Development of a prototype lateral flow immunoassay (LFI) for the rapid diagnosis of melioidosis

Raymond L Houghton, Dana E Reed, Mark A Hubbard, Michael J Dillon, Hongjing Chen, Bart J Currie, Mark Mayo, Derek S Sarovich, Vanessa Theobald, Direk Limmathurotsakul, Gumphol Wongsuvan, Narisara Chantratita, Sharon J Peacock, Alex R Hoffmaster, Brea Duval, Paul J Brett, Mary N Burtnick, David P Aucoin, Raymond L Houghton, Dana E Reed, Mark A Hubbard, Michael J Dillon, Hongjing Chen, Bart J Currie, Mark Mayo, Derek S Sarovich, Vanessa Theobald, Direk Limmathurotsakul, Gumphol Wongsuvan, Narisara Chantratita, Sharon J Peacock, Alex R Hoffmaster, Brea Duval, Paul J Brett, Mary N Burtnick, David P Aucoin

Abstract

Burkholderia pseudomallei is a soil-dwelling bacterium and the causative agent of melioidosis. Isolation of B. pseudomallei from clinical samples is the "gold standard" for the diagnosis of melioidosis; results can take 3-7 days to produce. Alternatively, antibody-based tests have low specificity due to a high percentage of seropositive individuals in endemic areas. There is a clear need to develop a rapid point-of-care antigen detection assay for the diagnosis of melioidosis. Previously, we employed In vivo Microbial Antigen Discovery (InMAD) to identify potential B. pseudomallei diagnostic biomarkers. The B. pseudomallei capsular polysaccharide (CPS) and numerous protein antigens were identified as potential candidates. Here, we describe the development of a diagnostic immunoassay based on the detection of CPS. Following production of a CPS-specific monoclonal antibody (mAb), an antigen-capture immunoassay was developed to determine the concentration of CPS within a panel of melioidosis patient serum and urine samples. The same mAb was used to produce a prototype Active Melioidosis Detect Lateral Flow Immunoassay (AMD LFI); the limit of detection of the LFI for CPS is comparable to the antigen-capture immunoassay (∼0.2 ng/ml). The analytical reactivity (inclusivity) of the AMD LFI was 98.7% (76/77) when tested against a large panel of B. pseudomallei isolates. Analytical specificity (cross-reactivity) testing determined that 97.2% of B. pseudomallei near neighbor species (35/36) were not reactive. The non-reactive B. pseudomallei strain and the reactive near neighbor strain can be explained through genetic sequence analysis. Importantly, we show the AMD LFI is capable of detecting CPS in a variety of patient samples. The LFI is currently being evaluated in Thailand and Australia; the focus is to optimize and validate testing procedures on melioidosis patient samples prior to initiation of a large, multisite pre-clinical evaluation.

Conflict of interest statement

RLH and HC are employed by a commercial company, InBios International. DPA has a patent submitted for the detection of B. pseudomallei capsular polysaccharide. Both of these competing interests do not alter our adherence to all PLOS NTDs policies on sharing data and materials.

Figures

Figure 1. Calculation of mAb 3C5 affinity…
Figure 1. Calculation of mAb 3C5 affinity for CPS.
A BIAcore X100 instrument was used to determine the affinity of mAb 3C5 for CPS. Biotinylated CPS was immobilized on the surface of a streptavidin sensor chip. Samples (two-fold serial dilution of mAb 3C5 [333–5.2 nM]) were injected over the sensor surface for 60 s, after which the mAb was allowed to passively dissociate for 120 s (left panel). The dissociation constant (KD) was determined using the steady-state model in BIAevaluation software (right panel).
Figure 2. Detection of purified CPS by…
Figure 2. Detection of purified CPS by antigen-capture ELISA.
mAb 3C5 was used in the capture phase of the ELISA at the concentrations listed. Following a wash and blocking step, purified CPS was serially diluted across the microtiter plate at the concentrations listed. The wells were then washed and HRP-labeled mAb 3C5 was used in the indicator phase to detect captured CPS. The ELISA was performed in triplicate and mean values are plotted.
Figure 3. Prototype Active Melioidosis Detect (AMD)…
Figure 3. Prototype Active Melioidosis Detect (AMD) LFI.
(A) Schematic of LFI components. (B) B. pseudomallei strain Bp82 colony grown on an agar plate was picked and suspended in 2 drops of lysis buffer. The lysate was added to the sample pad followed by three drops of LFI chase buffer (top LFI). The LFI was imaged following a 15 min run time. The same test condition were used with a colony of E. coli (bottom LFI).
Figure 4. Determination of the LOD of…
Figure 4. Determination of the LOD of the AMD LFI.
(A) Purified CPS was diluted in chase buffer at the indicated concentration and applied to the LFI sample pad. Results were photographed after 15 min. Purified CPS was also diluted in human control sera (B) and human control urine (C).
Figure 5. Prototype AMD LFI for detection…
Figure 5. Prototype AMD LFI for detection of B. pseudomallei CPS in melioidosis patient samples.
(A) Preliminary testing of a variety of archived patient samples from Australia and Thailand. (B) Detection of CPS in melioidosis patient urine samples (filtered) listed in Table 2. Urine (50 µl) was combined with 100 µl of chase buffer and applied to the sample pad. Note that samples that were positive by antigen-capture immunoassay (Table 2) were also positive by LFI and the levels of CPS detected between both assays are congruent.

References

    1. Wiersinga WJ, Currie BJ, Peacock SJ (2012) Melioidosis. N Engl J Med 367: 1035–1044.
    1. Currie BJ, Ward L, Cheng AC (2010) The epidemiology and clinical spectrum of melioidosis: 540 cases from the 20 year Darwin prospective study. PLoS neglected tropical diseases 4: e900.
    1. Limmathurotsakul D, Wongratanacheewin S, Teerawattanasook N, Wongsuvan G, Chaisuksant S, et al. (2010) Increasing incidence of human melioidosis in Northeast Thailand. Am J Trop Med Hyg 82: 1113–1117.
    1. Sirisinha S, Anuntagool N, Dharakul T, Ekpo P, Wongratanacheewin S, et al. (2000) Recent developments in laboratory diagnosis of melioidosis. Acta Trop 74: 235–245.
    1. Wongsuvan G, Limmathurotsakul D, Wannapasni S, Chierakul W, Teerawattanasook N, et al. (2009) Lack of correlation of Burkholderia pseudomallei quantities in blood, urine, sputum and pus. Southeast Asian J Trop Med Public Health 40: 781–784.
    1. Wuthiekanun V, Limmathurotsakul D, Wongsuvan G, Chierakul W, Teerawattanasook N, et al. (2007) Quantitation of B. Pseudomallei in clinical samples. Am J Trop Med Hyg 77: 812–813.
    1. Limmathurotsakul D, Jamsen K, Arayawichanont A, Simpson JA, White LJ, et al. (2010) Defining the true sensitivity of culture for the diagnosis of melioidosis using Bayesian latent class models. PLoS One 5: e12485.
    1. Wuthiekanun V, Dance DA, Wattanagoon Y, Supputtamongkol Y, Chaowagul W, et al. (1990) The use of selective media for the isolation of Pseudomonas pseudomallei in clinical practice. J Med Microbiol 33: 121–126.
    1. Inglis TJ, Merritt A, Chidlow G, Aravena-Roman M, Harnett G (2005) Comparison of diagnostic laboratory methods for identification of Burkholderia pseudomallei . J Clin Microbiol 43: 2201–2206.
    1. Nuti DE, Crump RB, Dwi Handayani F, Chantratita N, Peacock SJ, et al. (2011) Identification of circulating bacterial antigens by in vivo microbial antigen discovery. mBio Aug 16;2 (4)
    1. AuCoin DP (2012) In vivo microbial antigen discovery: finding the ‘needle in the haystack’. Expert Rev Mol Diagn 12: 219–221.
    1. Perry MB, MacLean LL, Schollaardt T, Bryan LE, Ho M (1995) Structural characterization of the lipopolysaccharide O antigens of Burkholderia pseudomallei . Infect Immun 63: 3348–3352.
    1. Ashdown LR (1979) An improved screening technique for isolation of Pseudomonas pseudomallei from clinical specimens. Pathology 11: 293–297.
    1. Peterman JH (1991) Immunochemistry of solid-phase immunoassay; Butler JE, editor. Boca Raton, FL.: CRC Press, Inc.
    1. AuCoin DP, Crump RB, Thorkildson P, Nuti DE, LiPuma JJ, et al. (2010) Identification of Burkholderia cepacia complex bacteria with a lipopolysaccharide-specific monoclonal antibody. Journal of medical microbiology 59: 41–47.
    1. Propst KL, Mima T, Choi KH, Dow SW, Schweizer HP (2010) A Burkholderia pseudomallei deltapurM mutant is avirulent in immunocompetent and immunodeficient animals: candidate strain for exclusion from select-agent lists. Infect Immun 78: 3136–3143.
    1. (2013): AOAC International.
    1. Heiss C, Burtnick MN, Wang Z, Azadi P, Brett PJ (2012) Structural analysis of capsular polysaccharides expressed by Burkholderia mallei and Burkholderia pseudomallei . Carbohydrate research 349: 90–94.
    1. Chantratita N, Wuthiekanun V, Limmathurotsakul D, Thanwisai A, Chantratita W, et al. (2007) Prospective clinical evaluation of the accuracy of 16S rRNA real-time PCR assay for the diagnosis of melioidosis. Am J Trop Med Hyg 77: 814–817.
    1. Limmathurotsakul D, Peacock SJ (2011) Melioidosis: a clinical overview. British medical bulletin 99: 125–139.
    1. Richardson LJ, Kaestli M, Mayo M, Bowers JR, Tuanyok A, et al. (2012) Towards a rapid molecular diagnostic for melioidosis: Comparison of DNA extraction methods from clinical specimens. Journal of microbiological methods 88: 179–181.
    1. Appassakij H, Silpapojakul KR, Wansit R, Pornpatkul M (1990) Diagnostic value of the indirect hemagglutination test for melioidosis in an endemic area. Am J Trop Med Hyg 42: 248–253.
    1. Kanaphun P, Thirawattanasuk N, Suputtamongkol Y, Naigowit P, Dance DA, et al. (1993) Serology and carriage of Pseudomonas pseudomallei: a prospective study in 1000 hospitalized children in northeast Thailand. J Infect Dis 167: 230–233.
    1. Cheng AC, Wuthiekanun V, Limmathurotsakul D, Chierakul W, Peacock SJ (2008) Intensity of exposure and incidence of melioidosis in Thai children. Trans R Soc Trop Med Hyg 102 Suppl 1: S37–39.
    1. Wuthiekanun V, Chierakul W, Langa S, Chaowagul W, Panpitpat C, et al. (2006) Development of antibodies to Burkholderia pseudomallei during childhood in melioidosis-endemic northeast Thailand. Am J Trop Med Hyg 74: 1074–1075.
    1. Cheng AC, O'Brien M, Freeman K, Lum G, Currie BJ (2006) Indirect hemagglutination assay in patients with melioidosis in northern Australia. Am J Trop Med Hyg 74: 330–334.
    1. Tandhavanant S, Wongsuvan G, Wuthiekanun V, Teerawattanasook N, Day NP, et al. (2013) Monoclonal antibody-based immunofluorescence microscopy for the rapid identification of Burkholderia pseudomallei in clinical specimens. Am J Trop Med Hyg 89: 165–168.
    1. Wuthiekanun V, Desakorn V, Wongsuvan G, Amornchai P, Cheng AC, et al. (2005) Rapid immunofluorescence microscopy for diagnosis of melioidosis. Clin Diagn Lab Immunol 12: 555–556.
    1. Chantratita N, Tandhavanant S, Wongsuvan G, Wuthiekanun V, Teerawatanasuk N, et al. (2013) Rapid Detection of Burkholderia pseudomallei in Blood Cultures Using a Monoclonal Antibody-Based Immunofluorescent Assay. Am J Trop Med Hyg 89: 971–972.
    1. Ekpo P, Rungpanich U, Pongsunk S, Naigowit P, Petkanchanapong V (2007) Use of protein-specific monoclonal antibody-based latex agglutination for rapid diagnosis of Burkholderia pseudomallei infection in patients with community-acquired septicemia. Clin Vaccine Immunol 14: 811–812.
    1. Wuthiekanun V, Anuntagool N, White NJ, Sirisinha S (2002) Short report: a rapid method for the differentiation of Burkholderia pseudomallei and Burkholderia thailandensis . Am J Trop Med Hyg 66: 759–761.
    1. Samosornsuk N, Lulitanond A, Saenla N, Anuntagool N, Wongratanacheewin S, et al. (1999) Short report: evaluation of a monoclonal antibody-based latex agglutination test for rapid diagnosis of septicemic melioidosis. Am J Trop Med Hyg 61: 735–737.
    1. Anuntagool N, Naigowit P, Petkanchanapong V, Aramsri P, Panichakul T, et al. (2000) Monoclonal antibody-based rapid identification of Burkholderia pseudomallei in blood culture fluid from patients with community-acquired septicaemia. J Med Microbiol 49: 1075–1078.
    1. Bartlett JG (2011) Diagnostic tests for agents of community-acquired pneumonia. Clin Infect Dis 52 Suppl 4: S296–304.
    1. Jarraud S, Descours G, Ginevra C, Lina G, Etienne J (2013) Identification of legionella in clinical samples. Methods Mol Biol 954: 27–56.
    1. Price EP, Sarovich DS, Mayo M, Tuanyok A, Drees KP, et al. (2013) Within-host evolution of Burkholderia pseudomallei over a twelve-year chronic carriage infection. MBio 4.
    1. Cuccui J, Milne TS, Harmer N, George AJ, Harding SV, et al. (2012) Characterization of the Burkholderia pseudomallei K96243 capsular polysaccharide I coding region. Infect Immun 80: 1209–1221.
    1. Sim BM, Chantratita N, Ooi WF, Nandi T, Tewhey R, et al. (2010) Genomic acquisition of a capsular polysaccharide virulence cluster by non-pathogenic Burkholderia isolates. Genome Biol 11: R89.
    1. Glass MB, Gee JE, Steigerwalt AG, Cavuoti D, Barton T, et al. (2006) Pneumonia and septicemia caused by Burkholderia thailandensis in the United States. J Clin Microbiol 44: 4601–4604.

Source: PubMed

3
Předplatit