ArticlePDF Available

Multiplex lateral flow detection and binary encoding enables a molecular colorimetric 7-segment display

Authors:

Abstract and Figures

Multiplex expansion in point-of-care diagnostics usually requires a linear increase of premium commodities such as reagents or space. Here we demonstrate the power of binary and molecular encoding to compress device operations. We describe the first colorimetric 7-segment display on a paper-based biosensor, providing compact and intuitive read-outs for multiplex detections.
Content may be subject to copyright.
Registered charity number: 207890
Featuring work from the Molecular Engineering Research
Group in the School of Science and Engineering at the
University of the Sunshine Coast, Queensland, Australia.
Title: Multiplex lateral fl ow detection and binary encoding
enables a molecular colorimetric 7-segment display
Binary and molecular encoding enables a digital-like 7 segment
display on a paper-based biosensor. The display operates without
batteries and wires, providing an e cient, compact, and intuitive
output for multiplexing applications.
As featured in:
See Jia Li and Joanne Macdonald,
Lab Chip, 2016, 16, 242.
www.rsc.org/loc
Lab on a Chip
COMMUNICATION
Cite this: Lab Chip,2016,16,242
Received 25th October 2015,
Accepted 24th November 2015
DOI: 10.1039/c5lc01323b
www.rsc.org/loc
Multiplex lateral flow detection and binary
encoding enables a molecular colorimetric
7-segment display
Jia Li
a
and Joanne Macdonald*
ab
Multiplex expansion in point-of-care diagnostics usually requires a
linear increase of premium commodities such as reagents or
space. Here we demonstrate the power of binary and molecular
encoding to compress device operations. We describe the first
colorimetric 7-segment display on a paper-based biosensor, pro-
viding compact and intuitive read-outs for multiplex detections.
Multiplexing is a critical parameter for increasing diagnostic
efficiency. The strategies that enable simultaneous analysis of
multiple samples are largely dependent on the underlying
diagnostic technology. Multiplex detection using enzyme-
linked immunosorbent assays, real-time polymerase chain
reactions, microarrays and/or bead-based methods (such as
the Bio-Rad Bio-Plex®Systems
1
and Luminex MagPix®
2
)
enable high-throughput and low volume processing, but
require non-portable equipment and trained personnel for
operation. For point-of-care settings, lateral flow devices
(LFDs) are ideal candidates, but few commercial devices
employ multiplexing. This is due to issues with specificity
and reproducibility, as well as expansion limits, as described
by Washburn, because the flow rate decreases with distance
from the conjugate pad.
3
Here we propose a novel solution
for expanding LFD multiplex detection that increases the effi-
ciency of detection without expanding the device dimensions
and consuming excess reagents. Our generic solution com-
presses multiplex LFD data by borrowing from computational
science and applying binary encoding to create signature pat-
terns of test dots. Thus, a sample is diagnosed based on the
set of test dots that appear on the device. By judicious
arrangement of test dots, we create a 7-segment LFD display
that simulates digital display of information for easy interpre-
tation by the end-user.
To develop a generic multiplex LFD, we first identified
twelve individual antigenantibody single-plex assays able to
operate using a sandwich immunoassay format. In this format
an analyte is sandwiched between a capture and detection anti-
body via tagged antigens (Fig. 1A). Gold nanoparticles (AuNPs)
conjugated to the capture antibody transform analyte detection
into a colorimetric signal. We chose fluorescein and anti-
fluorescein as the common AuNP conjugated capture antibody,
and identified an additional twelve commercially available anti-
genantibody combinations that could potentially be used as
detection antibodies (Fig. 1B). The analyte was a single-
stranded DNA corresponding to a segment of the L gene of
Rift Valley fever virus (RVFV).
4
Importantly, we kept the RVFV
DNA analyte sequence identical in all tests (apart from the 3
antigen labels) to eliminate any differences in behavior due
to subtle changes in nucleotide sequences.
5
In addition, use
of a dual-labeled synthetic analyte reduced the complexity of
interactions for our proof-of-concept demonstration (applied
implementation would utilize labeled oligonucleotide, anti-
body, or aptamer binding partners to capture an unlabeled
analyte). All twelve antigenantibody pairs were effective as
detection entities in the single-plex LFDs (Fig. 1 & S1), with
standard LFD sensitivities as low as 5 nM.
6
We note in partic-
ular the success of labels Alexa Fluor®488, Cascade Blue®,
Lucifer yellow, benzopyrene, BODIPY®FL, and dansyl, which
have not previously been applied in lateral flow detection.
After demonstrating successful operation of all twelve
detection antigenantibody combinations in a single-plex
LFD, we then attempted to combine these into a multiplex
LFD array using a dot-matrix (3 ×4) format. For the arrange-
ment of detection antibodies on the membrane, we hypothe-
sized that the antibodies furthest from the conjugate pad
would suffer the most from loss of reagents, and thus placed
the least sensitive detection antibodies closest to the conju-
gate pad (Fig. S2A). Specificity testing indicated three non-
specific reactions (Fig. S2B). To investigate the lack of
242 |Lab Chip,2016,16,242245 This journal is © The Royal Society of Chemistry 2016
a
Inflammation and Healing Research Cluster, Genecology Research Center, School
of Science and Engineering, University of the Sunshine Coast, Locked Bag 4,
Maroochydore DC, Queensland 4558, Australia. E-mail: jmacdon1@usc.edu.au
b
Division of Experimental Therapeutics, Department of Medicine, Columbia
University, New York, NY 10032, USA. E-mail: jm2236@columbia.edu
Electronic supplementary information (ESI) available. See DOI: 10.1039/
c5lc01323b
Lab Chip,2016,16,242245 | 243This journal is © The Royal Society of Chemistry 2016
binding for BODIPY®FL and dansyl antigenantibody pairs,
we reduced the LFD to a 7-dot array containing six detection
antibodies (chosen due to their superior specificity; see Fig.
S2), with a seventh detection antibody of either anti-
BodipyFL, anti-dansyl, or anti-Cy5 detection antibody (Fig. 2A;
anti-Cy5 was included as a comparative control). These
were tested with a single dual-labeled analyte containing
either 3BODIPY®FL, dansyl, or Cy®5 antigens, or with
an analyte reaction mix containing the corresponding seven
antigens. Interestingly, the BODIPY®FL antigen still did not
produce any observable test dots, whereas the dansyl test dots
appeared and showed specificity in the seven antigen-
antibody pairs system (Fig. 2). The lack of BODIPY®FL binding
(Fig. 2 & S2) is most likely due to the larger surface area
ablating detection, since BODIPY®FL was the least sensitive
in the single-plex assays (Fig. 1). However, the lack of dansyl
binding in the 3 ×4 array but successful binding in the 7-dot
array was consistent in multiple tests. This suggests the pres-
ence of other detection antibodies precluded dansyl binding,
despite it being placed in the first row of the 3 ×4 array. The
7-dot array that incorporated dansyl (Fig. 2), demonstrated
significant relative higher dot intensity (p<0.05) compared
to the Cy®5 (Fig. 1 and 2), and was used for all subsequent
demonstrations. These results support the notion that
specificity is a hindrance in expansion into multiplexing, but
with judicious design and repeated testing, compatible com-
binations can be identified that do not cross-react but
instead reproducibly give selective responses.
On a linear scale, our 7-dot LFD array is an improvement
on the current maximum multiplex detection that employs
antigenantibody reactions, since only penta-plex LFD has
been previously reported.
7
However, stacking the test lines
(or dots) along the flow path of the lateral flow device does
not provide an intuitive result output.
8
Inspired by the
electronic information display systems used in digital
watches and elevators, we considered development of a
proof-of-concept 7-segment LFD display. We devised a detec-
tion system to represent each segment of a 7-segment display
with one detection antibody (from the seven) (Fig. 3A and S3A).
Fig. 2 Hepta-plex lateral flow detection results for combinations of
seven antigenantibody pairs. A: Positioning of detection antibodies
deposited upon lateral flow strips. B: Hepta-plex lateral flow detection of
1μM dual-labeled analyte alone (containing either 3BODIPY®FL, Cy®5,
or dansyl), or combinations of dual-labeled analyte (1 μM) containing 3
biotin, Cascade Blue®, digoxigenin, dintrophenyl, tetramethylrhodamine
(TAMRA), and Texas Red®and either BODIPY®FL, Cy®5 or dansyl, as
indicated. Assay was repeated three times and a representative photo-
graph is shown. Experimental details are described in the ESI.
Fig. 3 7-Segment display of numbers on lateral flow strips. A:
Positioning of the detection antibodies to form the 7-segments of the
display. B: Addition of labeled analyte signature mixtures (1 μM each)
resulted in the successful appearance of numbers (0 to 9). The assay
was performed three times with similar results; a photograph of one
test is shown. Experimental details are described in the ESI.
Fig. 1 (A) Design of single-plex LFDs: a dual-labeled analyte (RVFV
DNA) was sandwiched between capture and detection antibodies due
to antigenantibody binding; AuNPs conjugated to the capture anti-
body (mouse anti-fluorescein antibody) enabled visualization of bind-
ing through the appearance of a red color at the test dot; rabbit anti-
mouse antibody, which can directly bind the capture antibody (mouse
anti-fluorescein), was deposited in parallel as a control. (B) Determina-
tion of detection limits for each single-plex assay: detection antibodies
corresponding to the 3antigen of dual-labeled RVFV were deposited
at the test dot; resultant assay color intensity (average and standard
deviation of 4 individual tests) was quantified using ImageJ software
and plotted against DNA concentration; a cut-off defined as three
standard deviations above the average was used to determine the limit
of detection for each assay. Experimental details are described in the
ESI.
Lab on a Chip Communication
244 |Lab Chip,2016,16,242245 This journal is © The Royal Society of Chemistry 2016
Initially we used duplicate test dots to represent each seg-
ment, however, this resulted in position effects where the
morphology of the second test dot was influenced by the first
test dot in the vertical segments (Fig. S3). We thus reduced
the vertical segments to only incorporate one test dot
(Fig. 3A). By addition of appropriate label mixtures, we con-
sistently demonstrated clear numbers (09) on the 7-segment
LFD display (Fig. 3B).
Our successful demonstration of the numbers 09 using
seven antigenantibody pairs on a LFD is the first digital-like
display of numbers on a paper-based biosensor. It operates
as a single-use static output that employs a pre-defined
molecular encoding strategy for information transfer. The
work is a significant advance on our previous molecular
7-segment display, based on molecular logic gates, which
required multiple additions of samples to each segment.
5
Here the solid-phase LFD enables a single addition of sample
to create a display, made possible through the novel combi-
nation of both molecular and binary encoding. This is nota-
bly different from other LFD computational strategies, which
showcase embedded molecular logic gates.
9,10
Our method
also advances previous multiplex LFDs, which use multiple
lines,
7,11
bi-directional,
12,13
parallel,
14,15
and multi-directional
systems.
16,17
Notably, intuitive read-outs were previously pro-
posed by Shen et al., where blood types on a paper-based bio-
sensor were displayed using letters deposited in different sec-
tions of the biosensor (not overlayed).
18,19
However,
expansion of these multiplex LFDs is hampered by either
space limitations within the original device dimension or the
requirement to consume more reagents as the device size
increases. In contrast, our novel combination of both binary
and molecular encoding demonstrates improved multiplexing
efficiency while minimizing excess reagent requirements.
The most compact LFD system to date is the microarray
LFD,
20,21
which similarly minimizes both reagent consump-
tion and device dimension. The largest demonstration to
date is detection of 384 recombinant protein antigens for
analysis of human protein atlas antibody cross-reactivities.
21
Here we provide further compaction by applying binary
encoding. Our generic antigenantibody method could theo-
retically detect 127 (2
7
1) discrete analytes using only 7
antigenantibody combinations. Noticeably, our system would
only apply to differentiation of discrete analytes: different
encoding schemes would need to be employed to differenti-
ate analyte mixtures. However, implementation of both
binary-encoded and traditional microarray LFD requires
external readers to interpret results. Our 7-segment LFD
intentionally restrict outputs to a numerical system to dem-
onstrate an intuitive read-out applicable to point-of-care diag-
nostics that does not require external reading devices.
In this study we used an artificial ideal analyte for proof-
of-concept demonstration of multiplex LFD displays. Impor-
tantly, these displays are generic and can be applied for the
detection of any candidate analyte (e.g. nucleic acids, proteins,
lipids, or small molecules) through the use of labeled binding
partners. Practically, this could be implemented via labeled
antibodies or aptamers for detection of their corresponding
antigens. In addition, we are also considering labeled
primers and probes for multiplex nucleic acid amplification,
which would additionally assist with detection of low copy
numbers. In this context, we note the advancement of iso-
thermal nucleic acid amplification technologies that offer
prospects for rapid point-of-care detection.
22,23
Determina-
tion of analyte concentration could be performed using
electronic reading devices (that assess intensity within a
defined test area regardless of dot morphology) and by pre-
training with known concentrations of analyte. Notably,
implementation of pattern displays requires the use of mix-
tures of labeled binding partners, such that an analyte must
combine with a unique subset of labels. This means that
multiple individual sub-reactions must work synchronously,
which ultimately increases confidence that can be drawn
from a successful display.
Conclusions
Our novel 7-segment LFD enables compaction of
multiplexing by borrowing from computational science and
employing a binary encoding scheme that moves beyond
space limitations. The easy-to-interpret results via a 7-seg-
ment display format are highly relevant to point-of-care appli-
cations. These displays are generic and can be applied for
multiplex detection of any candidates (e.g. nucleic acids, pro-
tein, lipid, or small molecules) if capture reagents incorpo-
rate our specific recognition labels. This is particularly ame-
nable for nucleic acid lateral flow detection, if upstream
nucleic acid amplification uses primers and probes that
incorporate the recognition labels.
22
Further improvements
could focus on employing advanced deposition techniques to
improve the performance and presentation of the outputs.
24
Acknowledgements
We thank John Bartlett, David McMillan, and Fabrice
Rossignol for support and advice, and Richard Burns for care-
ful reading of this manuscript. This work was funded by the
Queensland Government, Department of Science, Information
Technology, Innovation and the Arts (DSITIA, Australia), and
an internal HDR grant (University of the Sunshine Coast,
Australia).
Notes and references
1 I. Bio-Rad Laboratories, Bio-Rad Bio-Plex®Systems, http://
info.bio-rad.com/ww-Bio-Plex-Luminex-lp1.html?WT.srch=
1&WT.mc_id=aw-pqd-AU-bioplex_luminex_gold&WT.knsh_
id=631f362c-bf07-4e69-b01a-00fb1c8cb35f&gclid=COKLk_
mOjcgCFYiCvQodPpEJWA, Accessed September 23rd, 2015,
2015.
2 L. Corporation, Luminex MagPix®, http://www.luminexcorp.
com/clinical/instruments/magpix/, Accessed September 23rd,
2015, 2015.
3 E. W. Washburn, Phys. Rev., 1921, 17, 273283.
Lab on a ChipCommunication
Lab Chip,2016,16,242245 | 245This journal is © The Royal Society of Chemistry 2016
4 M. Euler, Y. Wang, O. Nentwich, O. Piepenburg, F. T. Hufert
and M. Weidmann, J. Clin. Virol., 2012, 54, 308312.
5 J. Macdonald, Y. Li, M. Sutovic, H. Lederman, K. Pendri, W.
Lu, B. L. Andrews, D. Stefanovic and M. N. Stojanovic, Nano
Lett., 2006, 6, 25982603.
6 J. Li, D. McMillan and J. Macdonald, Sens. Mater., 2015, 27,
549561.
7 P. Noguera, G. A. Posthuma-Trumpie, M. van Tuil, F. J. van
der Wal, A. de Boer, A. P. H. A. Moers and A. van
Amerongen, Anal. Bioanal. Chem., 2011, 399, 831838.
8 J. E. Poje, T. Kastratovic, A. R. Macdonald, A. C. Guillermo,
S. E. Troetti, O. J. Jabado, M. L. Fanning, D. Stefanovic and
J. Macdonald, Angew. Chem., Int. Ed., 2014, 53, 92229225.
9 J. Chen, Z. Fang, P. Lie and L. Zeng, Anal. Chem., 2012, 84,
63216325.
10 Y. Huang, W. Wen, D. Du, X. Zhang, S. Wang and Y. Lin,
Biosens. Bioelectron., 2014, 61, 598604.
11 L. Tian, T. Sato, K. Niwa, M. Kawase, A. C. Tanner and N.
Takahashi, BioMed Res. Int., 2014, 2014, 180323.
12 S. M. Hossain, C. Ozimok, C. Sicard, S. D. Aguirre, M. M. Ali,
Y. Li and J. D. Brennan, Anal. Bioanal. Chem., 2012, 403,
15671576.
13 S. M. Hossain, R. E. Luckham, M. J. McFadden and J. D.
Brennan, Anal. Chem., 2009, 81, 90559064.
14 D. a. C. Becton, BDEZ Flu A + B Test, http://www.bd.com/
ds/productCenter/256050.asp, Accessed January 12th, 2015.
15 A. Technologies, RAID, http://www.alexeter.com/biow/Products/
products/strips/RAID_DX.asp, Accessed January 12th, 2015.
16 C. Z. Li, K. Vandenberg, S. Prabhulkar, X. Zhu, L. Schneper,
K. Methee, C. J. Rosser and E. Almeide, Biosens. Bioelectron.,
2011, 26, 43424348.
17 K. L. Peters, I. Corbin, L. M. Kaufman, K. Zreibe, L. Blanes
and B. R. McCord, Anal. Methods, 2015, 7,6370.
18 M. Li, J. Tian, M. Al-Tamimi and W. Shen, Angew. Chem., Int.
Ed., 2012, 51, 54975501.
19 M. Li, W. L. Then, L. Li and W. Shen, Anal. Bioanal. Chem.,
2014, 406, 669677.
20 J. Gantelius, C. Hamsten, M. Neiman, J. M. Schwenk, A.
Persson and H. Andersson-Svahn, J. Microbiol. Methods,
2010, 82,1118.
21 J. Gantelius, T. Bass, R. Sjoberg, P. Nilsson and H.
Andersson-Svahn, Int. J. Mol. Sci., 2011, 12, 77487759.
22 J. Li and J. Macdonald, Biosens. Bioelectron., 2014, 64C,
196211.
23 A. James and J. Macdonald, Expert Rev. Mol. Diagn.,
2015, 15, 14751489.
24 J. Li, F. Rossignol and J. Macdonald, Lab Chip, 2015, 15,
25382558.
Lab on a Chip Communication
... Large-scale manufacturing of xLFTs requires the printing of multiple test spots in a single disposable strip, and thus, multiple dispensing nozzles with different quality-control measures, potentially increasing production costs. Furthermore, additional test lines complicate result interpretation, although this can be mitigated through the use of digital interpretation or test spot array patterns that are recognizable to users 166,167 . The development and commercialization of xLFTs are further limited by the need to validate multiple biomarkers with clinical importance, low market demand and a complicated regulatory approval pathway. ...
Article
The acceptability and feasibility of large-scale testing with lateral flow tests (LFTs) for clinical and public health purposes has been demonstrated during the COVID-19 pandemic. LFTs can detect analytes in a variety of samples, providing a rapid read-out, which allows self-testing and decentralized diagnosis. In this Review, we examine the changing LFT landscape with a focus on lessons learned from COVID-19. We discuss the implications of LFTs for decentralized testing of infectious diseases, including diseases of epidemic potential, the ‘silent pandemic’ of antimicrobial resistance, and other acute and chronic infections. Bioengineering approaches will play a key part in increasing the sensitivity and specificity of LFTs, improving sample preparation, incorporating nucleic acid amplification and detection, and enabling multiplexing, digital connection and green manufacturing, with the aim of creating the next generation of high-accuracy, easy-to-use, affordable and digitally connected LFTs. We conclude with recommendations, including the building of a global network of LFT research and development hubs to facilitate and strengthen future diagnostic resilience. The feasibility of large-scale testing with lateral flow tests has been demonstrated in the COVID-19 pandemic. This Review examines lessons learned from the COVID-19 pandemic to inform the design and bioengineering of next-generation lateral flow tests to strengthen future diagnostic resilience. Lateral flow tests (LFTs) were adopted at an unprecedented scale during the COVID-19 pandemic, enabling access to testing beyond healthcare settings.Only 0.4% of the 3 billion COVID-19 tests performed through to mid-2022 were conducted in low-income regions, raising ethical concerns and constraining our collective ability to respond to a pandemic.Key barriers to COVID-19 LFT development and adoption include lack of access to well characterized samples, limited accuracy, lack of connectivity, lack of evidence of cost-effectiveness, regulatory delays and centralized manufacturing capabilities.LFTs could also play an important part in the detection of other diseases of epidemic potential and antimicrobial resistance.Bioengineering approaches, such as the use of nano- and quantum materials, nucleic-acid-based LFTs, CRISPR and machine learning, will improve the sensitivity, specificity, multiplexing and connectivity features of LFTs.We recommend investing in an international LFT research and development hub network to spearhead the development of a pipeline of innovative bioengineering approaches to design next-generation LFTs. Lateral flow tests (LFTs) were adopted at an unprecedented scale during the COVID-19 pandemic, enabling access to testing beyond healthcare settings. Only 0.4% of the 3 billion COVID-19 tests performed through to mid-2022 were conducted in low-income regions, raising ethical concerns and constraining our collective ability to respond to a pandemic. Key barriers to COVID-19 LFT development and adoption include lack of access to well characterized samples, limited accuracy, lack of connectivity, lack of evidence of cost-effectiveness, regulatory delays and centralized manufacturing capabilities. LFTs could also play an important part in the detection of other diseases of epidemic potential and antimicrobial resistance. Bioengineering approaches, such as the use of nano- and quantum materials, nucleic-acid-based LFTs, CRISPR and machine learning, will improve the sensitivity, specificity, multiplexing and connectivity features of LFTs. We recommend investing in an international LFT research and development hub network to spearhead the development of a pipeline of innovative bioengineering approaches to design next-generation LFTs.
... To overcome this constraint, some equipment-free methods have been developed that allow quantitative or semi-quantitative measurements by PADs based on distance [217][218][219], count [220][221][222], time [223][224][225], and text [226][227][228] output instead of the color intensity. For instance, in a distance-based method reported by Cate et al. [229], the length of the color change on paper is correlated with the analyte concentration. ...
Article
Paper-based analytical devices (PADs) have shown great promise for point-of-care testing and on-site detection of analytes with chemical, biochemical, and environmental importance owing to their low cost, convenience, scalability, portability, and biocompatibility. The World Health Organization stated that sensors should meet the ASSURED criteria (affordable, sensitive, specific, user-friendly, rapid and robust, equipment-free, and deliverable). Paper-based optical sensors meet most of these criteria, making them in high demand and applicable in remote areas. Optical PADs outputs are obtained by different means, such as dyes, nanostructures, redox agents, and pH indicators. The outstanding physical and chemical characteristics of nanostructures, their intense signals, and tunable optical properties make them ideal for many sensing platforms, including paper-based ones. This review focuses primarily on paper-based nanosensors using various nanostructures to fabricate and produce optical signals for visualization. We describe the fundamentals and state of the art of PADs and comprehensively explain the following topics: paper types as the substrate of PADs, PAD fabrication approaches, nanostructure stabilization on PADs, signal acquisition, data handling, interpretation of results, sensing mechanisms, and application areas. We also discuss future trends and strategies to enable PADs to reach their full potential and increase their commercialization opportunities.
... Lateral flow dipstick detection of amplification (3) was performed using the Milenia Hybridetect II strips (Milenia Biotec, Giessen, Germany) pre-activated by adding 8 μL 0.4% casein in PBST to the sample pad (Rames and Macdonald, 2019). Amplicons (2 μL) were added to the lateral flow dipsticks, which were then placed into 100 μL running buffer (100 mM H 3 BO 3 , 100 mM Na 2 B 4 O 7 , 1% bovine serum albumin, 0.05% Tween 20, pH 8.8) (Li and Macdonald, 2016) for 5 min at room temperature. Completed lateral flow dipsticks were scanned using the Epson Perfection V39 Flatbed Scanner (Epson, New South Wales, Australia) and analysed as previously described (James et al., 2018). ...
Article
Insecticide resistance monitoring is essential in assessing the efficacy of vector control measures. However, gold standard PCR-based molecular analyses for insecticide resistance detection are often hindered by time-consuming sample processing, as well as considerable infrastructure and resourcing requirements. In this study, we combined a novel one-step sample preparation reagent with a rapid isothermal molecular test that detects a knock down resistance (kdr) mutation (F1534C) that enables pyrethroid resistance in Aedes aegypti mosquitoes. We trialled the rapid F1534C pyrethroid resistance test using insecticide resistant Ae. aegypti mosquito bodies and compared results to a conventional, allele-specific quantitative PCR (AS-qPCR) coupled with melt curve genotyping in corresponding mosquito heads. From a strain of Ae. aegypti established from an insecticide resistant population in Merida, Mexico (n = 27), all the mosquito bodies (n = 27) tested positive with the rapid F1534C test regardless of whether they were homozygous or heterozygous. To assess diagnostic test specificity, we confirmed that F1534 was not detected in laboratory-reared, fully susceptible Ae. aegypti mosquito bodies (n = 28) using the rapid F1534C test or the conventional AS-qPCR melt curve analysis. All corresponding mosquito heads (n = 28) were homozygous wild-type FF1534. The rapid F1534C test thus demonstrated 100% diagnostic sensitivity (95% CI: 87.23% to 100%) and 100% diagnostic specificity (95% CI: 87.66% to 100.00%) for detection of the F1534C pyrethroid resistant single nucleotide polymorphism (SNP) in both heterozygous and homozygous Ae. aegypti. In the collection of mutant mosquitoes from Mexico, CC1534 homozygous mutants occurred at a frequency of 74.1% (n = 20) and FC heterozygous mutants at a frequency of 25.9% (n = 7). The rapid F1534C test significantly reduced the sample processing and testing time from approximately 6 h for the AS-qPCR melt curve analysis to only 25 min. These results demonstrate significant potential for our approach to resistance testing as a field-based, low-resource, rapid alternative to time-consuming and expensive laboratory-based detection.
... After the RPA (DNA plasmid control) or RT-RPA (RNA transcripts and mosquitoes) incubation, 2 µL of the amplified RPA reaction mix was added to the sample pad of the lateral flow strip (Milenia Biotec, Giessen, Germany), which had been pre-activated by the addition of 8 µL 0.4% casein in PBST to the sample pad 36 . Strips were then placed in 100 µL running buffer (100 mM H 3 BO 3 , 100 mM Na 2 B 4 O 7 , 1% bovine serum albumin, 0.05% Tween 20, pH 8.8) 37 for five minutes at room temperature, analyzed visually and photographed using a digital camera (MultiDoc-ItTM Digital Imaging System: Upland, CA, USA) or scanned using the Epson Perfection V39 Flatbed Scanner (Epson, New South Wales, Australia). On visual analysis, a single control line depicted the absence of DENV and the appearance of two lines i.e., a test line along with the control line indicated the presence of DENV. ...
Article
Full-text available
The pantropic emergence of severe dengue disease can partly be attributed to the co-circulation of different dengue viruses (DENVs) in the same geographical location. Effective monitoring for circulation of each of the four DENVs is critical to inform disease mitigation strategies. In low resource settings, this can be effectively achieved by utilizing inexpensive, rapid, sensitive and specific assays to detect viruses in mosquito populations. In this study, we developed four rapid DENV tests with direct applicability for low-resource virus surveillance in mosquitoes. The test protocols utilize a novel sample preparation step, a single-temperature isothermal amplification, and a simple lateral flow detection. Analytical sensitivity testing demonstrated tests could detect down to 1,000 copies/µL of virus-specific DENV RNA, and analytical specificity testing indicated tests were highly specific for their respective virus, and did not detect closely related flaviviruses. All four DENV tests showed excellent diagnostic specificity and sensitivity when used for detection of both individually infected mosquitoes and infected mosquitoes in pools of uninfected mosquitoes. With individually infected mosquitoes, the rapid DENV-1, -2 and -3 tests showed 100% diagnostic sensitivity (95% CI = 69% to 100%, n=8 for DENV-1; n=10 for DENV 2,3) and the DENV-4 test showed 92% diagnostic sensitivity (CI: 62% to 100%, n=12) along with 100% diagnostic specificity (CI: 48–100%) for all four tests. Testing infected mosquito pools, the rapid DENV-2, -3 and -4 tests showed 100% diagnostic sensitivity (95% CI = 69% to 100%, n=10) and the DENV-1 test showed 90% diagnostic sensitivity (55.50% to 99.75%, n=10) together with 100% diagnostic specificity (CI: 48–100%). Our tests reduce the operational time required to perform mosquito infection status surveillance testing from > two hours to only 35 minutes, and have potential to improve accessibility of mosquito screening, improving monitoring and control strategies in low-income countries most affected by dengue outbreaks.
Article
Full-text available
Cervical cancer is a leading cause of death for women in low-resource settings despite being preventable through human papillomavirus (HPV) vaccination, early detection, and treatment of precancerous lesions. The World Health Organization recommends high-risk HPV (hrHPV) as the preferred cervical cancer screening strategy, which is difficult to implement in low-resource settings due to high costs, reliance on centralized laboratory infrastructure, and long sample-to-answer times. To help meet the need for rapid, low-cost, and decentralized cervical cancer screening, we developed tailed primer isothermal amplification and lateral flow detection assays for HPV16, HPV18, and HPV45 DNA. We translated these assays into a self-contained cartridge to achieve multiplexed detection of three hrHPV genotypes in a disposable cartridge. The developed test achieves clinically relevant limits of detection of 50–500 copies per reaction with extracted genomic DNA from HPV-positive cells. Finally, we performed sample-to-answer testing with direct lysates of HPV-negative and HPV-positive cell lines and demonstrated consistent detection of HPV16, HPV18, and HPV45 with 5000–50,000 cells/mL in < 35 min. With additional optimization to improve cartridge reliability, incorporation of additional hrHPV types, and validation with clinical samples, the assay could serve as a point-of-care HPV DNA test that improves access to cervical cancer screening in low-resource settings.
Chapter
Full-text available
Paper-based biosensor technology offers an incredible prospective for quick medical diagnosis and which is one of the best point-of-care (POC) biosensors. The main advantage of paper-based technology is that it is user-friendly, low-cost, simply and readily available (Hu et al 2017, Böhm et al 2018, Choi et al 2019). Many challenges are associated with these benefits in order to develop and commercialize it. Large scale manufacturing limitations and low performance are the main challenges connected with commercialization of paper-based biosensors (PBBs). PBBs propose an economic foundation for rapid and trouble-free recognition of a biomarker in quantifiable diagnostics and supplementary areas such as foodstuffs and ecological scrutinization. In view of the fact that paper is a multipurpose substrate that facilitates reflexive solutions currently and is biocompatible, it is appropriate to exploit it as a biochemical sensing platform. In this chapter we will discuss the state-of-the-art of paper-based technology and challenges in its commercialization and also enlighten the existing inclination of PBBs in stipulations of invention and recognition practices in medical diagnostics. Key features and schemes to boost performance and amplify commercialization are projected in this chapter.
Article
Full-text available
Multiplex detection is a smart and an emerging approach in point-of-care testing as it reduces analysis time and testing cost by detecting multiple analytes or biomarkers simultaneously which are crucial for disease detection at an early stage. Application of inexpensive substrate such as paper has immense potential and matter of research interest in the area of point of care testing for multiplexed analysis as it possesses several unique advantages. This study presents the use of paper, strategies adopted to refine the design created on paper and lateral flow strips to enhance the signal, increase the sensitivity and specificity of multiplexed biosensors. An overview of different multiplexed detection studies performed using biological samples has also been reviewed along with the challenges and advantages offered by multiplexed analysis.
Article
Molecular logic gate provides an intelligent option for simultaneous detection of biomarkers. Herein, a dual-mode DNA logic gate was proposed to portably and intelligently detect multiple microRNAs (miRNAs) by gas pressure biosensing and lateral flow assay (LFA). A platinum-coated gold nanoparticle (Au@PtNP) with catalase-like activity was used as a signal reporter to achieve a dual-signal readout. MiRNAs as the input initiated the cyclic strand displacement reaction (SDR) to enrich a large amount of Au@PtNPs. Thus, miRNA can be visually detected by a lateral flow strip (LFS) using the grayish-brown color of Au@PtNPs as output 1. Furthermore, Au@PtNP-catalyzed decomposition of H2O2 resulted in gas pressure as output 2, which was measured by a digital and handheld gas pressure meter. As a consequence, microRNA 21 (miR-21) was sensitively and reliably detected with the limit of detection (LOD) of 7.2 pM. The selectivity and real sample analysis were both satisfactory. Significantly, two-input and three-input AND logic gates were successfully developed to realize multiple detection of two miRNAs and three miRNAs, which provide a promising way for intelligent multi-input analysis. Predictably, with the advantages of portability, simplicity, and affordability, the dual-mode logic gate based on gas pressure biosensing and LFA offers a new perspective on the field of intelligent and portable biosensing and bioanalysis.
Article
Full-text available
The pantropic emergence of severe dengue disease can partly be attributed to the co-circulation of different dengue viruses (DENVs) in the same geographical location. Effective monitoring for circulation of each of the four DENVs is critical to inform disease mitigation strategies. In low resource settings, this can be effectively achieved by utilizing inexpensive, rapid, sensitive and specific assays to detect viruses in mosquito populations. In this study, we developed four rapid DENV tests with direct applicability for low-resource virus surveillance in mosquitoes. The test protocols utilize a novel sample preparation step, a single-temperature isothermal amplification, and a simple lateral flow detection. Analytical sensitivity testing demonstrated tests could detect down to 1,000 copies/µL of virus-specific DENV RNA, and analytical specificity testing indicated tests were highly specific for their respective virus, and did not detect closely related flaviviruses. All four DENV tests showed excellent diagnostic specificity and sensitivity when used for detection of both individually infected mosquitoes and infected mosquitoes in pools of uninfected mosquitoes. With individually infected mosquitoes, the rapid DENV-1, -2 and -3 tests showed 100% diagnostic sensitivity (95% CI = 69% to 100%, n=8 for DENV-1; n=10 for DENV 2,3) and the DENV-4 test showed 92% diagnostic sensitivity (CI: 62% to 100%, n=12) along with 100% diagnostic specificity (CI: 48–100%) for all four tests. Testing infected mosquito pools, the rapid DENV-2, -3 and -4 tests showed 100% diagnostic sensitivity (95% CI = 69% to 100%, n=10) and the DENV-1 test showed 90% diagnostic sensitivity (55.50% to 99.75%, n=10) together with 100% diagnostic specificity (CI: 48–100%). Our tests reduce the operational time required to perform mosquito infection status surveillance testing from > two hours to only 35 minutes, and have potential to improve accessibility of mosquito screening, improving monitoring and control strategies in low-income countries most affected by dengue outbreaks.
Article
Paper-fluidic devices are a popular platform for point-of-care diagnostics due to their low cost, ease of use, and equipment-free detection of target molecules. They are limited, however, by their lack of sensitivity and inability to incorporate more complex processes, such as nucleic acid amplification or enzymatic signal enhancement. To address these limitations, various valves have previously been implemented in paper-fluidic devices to control fluid obstruction and release. However, incorporation of valves into new devices is a highly iterative, time-intensive process due to limited experimental data describing the microscale flow that drives the biophysical reactions in the assay. In this paper, we tested and modeled different geometries of thermally actuated valves to investigate how they can be more easily implemented in an LFIA with precise control of actuation time, flow rate, and flow pattern. We demonstrate that bulk flow measurements alone cannot estimate the highly variable microscale properties and effects on LFIA signal development. To further quantify the microfluidic properties of paper-fluidic devices, micro-particle image velocimetry was used to quantify fluorescent nanoparticle flow through the membranes and demonstrated divergent properties from bulk flow that may explain additional variability in LFIA signal generation. Altogether, we demonstrate that a more robust characterization of paper-fluidic devices can permit fine-tuning of parameters for precise automation of multi-step assays and inform analytical models for more efficient design.
Article
Full-text available
Inkjet printing is emerging at the forefront of biosensor fabrication technologies. Parallel advances in both ink chemistry and printers have led to a biosensor manufacturing approach that is simple, rapid, flexible, high resolution, low cost, efficient for mass production, and extends the capabilities of devices beyond other manufacturing technologies. Here we review for the first time the factors behind successful inkjet biosensor fabrication, including printers, inks, patterning methods, and matrix types. We discuss technical considerations that are important when moving beyond theoretical knowledge to practical implementation. We also highlight significant advances in biosensor functionality that have been realised through inkjet printing. Finally, we consider future possibilities for biosensors enabled by this novel combination of chemistry and technology.
Article
Full-text available
In this paper the development of microfluidic paper-based analytical devices (µPADs) is described for the rapid, on-site detection of improvised explosives. Five lane µPADs were designed and printed using wax ink on chromatography paper to create hydrophobic channels. Each channel contains colorimetric reagents capable of reacting with one or more explosive compounds resulting in a specific colorimetric reaction. Two devices were prepared, each capable of performing five simultaneous analyses on a single µPAD. The first µPAD was developed to detect inorganic explosives such as black powder, flash powder, and ammonium nitrate. It detects nitrates, nitrites, chlorates, and perchlorate oxidizers, as well as ammonium. The second µPAD was developed to detect military explosives such as TNT and RDX along with other high explosives like urea nitrate. It also detects organic peroxides such as TATP and hydrogen peroxide. All experiments were performed by dissolving the explosives in deionized water or 50:50 acetone/ H2O as transport solvents with a detection time of around 5 minutes. Detection limits ranged from 0.39 – 19.8 µg of explosive compound. These two customized µPAD devices permit the on-site forensic testing of unknown explosives, thereby supplying law enforcement and military personnel with a resource for fast, easy detection of military, commercial, and homemade explosive components at low cost.
Article
Full-text available
Rapid, sensitive, on-site detection of bacteria without a need for sophisticated equipment or skilled personnel is extremely important in clinical settings and rapid response scenarios, as well as in resource-limited settings. Here, we report a novel approach for selective and ultra-sensitive multiplexed detection of Escherichia coli (non-pathogenic or pathogenic) using a lab-on-paper test strip (bioactive paper) based on intracellular enzyme (β-galactosidase (B-GAL) or β-glucuronidase (GUS)) activity. The test strip is composed of a paper support (0.5 × 8 cm), onto which either 5-bromo-4-chloro-3-indolyl-β-D: -glucuronide sodium salt (XG), chlorophenol red β-galactopyranoside (CPRG) or both and FeCl(3) were entrapped using sol-gel-derived silica inks in different zones via an ink-jet printing technique. The sample was lysed and assayed via lateral flow through the FeCl(3) zone to the substrate area to initiate rapid enzyme hydrolysis of the substrate, causing a change from colorless-to-blue (XG hydrolyzed by GUS, indication of nonpathogenic E. coli) and/or yellow to red-magenta (CPRG hydrolyzed by B-GAL, indication of total coliforms). Using immunomagnetic nanoparticles for selective preconcentration, the limit of detection was ~5 colony-forming units (cfu) per milliliter for E. coli O157:H7 and ~20 cfu/mL for E. coli BL21, within 30 min without cell culturing. Thus, these paper test strips could be suitable for detection of viable total coliforms and pathogens in bathing water samples. Moreover, inclusion of a culturing step allows detection of less than 1 cfu in 100 mL within 8 h, making the paper tests strips relevant for detection of multiple pathogens and total coliform bacteria in beverage and food samples.
Article
Full-text available
A response in writting: A low-cost bioactive paper device is designed to perform ABO and RhD blood typing tests, and the paper reports the results in writing. This idea was inspired by the vision of the British author, J. K. Rowling, through her novel "Harry Potter and the Chamber of Secrets" in which a piece of paper could be interrogated for information and unambiguous answers were received from the paper in writing.
Article
Isothermal molecular diagnostics are bridging the technology gap between traditional diagnostics and polymerase chain reaction-based methods. These new techniques enable timely and accurate testing, especially in settings where there is a lack of infrastructure to support polymerase chain reaction facilities. Despite this, there is a significant lack of uptake of these technologies in developing countries where they are highly needed. Among these novel isothermal technologies, recombinase polymerase amplification (RPA) holds particular potential for use in developing countries. This rapid nucleic acid amplification approach is fast, highly sensitive and specific, and amenable to countries with a high burden of infectious diseases. Implementation of RPA technology in developing countries is critically required to assess limitations and potentials of the diagnosis of infectious disease, and may help identify impediments that prevent adoption of new molecular technologies in low resource- and low skill settings. This review focuses on approaching diagnosis of infectious disease with RPA.
Article
We report the use of bioactive paper for typing of secondary human blood groups. Our recent work on using bioactive paper for human blood typing has led to the discovery of a new method for identifying haemagglutination of red blood cells. The primary human blood groups, i.e., ABO and RhD groups, have been successfully typed with this method. Clinically, however, many secondary blood groups can also cause fatal blood transfusion accidents, despite the fact that the haemagglutination reactions of secondary blood groups are generally weaker than those of the primary blood groups. We describe the design of a user-friendly sensor for rapid typing of secondary blood groups using bioactive paper. We also present mechanistic insights into interactions between secondary blood group antibodies and red blood cells obtained using confocal microscopy. Haemagglutination patterns under different conditions are revealed for optimization of the assay conditions.
Article
Penetration of Liquids into Cylindrical Capillaries.—The rate of penetration into a small capillary of radius r is shown to be: dldt=P(r2+4εr)8ηl, where P is the driving pressure, ε the coefficient of slip and η the viscosity. By integrating this expression, the distance penetrated by a liquid flowing under capillary pressure alone into a horizontal capillary or one with small internal surface is found to be the square root of (γrt·cosθ2η), where γ is the surface tension and θ the angle of contact. The quantity (γcosθ2η) is called the coefficient of penetrance or the penetrativity of the liquid.
Article
The first example of strip logic gates ("OR" and "AND" functions) for proteins and small molecules has been constructed on the basis of target-induced self-assembly of split aptamer fragments. Using thrombin and ATP as inputs, the corresponding split/integrated aptamers as molecular recognition elements, and gold nanoparticles as a tracer, the output signals can be directly visualized by observing the red bands on the test zones of the strips. The assay is simple, easy to perform, and cost-effective, allowing portable analysis at ambient temperature. The strip logic system is resistant to nonspecific interfering agents and can operate effectively even in human serum samples. Such logic strips hold great promise for application in intelligent point-of-care and in-field diagnostics.
Article
Detection of nucleic acids of Rift Valley fever virus (RVFV) has been shown to be useful in field diagnostics. To develop an isothermal 'recombinase polymerase amplification (RPA)' assay on an ESEquant tubescanner device. RPA was adapted for RNA amplification by first developing a two-step and then a one-step-RT-RPA protocol. Several RT enzymes were tested and the best sensitivity was achieved using Transcriptor (Roche). Finally an RT-RPA pellet containing a recombinant MuLV was tested in RVFV one-step-RT-RPA. The one-step-RT-RPA assay showed a sensitivity of 19 molecules detected as determined by probit analysis of eight runs using a RVFV S-segment based quantitative RNA standard and detected 20 different RVFV strains. The assays showed no cross detection of the human genome and several agents of a typical biothreat panel. It performed almost as good as the assay using glycerol buffer based Transcriptor albeit at a cost of 1-log(10) step in sensitivity. The presented combination of one-step-RT-RPA and portable fluorescence reading device could be a useful tool for field or point of care diagnostics.