R, Peto TJ, Lee SJ, Lwin KM, Suangkanarat P, Jeeyapant A, Vihokhern B, Wongsaen K, Van Hue D, Dong le T, Nguyen TU, Lubell Y, von Seidlein L, Dhorda M, Promnarate C, Snounou G, Malleret B, Renia L, Keereecharoen L, Singhasivanon P, Sirithiranont P, Chalk J, Nguon C, Hien TT, Day N, White NJ, Dondorp A, Nosten F, 2015. The epidemiology of subclinical malaria infections in South-East Asia: findings from cross-sectional surveys in Thailand-Myanmar border areas, Cambodia, and Vietnam. Malar J 14: 381.
    1. Doolan DL, Dobaño C, Baird JK, 2009. Acquired immunity to malaria. Clin Microbiol Rev 22: 13–36.
    1. Gupta SSR, Donnelly CA, Marsh K, Newbold C, 1999. Immunity to non-cerebral severe malaria is acquired after one or two infections. Nat Med 5: 340–343.
    1. Bashir IM, Otsyula N, Awinda G, Spring M, Schneider P, Waitumbi JN, 2013. Comparison of PfHRP-2/pLDH ELISA, qPCR and microscopy for the detection of Plasmodium events and prediction of sick visits during a malaria vaccine study. PLoS One 8: e56828.
    1. Waitumbi JN, Gerlach J, Afonina I, Anyona SB, Koros JN, Siangla J, Ankoudinova I, Singhal M, Watts K, Polhemus ME, Vermeulen NM, Mahoney W, Steele M, Domingo GJ, 2011. Malaria prevalence defined by microscopy, antigen detection, DNA amplification and total nucleic acid amplification in a malaria-endemic region during the peak malaria transmission season. Trop Med Int Health 16: 786–793.
    1. JP MJaG, 2013. Malaria rapid diagnostic tests: challenges and prospects. J Med Microbiol 62: 1491–1505.
    1. David PH, Hommel M, Miller LH, Udeinya IJ, Oligino LD, 1983. Parasite sequestration in Plasmodium falciparum malaria: spleen and antibody modulation of cytoadherence of infected erythrocytes. Proc Natl Acad Sci USA 80: 5075–5079.
    1. Wu L, van den Hoogen LL, Slater H, Walker PGT, Ghani AC, Drakeley CJ, Okell LC, 2015. Comparison of diagnostics for the detection of asymptomatic Plasmodium falciparum infections to inform control and elimination strategies. Nature 528: S86–S93.
    1. Tjitra E, Suprianto S, McBroom J, Currie BJ, Anstey NM, 2001. Persistent ICT malaria P.f/P.v panmalarial and HRP2 antigen reactivity after treatment of Plasmodium falciparum malaria is associated with gametocytemia and results in false-positive diagnoses of Plasmodium vivax in convalescence. J Clin Microbiol 39: 1025–1031.
    1. Iqbal J, Siddique A, Jameel M, Hira PR, 2004. Persistent histidine-rich protein 2, parasite lactate dehydrogenase, and panmalarial antigen reactivity after clearance of Plasmodium falciparum monoinfection. J Clin Microbiol 42: 4237–4241.
    1. Nyunt MH, Kyaw MP, Win KK, Myint KM, Nyunt KM, 2013. Field evaluation of HRP2 and pan pLDH-based immunochromatographic assay in therapeutic monitoring of uncomplicated falciparum malaria in Myanmar. Malar J 12: 123.
    1. Mishra B, Samantaray JC, Kumar A, Mirdha BR, 1999. Study of false positivity of two rapid antigen detection tests for diagnosis of Plasmodium falciparum malaria. J Clin Microbiol 37: 1233.
    1. Amoah LE, Abankwa J, Oppong A, 2016. Plasmodium falciparum histidine rich protein-2 diversity and the implications for PfHRP 2: based malaria rapid diagnostic tests in Ghana. Malar J 15: 101.
    1. Gamboa D, Ho MF, Bendezu J, Torres K, Chiodini PL, Barnwell JW, Incardona S, Perkins M, Bell D, McCarthy J, Cheng Q, 2010. A large proportion of P. falciparum isolates in the Amazon region of Peru lack pfhrp2 and pfhrp3: implications for malaria rapid diagnostic tests. PLoS One 5: e8091.
    1. Murillo Solano C, Akinyi Okoth S, Abdallah JF, Pava Z, Dorado E, Incardona S, Huber CS, Macedo de Oliveira A, Bell D, Udhayakumar V, Barnwell JW, 2015. Deletion of Plasmodium falciparum histidine-rich protein 2 (pfhrp2) and histidine-rich protein 3 (pfhrp3) genes in Colombian parasites. PLoS One 10: e0131576.

Source: PubMed

3
Subscribe