Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 1.
Published in final edited form as: Trends Analyt Chem. 2019 Dec 23;124:115782. doi: 10.1016/j.trac.2019.115782

Translating in vitro diagnostics from centralized laboratories to point-of-care locations using commercially-available handheld meters

JingJing Zhang a, Tian Lan b, Yi Lu c,*
PMCID: PMC7081941  NIHMSID: NIHMS1550125  PMID: 32194293

Abstract

There is a growing demand for high-performance point-of-care (POC) diagnostic technologies where in vitro diagnostics (IVD) is fundamental for prevention, identification, and treatment of many diseases. Over the past decade, a shift of IVDs from the centralized laboratories to POC settings is emerging. In this review, we summarize recent progress in translating IVDs from centralized labs to POC settings using commercially available handheld meters. After introducing typical workflows for IVDs and highlight innovative technologies in this area, we discuss advantages of using commercially available handheld meters for translating IVDs from centralized labs to POC settings. We then provide comprehensive coverage of different signal transduction strategies to repurpose the commercially-available handheld meters, including personal glucose meter, pH meter, thermometer and pressure meter, for detecting a wide range of targets by integrating biochemical assays with the meters for POC testing. Finally, we identify remaining challenges and offer future outlook in this area.

Keywords: In vitro diagnostics, Point-of-Care testing, Handheld meters, Signal transduction strategies, Personal glucose meter

Graphical Abstract

graphic file with name nihms-1550125-f0001.jpg

1. Introduction: Current trends in in vitro diagnostics: from centralized labs to POC setting

The general public has a growing demand for high-performance point-of-care (POC) diagnostic technologies where in vitro diagnostics (IVD) is fundamental for prevention, identification, and treatment of chronical, infectious and terminal diseases, in addition to maintaining healthy lives [1]. For example, qualitative or quantitative detections of clinically relevant biomarkers, such as small molecules, proteins, and nucleic acids, is of great importance to improve the diagnosis as well as to provide timely and effective treatment for heart disease [2], bacterial infection [3] or cancers in advanced stages [4]. Among the detection methods, biosensors, which is typically consists of a biorecognition element to bind its target and a signal transduction element to convert the binding event into a physiochemical detectable signal, play a dominant role, because of high affinity and selectivity of biorecognition elements, such as antibody-antigen, DNA/RNA hybridization, enzyme-substrate, and host-guest interactions for a wide range of biomarkers and other targets [5]. In addition, recent advances in biomedical research, nanotechnology, and engineering, have contributed to various signal transduction methods, including the use of electrochemical [6], fluorescent [7], colorimetric [8] or magnetic signal [9], as well as those methods using glucose [10], temperature [11], pH [12], time [13], distance [14], or pressure [15] as readout. These emerging techniques have continuously propelled the shift of IVDs from the traditional centralized laboratories that generally require expensive instruments, sophisticated sampling steps, and highly skills technicians, to POCTs where portable devices can be used by users with minimal or no training to carry out IVDs in resource limited settings, including remote areas and home [16, 17]. These POC tests (POCTs) are becoming increasingly important to meet the demand of personalized medicine [18].

The IVDs around the world are still dominated by analyses carried out in centralized labs, in which many automated analytical processes are emerging to enable the simultaneously analysis of various samples. While these central clinical labs can provide sensitive and selective diagnostic assays, such as high-throughput immunoassays, quantitative polymerase chain reaction (q-PCR (or RT-PCR)), flow cytometry, and mass spectrometry tests, they are often time-consuming, labor intensive and require sophisticated instruments and trained operators. In some cases, these limitations usually result in long turnaround time that not only delay the diagnosis but also prevent the effective monitoring of postoperative recovery. To overcome these limitations, many POCTs have been developed to improve patient care in many aspects of IVDs, such as capability of rapid detection of infection, personalized disease treatment, and self-management of chronic diseases and therapies beyond centralized lab and in many decentralized locations [19], such as the doctor’s office, or a remote clinical lab. While many of these cheaper and smaller POC tests provide a faster turnaround, many challenges remain before their delivery to end patient users for home testing. From the patient’s perspective, the increased desire for self-health management drives the need for the development of user-friendly, and reliable POC devices for telemedicine.

In this review we summarize recent progress in translating IVDs from centralized labs to POC settings using commercially available handheld meters. We first briefly introduce the typical workflow for IVDs and highlight several new innovative technologies in this topic. Next, we discuss advantages of using commercially available handheld meters for translating IVDs from centralized labs to POC settings. We then provide a comprehensive coverage of different signal transduction strategies to repurpose the commercially-available handheld meters, including personal glucose meter (PGM), pH meter, thermometer and pressure meter, for the detection of a wide range of targets by integrating biochemical assays with these commercially-available meters for POC testing. Finally, we offer our assessment of the future outlook and remaining challenges in POCTs.

2. General workflow for IVDs

From operation point of view, the IVDs of biological samples generally consists of four basic steps (Figure 1): (a) sample preparation including extraction, separation, and purification from raw biological samples such as blood, urine, or saliva; (b) target recognition via different interactions with tailored specificity; (c) transduction of recognition events into a readable qualitative or quantitative signal; (d) signal amplification of the binding events to increase detection sensitivity. To allow the extensive use of IVDs in medical applications, considerable efforts have been made to explore innovative solutions to improve the outcomes of these steps.

Figure 1.

Figure 1.

Schematic illustration of typical steps and strategies of in vitro diagnostics of biological samples.

2.1. Sample preparation

In IVDs, the sample preparation is an initial step to obtain analytical targets suitable for later steps in IVDs [20]. This step is extremely important for IVD of some biomarkers with low abundance in complex matrix. Based on the diversity of samples and targets, a variety of strategies for sample preparation have been developed, and extensively reviewed in the past few years [2023]. Taking nucleic acid targets in blood samples as an example, traditional laboratory methods include physiochemical lysis, centrifugation, and solid phase extraction and isolation [20]. Despite progress, few of these methods are available for IVDs in the POC setting, mainly due to the multiple centrifuge and wash steps. To address this issue, microfluidics-based technology has been introduced that can integrate multiple sampling processes into one miniaturized unit [24, 25]. Some lab-on-a-chip (LOC) systems or lab-in-a-drop (LID) systems have also been developed [24], and demonstrated great potentials in IVDs. For example, Bandodkar et al. [26] developed a battery-free, wireless electronic sensing platform that integrates chronometric microfluidic chips with embedded colorimetric sensors, which could simultaneously monitor multiple targets, such as sweat rate/loss, lactate, pH, chloride, and glucose. Microfluidic paper-based analytical devices (μPADs) have emerged in recent years as a promising candidate to address the growing need for integrated sampling process [27]. Using this approach, Govindarajan et al. [28] reported a microfluidic origami paper for POC extraction of bacterial DNA from raw viscous samples. The core idea of this μPADs-based sample preparation device is sequencing complex chemical and physical processing steps via sequential folding of a flat substrate pieces. This method offers the possibility of POC, room temperature, DNA extraction, followed by storage of the extraction device at room temperature for delivery to more centralized diagnostic facilities. In addition to these approaches, readers interested in the general topic of sample preparation can refer to recent relevant reviews [2023].

2.2. Target recognition

Once a biological sample has been prepared for an IVD test, it is analyzed by various techniques, including direct chemical or physical characterization and assays depending on affinity agents. Direct chemical or physical characterization involves instrumentations, including chromatographic equipment (e.g., HPLC and GC) and spectrometers. Various IVD assays will require affinity agents that specifically recognize the target; and today, target recognition in IVD assays most commonly includes antibody, enzymes, nucleic acid molecules and synthetic organic molecules. Among IVD assays, antibody-based immunoassays are the most common due to the wide range of targets being recognized with high selectivity and the presence of standardized assay formats, such as ELISA, latex agglutination and fluorescent polarization assays. Besides antibody, a large number of enzymes are also used directly as recognition molecules in IVD tests for metabolites and metal ion testing, such as glucose, lactate, K+ and Na+. These enzymes generate products and derivatives that can be quantified easily in the presence of the targets with high selectivity. Finally, as a result of recent advances in molecular diagnostics, nucleic acid probes (DNA or RNA) have been widely used to pinpoint the origins of various diseases and precise identification of infectious diseases through their highly specific nucleic acid hybridization. Although natural derived recognition molecules are widely used in IVD tests inside clinical laboratories, several limitations have limited their utility in the POC setting. For example, many antibodies and enzymes are generally considered to be less stable if they are not stored properly [29]; additionally, proteins purified from organisms often have large batch-to-batch variation, hence requiring careful calibrations, and limiting their use under POC settings.

Comparing to established affinity agents mentioned above, a range of new affinity agents that possess certain advantages have been discovered, isolated and tested for IVD applications as well as addressing some of the shortcomings of traditional antibodies and enzymes. For example, functional nucleic acids including aptamers and nucleic enzymes have been widely developed and tested as a substitute for antibodies and enzymes. they are widely regarded as more stable and economical replacement for antibodies as well as protein enzymes with a longer shelf-lives while possess the similar ability to recognize targets with high selectivity and activity. Additionally, since functional nucleic acids can be chemically synthesized, their batch-to-batch variation can be controlled more precisely. As a result, they have been used in numerous systems to develop IVD tests [3035].

2.3. Signal transduction

Common signals to transduce target recognition include colorimetric, fluorescent, and electrochemical readouts. From the perspective of POCT, one of the main advantages of colorimetric modality is that it can potentially provide the detection results using the naked eye or little instrumentation [30]. For instance, lateral flow assays with colorimetric readouts are one example and have catalyzed the emergence of many commercialized products [30], such as pregnancy tests. This type of diagnostic modality is very attractive due to its cost-effective, easy-to-use, and rapid sample-to-answer, however, in most cases, it cannot provide quantitative information that is often indicative of disease severity. Additionally, fluorescent modality based on fluorescent labels, can provide more quantitative information [36], but often relies on dedicated instruments and is affected by fluorophore concentration, lamp intensity, and detector gain, making it difficult to calibrate, maintain and use [37]. One alternative approach is to utilize fluorescence polarization (FP) in fluorescence assays [37], because it is strongly sensitive to the rate of rotation of the fluorescent tag that depends on the target-induced structural or conformational change. More detailed information related to fluorescence polarization/anisotropy signaling mechanisms can be found in a recent review [38]. Another common modality is based on electrochemical techniques [39], which is being researched in many aspects of POCT because of its superiority for miniaturization, portability, and cost-effective. In addition, with the advance of nanotechnology and electrical engineering, electrochemical modality can offer ultrasensitive detection in native biological samples. As an example, Hajian et al. [40] developed a graphene-based field-effect transistor that uses clustered regularly interspaced short palindromic repeats (CRISPR) technology to enable the digital detection of a target sequence within intact genomic material. In this method, the sensor can detect the presence and concentrations of genomic DNA containing the target gene within 15 min by a simple handheld reader, with a sensitivity of 1.7 fM and without the need for amplification. Overall, each of the above-mentioned modalities has its own pros and cons, and further optimization is needed to reach the ASSURED criteria for effective POCTs.

2.4. Signal amplification

Signal amplification is another key element for IVDs, because the majority of the biomarkers are in low abundance in biological samples. One of the most common strategies is enzyme-catalyzed signal amplification using enzyme conjugates [39], which possesses several advantages such as high substrate specificity and rapid catalysis to produce electrochemical or optical signals. Enzyme-linked immunosorbent assay (ELISA) is one such example that has been widely used in clinical IVDs. Signal amplification through the integration of nanomaterials is another important tool used in various molecular assays [41]. In this approach, due to their high surface-to-volume ratio and size-dependent optical or electrochemical properties, the major function of nanomaterials is to serve as nanocarriers to immobilize numerous bioreceptors (e.g. antibodies and DNAs) to provide more binding sites for analytes, or integrate with numbers of optical/electrochemical signaling labels to produce enhanced signals. For instance, by integration of enzyme- and nanomaterials-based signal amplification, Ren et al. developed a dual-functional HRP-SiO2@FFLuc NPs for in situ sequential detection and imaging of ATP and H2O2 in living mice [42]. Later, the same group reported fabrication of SiO2 nanourchins through assembly of two-pronged pair of primers on mesoporous SiO2 cores functionalized with Klenow DNA polymerase and DNA helicase [43]. Due to their high loading capacity, sufficient delivery of nucleic acid reagents into cells, efficient activity without any spatial mutual interference, and sustainable reactivity, the in situ amplified imaging of intracellular mRNAs in living cells was realized. Another new strategy of intracellular amplification-based dual-diagnosis and gene therapy were recently achieved through biodegradable nanosyringes (NSs), which were prepared by both covalent binding and electrostatic self-assembly of biomolecules onto free-standing silicon nanoneedles [44]. Recently, with the advance of DNA nanotechnology, DNA-based signal amplification strategies are emerging, and have demonstrated promising potentials for amplified biosensing for a broad spectrum of targets [45]. Some representative approaches include hybridization chain reaction (HCR), rolling circle amplification (RCA), nuclease-assisted target recycling, catalytic hairpin assembly (CHA) recycling, DNA nanomachines, and very recently CRISPR-based amplification. As a result of the discovery of collateral cleavage activity of several Cas proteins, a brand-new field is emerging and has revolutionized the IVDs for infectious diseases, with ultra-high sensitivity and selectivity. As an example, Chen et al. developed a method termed DNA endonuclease-targeted CRISPR trans reporter (DETECTR) by integrating Cas12a ssDNAse activation with isothermal amplification, which achieves attomolar sensitivity for DNA detection. DETECTR could enable rapid and specific detection of human papillomavirus in patient samples, thereby providing a simple platform for IVDs. Overall, the above-mentioned signal amplification strategies can also be combined together to produce more complex system to expand the scope of its application.

2.5. Multiplexing capability

Performing IVDs for multiple targets simultaneously from one single specimen is often clinically relevant, because measuring panels of biomarkers is expected to obtain more accurate diagnostic results than single biomarker [46]. To achieve a higher grade of multiplexing for IVDs, different methods have been developed [4749]: (1) employing separate POCT devices or regional separating analytes into multiple detection zones by microfluidic chips, droplets, or arrays; (2) the use of different labels on different analytes, such as fluorescent dyes, enzymes, or nanomaterials; (3) the combination of these two strategies. As an example, Gootenberg et al. [49] reported a multiplexed and portable nucleic acid detection platform by screening out four distinguished Cas effectors with cutting preferences to different nucleotide sequences. By applying Cas13, Cas12a, and Csm6 in the CRISPR-based sensors, they further extended the detection to four targets by leveraging orthogonal dinucleotide motifs, with reporters for LwaCas13a, PsmCas13b, CcaCas13b, and AsCas12a in FAM, TEX, Cy5, and HEX channels, respectively. Despite the success, new strategies for multiplex IVDs are still limited, and thus further implementation with other emerging technologies is desirable. More importantly, although striving for multiplex diagnostics is important, it should be noted that a simple, sensitive, selective, fast detection system is far more important than a fancy but complex system for practical applications [47].

3. Advantages of using commercially available handheld meters for POCTs

Leading the list of the above newly developed IVDs technologies are those for point-of-care testing, which has been widely applied in many fields, such as personal healthcare monitoring in a hospital, clinic, or at home, on-site environmental assessment, and food safety analysis [50]. These tests generally consist of only elementary manual to use with little or no sample pre-treatment, and provide rapid response to small sample volumes [4]. Additionally, the commercially-available POCT readers range from transportable instruments, benchtop analyzers, to handheld devices or even portable pocket-size meters. To date, POC diagnostics have been comprehensively reviewed in the past decade [5, 5153], from the development of new POC assays for diverse targets to the advancement in integration and miniaturization of POC devices.

The key criteria for such POC devices for IVDs are Affordable, Sensitive, Specific, User-friendly, Rapid and robust, Equipment-free and Deliverable to end-users (ASSURED) includes [16, 17]: (1) good usability, including simple and easy operation with eliminated or automated built-in maintenance functions; (2) sufficient accuracy, with simple or automated quality assessment; (3) accessibility for mobile healthcare and cloud-based data analysis; (4) multiplexing, that is simultaneous detection of different targets from a single sample. Overall, the emerging needs and demands for novel technologies for IVDs for diverse clinical applications is shifting the future of IVDs from central lab to point-of-care home testing.

Portable devices are growing in importance in applications of IVDs, however, despite many years of research, only a few portable meters are commercially available for non-specialists to make molecular measurements at home or in resource limited settings. A major challenge to develop such devices is the requirement of enormous financial and personnel expenses as well as time during the R&D process, making it difficult to be affordable to the general public. To address this issue, an alternative method is to integrate the well-developed molecular assays with current commercially available handheld meters through new signal transduction approaches. In this case, the developmental costs (e.g. time and financial) for a new POC device can be greatly decreased, while maintaining the advantages from both sides. Additionally, many existing portable devices have been marketed for decades (e.g., glucose meter, pH meter and thermometer), hence widely used and familiar to the general public. Directly make use of existing meters to the maximum extend can reduce the learning curve for any new POC IVD test significantly. Besides, by leveraging on existing platforms, any newly developed features can be easily introduced to the new POC IVD systems, such as wireless data transmission on some of today’s FDA approved smartphone enabled glucose meters. As a result, the adaptation of existing POC devices into a general measurement device via novel chemical or biological methods that can perform a wide range of common clinical tests could significantly increase the odds of bringing new POC IVD tests to reality.

4. Strategies to integrate molecular assays with commercially-available portable meters for POC testing

Some innovations in this field have been demonstrated to be versatile enough to support interchangeable biorecognition elements, ease of operation at a competitive cost, and remote detection of multiple, complex analytes. In the last decade, extensive signaling systems have been developed for quantitative analysis using commercially-available portable meters, such as personal glucose meter, pH meter, thermometer, and pressure meter. The advantages and disadvantages of these signaling systems are summarized in Table 1.

Table 1.

Advantages and disadvantages of various commercially-available portable meters adopted for POCT

Features PGM pH meter Thermometer Pressure meter
Wide availability
Low cost
Easy-to-use
Real-time, continuous monitoring
Small sample volume
Resistance to interference from
 Colored species
 Autofluorescence
 pH
 Glucose in biological matrix
 Temperature

4.1. Personal glucose meter

Personal glucose meter (PGM), firstly introduced around 1970, has become one of the most widely used POC devices that allows affordable detection of blood glucose levels [54]. With more than 40 years development, PGM displays unique features of low cost, portability, ease-of-use and rapid results, which benefits the diabetes around the globe. Despite the success, the common PGM can only detect blood glucose, which limits its application in molecular diagnosis. A major challenge in using a PGM as a general detector for non-glucose targets is to find a general method to link the presence and concentrations of other targets with a detectable glucose signal. From the perspective of the core element of a PGM, it is basically an amperometer that measures turnover of an electron-transfer mediator in the enzymatic oxidation of glucose, typically by glucose oxidase (GOx) or glucose dehydrogenase. Therefore, the detection of other targets is possible by replacing these glucose-responsive enzymes with other enzymes [55], such as lactate-responsive lactate dehydrogenase, cholesterol-responsive cholesterol oxidase and ethanol-responsive alcohol dehydrogenase. However, this method requires the change and modification of the PGM strips as well as the software of the PGM detector, thus limits its applicability in molecular diagnosis. An alternative method to expand the utility of PGMs to non-glucose targets was reported by Mogapatra et al. [56], where glucose was covalently linked to several enzyme substrates and subsequently applied for the detection of corresponding enzyme analytes. In this approach, the glucose can be released upon reaction of the substrate with the target enzyme, providing a rapid single-step assay for quantifying enzyme activities using a PGM. However, to apply this method, sophisticated synthesis of the enzyme substrates that covalently link to glucose is required. In addition, most of commercially-available PGMs are designed to monitor blood glucose, and thereby have a dynamic range of 0.6–33 mM for glucose, which is difficult for the detection of trace amount of biomarkers in blood. Therefore, a signal amplification method is necessary when integrating the molecular assays with a PGM. To meet the above challenges, three main signal transduction methodologies have been developed to repurpose the PGM for molecular diagnosis, including generation of glucose signal by enzymatic and non-enzymatic reactions, and generation of other PGM-detectable signals.

4.1.1. Generation of glucose signal by enzymatic reactions

The most common method to produce a PGM detectable signal is to use an enzyme to convert the PGM-inert substrate to the PGM-detectable glucose signal. For instance, in 2011, Xiang et al. reported breakthrough approaches to transform the selective binding of non-glucose targets with functional DNA-based receptors (e.g. aptamers, and DNAzymes) into a PGM-detectable glucose signal (Figure 2A) [10]. To this aim, invertase, which catalyzes the conversion of PGM-inert sucrose into glucose, was used as the signal transduction element, and conjugated with fDNAs via covalently bond. In the presence of a target, this fDNA-invertase conjugate is released into the solution due to a) the decrease of the melting temperature through the target-induced structure-switching of aptamers, or b) metal ion-dependent cleavage of DNAzyme substrate. In this case, the presence and concentrations of a target is correlated with the released amount of fDNA-invertase conjugate, and thereby is quantitatively related to the glucose generation by invertase-catalyzed reactions. More importantly, taking advantages of high enzymatic activity (kcat = 1540 s−1) of invertase [54], such a design allows the conversion of trace amount of targets into mM level of glucose that can be detected by commercially available PGMs. This concept has been further extended for the detection of many types of targets, including protein biomarkers [5766], DNAs [6776], miRNAs [7779], DNA oxidative damage [80], small organic molecules [65, 69, 8187], metal ions [8891], enzyme activities [9296], biotoxins [57, 97101], pathogens [102, 103], and cancer cells [104].

Figure 2.

Figure 2.

(A) Scheme of the method using a PGM to detect a wide range of targets beyond glucose. Reprinted from Xiang et al., 2011 [10] with permission of Nature Publishing Group. (B) Schematic illustration of detection process using glucose encapsulating liposome (GEL) on test trip device linked with a PGM. Reprinted from Zhao et al., 2017 [127] with permission of Elsevier. (C) Schematic illustration of NADH/PGM system for non-glucose target detection using NADH-dependent enzymes. Reprinted from Zhang et al., 2016 [129] with permission of Wiley. (D) Installing of logic gate on PGMs for the portable, resettable, and quantitative Point-of-Care detection of many targets in clinical care. Reprinted from Zhang et al., 2018 [130] with permission of Wiley.

Glucoamylase, or amyloglucosidase, which catalyzes the hydrolysis of PGM-inert amylose to glucose, is another alternative enzyme used in PGM-based sensors for non-glucose targets, including cocaine [105], ATP [105], hepatitis B surface antigen [106], DNA methyltransferase activity [107], miRNA [108, 109], microbial pathogen [110], thrombin [111], and C-reactive protein [111]. For instance, Yan et al. [105] developed a target-responsive “sweet” DNA hydrogel with entrapped glucoamylase for PGM-based portable and quantitative detection of non-glucose targets. In this approach, the presence of target molecule causes the disassembly of DNA hydrogel to release the entrapped glucoamylase, which subsequently catalyzes the hydrolysis of amylose to generate large amount of glucose for sensitive detection of the model target, cocaine and ATP.

Alkaline phosphatase (ALP), which catalyzes the hydrolysis of PGM-inert glucose-1-phosphate to glucose, has also been used to generate glucose signals for PGM-based sensors [112, 113]. For instance, Zhang et al. [112] have developed a simple method to link the enzymatic activity of galactose-1-phosphate uridyltransferase to the production of glucose, which allows POC diagnosis of galactosemia in authentic human clinical samples. Given the presence of ALP in numerous enzymatic assays for clinical diagnostics, this method can be adapted to quantify a wide range of enzyme activities using a PGM.

In addition to the above mentioned enzymes, other types of enzymes, such as β-galactosidase [114, 115], hexokinase [116, 117], and microbes-containing enzymes [118, 119], has also been integrated with PGMs for the development of portable sensors. Table 2 summarizes the analytical performance of some representative reports discussed above focusing on enzyme-based PGM sensors.

Table 2.

Analytical performance of some representative enzyme-based PGM sensors.

Enzyme Type Real Samples Analyte Assay Format Amplification method Dynamic range LOD ref
Invertase human serum cocaine sandwich enzymatic 0–500 μM 3.4 μM [10]
adenosine 0–1,000 μM 18 μM
IFN-γ 0–400 nM 2.6 nM
UO22+ 0–200 nM 9.1 nM
human serum streptavidin sandwich enzymatic 0–80 nM 4.0 nM [57]
PSA 0–100 ng/mL 1.5 ng/mL
biotin competitive 0–96 μM 1.5 μM
OTA 0–1000 nM 17 nM
human serum CEA sandwich magnetic beads 0–1.0 ng/mL 0.05 ng/mL [58]
N.A. insulin sandwich enzymatic 0–10 nM 1 nM [59]
HbA1c 0–200 mg/L 3 mg/L
human serum AFP sandwich enzymatic 0.5–50 ng/mL 0.18 ng/mL [60]
human serum PDGF-BB sandwich CXR, CAMB 0.316 fM-1 pM 0.11 fM [63]
human serum VEGF sandwich HCR 3–100 pg/mL 1.2 pg/mL [64]
human serum 8-OHdG competitive AuNPs 0.1–100 ng/mL 0.23 ng/mL [65]
PSA 1–100 ng/mL 1.26 ng/mL
human serum AFP sandwich BRCA 0.1–100 ng/mL 87 pg/mL [66]
N.A. HBV DNA sandwich enzymatic 0–50 nM 40 pM [67]
N.A. HIV DNA sandwich Fe3O4/Au NP 0.5 pM-1 nM 0.5 pM [68]
human serum DNA sandwich ICSDPR 5.0–1000 fM 0.5 pM [71]
cell lysate miRNA-21 competitive HNFs 0.5–100 nM 0.41 nM [78]
milk melamine competitive enzymatic 0–500 μM 0.53 μM [82]
FBS amino acids Label-free enzymatic 0–20 μM 0.1 μM [87]
N.A. Pb2+ competitive enzymatic 0–1000 nM 16 nM [88]
UO22+ 0–1000 nM 5.0 nM
cancer cell telomerase sandwich DNA machine 0.1 fM-1 nM 0.1 fM [94]
drinking water MC-LR sandwich GO-AuNPs 0.60–100.00 ng/mL 0.10 ng/mL [100]
human serum SW620 cell sandwich GO 1,560–100,000/mL 560 cells [103]
Glucoamylase urine cocaine label-free hydrogel 0–750 μM 4.4 μM [105]
ATP 0–1 mM N.A.
human serum DNA MTase sandwich liposome 0.005 to 0.1 U/mL 0.002 U/L [107]
cancer cells miRNA-21 sandwich P-ERCA 0.5–10 fM 0.1 fM [109]
lake water E. coli competitive Fe3O4@SiO2 20–105 bacteria/mL 40 bacteria/mL [110]
human serum thrombin sandwich liposome 0–200 nM 12.8 nM [111]
CRP 0–500 ng/mL 13 ng/mL
ALP human blood GALT label-free enzymatic 0–43.5 μmol h−1 (g Hb)−1 N.A. [112]
Galactosidase potable water E. coli label-free enzymatic 20–2×109 CFU/mL N.A. [115]
Hexokinase tap water, human serum, bovine urine ATP label-free enzymatic 0.05–0.4 μM 49 nM [116]

Abbreviations: IFN-γ = interferon-gamma; PSA = prostate-specific antigen; OTA = ochratoxin A; CEA = carcinoembryonic antigen. HbA1c = glycated hemoglobin; PDGF-BB = platelet-derived growth factor BB; CXR = cation exchange reaction CAMB = catalytic and molecular beacon; VEGF = vascular endothelial growth factor; 8-OHdG = 8-hydroxy-2’-deoxyguanosine; AuNPs = gold nanoparticles; BRCA = Backfilling rolling cycle amplification; HBV = hepatitis B virus; HIV = human immuno-deficiency virus; ICSDPR =isothermal circular strand-displacement polymerization reaction; HNFs = hybrid nanoflowers; FBS = fetal bovine serum; MC-LR = Microcystin-LR; GO-AuNPs = graphene oxide-gold nanoparticles; SW620 = human colorectal cancer cell; DNA MTase = DNA methyltransferase; P-ERCA = padlock exponential rolling circle amplification; ALP = Alkaline phosphatase; GALT = galactose-1-phosphate uridyltransferase; CRP = C-reactive protein; N.A. = Not Applicable; LOD = limit of detection;

4.1.2. Generation of glucose signal by non-enzymatic reactions

Instead of using enzymes to regulate the glucose signal for PGM-based sensing, another elegant method to generate a target-responsive glucose signal is to directly release encapsulated glucose from nanocontainers [120], such as mesoporous silica nanoparticles [121125], liposome [126, 127], and TiO2 nanotubes [128]. Compared with enzymatic generation of glucose signals, such a non-enzymatic controllable release system minimized the interferences from temperature and pH of solution, while maintains the advantages of easy operation and signal amplification. For instance, Fu et al. [122] reported the use of DNAzyme-capped mesoporous silica nanoparticles (MSNs) as glucose nanocontainers, and developed a PGM-based portable sensor for sensitive detection of lead ions. In this system, the presence of target Pb2+ ion catalyzes the cleavage of the DNAzymes, thereby opening the molecular gates and releasing encapsulated glucose from the MSNs. The released glucose molecules can be monitored by an external PGM, which is related to the presence and concentrations of the target Pb2+ ions. Using similar strategy, Gao et al. [123] designed a polystyrene microsphere-gated magnetic mesoporous silica nanocontainer for encapsulation of glucose, and combined with a competitive immunoassay for sensitive detection of brevetoxin B. Similar MSNs-based platform has also been applied for the detection of telomerase activity in cell extract using telomeric wrapping DNA as the gate.

Another non-enzymatic glucose generation system was recently developed by Zhao et al. [126], where antibody-tagged liposome was used as glucose container. The presence of target antigen leads to the capture of antibody-tagged liposome to form a sandwich complex, which can be lysed to release the encapsulated glucose for a PGM-based detection of protein biomarker. To simplify the detection process, the same group further developed an integrated disposable immunochromatographic strip with PGM readout for rapid quantification of phosphorylated proteins (Figure 2B) [127]. Later, TiO2 nanotube arrays were also used as a nanocontainer and integrated with PGMs for portable and sensitive detection of cocaine [128]. Very recently, a 3D paper-based device was integrated with PGMs for highly sensitive and portable detection of silver ion [120]. The silver ion-triggered self-growing of Ag particles in the nanoporous membranes (NMs) of μPADs can partially block the pores of the NM, and as the solution flows through the NM and re-dissolves the glucose powder, rapid and efficient glucose signal generation can be achieved.

4.1.3. Generation of other PGM-detectable signals

When performing a molecular assay in blood or urine samples, the interference of endogenous glucose to PGM signals is problematic for many targets. To overcome this limitation, Zhang et al. [129] developed a new PGM-based signaling strategy based on its dose-dependent response to nicotinamide coenzymes (e.g. NADH) (Figure 2C). Taking advantage of this feature, a series of enzymatic reactions have been applied to link the target substrate to the consumption or production of NADH that could be monitored using a PGM. More importantly, an innovative strategy was developed to overcome the background glucose issue by employing an enzyme hexokinase that can efficiently convert the endogenous glucose to PGM-inert phosphorylated glucose during the PGM assay. As NADH is a functionally important cofactor associated with many clinical tests, this method can be readily expanded for the detection of a broad range of targets using various NADH-dependent enzymes. The invention of NADH-based signaling strategy is a milestone in the development of PGM-based practical sensors, allowing millions of people to use a single portable device to monitor not only their blood glucose levels, but also other relevant biomarkers.

Benefiting from the above glucose and NADH–based signal transduction strategies, a PGM-based biocomputing platform was recently developed by Zhang et al. [130], possessing logic capability for multi-parameter sensing of many clinically relevant targets (Figure 2D). In this approach, a series of metal ions, disease-related metabolites, coenzymes, and native enzymes have been applied as inputs, while glucose and/or NADH signal on a BGM was used as outputs. A complete set of seven binary logic gates (YES, NOT, OR, NOR, NAND, INHIBIT, and Concatenated) were constructed on PGMs for the monitoring of multiple biological substances, showing great promise for POC diagnostics of diseases, such as hyponatremia and hypernatremia. Significantly, this method will not only broaden the clinical applications of the BGM, but also open a new avenue in making PGM a logic-gate responsive POC device for practical applications.

In addition to the above PGM-based signaling strategies, other electroactive species, including paracetamol- or catechol-bearing compounds [131], reduced glutathione (GSH) [132], thiocholine [133], have also been demonstrated to generate a PGM-detectable signals for direct detection of β-galactosidase [131], α-mannosidase [131], influenza viruses [131], Streptococcus pneumoniae [131], and E. coli [131], intracellular glutathione and serum triglyceride levels [132], as well as organophosphorus pesticide [133].

In general, repurposing PGMs as a general measurement device may help in rapid practical translation of traditional diagnostic assays by bypassing costly device development process and sensor prototype validation.

4.2. pH meter

pH meter is another important type of commercialized portable meter, which measures the hydrogen ion activity in solutions. The obtained pH value indicates the degree of the acidity or alkalinity, with the unique features of pocket-size portability, low cost, easy operation and reliable high sensitivity (0.001 unit of pH change). As a result, numerous pH meters have been developed for diverse applications, such as healthcare and clinical assessment, water quality evaluation, and soil analysis. However, most of the commercialized pH meters are still limited to only detect a single “target”, pH. To reach the full potential of pH meters for molecular diagnostic applications, there is a need to find a method that can link the binding of any targets with a change of pH so any pH meter can be used to measure the presence and concentrations of targets of interests. To date, different signal transduction strategies based on pH meter have been developed, and can be categorized into two types, as detailed in the following sections.

4.2.1. Generation of pH change by enzymatic reactions

To generate a pH change of the assay solutions, the most used method is based on the catalytic reaction of bioactive enzymes that can produce the acid or alkaline species, such as glucose oxidase (GOx) [134], urease, and acetylcholinesterase (AchE) [135]. By applying these enzymes as labels and further integration with sandwich or competitive assay format (see section 2.2), the obtained sensors are able to transform the binding of target into a target-dependent pH change that can be detected using a pH meter. For example, Ye et al. [136] proposed a portable biosensor for on-site detection of a food pathogen through the combination of sandwich immunosensor and ubiquitous pH meter (Figure 3A). In this system, ConA-GOx-CaHPO4 organic–inorganic hybrid nanoflowers were first prepared through a modified biomineralization process and applied as a signaling tag for Escherichia coli (E. coli) O157:H7 due to its high affinity to lipopolysaccharides O-antigen of E. coli. The readout signal was generated by the resulting pH decrease through the GOx-catalysis of glucose into gluconic acid after the sandwich immunoreaction. Using similar strategy, a series of pH meter-based portable biosensors have been developed for the detection of clinically relevant protein biomarkers, such as troponin I [135], human oncogenic protein [137, 138], vascular endothelial growth factor (VEGF) [134], carcinoembryonic antigen (CEA) [139], neuron-specific enolase (NSE) [140], and squamous cell carcinoma antigen (SCCA) [141]. In addition to the detection of proteins based on immunoreactions, Xie et al. [142] developed a pH meter based platform for the facile and sensitive detection of genomic DNA by directly measuring released hydrogen ions during the loop mediated isothermal amplification (LAMP) reaction catalyzed by DNA polymerase. Recently, to extend the targets to heavy metal ions, Tang et al. [143] designed a sensitive and portable pH meter based magnetic detection method for lead ion (Pb2+) by integration of Pb2+-specific DNAzyme, GOx with rolling circle amplification.

Figure 3.

Figure 3.

(A) Illustration of the synthetic process for Con A-GOx-CaHPO4 organic-inorganic hybrid nanoflowers and the corresponding scheme for immunoassay of E. coli O157:H7. Reprinted from Ye et al., 2016 [136] with permission of Wiley. (B) Schematic representation for the screening of thrombin inhibitors (lower four sensing areas) and detection of thrombin (upper four sensing areas) using a single eight-zone self-powered, portable, and light-addressable photoelectrochemical sensor. Reprinted from Wang et al., 2018 [147] with permission of ACS. (C) Translating molecular detections into a simple temperature test using a target-responsive smart thermometer. Reprinted from Zhang et al., 2018 [11] with permission of RSC. (D) Principle of the temperature-based immunosensor based on the exothermic reaction between H2O and CaO using a common thermometer as readout. Reprinted from Ma et al., 2019 [151] with permission of ACS.

While the above sensors measure the decrease of the pH value in solution, urease, which catalyzes the hydrolysis of urea into ammonia that raises the pH value [12], has also been applied for the development of portable pH meter-based sensors [144]. For instance, taking advantage of a bacteria-specific RNA-cleaving DNAzyme probe as the molecular recognition element and the urease as a label, Tram et al. [12] translated the detection of bacteria into a pH increase, which can be readily detected using a litmus dye or pH paper. Recently, by replacing the DNAzymes with aptamers [144], antibodies [145], or telomerase specific primer [146], the portable detection of aflatoxin B1 in food samples or telomerase activity in cells have also been achieved.

Based on these studies, we summarize the analytical performance of some representative reports discussed above focusing on enzyme-based pH meter sensors, as shown in Table 3.

Table 3.

Analytical performance of some representative enzyme-based pH meter sensors.

Enzyme Type Real Samples Analyte Assay Format Amplification method Dynamic range LOD ref
GOx human serum VEGF sandwich HCR 0.8–480 pg/mL 0.5 pg/mL [134]
human serum HOP sandwich microparticles 0.620–40 pg/mL 0.57 pg/mL [137]
human serum HOP sandwich microbeads 12.5–200 pg/mL 5.5 pg/mL [138]
human serum CEA sandwich AuHM 0.1–100 ng/mL 0.062 ng/mL [139]
human serum NSE sandwich liposomes 0.01–100 ng/mL 8.9 pg/mL [140]
human serum SCCA sandwich HCR 0.01–10 ng/mL 5.7 pg/mL [141]
drinking water Pb2+ label-free RCA 1.0–100 nM 0.91 nM [143]
AchE human serum troponin I sandwich enzymatic 0–100 ng/mL 10 pg/mL [135]
DNA polymerase silkworm egg genomic DNA label-free LAMP 0.5 pg/μL-50 ng/μL N.A. [142]
Urease Culture medium bacterial sandwich enzymatic 5 – 5 × 105 cells single CFU [12]
corn AFB, label-free hydrogel 0.2–20 μM 0.1 μM [144]
corn AFB, sandwich enzymatic 0.62–23.42 ng/mL 0.2 ng/mL [145]
human serum telomerase sandwich enzymatic 50–10,000 cells 20 HeLa cells. [146]

Abbreviations: GOx = glucose oxidase; VEGF = Vascular endothelial growth factor; HCR = hybridization chain reaction; HOP = human oncogenic protein; CEA = Carcinoembryonic antigen; AuHM = gold hollow microspheres; NSE = neuron-specific enolase; SCCA = squamous cell carcinoma antigen; RCA = rolling circle amplification; AchE = acetylcholinesterase; LAMP = Loop mediated isothermal amplification; N.A. = Not Applicable; LOD = limit of detection; CFU = Colony-Forming Units; AFB1 = aflatoxin B1;

4.2.2. Generation of pH change by non-enzymatic reactions

To measure the hydrogen ions concentration, the pH meter records voltage differences between two electrodes [147]. Based on this mechanism, Wang et al. [147] developed a portable, self-powered, and light-addressable photoelectrochemical (PEC) sensor for high-throughput screening of thrombin inhibitor drugs using a pH meter as signal readout (Figure 3B). The sensor was fabricated by immobilizing biotin-modified thrombin-cleavable peptides on eight separated sensing zones of a single gold film electrode, with a platinum (Pt) wire counter electrode that forms a self-powered PEC sensing platform. The presence of thrombin cleaves the biotin-modified peptides, leading to the decreased binding sites for streptavidin-labeled fullerene (C60) PEC bioprobes. Due to the high conversion efficiency of C60-based PEC bioprobe, the proposed thrombin sensor exhibits a wide detection range from 0.1 pM to 1.0 nM, with detection limit of 0.05 pM using a portable pH meter. In addition, the detection of thrombin and evaluation of inhibitor activity in serum samples have also been demonstrated.

4.3. Thermometer

The thermometer is one of the most commercial-available, easy-to-use and quantitative devices for portable measurements, which plays an important role in environmental monitoring and medical diagnostics. However, a major challenge in using a thermometer for the detection of other targets beyond temperature is to develop a method that can transform the binding of any targets into a temperature change so that the presence and concentration of a target can be detected using a thermometer. To address this issue, conventional calorimetric biosensors were developed [148], which utilize enzymatic reactions to generate heat and correlate it with the concentration of the enzyme substrate. However, these enzymatic reactions usually produce subtle change of temperature so that can only be detected using a highly sensitive and expensive instrument, which limits its application for POCT in resource-limited settings. As a result, the signal amplification is crucial for the successful development of thermometer-based portable sensors. To this aim, several exothermic reactions have been combined with the construction of thermometer-based biosensors in recent years for efficient signal amplification.

4.3.1. Generation of temperature signal by laser-assisted exothermic reaction

Photothermal effect is a common phenomenon associated with electromagnetic radiation, which is produced by the laser photoexcitation of a material, thereby producing thermal energy with an increase of temperature [149]. Using the laser-assisted exothermic reaction, Zhang et al. [11] translated molecular detections into a simple temperature test using a target-responsive smart thermometer (Figure 3C). The sensor system is based on the target-induced release of DNA–phospholipase A2 enzyme conjugate from a functional DNA duplex immobilized on magnetic beads, which subsequent catalyzes the hydrolysis of liposome-indocyanine green to produce a change of temperature signal under the NIR-laser irradiation. Considering the low cost and facile incorporation of the system with suitable functional DNAs to recognize many targets, the system makes the thermometer an affordable and pocket-size meter for the detection and quantification of a wide range of targets.

4.3.2. Generation of temperature signal by non-laser exothermic reaction

In addition to the laser-assisted exothermic reaction, the dissolution of some inorganic oxide in water can also generate a large amount of heat to change the solution temperature. Based on this phenomenon, Gao et al. [150] reported an exothermic chip that could generate a temperature change based on a target-responsive aptamer-modified hydrogel. The presence of an analyte could increase the capillary flow rate for the acceleration of NaOH dissolution in water, resulting in a quick increase of temperature that can be measured using a forehead thermometer. Using this method, heavy metal ions (Hg2+ and Pb2+) in different real samples are quantitatively analyzed. Recently, Ma et al. [151] further designed a temperature-based immunosensor (Figure 3D), which transduces the antibody–antigen recognition event into the temperature change with a commercialized thermometer as signal readout. By taking advantages of high catalysis efficiency of nanozyme and self-heating reaction between CaO and H2O, the quantitative detection of CEA can be realized without the assistant of any expensive instruments or laser sources other than a simple thermometer.

4.4. Pressure Meter

Pressure meter is also a common portable device for measuring pressure generated by gases or liquids during many chemical or biochemical reactions. With the advantages of high sensitivity and portability, pressure meter is emerging as a promising candidate for POC diagnostics. Yang’s group pioneered the development of pressure-based biosensors by translating the molecular recognition signal into a measurable pressure signal in a sealed device through a gas-generation reaction. For example, Zhu et al. designed pressure-based bioassays by integrating a sandwich immunoassay with a common gas-generation reaction catalyzed by a catalase or Pt nanoparticles [152]. The presence of a target results in the formation of the sandwich immunocomplex that triggers the decomposition of substrate H2O2 to H2O and O2, leading to significant pressure increase in the reaction chamber that can be measured using a pressure meter. Taking advantages of the high liquid to gas volume expansion and the excellent catalytic properties of catalase/PtNP, this pressure-based method was able to provide more than 1010 folds of signal enhancement with a few minutes of reaction. Taking advantage of this feature, Yang and other groups has developed a series of pressure meter-based biosensors for the detection of diverse targets, including PSA, Influenza A/H5N1 virus (H5N1) [152], C-reactive protein (CRP) [153], cocaine, Pb2+, OTA [154], cancer cells [155, 156], telomerase activity [157], myoglobin [158, 159], miR-21 [160], Hg2+ [161], and pathogenic bacteria [162].

4.5. Other meters

In addition to the above mentioned portable sensors that utilize glucose, pH or temperature as the signal readout, some important metabolites, such as ATP [163, 164], uric acid [165], have also been applied as the signal readout that can be quantified using a commercial-available portable meter. For instance, Chen et al. [164] integrated the double-enzymes-mediated bioluminescent reaction, conjugated nanoparticles, and portable ATP detector to construct a rapid, highly sensitive, and quantitative POCT assay for proteins. The proposed sensor possesses the features of high sensitivity (femtomolar level of biomarkers), fast sample-in-answer-out (< 1h), and convenient operation (portable ATP detector), which broadens the range of portable sensors in molecular diagnosis.

5. Conclusion and perspectives

In summary, we have witnessed a rapid growth in developing IVDs in the past decade. At the fundamental level, we briefly discussed the typical workflow for IVDs from sample preparation to target recognition, signal transduction, signal amplification and multiplex capability, and highlighted several new innovative technologies in each element of the workflow. Owing to the demand for personalized medicine, IVDs is shifting from centralized lab to POCT. POC IVDs developed for resource-limited settings is becoming widespread acceptance for doctor’s office, clinic, or home use, and these POCTs will play an important role in reducing medical costs for everyone. From the perspective of product development, the adaptation of existing POCT devices for detection and management of new targets can significantly increase the odds of bringing new POC tests to reality. To date, various commercially-available portable meters have been repurposed to broad their utility in POC IVDs through the integration of many innovative signal transduction strategies.

Despite the significant progress, several issues related to current and future practical applications in POC IVDs still need to be addressed. The first issue is reagents storage and release in POC devices. To integrate molecular assays with existing POC devices for home monitoring, it is necessary to have all necessary sensor reagents pre-stored in testing strips or cartridges of POC devices. Some microfluidic chips have been developed for reagents storage, which could to some extent eliminate the manual reagents introduction, simplify the detection process, and minimize the contamination risk, making POC devices much friendly to end-users. Deng et al. [166] recently provided an excellent and timely overview of the advances in reagents storage and release in self-contained POC devices. However, the reagents involved in POC testing vary widely, and thus each of the storage and release strategy requires further optimization for new conditions. Future advances in microfluidic technologies and engineering strategies, as well as the creation of new stable reagents, and innovation in nanotechnology, will provide more opportunities tackle the limitations in this aspect.

Another consideration from a POC perspective for home use is user competency, which is one of potential source of error in POC tests. Compared with molecular testing in centralized lab, POC testing by patients may have higher error rates due to the lack of training during sample collection and analysis. It is therefore important to develop more automatic platforms for POC diagnostics, because a high level of automation in sampling process will facilitate the generation of accurate results for POC tests at home. Current existing portable meters, such as PGMs, have significantly improved analytical performance, and have been engineered to eliminate many of the potential error-generating steps in the testing process.

Finally, the connectivity is another important aspect in current and future POC diagnostics, which is vital for effective management of testing results and quality assurance, particularly in the new era of telemedicine. With the advance of mobile technology, mobile POC devices are emerging to broaden the current POC diagnostic landscape. A broad collaboration of experts in chemistry, engineering, and computer science is desired to push the community toward the development of more effective POC devices.

Highlights.

  • Typical workflow for in vitro diagnostics and innovative technologies to improve the workflow are summarized.

  • Advantages of using commercially available handheld meters for POCT are highlighted.

  • Comprehensive coverage of signal transduction strategies to repurpose the commercially-available handheld meters for POCT are presented.

  • Future challenges and prospective of the applying POCT are discussed.

6. Acknowledgements

We thank the US National Institute of Health (GM124316, MH110975, and HL142326A) for financial support. We would also like to thank Dr. Ana Sol Peinetti for proof reading and editing of the manuscript.

List of Abbreviations:

POC

Point-of-care

IVD

In vitro diagnostics

PCR

Polymerase chain reaction

LOC

Lab-on-a-chip

LID

Lab-in-a-drop

μPADs

Microfluidic paper-based analytical devices

CRISPR

Clustered regularly interspaced short palindromic repeats

ASSURED

Affordable, Sensitive, Specific, User-friendly, Rapid and robust, Equipment-free and Deliverable to end-users

IFN-γ

Interferon-gamma

ELISA

Enzyme-linked immunosorbent assay

CHA

Catalytic hairpin assembly

DETECTR

DNA endonuclease-targeted CRISPR trans reporter

R&D

Research and development

PGM

Personal glucose meter

MSNs

Mesoporous silica nanoparticles

NMs

Nanoporous membranes

NADH

Nicotinamide adenine dinucleotide

GSH

reduced glutathione

PSA

prostate-specific antigen

OTA

Ochratoxin A

CEA

Carcinoembryonic antigen

HbA1c

Glycated hemoglobin

PDGF-BB

Platelet-derived growth factor BB

CXR

Cation exchange reaction

CAMB

Catalytic and molecular beacon

VEGF

Vascular endothelial growth factor

8-OHdG

8-hydroxy-2’-deoxyguanosine

AuNPs

Gold nanoparticles

BRCA

Backfilling rolling cycle amplification

HBV

Hepatitis B virus

HIV

Human immuno-deficiency virus

ICSDPR

Isothermal circular strand-displacement polymerization reaction

HNFs

Hybrid nanoflowers

FBS

Fetal bovine serum

MC-LR

Microcystin-LR

GO-AuNPs

Graphene oxide-gold nanoparticles

SW620

Human colorectal cancer cell

DNA MTase

DNA methyltransferase

P-ERCA

Padlock exponential rolling circle amplification

ALP

Alkaline phosphatase

GALT

Galactose-1-phosphate uridyltransferase

CRP

C-reactive protein

N.A.

Not Applicable

LOD

Limit of detection

ConA

Concanavalin A

GOx

Glucose oxidase

HCR

Hybridization chain reaction

HOP

Human oncogenic protein

CEA

Carcinoembryonic antigen

AuHM

Gold hollow microspheres

NSE

Neuron-specific enolase

SCCA

Squamous cell carcinoma antigen

RCA

Rolling circle amplification

AchE

Acetylcholinesterase

LAMP

Loop mediated isothermal amplification

CFU

Colony-Forming Units

AFB1

Aflatoxin B1

PEC

Photoelectrochemical

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of Interest

Yi Lu is a co-Founder of GlucoSentient, Inc.

7. References

  • [1].Tang L, Li J, Plasmon-Based Colorimetric Nanosensors for Ultrasensitive Molecular Diagnostics, ACS. Sens 2 (2017) 857–875. 10.1021/acssensors.7b00282. [DOI] [PubMed] [Google Scholar]
  • [2].Kramer F, Just S, Zeller T, New perspectives: systems medicine in cardiovascular disease, BMC Syst. Biol 12 (2018) 57 10.1186/s12918-018-0579-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Caliendo AM, Gilbert DN, Ginocchio CC, Hanson KE, May L, Quinn TC, Tenover FC, Alland D, Blaschke AJ, Bonomo RA, Carroll KC, Ferraro MJ, Hirschhorn LR, Joseph WP, Karchmer T, MacIntyre AT, Reller LB, Jackson AF, Infectious A Diseases Society of, Better tests, better care: improved diagnostics for infectious diseases, Clin Infect Dis 57 Suppl 3 (2013) S139–170. 10.1093/cid/cit578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Gubala V, Harris LF, Ricco AJ, Tan MX, Williams DE, Point of care diagnostics: status and future, Anal. Chem 84 (2012) 487–515. 10.1021/ac2030199. [DOI] [PubMed] [Google Scholar]
  • [5].Song Y, Lin B, Tian T, Xu X, Wang W, Ruan Q, Guo J, Zhu Z, Yang C, Recent Progress in Microfluidics-Based Biosensing, Anal. Chem 91 (2019) 388–404. 10.1021/acs.analchem.8b05007. [DOI] [PubMed] [Google Scholar]
  • [6].Zhang JJ, Zheng TT, Cheng FF, Zhang JR, Zhu JJ, Toward the Early Evaluation of Therapeutic Effects: An Electrochemical Platform for Ultrasensitive Detection of Apoptotic Cells, Anal. Chem 83 (2011) 7902–7909. 10.1021/ac201804b. [DOI] [PubMed] [Google Scholar]
  • [7].Zhang JJ, Cheng FF, Li JJ, Zhu JJ, Lu Y, Fluorescent nanoprobes for sensing and imaging of metal ions: Recent advances and future perspectives, Nano Today 11 (2016) 309–329. 10.1016/j.nantod.2016.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Zhang JJ, Cheng FF, Zheng TT, Zhu JJ, Versatile aptasensor for electrochemical quantification of cell surface glycan and naked-eye tracking glycolytic inhibition in living cells, Biosens. Bioelectron 89 (2017) 937–945. 10.1016/j.bios.2016.09.087. [DOI] [PubMed] [Google Scholar]
  • [9].Xu WC, Xing H, Lu Y, A smart T-1-weighted MRI contrast agent for uranyl cations based on a DNAzyme-gadolinium conjugate, Analyst 138 (2013) 6266–6269. 10.1039/c3an01182h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Xiang Y, Lu Y, Using personal glucose meters and functional DNA sensors to quantify a variety of analytical targets, Nat. Chem 3 (2011) 697–703. 10.1038/Nchem.1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Zhang JJ, Xing H, Lu Y, Translating molecular detections into a simple temperature test using a target-responsive smart thermometer, Chemical Science 9 (2018) 3906–3910. 10.1039/c7sc05325h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Tram K, Kanda P, Salena BJ, Huan SY, Li YF, Translating Bacterial Detection by DNAzymes into a Litmus Test, Angew. Chem. Int. Ed. Engl 53 (2014) 12799–12802. 10.1002/anie.201407021. [DOI] [PubMed] [Google Scholar]
  • [13].Zhang Y, Zhou CB, Nie JF, Le SW, Qin Q, Liu F, Li YP, Li JP, Equipment-Free Quantitative Measurement for Microfluidic Paper-Based Analytical Devices Fabricated Using the Principles of Movable-Type Printing, Anal. Chem 86 (2014) 2005–2012. 10.1021/ac403026c. [DOI] [PubMed] [Google Scholar]
  • [14].Tian T, Li JX, Song YL, Zhou LJ, Zhu Z, Yang CYJ, Distance-based microfluidic quantitative detection methods for point-of-care testing, Lab. Chip 16 (2016) 1139–1151. 10.1039/c5lc01562f. [DOI] [PubMed] [Google Scholar]
  • [15].Liu D, Tian T, Chen XF, Lei ZC, Song YL, Shi YZ, Ji TH, Zhu Z, Yang L, Yang CY, Gas-generating reactions for point-of-care testing, Analyst 143 (2018) 1294–1304. 10.1039/c8an00011e. [DOI] [PubMed] [Google Scholar]
  • [16].Connelly JT, Rolland JP, Whitesides GM, “Paper Machine” for Molecular Diagnostics, Anal. Chem 87 (2015) 7595–7601. 10.1021/acs.analchem.5b00411. [DOI] [PubMed] [Google Scholar]
  • [17].Luppa PB, Bietenbeck A, Beaudoin C, Giannetti A, Clinically relevant analytical techniques, organizational concepts for application and future perspectives of point-of-care testing, Biotechnol. Adv 34 (2016) 139–160. 10.1016/j.biotechadv.2016.01.003. [DOI] [PubMed] [Google Scholar]
  • [18].Nayak S, Blumenfeld NR, Laksanasopin T, Sia SK, Point-of-Care Diagnostics: Recent Developments in a Connected Age, Anal. Chem 89 (2017) 102–123. 10.1021/acs.analchem.6b04630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Chen H, Liu K, Li Z, Wang P, Point of care testing for infectious diseases, Clin. Chim. Acta 493 (2019) 138–147. 10.1016/j.cca.2019.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Chen Y, Xia L, Liang R, Lu Z, Li L, Huo B, Li G, Hu Y, Advanced materials for sample preparation in recent decade, Trac-Trend. Anal. Chem 120 (2019) 115652 [Google Scholar]
  • [21].Prata JC, da Costa JP, Duarte AC, Rocha-Santos T, Methods for sampling and detection of microplastics in water and sediment: A critical review, Trac-Trend. Anal. Chem 110 (2019) 150–159. 10.1016/j.trac.2018.10.029. [DOI] [Google Scholar]
  • [22].Li M, Xiong Y, Qing G, Smart bio-separation materials, Trac-Trend. Anal. Chem (2019). [Google Scholar]
  • [23].Petralia S, Conoci S, PCR Technologies for Point of Care Testing: Progress and Perspectives, ACS. Sens 2 (2017) 876–891. 10.1021/acs.sensors.7b00299. [DOI] [PubMed] [Google Scholar]
  • [24].Koo KM, Wee EJH, Wang Y, Trau M, Enabling miniaturised personalised diagnostics: from lab-on-a-chip to lab-in-a-drop, Lab. Chip 17 (2017) 3200–3220. 10.1039/c7lc00587c. [DOI] [PubMed] [Google Scholar]
  • [25].Yeh EC, Fu CC, Hu L, Thakur R, Feng J, Lee LP, Self-powered integrated microfluidic point-of-care low-cost enabling (SIMPLE) chip, Sci. Adv 3 (2017) e1501645 10.1126/sciadv.1501645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Bandodkar AJ, Gutruf P, Choi J, Lee K, Sekine Y, Reeder JT, Jeang WJ, Aranyosi AJ, Lee SP, Model JB, Ghaffari R, Su CJ, Leshock JP, Ray T, Verrillo A, Thomas K, Krishnamurthi V, Han S, Kim J, Krishnan S, Hang T, Rogers JA, Battery-free, skin-interfaced microfluidic/electronic systems for simultaneous electrochemical, colorimetric, and volumetric analysis of sweat, Sci. Adv 5 (2019) eaav3294 10.1126/sciadv.aav3294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Liu H, Xiang Y, Lu Y, Crooks RM, Aptamer-based origami paper analytical device for electrochemical detection of adenosine, Angew. Chem. Int. Ed. Engl 51 (2012) 6925–6928. 10.1002/anie.201202929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Govindarajan AV, Ramachandran S, Vigil GD, Yager P, Bohringer KF, A low cost point-of-care viscous sample preparation device for molecular diagnosis in the developing world; an example of microfluidic origami, Lab. Chip 12 (2012) 174–181. 10.1039/c1lc20622b. [DOI] [PubMed] [Google Scholar]
  • [29].Goode JA, Rushworth JV, Millner PA, Biosensor Regeneration: A Review of Common Techniques and Outcomes, Langmuir 31 (2015) 6267–6276. 10.1021/la503533g. [DOI] [PubMed] [Google Scholar]
  • [30].Chen AL, Yang SM, Replacing antibodies with aptamers in lateral flow immunoassay, Biosens. Bioelectron 71 (2015) 230–242. 10.1016/j.bios.2015.04.041. [DOI] [PubMed] [Google Scholar]
  • [31].Zhou WH, Saran R, Liu JW, Metal Sensing by DNA, Chem. Rev 117 (2017) 8272–8325. 10.1021/acs.chemrev.7b00063. [DOI] [PubMed] [Google Scholar]
  • [32].Wang W, Yu S, Huang S, Bi S, Han H, Zhang J-R, Lu Y, Zhu J-J, Bioapplications of DNA nanotechnology at the solid–liquid interface, Chem. Soc. Rev 48 (2019) 4892–4920. 10.1039/C8CS00402A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Hwang K, Mou Q, Lake RJ, Xiong M, Holland B, Lu Y, Metal-Dependent DNAzymes for the Quantitative Detection of Metal Ions in Living Cells: Recent Progress, Current Challenges, and Latest Results on FRET Ratiometric Sensors, Inorg. Chem (2019). 10.1021/acs.inorgchem.9b01280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Li L, Xing H, Zhang J, Lu Y, Functional DNA Molecules Enable Selective and Stimuli-Responsive Nanoparticles for Biomedical Applications, Acc. Chem. Res 52 (2019) 2415–2426. 10.1021/acs.accounts.9b00167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Zhang J, Lan T, Lu Y, Molecular Engineering of Functional Nucleic Acid Nanomaterials toward In Vivo Applications, Adv. Healthc. Mater 8 (2019) e1801158 10.1002/adhm.201801158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Zhang J, Smaga LP, Satyavolu NSR, Chan J, Lu Y, DNA Aptamer-Based Activatable Probes for Photoacoustic Imaging in Living Mice, J. Am. Chem. Soc 139 (2017) 17225–17228. 10.1021/jacs.7b07913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Lopez A, Liu JW, Fluorescence Polarization for Probing DNA Adsorption by Nanomaterials and Fluorophore/DNA Interactions, Langmuir 35 (2019) 9954–9961. 10.1021/acs.langmuir.9b01678. [DOI] [PubMed] [Google Scholar]
  • [38].Perrier S, Guieu V, Chovelon B, Ravelet C, Peyrin E, Panoply of Fluorescence Polarization/Anisotropy Signaling Mechanisms for Functional Nucleic Acid-Based Sensing Platforms, Anal. Chem 90 (2018) 4236–4248. 10.1021/acs.analchem.7b04593. [DOI] [PubMed] [Google Scholar]
  • [39].Wen W, Yan X, Zhu CZ, Du D, Lin YH, Recent Advances in Electrochemical Immunosensors, Anal. Chem 89 (2017) 138–156. 10.1021/acs.analchem.6b04281. [DOI] [PubMed] [Google Scholar]
  • [40].Hajian R, Balderston S, Tran T, DeBoer T, Etienne J, Sandhu M, Wauford NA, Chung JY, Nokes J, Athaiya M, Paredes J, Peytavi R, Goldsmith B, Murthy N, Conboy IM, Aran K, Detection of unamplified target genes via CRISPR-Cas9 immobilized on a graphene field-effect transistor, Nat. Biomed. Eng 3 (2019) 427–437. 10.1038/s41551-019-0371-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Huang JY, Lin HT, Chen TH, Chen CA, Chang HT, Chen CF, Signal Amplified Gold Nanoparticles for Cancer Diagnosis on Paper-Based Analytical Devices, ACS. Sens 3 (2018) 174–182. 10.1021/acssensors.7b00823. [DOI] [PubMed] [Google Scholar]
  • [42].Ren H, Long Z, Cui M, Shao K, Zhou K, Ouyang J, Na N, Dual-Functional Nanoparticles for In Situ Sequential Detection and Imaging of ATP and H2 O2, Small 12 (2016) 3920–3924. 10.1002/smll.201601571. [DOI] [PubMed] [Google Scholar]
  • [43].Shen XT, Wang Y, Zhang Y, Ouyang J, Na N, Unique SiO2 Nanourchins Enable Amplification in Living Cells for In Situ Imaging of mRNAs, Advanced Functional Materials 28 (2018). [Google Scholar]
  • [44].Shen X, Zhang Y, Sun J, Lu H, Ouyang J, Na N, Biodegradable nanosyringes for intracellular amplification-based dual-diagnosis and gene therapy in single living cells, Chem. Sci 10 (2019) 6113–6119. 10.1039/c9sc01894h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Yang Z, Loh KY, Chu Y-T, Feng R, Satyavolu NSR, Xiong M, Nakamata Huynh SM, Hwang K, Li L, Xing H, Zhang X, Chemla YR, Gruebele M, Lu Y, Optical Control of Metal Ion Probes in Cells and Zebrafish Using Highly Selective DNAzymes Conjugated to Upconversion Nanoparticles, J. Am. Chem. Soc 140 (2018) 17656–17665. 10.1021/jacs.8b09867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Rusling JF, Multiplexed Electrochemical Protein Detection and Translation to Personalized Cancer Diagnostics, Anal. Chem 85 (2013) 5304–5310. 10.1021/ac401058v. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Li Y, Liu L, Liu G, CRISPR/Cas Multiplexed Biosensing: A Challenge or an Insurmountable Obstacle?, Trends. Biotechnol 37 (2019) 792–795. 10.1016/j.tibtech.2019.04.012. [DOI] [PubMed] [Google Scholar]
  • [48].Bruch R, Urban GA, Dincer C, CRISPR/Cas Powered Multiplexed Biosensing, Trends. Biotechnol 37 (2019) 791–792. 10.1016/j.tibtech.2019.04.005. [DOI] [PubMed] [Google Scholar]
  • [49].Gootenberg JS, Abudayyeh OO, Kellner MJ, Joung J, Collins JJ, Zhang F, Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6, Science 360 (2018) 439–444. 10.1126/science.aaq0179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Sahlgren C, Meinander A, Zhang HB, Cheng F, Preis M, Xu CL, Salminen TA, Toivola D, Abankwa D, Rosling A, Sen Karaman D, Salo-Ahen OMH, Osterbacka R, Eriksson JE, Willfor S, Petre I, Peltonen J, Leino R, Johnson M, Rosenholm J, Sandler N, Tailored Approaches in Drug Development and Diagnostics: From Molecular Design to Biological Model Systems, Adv. Healthc. Mater 6 (2017). 10.1002/adhm.201700258. [DOI] [PubMed] [Google Scholar]
  • [51].Zarei M, Advances in point-of-care technologies for molecular diagnostics, Biosens. Bioelectron 98 (2017) 494–506. 10.1016/j.bios.2017.07.024. [DOI] [PubMed] [Google Scholar]
  • [52].Choi JR, Tang R, Wang S, Wan Abas WA, Pingguan-Murphy B, Xu F, Paper-based sample-to-answer molecular diagnostic platform for point-of-care diagnostics, Biosens. Bioelectron 74 (2015) 427–439. 10.1016/j.bios.2015.06.065. [DOI] [PubMed] [Google Scholar]
  • [53].Song YJ, Huang YY, Liu XW, Zhang XJ, Ferrari M, Qin LD, Point-of-care technologies for molecular diagnostics using a drop of blood, Trends. Biotechnol 32 (2014) 132–139. 10.1016/j.tibtech.2014.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Lan T, Zhang JJ, Lu Y, Transforming the blood glucose meter into a general healthcare meter for in vitro diagnostics in mobile health, Biotechnol. Adv 34 (2016) 331–341. 10.1016/j.biotechadv.2016.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Nie Z, Deiss F, Liu X, Akbulut O, Whitesides GM, Integration of paper-based microfluidic devices with commercial electrochemical readers, Lab. Chip 10 (2010) 3163–3169. 10.1039/C0LC00237B. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Mohapatra H, Phillips ST, Reagents and assay strategies for quantifying active enzyme analytes using a personal glucose meter, Chem. Commun 49 (2013) 6134–6136. 10.1039/C3CC43702G. [DOI] [PubMed] [Google Scholar]
  • [57].Xiang Y, Lu Y, Portable and Quantitative Detection of Protein Biomarkers and Small Molecular Toxins Using Antibodies and Ubiquitous Personal Glucose Meters, Anal. Chem 84 (2012) 4174–4178. 10.1021/ac300517n. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Su J, Xu J, Chen Y, Xiang Y, Yuan R, Chai YQ, Personal glucose sensor for point-of-care early cancer diagnosis, Chem. Commun 48 (2012) 6909–6911. 10.1039/c2cc32729e. [DOI] [PubMed] [Google Scholar]
  • [59].Xiang Y, Lan T, Lu Y, Using the Widely Available Blood Glucose Meter to Monitor Insulin and HbA1c, J. Diabetes Sci. Technol 8 (2014) 855–858. 10.1177/1932296814532875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Zhu X, Zheng H, Xu H, Lin R, Han Y, Yang G, Lin Z, Guo L, Qiu B, Chen G, A reusable and portable immunosensor using personal glucose meter as transducer, Anal. Methods 6 (2014) 5264–5268. 10.1039/C4AY00490F. [DOI] [Google Scholar]
  • [61].Hun X, Xu Y, Luo X, Peptide-based biosensor for the prostate-specific antigen using magnetic particle-bound invertase and a personal glucose meter for readout, Microchimica Acta 182 (2015) 1669–1675. 10.1007/s00604-015-1483-y. [DOI] [Google Scholar]
  • [62].Ismail NF, Lim TS, Site-specific scFv labelling with invertase via Sortase A mechanism as a platform for antibody-antigen detection using the personal glucose meter, Sci. Rep 6 (2016). 10.1038/srep19338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Hong L, Zhou F, Shi DM, Zhang XJ, Wang GF, Portable aptamer biosensor of platelet-derived growth factor-BB using a personal glucose meter with triply amplified, Biosens. Bioelectron 95 (2017) 152–159. 10.1016/j.bios.2017.04.023. [DOI] [PubMed] [Google Scholar]
  • [64].Zhu X, Kou FX, Xu HF, Lin LP, Yang GD, Lin ZY, A highly sensitive aptamer-immunoassay for vascular endothelial growth factor coupled with portable glucose meter and hybridization chain reaction, Sens. Actuators. B-Chem 253 (2017) 660–665. 10.1016/j.snb.2017.06.174. [DOI] [Google Scholar]
  • [65].Zhu XN, Sarwar M, Zhu JJ, Zhang CX, Kaushik A, Li CZ, Using a glucose meter to quantitatively detect disease biomarkers through a universal nanozyme integrated lateral fluidic sensing platform, Biosens. Bioelectron 126 (2019) 690–696. 10.1016/j.bios.2018.11.033. [DOI] [PubMed] [Google Scholar]
  • [66].Ge LL, Li B, Xu HX, Pu WY, Kwok HF, Backfilling rolling cycle amplification with enzyme-DNA conjugates on antibody for portable electrochemical immunoassay with glucometer readout, Biosens. Bioelectron 132 (2019) 210–216. 10.1016/j.bios.2019.02.051. [DOI] [PubMed] [Google Scholar]
  • [67].Xiang Y, Lu Y, Using Commercially Available Personal Glucose Meters for Portable Quantification of DNA, Anal. Chem 84 (2012) 1975–1980. 10.1021/ac203014s. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Xu J, Jiang BY, Xie JQ, Xiang Y, Yuan R, Chai YQ, Sensitive point-of-care monitoring of HIV related DNA sequences with a personal glucometer, Chem. Commun 48 (2012) 10733–10735. 10.1039/c2cc35941c. [DOI] [PubMed] [Google Scholar]
  • [69].Zhou ZJ, Xiang Y, Tong AJ, Lu Y, Simple and Efficient Method to Purify DNA-Protein Conjugates and Its Sensing Applications, Anal. Chem 86 (2014) 3869–3875. 10.1021/ac4040554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Wang Q, Wang H, Yang X, Wang K, Liu F, Zhao Q, Liu P, Liu R, Multiplex detection of nucleic acids using a low cost microfluidic chip and a personal glucose meter at the point-of-care, Chem. Commun 50 (2014) 3824–3826. 10.1039/c4cc00133h. [DOI] [PubMed] [Google Scholar]
  • [71].Xu XT, Liang KY, Zeng JY, Portable and sensitive quantitative detection of DNA based on personal glucose meters and isothermal circular strand-displacement polymerization reaction, Biosens. Bioelectron 64 (2015) 671–675. 10.1016/j.bios.2014.09.094. [DOI] [PubMed] [Google Scholar]
  • [72].Xu XT, Liang KY, Zeng JY, Portable and sensitive quantitative detection of DNA using personal glucose meters and exonuclease III-assisted signal amplification, Analyst 139 (2014) 4982–4986. 10.1039/c4an00905c. [DOI] [PubMed] [Google Scholar]
  • [73].Fang J, Guo YC, Yang YJ, Yu W, Tao YY, Dai T, Yuan CJ, Xie GM, Portable and sensitive detection of DNA based on personal glucose meters and nanogold-functionalized PAMAM dendrimer, Sens. Actuators. B-Chem. 272 (2018) 118–126. 10.1016/j.snb.2018.05.086. [DOI] [Google Scholar]
  • [74].Shan YK, Zhang Y, Kang WJ, Wang B, Li JH, Wu XP, Wang SY, Liu F, Quantitative and selective DNA detection with portable personal glucose meter using loop-based DNA competitive hybridization strategy, Sens. Actuators. B-Chem. 282 (2019) 197–203. 10.1016/j.snb.2018.11.062. [DOI] [PubMed] [Google Scholar]
  • [75].Dong Q, Liu QY, Guo LL, Li D, Shang XD, Li BL, Du Y, A signal-flexible gene diagnostic strategy coupling loop-mediated isothermal amplification with hybridization chain reaction, Anal. Chim. Acta 1079 (2019) 171–179. 10.1016/j.aca.2019.06.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Jia YJ, Sun FF, Na N, Ouyang J, Detection of p53 DNA using commercially available personal glucose meters based on rolling circle amplification coupled with nicking enzyme signal amplification, Anal. Chim. Acta 1060 (2019) 64–70. 10.1016/j.aca.2019.01.061. [DOI] [PubMed] [Google Scholar]
  • [77].Gu Y, Zhang TT, Huang ZF, Hu SW, Zhao W, Xu JJ, Chen HY, An exploration of nucleic acid liquid biopsy using a glucose meter, Chem. Sci 9 (2018) 3517–3522. 10.1039/c8sc00627j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Wu TT, Yang YM, Cao Y, Song YC, Xu LP, Zhang XJ, Wang ST, Bioinspired DNA-Inorganic Hybrid Nanoflowers Combined with a Personal Glucose Meter for Onsite Detection of miRNA, ACS. Appl. Mater. Inter 10 (2018) 42050–42057. 10.1021/acsami.8b15917. [DOI] [PubMed] [Google Scholar]
  • [79].Wu TT, Cao Y, Yang YM, Zhang XY, Wang ST, Xu LP, Zhang XJ, A three-dimensional DNA walking machine for the ultrasensitive dual-modal detection of miRNA using a fluorometer and personal glucose meter, Nanoscale 11 (2019) 11279–11284. 10.1039/c9nr03588e. [DOI] [PubMed] [Google Scholar]
  • [80].Zhu XN, Sarwar M, Yue QL, Chen CY, Li CZ, Biosensing of DNA oxidative damage: a model of using glucose meter for non-glucose biomarker detection, Int. J. Nanomed 12 (2017) 979–987. 10.2147/Ijn.S125437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Chen S, Zhang J, Gan N, Hu F, Li T, Cao Y, Pan D, An on-site immunosensor for ractopamine based on a personal glucose meter and using magnetic β-cyclodextrin-coated nanoparticles for enrichment, and an invertase-labeled nanogold probe for signal amplification, Microchimica Acta 182 (2015) 815–822. 10.1007/s00604-014-1392-5. [DOI] [Google Scholar]
  • [82].Gu CM, Lan T, Shi HC, Lu Y, Portable Detection of Melamine in Milk Using a Personal Glucose Meter Based on an in Vitro Selected Structure-Switching Aptamer, Anal. Chem 87 (2015) 7676–7682. 10.1021/acs.analchem.5b01085. [DOI] [PubMed] [Google Scholar]
  • [83].Hun X, Xu YQ, Xie GL, Luo XL, Aptamer biosensor for highly sensitive and selective detection of dopamine using ubiquitous personal glucose meters, Sens. Actuators. B-Chem 209 (2015) 596–601. 10.1016/j.snb.2014.11.135. [DOI] [Google Scholar]
  • [84].Zhang JJ, Shen Z, Xiang Y, Lu Y, Integration of Solution-Based Assays onto Lateral Flow Device for One-Step Quantitative Point-of-Care Diagnostics Using Personal Glucose Meter, ACS. Sens 1 (2016) 1091–1096. 10.1021/acssensors.6b00270. [DOI] [Google Scholar]
  • [85].Li F, Zhang RH, Kang HB, Hu YG, Liu YH, Zhu JY, Facile and sensitive detection of clenbuterol in pork using a personal glucose meter, Anal. Methods 9 (2017) 6507–6512. 10.1039/c7ay01826f. [DOI] [Google Scholar]
  • [86].Qiu Y, Gu CM, Li B, Shi HC, Aptameric detection of quinine in reclaimed wastewater using a personal glucose meter, Anal. Methods 10 (2018) 2931–2938. 10.1039/c8ay00585k. [DOI] [Google Scholar]
  • [87].Jang YJ, Lee KH, Yoo TH, Kim DM, Interfacing a Personal Glucose Meter with Cell-Free Protein Synthesis for Rapid Analysis of Amino Acids, Anal. Chem 91 (2019) 2531–2535. 10.1021/acs.analchem.8b05526. [DOI] [PubMed] [Google Scholar]
  • [88].Xiang Y, Lu Y, An invasive DNA approach toward a general method for portable quantification of metal ions using a personal glucose meter, Chem. Commun 49 (2013) 585–587. 10.1039/c2cc37156a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Huang S, Wang WJ, Cheng FF, Yao HQ, Zhu JJ, Highly sensitive detection of mercury ion based on T-rich DNA machine using portable glucose meter, Sens. Actuators. B-Chem 242 (2017) 347–354. 10.1016/j.snb.2016.10.123. [DOI] [Google Scholar]
  • [90].Ming JJ, Fan WB, Jiang TF, Wang YH, Lv ZH, Portable and sensitive detection of copper(II) ion based on personal glucose meters and a ligation DNAzyme releasing strategy, Sens. Actuators. B-Chem 240 (2017) 1091–1098. 10.1016/j.snb.2016.09.091. [DOI] [Google Scholar]
  • [91].Zeng LW, Gong JY, Rong PS, Liu CS, Chen JH, A portable and quantitative biosensor for cadmium detection using glucometer as the point-of-use device, Talanta 198 (2019) 412–416. 10.1016/j.talanta.2019.02.045. [DOI] [PubMed] [Google Scholar]
  • [92].Zhu X, Xu H, Lin R, Yang G, Lin Z, Chen G, Sensitive and portable detection of telomerase activity in HeLa cells using the personal glucose meter, Chem. Commun 50 (2014) 7897–7899. 10.1039/c4cc03553d. [DOI] [PubMed] [Google Scholar]
  • [93].Yang WX, Lu XH, Wang YC, Sun SJ, Liu CH, Li ZP, Portable and sensitive detection of protein kinase activity by using commercial personal glucose meter, Sens. Actuators. B-Chem 210 (2015) 508–512. 10.1016/j.snb.2015.01.027. [DOI] [Google Scholar]
  • [94].Wang WJ, Huang S, Li JJ, Rui K, Zhang JR, Zhu JJ, Coupling a DNA-Based Machine with Glucometer Readouts for Amplified Detection of Telomerase Activity in Cancer Cells, Sci. Rep 6 (2016). 10.1038/srep23504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Liu CX, Zhang SS, Li X, Xue QW, Jiang W, Multi-code magnetic beads based on DNAzyme-mediated double-cycling amplification for a point-of-care assay of telomerase activity, Analyst 144 (2019) 4241–4249. 10.1039/c9an00589g. [DOI] [PubMed] [Google Scholar]
  • [96].Chen Y, Meng XZ, Gu HW, Yi HC, Sun WY, A dual-response biosensor for electrochemical and glucometer detection of DNA methyltransferase activity based on functionalized metal-organic framework amplification, Biosens. Bioelectron 134 (2019) 117–122. 10.1016/j.bios.2019.03.051. [DOI] [PubMed] [Google Scholar]
  • [97].Gu CM, Long F, Zhou XH, Shi HC, Portable detection of ochratoxin A in red wine based on a structure-switching aptamer using a personal glucometer, RSC. Adv 6 (2016) 29563–29569. 10.1039/c5ra27880e. [DOI] [Google Scholar]
  • [98].Yang XS, Shi DM, Zhu SM, Wang BJ, Zhang XJ, Wang GF, Portable Aptasensor of Aflatoxin B1 in Bread Based on a Personal Glucose Meter and DNA Walking Machine, ACS. Sens 3 (2018) 1368–1375. 10.1021/acssensors.8b00304. [DOI] [PubMed] [Google Scholar]
  • [99].Di Giovanni S, Zambrini V, Varriale A, D’Auria S, Sweet Sensor for the Detection of Aflatoxin M1 in Whole Milk, ACS Omega 4 (2019) 12803–12807. 10.1021/acsomega.9b01300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Zhao LF, Teng L, Zhang J, Li H, Point-of-care detection of Microcystin-LR with a personal glucose meter in drinking water source, Chinese. Chem. Lett 30 (2019) 1035–1037. 10.1016/j.cclet.2019.01.001. [DOI] [Google Scholar]
  • [101].Qiu SY, Yuan LJ, Wei YH, Zhang DW, Chen QL, Lin ZY, Luo LG, DNA template-mediated click chemistry-based portable signal-on sensor for ochratoxin A detection, Food. Chem 297 (2019). 10.1016/j.foodchem.2019.05.203. [DOI] [PubMed] [Google Scholar]
  • [102].Joo J, Kwon D, Shin HH, Park KH, Cha HJ, Jeon S, A facile and sensitive method for detecting pathogenic bacteria using personal glucose meters, Sens. Actuators. B-Chem 188 (2013) 1250–1254. 10.1016/j.snb.2013.08.027. [DOI] [Google Scholar]
  • [103].Huang H, Zhao G, Dou W, Portable and quantitative point-of-care monitoring of Escherichia coli O157:H7 using a personal glucose meter based on immunochromatographic assay, Biosens. Bioelectron 107 (2018) 266–271. 10.1016/j.bios.2018.02.027. [DOI] [PubMed] [Google Scholar]
  • [104].Zhou J, Duan LX, Huang JS, Zuo ZK, Tang TL, Cao D, Ye F, Zhou SW, Zhou YY, Zhang LY, Portable detection of colorectal cancer SW620 cells by using a personal glucose meter, Anal. Biochem 577 (2019) 110–116. 10.1016/j.ab.2019.04.018. [DOI] [PubMed] [Google Scholar]
  • [105].Yan L, Zhu Z, Zou Y, Huang YS, Liu DW, Jia SS, Xu DM, Wu M, Zhou Y, Zhou S, Yang CJ, Target-Responsive “Sweet” Hydrogel with Glucometer Readout for Portable and Quantitative Detection of Non-Glucose Targets, J. Am. Chem. Soc 135 (2013) 3748–3751. 10.1021/ja3114714. [DOI] [PubMed] [Google Scholar]
  • [106].Taebi S, Keyhanfar M, Noorbakhsh A, A novel method for sensitive, low-cost and portable detection of hepatitis B surface antigen using a personal glucose meter, J. Immunol. Methods 458 (2018) 26–32. 10.1016/j.jim.2018.04.001. [DOI] [PubMed] [Google Scholar]
  • [107].Zhang YN, Xue QW, Liu JF, Wang HS, Magnetic bead-liposome hybrids enable sensitive and portable detection of DNA methyltransferase activity using personal glucose meter, Biosens. Bioelectron 87 (2017) 537–544. 10.1016/j.bios.2016.08.103. [DOI] [PubMed] [Google Scholar]
  • [108].Si YM, Li LL, Wang NN, Zheng J, Yang RH, Li JS, Oligonucleotide Cross-Linked Hydrogel for Recognition and Quantitation of MicroRNAs Based on a Portable Glucometer Readout, ACS. Appl. Mater. Inter 11 (2019) 7792–7799. 10.1021/acsami.8b21727. [DOI] [PubMed] [Google Scholar]
  • [109].Xue QW, Kong YC, Wang HS, Jiang W, Liposome-encoded magnetic beads initiated by padlock exponential rolling circle amplification for portable and accurate quantification of microRNAs, Chem. Commun 53 (2017) 10772–10775. 10.1039/c7cc05686a. [DOI] [PubMed] [Google Scholar]
  • [110].Wang ZZ, Chen ZW, Gao N, Ren JS, Qu XG, Transmutation of Personal Glucose Meters into Portable and Highly Sensitive Microbial Pathogen Detection Platform, Small 11 (2015) 4970–4975. 10.1002/smll.201500944. [DOI] [PubMed] [Google Scholar]
  • [111].Lin BQ, Liu D, Yan JM, Qiao Z, Zhong YX, Yan JW, Zhu Z, Ji TH, Yang CJ, Enzyme-Encapsulated Liposome-Linked Immunosorbent Assay Enabling Sensitive Personal Glucose Meter Readout for Portable Detection of Disease Biomarkers, ACS. Appl. Mater. Inter 8 (2016) 6890–6897. 10.1021/acsami.6b00777. [DOI] [PubMed] [Google Scholar]
  • [112].Zhang JJ, Xiang Y, Novak DE, Hoganson GE, Zhu JJ, Lu Y, Using a Personal Glucose Meter and Alkaline Phosphatase for Point-of-Care Quantification of Galactose-1-Phosphate Uridyltransferase in Clinical Galactosemia Diagnosis, Chem-Asian J. 10 (2015) 2221–2227. 10.1002/asia.201500642. [DOI] [PubMed] [Google Scholar]
  • [113].Ahn JK, Kim HY, Lee CY, Park KS, Park HG, Label-free and washing-free alkaline phosphatase assay using a personal glucose meter, J. Biol. Eng 13 (2019). 10.1186/s13036-019-0182-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Wang Q, Wang H, Yang XH, Wang KM, Liu RJ, Li Q, Ou JQ, A sensitive one-step method for quantitative detection of alpha-amylase in serum and urine using a personal glucose meter, Analyst 140 (2015) 1161–1165. 10.1039/c4an02033b. [DOI] [PubMed] [Google Scholar]
  • [115].Chavali R, Gunda NSK, Naicker S, Mitra SK, Detection of Escherichia coli in potable water using personal glucose meters, Anal. Methods 6 (2014) 6223–6227. 10.1039/c4ay01249f. [DOI] [Google Scholar]
  • [116].Ahn JK, Kim HY, Park KS, Park HG, A Personal Glucose Meter for Label-Free and Washing-Free Biomolecular Detection, Anal. Chem 90 (2018) 11340–11343. 10.1021/acs.analchem.8b02014. [DOI] [PubMed] [Google Scholar]
  • [117].Bai J, Liu LY, Han YM, Jia CC, Liang CX, One-step detection of hexokinase activity using a personal glucose meter, Anal. Methods 10 (2018) 2075–2080. 10.1039/c8ay00498f. [DOI] [Google Scholar]
  • [118].Fang DY, Gao GY, Yu Y, Shen J, Zhi JF, Adaptive use of a personal glucose meter (PGM) for acute biotoxicity assessment based on the glucose consumption of microbes, Analyst 141 (2016) 3004–3011. 10.1039/c5an02478a. [DOI] [PubMed] [Google Scholar]
  • [119].Kwon D, Lee H, Yoo H, Hwang J, Lee D, Jeon S, Facile method for enrofloxacin detection in milk using a personal glucose meter, Sens. Actuators. B-Chem 254 (2018) 935–939. 10.1016/j.snb.2017.07.118. [DOI] [Google Scholar]
  • [120].Xiao WC, Gao YM, Zhang Y, Li J, Liu ZY, Nie JF, Li JP, Enhanced 3D paper-based devices with a personal glucose meter for highly sensitive and portable biosensing of silver ion, Biosens. Bioelectron 137 (2019) 154–160. 10.1016/j.bios.2019.05.003. [DOI] [PubMed] [Google Scholar]
  • [121].Lu MH, Bioresponsive-controlled release of methylene blue from magnetic mesoporous silica from the electrochemical detection of telomerase activity, Analyst 142 (2017) 3477–3483. 10.1039/c7an01127j. [DOI] [PubMed] [Google Scholar]
  • [122].Fu LB, Zhuang JY, Lai WQ, Que XH, Lu MH, Tang DP, Portable and quantitative monitoring of heavy metal ions using DNAzyme-capped mesoporous silica nanoparticles with a glucometer readout, J. Mater. Chem. B 1 (2013) 6123–6128. 10.1039/c3tb21155j. [DOI] [PubMed] [Google Scholar]
  • [123].Gao ZQ, Tang DP, Xu MD, Chen GN, Yang HH, Nanoparticle-based pseudo hapten for target-responsive cargo release from a magnetic mesoporous silica nanocontainer, Chem. Commun 50 (2014) 6256–6258. 10.1039/c4cc01511h. [DOI] [PubMed] [Google Scholar]
  • [124].Liang XL, Wang L, Wang D, Zeng LW, Fang ZY, Portable and quantitative monitoring of mercury ions using DNA-gated mesoporous silica nanoparticles using a glucometer readout, Chem. Commun 52 (2016) 2192–2194. 10.1039/c5cc08611f. [DOI] [PubMed] [Google Scholar]
  • [125].Tan QQ, Zhang RR, Kong RM, Kong WS, Zhao WZ, Qu FL, Detection of glutathione based on MnO2 nanosheet-gated mesoporous silica nanoparticles and target induced release of glucose measured with a portable glucose meter, Microchimica Acta 185 (2018). 10.1007/s00604-017-2603-7. [DOI] [PubMed] [Google Scholar]
  • [126].Zhao YT, Du D, Lin YH, Glucose encapsulating liposome for signal amplification for quantitative detection of biomarkers with glucometer readout, Biosens. Bioelectron 72 (2015) 348–354. 10.1016/j.bios.2015.05.028. [DOI] [PubMed] [Google Scholar]
  • [127].Zhao YT, Chen X, Lin S, Du D, Lin YH, Integrated immunochromatographic strip with glucometer readout for rapid quantification of phosphorylated proteins, Anal. Chim. Acta 964 (2017) 1–6. 10.1016/j.aca.2017.01.011. [DOI] [PubMed] [Google Scholar]
  • [128].Li GZ, Tang DP, Bioresponsive controlled glucose release from TiO2 nanotube arrays: a simple and portable biosensing system for cocaine with a glucometer readout, J. Mater. Chem. B 5 (2017) 5573–5579. 10.1039/c7tb00670e. [DOI] [Google Scholar]
  • [129].Zhang JJ, Xiang Y, Wang M, Basu A, Lu Y, Dose-Dependent Response of Personal Glucose Meters to Nicotinamide Coenzymes: Applications to Point-of-Care Diagnostics of Many Non-Glucose Targets in a Single Step, Angew. Chem. Int. Ed. Engl 55 (2016) 732–736. 10.1002/anie.201507563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Zhang JJ, Lu Y, Biocomputing for Portable, Resettable, and Quantitative Point-of-Care Diagnostics: Making the Glucose Meter a Logic-Gate Responsive Device for Measuring Many Clinically Relevant Targets, Angew. Chem. Int. Ed. Engl 57 (2018) 9702–9706. 10.1002/anie.201804292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Das A, Cui XK, Chivukula V, Iyer SS, Detection of Enzymes, Viruses, and Bacteria Using Glucose Meters, Anal. Chem 90 (2018) 11589–11598. 10.1021/acs.analchem.8b02960. [DOI] [PubMed] [Google Scholar]
  • [132].Xu JQ, Qiao XJ, Zhang JJ, Cheng N, Sheng QL, Zheng JB, Cao W, Yue TL, Lu Y, Point-of-care monitoring of intracellular glutathione and serum triglyceride levels using a versatile personal glucose meter, Anal. Methods 11 (2019) 1849–1856. 10.1039/c8ay02709a. [DOI] [Google Scholar]
  • [133].Tang WZ, Yang JX, Wang F, Wang JL, Li ZH, Thiocholine-triggered reaction in personal glucose meters for portable quantitative detection of organophosphorus pesticide, Anal. Chim. Acta 1060 (2019) 97–102. 10.1016/j.aca.2019.01.051. [DOI] [PubMed] [Google Scholar]
  • [134].Xu HF, Kou FX, Ye HZ, Wang ZW, Huang SX, Liu XX, Zhu X, Lin ZY, Chen GN, Highly sensitive antibody-aptamer sensor for vascular endothelial growth factor based on hybridization chain reaction and pH meter/indicator, Talanta 175 (2017) 177–182. 10.1016/j.talanta.2017.04.073. [DOI] [PubMed] [Google Scholar]
  • [135].Kwon D, Joo J, Lee S, Jeon S, Facile and Sensitive Method for Detecting Cardiac Markers using Ubiquitous pH Meters, Anal. Chem 85 (2013) 12134–12137. 10.1021/ac403329w. [DOI] [PubMed] [Google Scholar]
  • [136].Ye RF, Zhu CZ, Song Y, Lu Q, Ge XX, Yang X, Zhu MJ, Du D, Li H, Lin YH, Bioinspired Synthesis of All-in-One Organic-Inorganic Hybrid Nanoflowers Combined with a Handheld pH Meter for On-Site Detection of Food Pathogen, Small 12 (2016) 3094–3100. 10.1002/smll.201600273. [DOI] [PubMed] [Google Scholar]
  • [137].Zhang Y, Yang JN, Nie JF, Yang JH, Gao D, Zhang L, Li JP, Enhanced ELISA using a handheld pH meter and enzyme-coated microparticles for the portable, sensitive detection of proteins, Chem. Commun 52 (2016) 3474–3477. 10.1039/c5cc09852a. [DOI] [PubMed] [Google Scholar]
  • [138].Le SW, Zhou H, Nie JF, Cao CH, Yang JN, Pan HC, Li JP, Zhang Y, Fabrication of paper devices via laser-heating-wax-printing for high-tech enzyme-linked immunosorbent assays with low-tech pen-type pH meter readout, Analyst 142 (2017) 511–516. 10.1039/c6an02422j. [DOI] [PubMed] [Google Scholar]
  • [139].Jiang Y, Su ZY, Zhang J, Cai MJ, Wu LL, A novel electrochemical immunoassay for carcinoembryonic antigen based on glucose oxidase-encapsulated nanogold hollow spheres with a pH meter readout, Analyst 143 (2018) 5271–5277. 10.1039/c8an01436a. [DOI] [PubMed] [Google Scholar]
  • [140].Liang JM, Wang J, Zhang LW, Wang SJ, Yao CP, Zhang Z, Glucose oxidase-loaded liposomes for in situ amplified signal of electrochemical immunoassay on a handheld pH meter, New. J. Chem 43 (2019) 1372–1379. 10.1039/c8nj05637d. [DOI] [Google Scholar]
  • [141].Chen JC, Xue HF, Chen QY, Lin Y, Tang DP, Zheng JW, Enzyme-conjugated hybridization chain reaction for magneto-controlled immunoassay of squamous cell carcinoma antigen with pH meter, Chinese. Chem. Lett 30 (2019) 1631–1634. 10.1016/j.cclet.2019.03.045. [DOI] [Google Scholar]
  • [142].Xie SB, Yuan YL, Song Y, Zhuo Y, Li T, Chai YQ, Yuan R, Using the ubiquitous pH meter combined with a loop mediated isothermal amplification method for facile and sensitive detection of Nosema bombycis genomic DNA PTP1, Chem. Commun 50 (2014) 15932–15935. 10.1039/c4cc06449f. [DOI] [PubMed] [Google Scholar]
  • [143].Tang DP, Xia BY, Tang Y, Zhang J, Zhou Q, Metal-ion-induced DNAzyme on magnetic beads for detection of lead(II) by using rolling circle amplification, glucose oxidase, and readout of pH changes, Microchimica Acta 186 (2019). 10.1007/s00604-019-3454-1. [DOI] [PubMed] [Google Scholar]
  • [144].Zhao MM, Wang PL, Guo YJ, Wang LX, Luo F, Qiu B, Guo LH, Su XO, Lin ZY, Chen GN, Detection of aflatoxin B-1 in food samples based on target-responsive aptamer-cross-linked hydrogel using a handheld pH meter as readout, Talanta 176 (2018) 34–39. 10.1016/j.talanta.2017.08.006. [DOI] [PubMed] [Google Scholar]
  • [145].Chen JJ, Wang LX, Zhao MM, Huang D, Luo F, Huang LY, Qiu B, Guo LH, Lin ZY, Chen GN, Enzyme-linked immunosorbent assay for aflatoxin B-1 using a portable pH meter as the readout, Anal. Methods 10 (2018) 3804–3809. 10.1039/c8ay01030g. [DOI] [Google Scholar]
  • [146].Wang LX, Chen CQ, Huang HW, Huang D, Luo F, Qiu B, Guo LH, Lin ZY, Yang HH, Sensitive detection of telomerase activity in cancer cells using portable pH meter as readout, Biosens. Bioelectron 121 (2018) 153–158. 10.1016/j.bios.2018.08.069. [DOI] [PubMed] [Google Scholar]
  • [147].Wang J, Song MM, Hu CG, Wu KB, Portable, Self-Powered, and Light-Addressable Photoelectrochemical Sensing Platforms Using pH Meter Readouts for High-Throughput Screening of Thrombin Inhibitor Drugs, Anal. Chem 90 (2018) 9366–9373. 10.1021/acs.analchem.8b01979. [DOI] [PubMed] [Google Scholar]
  • [148].Zhou S, Zhao Y, Mecklenburg M, Yang D, Xie B, A novel thermometric biosensor for fast surveillance of beta-lactamase activity in milk, Biosens. Bioelectron 49 (2013) 99–104. 10.1016/j.bios.2013.05.005. [DOI] [PubMed] [Google Scholar]
  • [149].Cai GN, Yu ZZ, Tong P, Tang DP, Ti3C2 MXene quantum dot-encapsulated liposomes for photothermal immunoassays using a portable near-infrared imaging camera on a smartphone, Nanoscale 11 (2019) 15659–15667. 10.1039/c9nr05797h. [DOI] [PubMed] [Google Scholar]
  • [150].Gao BB, Liu H, Gu ZZ, An exothermic chip for point-of-care testing using a forehead thermometer as a readout, Lab. Chip 16 (2016) 525–531. 10.1039/c5lc01136a. [DOI] [PubMed] [Google Scholar]
  • [151].Ma X, Wang Z, He S, Chen C, Luo F, Guo L, Qiu B, Lin Z, Chen G, Hong G, Development of an Immunosensor Based on the Exothermic Reaction between H2O and CaO Using a Common Thermometer as Readout, ACS. Sens (2019). 10.1021/acssensors.9b00968. [DOI] [PubMed] [Google Scholar]
  • [152].Zhu Z, Guan Z, Liu D, Jia S, Li J, Lei Z, Lin S, Ji T, Tian Z, Yang CJ, Translating Molecular Recognition into a Pressure Signal to enable Rapid, Sensitive, and Portable Biomedical Analysis, Angew. Chem. Int. Ed. Engl 54 (2015) 10448–10453. 10.1002/anie.201503963. [DOI] [PubMed] [Google Scholar]
  • [153].Ji T, Liu D, Liu F, Li J, Ruan Q, Song Y, Tian T, Zhu Z, Zhou L, Lin H, Yang C, Wang D, A pressure-based bioassay for the rapid, portable and quantitative detection of C-reactive protein, Chem. Commun 52 (2016) 8452–8454. 10.1039/c6cc03705d. [DOI] [PubMed] [Google Scholar]
  • [154].Liu D, Jia S, Zhang H, Ma Y, Guan Z, Li J, Zhu Z, Ji T, Yang CJ, Integrating Target-Responsive Hydrogel with Pressuremeter Readout Enables Simple, Sensitive, User-Friendly, Quantitative Point-of-Care Testing, ACS. Appl. Mater. Inter 9 (2017) 22252–22258. 10.1021/acsami.7b05531. [DOI] [PubMed] [Google Scholar]
  • [155].Ding E, Hai J, Li T, Wu J, Chen F, Wen Y, Wang B, Lu X, Efficient Hydrogen-Generation CuO/Co3O4 Heterojunction Nanofibers for Sensitive Detection of Cancer Cells by Portable Pressure Meter, Anal. Chem 89 (2017) 8140–8147. 10.1021/acs.analchem.7b01951. [DOI] [PubMed] [Google Scholar]
  • [156].Wang ZY, Hai J, Li TR, Ding EL, He JX, Wang BD, Pressure and Fluorescence Dual Signal Readout CuO-NiO/C Heterojunction Nanofibers-Based Nanoplatform for Imaging and Detection of Target Cancer Cells in Blood, Acs Sustainable Chemistry & Engineering 6 (2018) 9921–9929. 10.1021/acssuschemeng.8b01166. [DOI] [Google Scholar]
  • [157].Wang Y, Yang L, Li B, Yang CJ, Jin Y, Point-of-Care Assay of Telomerase Activity at Single-Cell Level via Gas Pressure Readout, Anal. Chem 89 (2017) 8311–8318. 10.1021/acs.analchem.7b01529. [DOI] [PubMed] [Google Scholar]
  • [158].Song Y, An Y, Liu W, Hou W, Li X, Lin B, Zhu Z, Ge S, Yang HH, Yang C, Centrifugal micropipette-tip with pressure signal readout for portable quantitative detection of myoglobin, Chem. Commun 53 (2017) 11774–11777. 10.1039/c7cc07231g. [DOI] [PubMed] [Google Scholar]
  • [159].Huang D, Lin B, Song Y, Guan Z, Cheng J, Zhu Z, Yang C, Staining Traditional Colloidal Gold Test Strips with Pt Nanoshell Enables Quantitative Point-of-Care Testing with Simple and Portable Pressure Meter Readout, ACS. Appl. Mater. Inter 11 (2019) 1800–1806. 10.1021/acsami.8b15562. [DOI] [PubMed] [Google Scholar]
  • [160].Shi L, Lei J, Zhang B, Li B, Yang CJ, Jin Y, Ultrasensitive and Facile Detection of MicroRNA via a Portable Pressure Meter, ACS. Appl. Mater. Inter 10 (2018) 12526–12533. 10.1021/acsami.8b02551. [DOI] [PubMed] [Google Scholar]
  • [161].Lei J, Shi L, Li B, Yang CJ, Jin Y, Ultrasensitive and portable assay of mercury (II) ions via gas pressure as readout, Biosens. Bioelectron 122 (2018) 32–36. 10.1016/j.bios.2018.09.020. [DOI] [PubMed] [Google Scholar]
  • [162].Li J, Jiang H, Rao X, Liu Z, Zhu H, Xu Y, Point-of-Care Testing of Pathogenic Bacteria at the Single-Colony Level via Gas Pressure Readout Using Aptamer-Coated Magnetic CuFe2O4 and Vancomycin-Capped Platinum Nanoparticles, Anal. Chem 91 (2019) 1494–1500. 10.1021/acs.analchem.8b04584. [DOI] [PubMed] [Google Scholar]
  • [163].Liu P, Fang X, Cao H, Gu M, Kong J, Deng A, Magnetic-bioluminescent-nanoliposomes for ultrasensitive and portable detection of protein biomarkers in blood, Anal. Chim. Acta 1039 (2018) 98–107. 10.1016/j.aca.2018.07.039. [DOI] [PubMed] [Google Scholar]
  • [164].Chen YP, Xianyu YL, Wu J, Dong ML, Zheng WS, Sun JS, Jiang XY, Double-Enzymes-Mediated Bioluminescent Sensor for Quantitative and Ultrasensitive Point-of-Care Testing, Anal. Chem 89 (2017) 5422–5427. 10.1021/acs.analchem.7b00239. [DOI] [PubMed] [Google Scholar]
  • [165].Wang Q, Liu F, Yang XH, Wang KM, Liu P, Liu JB, Huang J, Wang H, Using personal uric acid meter and enzyme-DNA conjugate for portable and quantitative DNA detection, Sens. Actuators. B-Chem 186 (2013) 515–520. 10.1016/j.snb.2013.06.056. [DOI] [Google Scholar]
  • [166].Deng JQ, Jiang XY, Advances in Reagents Storage and Release in Self-Contained Point-of-Care Devices, Adv. Mater. Technol 4 (2019). 10.1002/admt.201800625. [DOI] [Google Scholar]

RESOURCES