
Citation: Sanna Turunen, Susanna Kaisto, Ilya Skovorodkin, Vladimir Mironov, Tomi Kalpio, Seppo Vainio, Aleksandra Rak-Raszewska. 3D bioprinting of the kidney—hype or hope?[J]. AIMS Cell and Tissue Engineering, 2018, 2(3): 119-162. doi: 10.3934/celltissue.2018.3.119
[1] | Lucia Spicuzza, Davide Campagna, Chiara Di Maria, Enrico Sciacca, Salvatore Mancuso, Carlo Vancheri, Gianluca Sambataro . An update on lateral flow immunoassay for the rapid detection of SARS-CoV-2 antibodies. AIMS Microbiology, 2023, 9(2): 375-401. doi: 10.3934/microbiol.2023020 |
[2] | CB Harder, S Persson, J Christensen, A Ljubic, EM Nielsen, J Hoorfar . Molecular diagnostics of Salmonella and Campylobacter in human/animal fecal samples remain feasible after long-term sample storage without specific requirements. AIMS Microbiology, 2021, 7(4): 399-414. doi: 10.3934/microbiol.2021024 |
[3] | Alrayan Abass Albaz, Misbahuddin M Rafeeq, Ziaullah M Sain, Wael Abdullah Almutairi, Ali Saeed Alamri, Ahmed Hamdan Aloufi, Waleed Hassan Almalki, Mohammed Tarique . Nanotechnology-based approaches in the fight against SARS-CoV-2. AIMS Microbiology, 2021, 7(4): 368-398. doi: 10.3934/microbiol.2021023 |
[4] | Anil K. Persad, Michele L. Williams, Jeffrey T. LeJeune . Rapid loss of a green fluorescent plasmid in Escherichia coli O157:H7. AIMS Microbiology, 2017, 3(4): 872-884. doi: 10.3934/microbiol.2017.4.872 |
[5] | Dharmender Kumar, Lalit Batra, Mohammad Tariq Malik . Insights of Novel Coronavirus (SARS-CoV-2) disease outbreak, management and treatment. AIMS Microbiology, 2020, 6(3): 183-203. doi: 10.3934/microbiol.2020013 |
[6] | T. Amrouche, M. L. Chikindas . Probiotics for immunomodulation in prevention against respiratory viral infections with special emphasis on COVID-19. AIMS Microbiology, 2022, 8(3): 338-356. doi: 10.3934/microbiol.2022024 |
[7] | Umar Shahbaz, Nazira Fatima, Samra Basharat, Asma Bibi, Xiaobin Yu, Muhammad Iftikhar Hussain, Maryam Nasrullah . Role of vitamin C in preventing of COVID-19 infection, progression and severity. AIMS Microbiology, 2022, 8(1): 108-124. doi: 10.3934/microbiol.2022010 |
[8] | Elena Kotenkova, Dagmara Bataeva, Mikhail Minaev, Elena Zaiko . Application of EvaGreen for the assessment of Listeria monocytogenes АТСС 13932 cell viability using flow cytometry. AIMS Microbiology, 2019, 5(1): 39-47. doi: 10.3934/microbiol.2019.1.39 |
[9] | Paola Brun . The profiles of dysbiotic microbial communities. AIMS Microbiology, 2019, 5(1): 87-101. doi: 10.3934/microbiol.2019.1.87 |
[10] | Luciana C. Gomes, Joana M. R. Moreira, José D. P. Araújo, Filipe J. Mergulhão . Surface conditioning with Escherichia coli cell wall components can reduce biofilm formation by decreasing initial adhesion. AIMS Microbiology, 2017, 3(3): 613-628. doi: 10.3934/microbiol.2017.3.613 |
Successful operation of any clinical laboratory of the world today is unlikely to be possible without test systems based on the lateral flow immunoassay (LFIA) method, which have been used in clinical diagnostics for already 60 years. In some countries of the world, these diagnostic platforms are known as immunochromatographic tests. They are currently a relevant and promising alternative to the existing analytical instrument technologies. These devices are considered as simplified formats of modern biosensors, in which the recognition element is located on the surface of a porous membrane and result is visualized within a few minutes [1]–[4].
These rapid, inexpensive, reliable, and easy-to-use diagnostic platforms have proven their high efficiency in case of limited resources and lack of specially trained personnel. Currently, LFIA tests represent the most promising and dynamically developing segment of the market of rapid in vitro diagnostic tools, with an annual cumulative growth in global production of 7.7% [1],[2],[4],[5].
The growing popularity of these test systems for medical care or diagnostics in developing countries, medical institutions, emergency situations, as well as for individual use by patients monitoring their health at home are the major factors that contribute to the continuous development and improvement of this method and to the invention of new-generation formats [2],[6]–[8].
The principle of diagnostics based on lateral flow immunoassay (paper chromatography) was first proposed in 1959 by the biophysicist Rosalyn S. Yalow and the physician/endocrinologist Solomon A. Berson (Figure 1).
The first designed system using paraffin paper was a rapid test to determine insulin in human blood plasma [9]. The new principle, soon named LFIA, became a breakthrough technology not only in diagnosis of diabetes mellitus. The formats of new tests progressed rapidly, the paper was replaced by nitrocellulose, and soon the range of clinical laboratory diagnostics was extended by numerous test systems for determining other minor blood analytes (hormones, enzymes, vitamins, and markers of infectious process). As the technology developed, the range of its applications expanded to diagnostics of infectious diseases, cardiovascular diseases [10],[11], cancer biomarkers [12], food pathogens [13], and veterinary diagnostics [14].
Over the following 60 years, several variants of LFIA design were proposed that simplified the method and simultaneously made the test systems more sensitive and selective, affordable, and easy to handle. This allowed their use not only by laboratory staff, but also by other medical specialists and individually by patients for self-monitoring of their health [7],[15]–[17].
The invention of LFIA platforms was mediated by the development of patient-oriented technologies, the shift in the paradigm of patient care culture, and the increasing need for rapidly obtained laboratory information to make urgent decisions in emergency medicine, as well as by the recently introduced global concept of ‘point-of-care testing’ (testing at the site of care) [18]–[20].
To date, the classical microbiological and immunoserological methods, as well as modern diagnostic platforms such as enzyme immunoassay (EIA) and chemiluminescence assay, polymerase chain reaction (PCR), flow cytometry, and mass spectrometry (MALDI), have been used to accurately identify molecular markers. However, these diagnostic tools requiring expensive equipment, long testing time, and qualified personnel are not always available for small local hospitals, especially in conditions of limited budget and decentralized infrastructure of medical units [3],[11],[20],[21].
The potential of these recently introduced efficient technologies consists in the continuous development and improvement of the existing numerous LFIA-platforms, as well as in the creation of multiplex formats and complication of diagnostic goals (such as, e.g., cancer screening). Moreover, the absence of need for special temperature storage conditions contributes to the expansion of the range of their use in developing countries and in sparsely populated and remote regions [15],[16],[21],[22].
Over the decades of application, these test systems have passed the test of time and confirmed their wide availability, high speed of detection, ease of operation and readout of results, and efficient and reliable diagnosis of diseases [21],[23]–[25]. The cost efficiency and easy-to-handle property of these portable diagnostic systems are fully consistent with the world's modern concept of ‘point-of-care testing’ (laboratory testing at the site of treatment). For the above reasons, LFIA have not lost their value today [23]–[25].
Depending on the recognition elements used, LFIA platforms are divided into different types and design formats. Among the formats of this diagnostic strategy are qualitative, semi-quantitative, and quantitative test systems for identifying specific antigens [26],[27], antibodies [28],[29], and fragments of nucleic acids (amplicons) which can be formed during a polymerase chain reaction [12],[30].
The principle of LFIA is simple. A typical test system consists of a plastic base (substrate) coated with overlapping layers of porous membranes containing recognition molecules to interact with the target molecule (Figure 2).
Porous membranes are one of the most important elements of a test system, predominantly made of nitrocellulose. The key parameters that characterize properties of this material are the capillary forces and the ease of binding and subsequent immobilization of proteins involved in further reactions. The pore size of the membranes is from 0.05 to 12 µm, which provides the required rate, time, and uniformity of capillary flow, the most important characteristics determining the quality of test systems [3],[31]–[34].
A liquid sample (biosubstrates) to be analyzed is placed on a sample pad impregnated with a buffer solution, proteins, and surfactants (Figure 2). This part of the test system performs several important functions: even distribution of sample and direction of its movement to the conjugate at a certain rate. Furthermore, the pad acts as a filter to remove unwanted elements of biosubstrates such as red blood cells [10],[35]–[37].
Then the sample is moved by capillary forces along the strip to the pad to release the conjugate containing specific antibodies. The quality of specific antibodies used in the systems and their purification is the most important condition for optimal performance of tests. In LFIA, monoclonal antibodies derived from a hybrid mouse cell line, bound to stained or fluorescent particles (labels), are typically used [2],[32],[38]–[40].
The main requirements for the materials used in LFIA test systems as labels (the list of which is quite wide: they are colloidal gold nanoparticles, colored latex, magnetic and carbon nanoparticles, quantum dots, phosphors, fluorophores, enzymes, etc.) are high stability and low cost.
In addition, the tagging material must be found in very low concentrations and must retain its properties when bound to biorecognition molecules.
The materials listed above fully comply with these requirements; in addition, latex can be made in various colors and used in this form in multiplex systems [3],[13],[41]–[43].
Then the conjugated antibodies bind to the target analyte and migrate to the recognition zone. This part of the strip contains biological components that react with the formed analyte–antibody complex, which is manifested as a colored line in the test zone, while the line in the control zone indicates the correct flow of the substrate [32],[34],[36]. The color intensity of the test line, which is proportional to the analyte content in the sample, is evaluated visually or using special equipment (reader) [44].
To maintain the capillary effect, a cellulose absorbent pad is placed at the distal end of the test strip, which is necessary to remove excess reagents and prevent the reverse flow of the liquid. The application of this pad allows use of a large sample size, thus, increasing the sensitivity of the test [35],[38],[40].
Among other factors that affect test's sensitivity and specificity are chemicals present in biosubstrates that can bind to the system's components and mediate false positives. Sensitivity of a test system is limited by the constant of dissociation of the antibody–antigen conjugate and by colorimetric detection [36],[39]. Therefore, the modern strategies implemented by manufacturers to overcome these limitations are aimed at improving the characteristics of labels used (fluorescent, paramagnetic), which cannot be detected visually but require special devices, i.e. readers, for quantitative analysis [21],[29],[36]. Furthermore, automated detection reduces time expenditures and improves interpretation of results [18],[40].
The modern multiplex LFIA formats are capable of detecting several analytes at a time in a single sample (for example, test systems for detecting narcotic drugs in urine). In this case, the system includes one pad for applying a sample, several (according to the number of tested substances) pads to release conjugate, containing specific antibodies to each of the target analytes, and the same number of test zones to readout the result [21],[26],[29],[43].
Currently, these easy-to-use rapid tests are widely applied to confirm presence or absence of target analytes (e.g., antigens, antibodies, biochemical markers, or amplicons) in biological substrates (urine, serum or plasma, or whole blood) not only in human medicine, but also in environmental research, agriculture, and veterinary medicine [2]–[4],[12]–[14],[18]–[22]. In these fields, the above tests are used to verify pathogens, specific proteins, enzymes, detect chemicals, narcotic drugs, toxins, pollutants, and other substances [10],[14],[23]–[26].
Studies based on LFIA tests are performed at clinical laboratories, sites of medical care, or at home by medical staff or patients themselves. Use of a visual label in the form of gold or carbon nanoparticles or colored latex allows visual qualitative or quantitative (with special equipment available) testing within a few minutes [28],[32],[44],[45].
The assay is based on the antigen–antibody reaction, with biorecognition molecules such as aptamers (the artificial nucleic acids), nucleic acids, proteins or antiligands (specifically aimed at binding target analytes), molecular beacons (special DNA hairpin structure with fluorophore at one end and quencher at the other end), used as antibody, which allows detection of even minor concentrations in the presence of structurally bound molecules [33],[45],[46].
The technological flexibility of the LFIA formats provided creation of many types of test systems based on two key approaches that have gained the greatest popularity among specialists over the past decades:
-noncompetitive (direct) assay (sandwich format) is used to identify analytes having a high molecular weight (HMW) with several antigenic determinants (e.g., р24 antigen, a protein of the HIV nucleotide wall). In this design, result is positive if a color line, absent in case of negative result, appears in the test zone [19],[26],[29]. The most widely known examples of the sandwich format are pregnancy test systems that detect an elevated level of chorionic gonadotropin (from 10 to 25 mIU/mL, depending on sensitivity of tests) in woman's urine at an early pregnancy stage [47];
-competitive assay (inhibition format) is designed for analytes having a low molecular weight (LMW) with a single antigenic site. In this test format, the target analyte blocks the binding sites of antibodies located on the test line, preventing their interaction with the conjugate. In this design, therefore, conclusion is positive in the absence of color line and negative when a color line of any intensity appears in the test zone [48]. The typical examples of this format are drug and toxin test systems.
Each of these formats has advantages and disadvantages depending on analytes being tested, ranges of their critical concentrations, and design of the test system used. The sandwich format usually shows a higher analytical sensitivity (picograms of analyte per 1 mL) compared to competitive test systems (nanograms per 1 mL) [26].
However, at a high concentration of analyzed substance, sandwich systems may show a false negative result associated with the ‘high-dose effect’. The test systems based on the competitive format lack this drawback [47],[48].
Since the 1960s, LFIA test systems have held a firm place in the range of medical diagnostic methods as the most rapid, cost-effective, and simplest tools. Their efficiency has increased significantly with the development of readout technologies and the invention of devices providing transduction of color intensity of indicator lines into a semi-quantitative and quantitative result. In the former case, the result is shown as low, medium, or high; in the latter case, numerical values of analyte concentration depending on the density of test line are displayed [32],[35]–[39].
An example of the quantitative LFIA format is the Seralite-FLC test system for detecting free kappa-(κ-) and lambda- (λ-) chains in blood serum, which displays a numerical value of analyte content in mg/L, as well as the κ/λ ratio, within 10 min [37],[49]. This test system is designed on the basis of highly specific monoclonal antibodies against κ and λ, showing no cross-reactivity with other specific blood proteins [49].
Detection and control of infectious diseases is a serious healthcare issue. LFIA testing can be successfully used in diagnostics of infectious diseases [6],[16]–[18],[22],[32].
One of the key trends of development of the versatile LFIA technology is the designing of test systems in a multiplex (multi-purpose) format, which allows detection of several bacterial or viral targets at a time in a single test. This technology is a novel diagnostic approach providing wide opportunities for verification of causative agents [6],[8],[17],[32],[50] (Table 1).
Infectious agent | Detectable marker of infectious contamination (target analyte) | Format of test systems LFIA | Refs |
Dengue virus | Dengue non-structural protein 1 (NS1) | Magneto-enzyme | [51],[52] |
Zika virus | Zika virus nonstructural protein 1 (NS1) | Smartphone-based fluorescent | [53] |
Chikungunya virus | SD Bioline, IgM OnSite, IgM |
Chromatographic Chromatographic |
[54] |
Yellow fever (YF) virus | YF non-structural protein 1 (NS1) | Chromatographic | [55] |
Ebola virus | Antigen Ebola virus ReEBOV VP40 | Magneto-enzyme | [56] |
Dengue virus | Ig G/ IgM | Multiplex | [42] |
Yellow fever (YF) virus | Ig G/ IgM | Multiplex | [42] |
Ebola virus | Ig G/ IgM | Multiplex | [42] |
Human immunodeficiency virus (HIV) | Anti-HIV IgG | Multiplex | [41] |
Hepatitis C virus (HCV) | Anti-HCV IgG | Multiplex | [41] |
Hepatitis B virus (HBV) | Hepatitis B e-antigen (HBeAg) | Monoplex | [57] |
Human Immunodeficiency Virus (HIV-1) | HIV-1 p24 antigen | Monoplex | [58],[59] |
Foot-and-mouth disease virus | Antigen detection for all 7 serotypes for types O, A, C and Asia1 | Multiplex | [60] |
Respiratory viruses | Human adenovirus, influenza A H1N1 virus | Magnetic SERS-based LFIA (FeO @ Ag) | [61] |
Newcastle disease virus (NDV) | Amplification (RPA)-nucleic acid lateral flow (NALF) immunoassay | Multiplex RPA-NALF | [62] |
Infectious bronchitis virus (IBV) | Amplification (RPA)-nucleic acid lateral flow (NALF) immunoassay | Multiplex RPA-NALF | [62] |
Human Polyomavirus BK (BKV) | DNA BKV | Monoplex sandwich-type | [63] |
Bordetella pertussis | Anti-toxin pertussis IgG | Fluorescent Eu-nanoparticle reporters | [64] |
Staphylococcus aureus | Staphylococcal enterotoxin B | SERS-based lateral flow immunoassay | [65] |
Streptococcus pyogenes, Group A | S. pyogenes (A) | SERS-based lateral flow immunoassay | [66] |
Yersinia pestis | F1 capsular antigen | Monoplex | [67] |
Escherichia coli O157: H7 | E. coli O157: H7 | Sandwich models | [68] |
Listeria monocytogenes | L. monocytogenes | SERS-based lateral flow immunoassay | [69] |
Salmonella typhimurium | S. typhimurium | SERS-based lateral flow immunoassay | [69] |
Helicobacter pylori (HpSA-test) | Antigen H. pylori HpSA | Monoplex | [70] |
The first commercial combined LFIA test system was successfully demonstrated by C.S. Jørgensen with co-authors in 2015 [25]. It allowed efficient detection of Streptococcus pneumoniae and Legionella pneumophila antigens in the urine. This even more increased the popularity of the versatile LFIA technology, which, in the sandwich assay format, is equally efficient for both HMW antigens of microorganisms and antibodies to them in biosubstrates and for LMW analytes [8],[16],[23],[24].
Today, test systems based on the immunoassay technology, in both the standard and multiplex formats, represent a major segment of the market of laboratory-based rapid diagnostics [18],[23],[24],[32]. Many of them are successfully used within the framework of the ‘point-of-care testing’ (POC) program [3],[8],[11],[15],[71].
Result of immunochromatographic tests can be verified with the naked eye. In addition, these tests have advantage such as low cost of operation, easy handling, and operation without additional equipment [23],[27],[28],[32].
When molecular markers of an infectious process are detected using LFIA, this usually requires confirmation by an independent method. Therefore, immunoassay technologies are suitable mostly for primary screening [6],[8],[17],[72]. In addition, despite the obvious attractiveness of the LFIA formats, their limitations a great while held back the expansion of the practical use of these diagnostic platforms for clinical laboratory diagnostics (Table 2).
Advantage | Refs | Limitations | Refs |
|
[3],[27],[40] |
|
[27],[29],[40],[62] |
|
[23],[32],[40] |
|
[23],[28],[32],[47] |
|
[12],[31],[37] |
|
[10],[29],[37],[42] |
|
[3],[18],[28],[32] |
|
[3],[13],[32],[54] |
|
[4],[10],[13] |
|
[4],[28],[51],[55] |
|
[13],[24],[26] |
|
[13],[33],[52],[59] |
|
[12],[25],[33],[35] |
|
[12],[23],[42],[58] |
|
[23],[28],[37],[38] |
|
[23],[28],[47],[62] |
However, modern platforms of immunochromatographic systems, devoid of many drawbacks, have successfully proven themselves not only in medicine, but also in veterinary and environmental research. In addition, the high sensitivity, safety, and ease of use of LFIA test kits are critical in the elimination of epidemic outbreaks of dangerous infections.
R. Nouvellet et al. [73] give an example of the successful application of chromatographic analysis in the elimination of the Ebola epidemic in 2014–2015 in West Africa. At the initial stage of the epidemic, according to the WHO recommendation, the diagnosis of Ebola was based solely on the results of reverse transcriptional polymerase chain reaction (RT-PCR), which detected viral RNA in serum or plasma. However, these diagnostics was slow and expensive (2–6 hours at a cost of US 100$), and blood storage required maintaining a cold chain, which was problematic in Africa. The extreme nature of the epidemic has prompted WHO to call for fast and inexpensive test systems that do not require special instruments and equipment. Such test systems are chromatographic strips for the detection of the Ebola antigen ReEBOV VP40. Ultimately, it was recognized that rapid and accurate diagnostics was critical to successfully containing and eliminating the Ebola outbreak [73].
Further research is currently conducted to address some of the major drawbacks of LFIA test systems, especially as regards obtaining quantitative results and documenting them. They can be digitized using scanners or cameras with special software that would allow also recording the result and transmitting it at a distance. However, technological improvements will require more sophisticated hardware and will eventually increase the cost and duration of analysis.
Analyzing the advantages and limitations of LFIA technology, many authors for many years have pointed out a significant drawback (‘key failure’) of these diagnostic tools–visual assessment and qualitative conclusion (yes/no), which limits the objectivity and informational value of these analyzes [6],[74].
Advanced strategies of the development of LFIA testing technologies are capable of providing reliable quantitative information about the content of the target analyte in biosubstrates. In 2019, a fairly detailed reviews on this topic was published [6],[74],[75], so the author will limit himself to only a conceptual discussion of these modern approaches.
Traditionally, LFIA tests were considered as diagnostic tools for the qualitative analysis of the presence (or absence) of the desired analyte (marker) or its content exceeding a certain threshold. The result was assessed visually by comparing the staining of the test area with the control area [76].
The need to improve immunochromatographic technologies was associated with an increase in the relevance of a quantitative assessment of the content of markers of the pathological process during dynamic monitoring of the effectiveness of therapy, the patient's condition or the environment. At the same time, the inclusion of instrumental registration in LFIA tests did not make them more complicated.
In recent years, technological designs of various detecting devices have been proposed for quantitative assessment of immunochromatography, based on various principles of detection, ranging from the first reflectometric instruments to modern portable digital cameras [75],[76]. These detectors allow not only to quantitatively analyze biological samples, but also to store and transmit on-line results for a remote objective conclusion [74],[77],[78].
An important advantage of the LFIA technologies that are currently being actively developed are sensor and array-based platforms. The specialized quantitative analysis tools LFIA tests are divided according to the principles of recording the result. Thus, in the last decade, commercial models of open and closed detectors have appeared, based on optical data processing, which allow reading not only color stripes, but also fluorescent labels [6],[76],[79] (Table 3). In addition, the advent of related software products for smartphones allows these individual mobile devices to be used to obtain rapid quantitative results without the use of additional equipment [75],[80]. The use of mobile device platforms in quantitative LFIA formats to accurately record test results increases the efficiency of the clinic, epidemiological surveillance and disease control at the system level, and ultimately reduces the time it takes to initiate specialized treatment. In cases of suspected bacterial or viral infections, the use of mobile devices in LFIA testing allows for the rapid detection of isolated cases and a timely public health response.
For example, A. Nsabimana and colleagues in a recent study assessed the feasibility and effectiveness of this smart technology LFIA platforms for recording quantitative HIV test results in 2,190 patients in urban and rural Rwanda (East Africa) in 3 hospitals.
In recent years, with the emergence advent of related software products for smartphones allows these personal mobile devices to be used to obtain rapid quantitative results without the use of additional equipment [75],[80]. The use of mobile device in quantitative LFIA formats to accurately record test results increases the efficiency of the clinic, epidemiological surveillance and disease control at the system level, and ultimately reduces the time it takes to initiate specialized treatment. In cases of suspected bacterial or viral infections, the use of mobile devices in LFIA testing allows for the rapid detection of isolated cases and a timely and timely public health response.
For example, A. Nsabimana and colleagues [81] in a recent study assessed the feasibility and effectiveness of this smart technology LFIA platforms for recording quantitative HIV test results in 2,190 patients in urban and rural Rwanda (East Africa) in 3 hospitals.
Detection platforms principle | Company/Country | Detector Model | Mode of Measurements | Company website |
Optical | Axxin/Australia | AXXIN AX-2X | Colorimetry, fluorimetry | axxin.com |
Bio-AMD/United Kingdom | Digital Strip Reader | Colorimetry | bioamd.com | |
BioAssay Works/United States | Cube-Reader | Colorimetry | bioassayworks.com | |
Hamamatsu/Japan | Immunochromato-Reader C11787 | Colorimetry, fluorimetry | hamamatsu.com | |
Magnetic Labels (Magneto-enzyme) | Magna BioSciences/United States | MICT® Bench-Top System | Open System | magnabiosciences.com |
Magnasense Technologies/Finland | Magnasense's Magnetometric Reader | Open System | magnasense.com | |
VWR International/United States | FoodChek™ MICT System | Closed System | vwr.com |
As an alternative to mobile devices, it is proposed to use standard office scanners, which are already actively used in the quantitative analysis of the results of electrophoresis of protein fractions of blood [78],[82].
In modern formats of immunochromatographic test systems, magnetic particles are used as labels. Their registration in a magnetic field is not affected by the color of the bioassay or the colored components of the substrates. Magnetic recording detectors for quantitative sidestream analysis are more efficient and sensitive and have proven themselves in the commercial market [51],[61] (Table 4).
Detecting Device and Construction | Target Analytes | Range of Concentration Measured | Refs |
Dual LFIA with iPhone 5s | Salmonella enteritidis | 20–107 CFU/mL | [7] |
E. coli O157:H7 | 34–107 CFU/mL | ||
UC-LFS platform | Brain natriuretic peptide | 5–100 pg/mL | [31] |
Suppression of tumorigenicity 2 | 1–25 ng/mL | ||
Smartphone's ambient-light-sensor-based reader (SPALS-reader) | |||
Cadmium ion | 0.16–50 ng/mL | [35] | |
Clenbuterol | 0.046–1 ng/mL | ||
Porcine epidemic diarrhea virus | 0.055–20 µg/mL | ||
Electrochemical detection | Human chorionic gonadotrophin (HCG) | 25–50 mIU HCG in serum | [82] |
Magneto-enzyme | Dengue virus non-structural protein 1 (NS1) | 0, 1–0,25 ng/ml | [51] |
Magnetic SERS (Raman scattering-based LFIA) | Human adenovirus, | from 50 PFU/mL | [61] |
Influenza A H1N1 virus | from 10 PFU/mL | ||
Raman scattering-based LFIA (SERS-LFIA) | Streptococcus pyogenes, Group A | 0,2–100 KOE/mL | [66] |
Listeria monocytogenes | 102–107 KOE/mL | [69] |
Electrically conductive metal nanoparticles or oxidizing enzymes can be used as markers in the design of modern test strips. The principle of signal measurement in such systems is based on fluctuations in current, voltage or resistance (amperometry, potentiometry or conductometry) [82].
And, finally, surface enhanced Raman spectroscopy (SERS) is increasingly used in modern models of quantitative detection of LFIA to increase the sensitivity of the immunochromatographic method [61],[65],[69]. These formats allowing the detection of specific biomarkers of infections (antibodies, peptides, or DNA) conjugated with gold nanoparticles [65],[69].
Modern SERS platforms for sandwich immunoassay using paper test strips have proven to be in great demand for the quantitative detection of infectious diseases and, in particular, viral infections, including COVID-19.
At the end of 2019, a new coronavirus infection COVID-19 appeared in China, the subsequent spread of which around the world assumed the character of a pandemic [83]–[85]. In this regard, the creation of platforms for the effective diagnosis of this disease has become especially relevant for public health [84],[86]–[88].
It is known that COVID-19 belongs to the Coronaviridae family of RNA-containing coronaviruses that cause acute respiratory infections in humans of varying severity (from asymptomatic or mild to severe pneumonia) [84],[86],[87]. In this case, the main pathogenetic targets are the respiratory system, the gastrointestinal tract, the liver and the central nervous system, the defeat of which has become a characteristic feature and others coronavirus infections (Middle East Respiratory Syndrome, MERS, and Severe Acute Respiratory Syndrome, SARS) [85],[88],[89].
Before the emergence in the XXI century of epidemic outbreaks of infections (SARS in 2002–2004 and MERS in 2012 caused by β-coronaviruses MERS-CoV [80],[81],[84] and SARS-CoV [86],[87], these pathogens were not considered highly pathogenic to humans. They circulated in the human population, causing only sporadic cases of diseases that occur, as a rule, in a mild form [85],[88]. Epidemic outbreaks of SARS and MERS, and especially COVID-19, have changed the current view of the pathogenicity of coronaviruses and the epidemiology of new infections, as well as the potential perspective of emergence new outbreaks [86],[89]–[91].
The etiological cause of the 2019 pandemic was the new SARS-CoV-2 virus (Severe acute respiratory syndrome coronavirus 2), which in half a year caused hundreds of thousands of deaths and caused disease in millions of the world's inhabitants, caused chaos and a drop in the level of the world economy, and in the world community–fear and anxiety for the future of mankind. According to a WHO report, as of July 1, 2020, the number of COVID-19 cases in the world exceeded 10 million, and the quantity of deaths was over 500 thousand [90].
Like other β-coronaviruses, the SARS-CoV-2 gene contains specific RNA sequences encoding 27 proteins. Among them, 15 non-structural proteins that provide virus replication [87],[92]–[94] and 12 structural proteins are distinguished. For SARS-CoV-2, antibodies have been detected that recognize three of the four SARS-CoV-2 proteins exposed on the surface of the viral capsid: the nucleocapsid (N), envelope (E), and spike (S) proteins [95].
One of the most important directions in the strategy of combating a new infection has become the need for mass laboratory screening of populations at high risk of infection. The need for timely and high-quality laboratory diagnosis of patients infected with SARS-CoV-2 has become the main priority in eliminating the pandemic and introducing quarantine measures [87],[90],[96],[97].
In these conditions, the creation of quick, effective and inexpensive tools for the diagnosis of COVID-19, on the one hand, has become an essential component of the fight against a new infection, and on the other, it has been focused on the experience of eliminating previous coronavirus infections SARS and MERS [86],[88],[91],[92].
However, both in cases of elimination of SARS and MERS infections, and in the diagnosis of COVID-19, public health faces the same task. It is associated with determining the role and place of various diagnostic platforms for screening, diagnosis and monitoring of new coronavirus infections: RT-PCR, RT-LAMP, ELISA and LFIA, taking into account the advantages [87],[89],[91],[98].
The isolation of the pathogen culture in its pure form, which is the gold standard for the diagnosis of viral infections, is a laborious and lengthy process associated with working in a special laboratory that has the appropriate permission to work with biologically dangerous pathogens [94],[98].
Molecular testing of real-time polymerase chain reaction (RT-PCR) coronavirus infections, which is widely used in diagnostic virology, is a highly sensitive and recognized biomedical method [29],[87],[92],[98]. However, the full cycle of the study takes about 2 hours, sample preparation/extraction is required which takes 2–3 hours, and it is necessary special equipment, as well as trained specialists in laboratory diagnostics, which limits its use in COVID-19, despite the fact that PCR diagnostics are used throughout the world during a pandemic to detect SARS-CoV-2 [89],[93],[97].
The main disadvantages of this method in the diagnosis of infections are derived from the principle of the RT-PCR method, which provides for the detection of coronavirus RNA of only a specific type (SARS-CoV-2) and at a certain period of the disease [80],[87],[92]. Therefore, the results may be negative when examining convalescing patients cleared of the pathogen, as well as in patients with other infections [88],[98]. In addition, a false-negative result may be due to the uneven distribution of the virus in the respiratory system (sputum, nasopharyngeal secretion, pharynx), as well as (which seems inevitable in conditions of a huge flow of biological samples) ignoring the standards for sample collection [29],[86],[93],[98].
Recently, for screening and monitoring studies in coronavirus infections, the faster (30 mins–1 h) and economical molecular method of isothermal amplification of nucleic acids (LAMP and RT-LAMP) has become increasingly popular [34],[89],[99]–[101]. This diagnostic technology involves the use of simple-to-operate equipment with the ability to visually evaluate the result [69],[82]. This diagnostic method was proposed in 2000 and managed to prove itself in epidemics of SARS and MERS infections [69],[79],[80], as well as during the COVID-19 pandemic as a screening tool for remote regions and rural hospitals [86],[88].
The disadvantages of LAMP testing are the researchers' lack of sufficient experience in using the method in the conditions of epidemic outbreaks and emergency situations associated with SARS, MERS and the COVID-19 pandemic, as well as of the clinical interpretation of the results [34],[89],[95]. Like RT-PCR, RT-LAMP only detects the presence of viral genetic material and does not indicate the facts of previous infection and subsequent recovery of the patient [85],[86].
On the contrary, serological tests in the most common formats of enzyme-linked immunosorbent assay (ELISA) and of rapid tests LFIA, are widely known and have proven themselves as simple and cost-effective diagnostic platforms for coronavirus infections [83]–[89]. In contrast to molecular genetic diagnostic platforms, serological tests can detect not only viral contagion of patients, but also evaluate the response of the immune system and provide the necessary information for making organizational decisions [89],[91],[92],[96],[97].
Based on immuno-serological testing, these diagnostic technologies can provide faster and more detailed epidemiological information. For example, a full cycle of an ELISA study takes at least several hours to complete and requires special equipment in a clinical laboratory [83]–[86], while analysis using LFIA test systems requires 10 to 20 minutes [93]–[96],[98]–[100].
In the context of the diagnosis of SARS, MERS, and COVID-19 infections, LFIA rapid tests were most often used to test for the presence of patient antibodies (IgG and IgM) or viral antigens [93],[95],[98],[100]. Thus, using these tests, it is possible to identify not only infected patients, but also conduct retrospective diagnostics for those patients who have suffered asymptomatic disease, have recovered and currently have a certain degree of immune defense [97],[99],[100].
For example, J. Wu et al. [88] performed a retrospective study of the dynamics of the appearance of antibodies to SARS-CoV-2, the time-dependent sensitivity of four LFIA test systems in patients, to diagnose the significance of rapid serological tests in the management of patients with COVID-19, the diagnosis of which was established by molecular testing (RT-PCR). It turned out that 3 weeks after the onset of symptoms of the disease, all tests revealed antibodies (IgM and IgG), and the sensitivity and specificity was 100%. Moreover, in patients with COVID-19 complicated by pneumonia, an earlier appearance of antibodies against SARS-CoV-2 was detected [88].
In another study, Z. Chen and colleagues [92] report the development and testing of a new LFIA system for the detection of anti-SARV-CoV-2 IgG antibodies in human serum. The design features of the new test system are associated with the use of lanthanide-doped polystyrene nanoparticles and the recombinant nucleocapsid phosphoprotein SARS-CoV-2, placed on a nitrocellulose membrane to capture specific antibodies. The whole analysis process takes 10 minutes [92].
In accordance with the LFIA technology, infectious markers can be detected in the blood (antibodies), saliva, and the upper respiratory tract secretion throughout the infection cycle (contamination, incubation period, high season, recovery) [94],[101]–[103]. Unlike molecular methods, the use of LFIA in-house rapid tests does not require special training of specialists and can be used for screening studies in the field, at railway stations, airports, at unequipped diagnostic points, and in rural hospitals [95],[97],[104]–[106].
Despite the fact that the use of individual LFIA test systems turned out to be more expensive compared to ELISA in practical applications for the diagnosis of SARS, MERS and COVID-19 infections, their use is justified by the clinical advantages of this diagnostic platform [97],[100],[107].
During the fight against the COVID-19 pandemic, many different diagnostic systems were developed and proposed based on the principles of side-stream immunoassay, which differ in sensitivity and specificity [98],[108]–[111]. Some of the proposed platforms passed the necessary expertise and received permission for use in clinical practice (Table 5). Reliable efficacy and high sensitivity in detecting SARS-CoV-2 specific antibodies using immunochromatographic test systems shows that LFIA technologies can be a useful diagnostic tool in addition to molecular methods for diagnosing COVID-19. The international experience of using serological tests based on lateral flow immunoassay to eliminate epidemic outbreaks of coronavirus infections has shown the importance and necessity of this diagnostic tool. The most rational use of LFIA is mass screening of the population from risk groups, as well as patients with asymptomatic form of the disease. All positive results must be verified by quantitative molecular genetic methods.
Country-developers and producer companies | Sensitivity/ specificity of the test-systems (%) | Description of the test system | Biosubstrates used for diagnosis, analysis time | Links |
US / China, Cellex Inc. | 93,8/95,6 | IgM/IgG is detected by SARS-CoV-2 protein nucelocapside | Serum, plasma or whole blood (K2-EDTA, sodium citrate), 20 min | [113] |
US, ChemBio | 92,7 (IgM) и 95,9 (IgG)/99,0 (IgM и IgG) | IgM/IgG is detected by SARS-CoV-2 protein nucelocapside | Finger or vein whole blood, serum and plasma (lithium heparin, K2-EDTA), 15 min | [114] |
US, Autobio Diagnostics Co. Ltd. (+ Hardy Diagnostics) | 95,7 (IgM) и 99,0 (IgG)/99,0 (IgM и IgG) | IgM/IgG is detected by SARS-CoV-2 antigens | Finger or vein whole blood, serum and plasma (heparin, K2-EDTA), 15 min | [115] |
US / China, Healgen Scientific LLC | 96,7 (IgG), 86,7 (IgM), 96,7 comb./98,0 (IgG), 99,0 (IgM), 97,0 comb. | IgM/IgG is detected by SARS-CoV-2 antigens | Finger or vein whole blood, serum and plasma (heparin, K2-EDTA), 10 min | [116] |
China, Hangzhou Biotest Biotech Co., Ltd | 92.5 (IgM), 91.56 (IgG)/98.1 (IgM), 99.52 (IgG) | IgM/IgG is detected by SARS-CoV-2 recombinant spike protein receptor binding domain | Finger or vein whole blood, serum and plasma (heparin, K2-EDTA), up to 20 min | [117] |
China, Biohit Healthcare (Heifei) Co. Ltd. | 33.0 (IgM, days 1–7), 56.6 (IgG days 8–14), 83.0 (IgM days 8–14), 96.2 (IgG days 15+), 97.7 (IgM days 15 +)/99.5 (IgM), 100.0 (IgG) | IgM/IgG is detected antibodies by SARS-CoV-2 recombinant N-protein antigen and mouse anti human IgM/IgG antibody | Samples for human serum, plasma or whole blood (heparin, K2-EDTA, and sodium citrate), 15 min | [118] |
China, Hangzhou Laihe Biotech Co., Ltd | 100,0 (IgM, 0–6 days), 85.7 (IgM, 7–14 days), 76,0 (IgG, 7–14 days), 99.25 (IgM, 14+ days), 98.5 (IgG, 14+ days) / 99.43 | IgM/IgG is detected by SARS-CoV-2 antibodies. The target antigen is the S1 region of the spike protein. | Samples for human serum, plasma or whole blood (heparin, K2-EDTA, and sodium citrate), 15 min | [119] |
US / China, Aytu Biosciences / Orient Gene Biotech | 87.9 (IgM) & 97.2 (IgG)/100,0 for IgG and IgM | IgM/IgG is detected antibodies by SARS-CoV-2 antigen. | Samples for human serum, plasma or whole blood, 10 min | [120] |
Note: * - according to the Center for Health Security at J. Hopkins University
Some of the above-discussed innovations are associated with variations in the nature of labels used, as well as with technical improvements in the quantitative format of conclusion made. Some of the new strategies are based on a combination of colloidal gold nanoparticles with enzyme (such as horseradish peroxidase), which causes a catalytic amplification of signal [50],[51]. Other methods of signal amplification (1000-fold or more), as well as increase in sensitivity of test systems, are associated with the use of laser detection (plasmon resonance, surface-enhanced Raman scattering), chemiluminescent or fluorescent labels [12],[30],[45],[65].
A noteworthy format of LFIA to detect increase in myoglobin concentration, based on a sandwich system, was proposed by К. Edwards with co-authors [43]. In the proposed system, immobilized antibodies conjugate with streptavidin and are detected with a specific fluorescent dye (sulforodamine B) encapsulated in liposomes, which facilitates signal generation [43].
A number of promising innovations for multiplexing of the LFIA technology have been successfully tested in recent years [21],[44],[47]. Thus, there are test systems that include colloidal gold nanoparticles and oligonucleotides for the simultaneous detection of antigens and antibodies [21],[44] and the use of two conjugate pads for the simultaneous detection of two proteins [44]. Furthermore, a combination of LFIA with electronic computing units provides a response in the format of ‘OR’ and ‘AND’ logic gates [47].
As biomedical applications expanded, the requirements for LFIA systems grew steadily. They primarily concerned the improvement of sensitivity, reproducibility and the possibility of multiplexing, as well as increasing the objectivity of the quantitative assessment of results, which has recently been associated with laboratory information systems.
Modern technological trends in the improvement of immunochromatographic test systems are associated with obtaining high-sensitivity results with low constant of variance (CV). Improvements in sensitivity would allow assay LFIA systems to be applied in areas where larger clinical immunoassay systems, and methodologies such as PCR, are considered the gold standards. It is known that PCR diagnostics is of key importance for the diagnosis of bacterial and viral infections, biomedical research, food safety assessment, and environmental monitoring. At the same time, there was an urgent need for a simple, fast and cost-effective method without the use of complex and expensive equipment and reagents that are not quite available in conventional laboratories to detect fragments of nucleic acids (NA). This has led to the creation of paper-based analytical platforms that combine highly sensitive molecular genetic technologies with the speed and convenience of lateral flow technologies.
Over the past 10–15 years, a significant number of platforms have been proposed that have been developed for the detection of HA fragments using lateral flow technology and PCR [121]–[125]. These methods have simplified PCR technology, eliminating electrophoresis for confirming the presence of nucleic acid after DNA amplification, and of purchase of expensive equipment for molecular genetic analysis [126]–[128].
The very idea of detecting DNA fragments using LFIA analytical tools is not new and was successfully implemented in practice at the end of the 20th century [129]. Today, this technology is actively used in many areas of biomedicine, such as veterinary diagnostics, food and environmental monitoring, and diagnostics of plant diseases [122],[124],[125].
In new technologies that combine the capabilities of PCR and the advantages of LFIA, an amplified double-stranded sequence that is specific to the target microorganism is captured on paper immunochromatographic strips in the antibody-antigen format. In this pair, the antibody is specific to the label (for example, to biotin or streptavidin) and the Antigen is a labeled amplicon. The use of nitrocellulose strip as an immunosorbent and analytical platform allows one-step, fast and inexpensive analyzes (123,126,127). NA detection is performed using primers with two different labels (for example, streptavidin + or biotin +), and reporters (for example, avidin-labeled gold nanoparticles) provide visualization with the naked eye.
The developed new formats that constructively combine PCR and LFIA technologies are presented in two types: Nucleic Acid Lateral Flow Immunoassay (NALFIA) and Nucleic Acid Lateral Flow (NALF). The fundamental difference between these formats lies in the method of NA determination: direct, using reporter oligonucleotide probes (NALF) or previously labeled NA with hapten labels (digoxigenin, fluorescein, biotin, using reporter antibodies or streptavidin. Accordingly, the new analytical platforms were called PCR-NALF and PCR-NALFIA [122],[124],[126],[127].
Thus, M. Jauset-Rubio with colleagues [130] are report on the development of a point-of-care PCR-NALF test for the direct detection of isothermally amplified DNA. The detection limit (1 × 10−11 M or 190 amol), which is equivalent to 8.67 × 105 DNA copies, while the entire study cycle (amplification and detection) lasted less than 15 minutes at 37 °C.
In another study, S. Pecchia & D. Da Lio [126] proposed a test system in the PCR-NALFIA format for the detection of Macrophomina phaseolina in different types of infected soils and for pathogen detection and identification in plant tissues.
Thus, the evolution of LFIA technology occurs both in the direction of improving the analytical performance of these diagnostic tools and the emergence of new platforms that combine the advantages of various methods [124],[126],[130]. The possibilities of quantitative detection, analysis multiplexing, as well as the emergence of modern LFIA formats combined with PCR, create unprecedented laboratory diagnostic opportunities for the development of the POC concept. The future perspective is possibly related to the opportunity of a complete replacement of the PCR method, which requires trained specialists and special equipment, with LFIA platforms with the function of recombinase polymerase amplification.
Over the 60-year history of application of immunoassay, diagnostic technologies have become an indispensable tool in medicine, veterinary, and ecology, firmly occupying the position of the most demanded and popular rapid tests that fully conform to the modern global concept of ‘point-of-care testing’ [15],[16],[27].
The principle of the lateral flow immunoassay method by R. Yalow and S. Berson has remained unchanged for the past decades, despite the numerous latest LFIA formats proposed to improve its sensitivity and specificity. Immunoassay now becomes increasingly widespread in the world's healthcare systems. Today, this does not necessarily mean substitution of centralized laboratories by ‘point-of-care testing’ technologies, as LFIA platforms occupy only certain position in diagnostics of emergency conditions and monitoring of patients' health.
The main advantages of the method–simplicity and availability combined with its high efficiency–have always been decisive when choosing between tools for diagnostic screening in conditions of limited budget and low access to well-equipped laboratories or medical units. However, the simplicity of LFIA platforms contradicts the complex goal of optimizing the method to make it more sensitive, multiplexed, and quantitative. Therefore, the modern strategies implemented by designers of the method focus on empirical selection of materials for membranes, purity of reagents (antibodies, buffer systems, blocking reagents), and design of test systems [50].
The latest innovations aimed at improving the analytical characteristics of the LFIA technology are interesting, promising, and can provide these platforms with additional advantages (e.g., integration into the ‘chip-based laboratory’ design) [66]. Nevertheless, most of them increase cost of test systems, their complexity, and, thus, reduce availability of these remarkable technologies, a property that has made them popular and attractive for the recent six decades.
[1] | European Commision (2014) Journalist Workshop on organ donation and transplantation, Recent facts and figures. |
[2] |
Weiner DE (2009) Public health consequences of chronic kidney disease. Clin Pharmacol Ther 86: 566–569. doi: 10.1038/clpt.2009.137
![]() |
[3] |
Hesuani Y, Pereira FDAS, Parfenov V, et al. (2016) Design and Implementation of Novel Multifunctional 3D Bioprinter. 3D Print Addit Manuf 3: 64. doi: 10.1089/3dp.2015.0040
![]() |
[4] |
Colaco M, Igel DA, Atala A (2018) The potential of 3D printing in urological research and patient care. Nat Rev Urol 15: 213–221. doi: 10.1038/nrurol.2018.6
![]() |
[5] |
Cui X, Breitenkamp K, Lotz M, et al. (2012) Synergistic action of fibroblast growth factor-2 and transforming growth factor-beta1 enhances bioprinted human neocartilage formation. Biotechnol Bioeng 109: 2357–2368. doi: 10.1002/bit.24488
![]() |
[6] |
Murphy SV, Atala A (2014) 3D bioprinting of tissues and organs. Nat Biotechnol 32: 773–785. doi: 10.1038/nbt.2958
![]() |
[7] |
Skardal A, Atala A (2015) Biomaterials for integration with 3-D bioprinting. Ann Biomed Eng 43: 730–746. doi: 10.1007/s10439-014-1207-1
![]() |
[8] |
Bulanova EA, Koudan EV, Degosserie J, et al. (2017) Bioprinting of functional vascularized mouse thyroid gland construct. Biofabrication 9: 034105. doi: 10.1088/1758-5090/aa7fdd
![]() |
[9] | Rak-Raszewska A, Hauser PV, Vainio S (2015) Organ In Vitro Culture: What Have We Learned about Early Kidney Development? Stem Cells Int 2015: 959807. |
[10] |
Grobstein C (1953) Inductive epitheliomesenchymal interaction in cultured organ rudiments of the mouse. Sci 118: 52–55. doi: 10.1126/science.118.3054.52
![]() |
[11] |
Grobstein C (1956) Inductive tissue interaction in development. Adv Cancer Res 4: 187–236. doi: 10.1016/S0065-230X(08)60725-3
![]() |
[12] |
Saxen L, Wartiovaara J (1966) Cell contact and cell adhesion during tissue organization. Int J Cancer 1: 271–290. doi: 10.1002/ijc.2910010307
![]() |
[13] |
Herrera M, Mirotsou M (2014) Stem cells: potential and challenges for kidney repair. Am J Physiol Renal Physiol 306: F12–23. doi: 10.1152/ajprenal.00238.2013
![]() |
[14] |
Auerbach R, Grobstein C (1958) Inductive interaction of embryonic tissues after dissociation and reaggregation. Exp Cell Res 15: 384–397. doi: 10.1016/0014-4827(58)90039-9
![]() |
[15] |
Unbekandt M, Davies JA (2010) Dissociation of embryonic kidneys followed by reaggregation allows the formation of renal tissues. Kidney Int 77: 407–416. doi: 10.1038/ki.2009.482
![]() |
[16] |
Junttila S, Saarela U, Halt K, et al. (2015) Functional genetic targeting of embryonic kidney progenitor cells ex vivo. J Am Soc Nephrol 26: 1126–1137. doi: 10.1681/ASN.2013060584
![]() |
[17] |
Lancaster MA, Knoblich JA (2014) Organogenesis in a dish: modeling development and disease using organoid technologies. Sci 345: 1247125. doi: 10.1126/science.1247125
![]() |
[18] |
Schutgens F, Verhaar MC, Rookmaaker MB (2016) Pluripotent stem cell-derived kidney organoids: An in vivo-like in vitro technology. Eur J Pharmacol 790: 12–20. doi: 10.1016/j.ejphar.2016.06.059
![]() |
[19] |
Xinaris C, Benedetti V, Rizzo P, et al. (2012) In vivo maturation of functional renal organoids formed from embryonic cell suspensions. J Am Soc Nephrol 23: 1857–1868. doi: 10.1681/ASN.2012050505
![]() |
[20] | Xinaris C, Benedetti V, Novelli R, et al. (2015) Functional Human Podocytes Generated in Organoids from Amniotic Fluid Stem Cells. J Am Soc Nephrol 27: 1400–1411. |
[21] |
Bantounas I, Ranjzad P, Tengku F, et al. (2018) Generation of Functioning Nephrons by Implanting Human Pluripotent Stem Cell-Derived Kidney Progenitors. Stem Cell Reports 10: 766–779. doi: 10.1016/j.stemcr.2018.01.008
![]() |
[22] |
Bertram JF, Douglas-Denton RN, Diouf B, et al. (2011) Human nephron number: implications for health and disease. Pediatr Nephrol 26: 1529–1533. doi: 10.1007/s00467-011-1843-8
![]() |
[23] | Ogunlade O, Connell JJ, Huang JL, et al. (2017) In vivo 3-dimensional photoacoustic imaging of the renal vasculature in preclinical rodent models. Am J Physiol Renal Physiol 314: F1145–1153. |
[24] |
Wilmer MJ, Saleem MA, Masereeuw R, et al. (2010) Novel conditionally immortalized human proximal tubule cell line expressing functional influx and efflux transporters. Cell Tissue Res 339: 449–457. doi: 10.1007/s00441-009-0882-y
![]() |
[25] | Saleem MA, O'Hare MJ, Reiser J, et al. (2002) A conditionally immortalized human podocyte cell line demonstrating nephrin and podocin expression. J Am Soc Nephrol 13: 630–638. |
[26] |
Rak-Raszewska A, Wilm B, Edgar D, et al. (2012) Development of embryonic stem cells in recombinant kidneys. Organog 8: 125–136. doi: 10.4161/org.22597
![]() |
[27] |
Takasato M, Er PX, Becroft M, et al. (2014) Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nat Cell Biol 16: 118–126. doi: 10.1038/ncb2894
![]() |
[28] |
Taguchi A, Kaku Y, Ohmori T, et al. (2014) Redefining the in vivo origin of metanephric nephron progenitors enables generation of complex kidney structures from pluripotent stem cells. Cell Stem Cell 14: 53–67. doi: 10.1016/j.stem.2013.11.010
![]() |
[29] |
Kang M, Han YM (2014) Differentiation of human pluripotent stem cells into nephron progenitor cells in a serum and feeder free system. PLoS One 9: e94888. doi: 10.1371/journal.pone.0094888
![]() |
[30] |
Toyohara T, Mae S, Sueta S, et al. (2015) Cell Therapy Using Human Induced Pluripotent Stem Cell-Derived Renal Progenitors Ameliorates Acute Kidney Injury in Mice. Stem Cells Transl Med 4: 980–992. doi: 10.5966/sctm.2014-0219
![]() |
[31] |
Takasato M, Er PX, Chiu HS, et al. (2015) Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nat 526: 564–568. doi: 10.1038/nature15695
![]() |
[32] |
Freedman BS, Brooks CR, Lam AQ, et al. (2015) Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat Commun 6: 8715. doi: 10.1038/ncomms9715
![]() |
[33] |
Morizane R, Lam AQ, Freedman BS, et al. (2015) Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nat Biotechnol 33: 1193–1200. doi: 10.1038/nbt.3392
![]() |
[34] | Morizane R, Bonventre JV (2017) Generation of nephron progenitor cells and kidney organoids from human pluripotent stem cells. Nat Protoc 12: 195–207. |
[35] |
Xia Y, Sancho-Martinez I, Nivet E, et al. (2014) The generation of kidney organoids by differentiation of human pluripotent cells to ureteric bud progenitor-like cells. Nat Protoc 9: 2693–2704. doi: 10.1038/nprot.2014.182
![]() |
[36] |
He Y, Yang F, Zhao H, et al. (2016) Research on the printability of hydrogels in 3D bioprinting. Sci Rep 6: 29977. doi: 10.1038/srep29977
![]() |
[37] |
Aljohani W, Ullah MW, Zhang X, et al. (2018) Bioprinting and its applications in tissue engineering and regenerative medicine. Int J Biol Macromol 107: 261–275. doi: 10.1016/j.ijbiomac.2017.08.171
![]() |
[38] |
Jose RR, Rodriguez MJ, Dixon TA, et al. (2016) Evolution of Bioinks and Additive Manufacturing Technologies for 3D Bioprinting. ACS Biomater Sci Eng 2: 1662–1678. doi: 10.1021/acsbiomaterials.6b00088
![]() |
[39] |
Garreta E, Oria R, Tarantino C, et al. (2017) Tissue engineering by decellularization and 3D bioprinting. Mater Today 20: 166–178. doi: 10.1016/j.mattod.2016.12.005
![]() |
[40] | Murphy SV, Skardal A, Atala A (2013) Evaluation of hydrogels for bio-printing applications. J Biomed Mater Res A 101: 272–284. |
[41] |
Cui H, Nowicki M, Fisher JP, et al. (2017) 3D Bioprinting for Organ Regeneration. Adv Healthc Mater 6: 1601118. doi: 10.1002/adhm.201601118
![]() |
[42] |
Peppas NA, Bures P, Leobandung W, et al. (2000) Hydrogels in pharmaceutical formulations. Eur J Pharm Biopharm 50: 27–46. doi: 10.1016/S0939-6411(00)00090-4
![]() |
[43] |
Drury JL, Mooney DJ (2003) Hydrogels for tissue engineering: scaffold design variables and applications. Biomater 24: 4337–4351. doi: 10.1016/S0142-9612(03)00340-5
![]() |
[44] |
Peloso A, Tamburrini R, Edgar L, et al. (2016) Extracellular matrix scaffolds as a platform for kidney regeneration. Eur J Pharmacol 790: 21–27. doi: 10.1016/j.ejphar.2016.07.038
![]() |
[45] |
Pati F, Jang J, Ha DH, et al. (2014) Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink. Nat Commun 5: 3935. doi: 10.1038/ncomms4935
![]() |
[46] |
Skardal A, Devarasetty M, Kang HW, et al. (2015) A hydrogel bioink toolkit for mimicking native tissue biochemical and mechanical properties in bioprinted tissue constructs. Acta Biomater 25: 24–34. doi: 10.1016/j.actbio.2015.07.030
![]() |
[47] |
Jang J, Kim TG, Kim BS, et al. (2016) Tailoring mechanical properties of decellularized extracellular matrix bioink by vitamin B2-induced photo-crosslinking. Acta Biomater 33: 88–95. doi: 10.1016/j.actbio.2016.01.013
![]() |
[48] |
Beamish JA, Chen E, Putnam AJ (2017) Engineered extracellular matrices with controlled mechanics modulate renal proximal tubular cell epithelialization. PLoS One 12: e0181085. doi: 10.1371/journal.pone.0181085
![]() |
[49] |
Chen WC, Lin HH, Tang MJ (2014) Regulation of proximal tubular cell differentiation and proliferation in primary culture by matrix stiffness and ECM components. Am J Physiol Renal Physiol 307: F695–707. doi: 10.1152/ajprenal.00684.2013
![]() |
[50] |
Homan KA, Kolesky DB, Skylar-Scott MA, et al. (2016) Bioprinting of 3D Convoluted Renal Proximal Tubules on Perfusable Chips. Sci Rep 6: 34845. doi: 10.1038/srep34845
![]() |
[51] |
Kleinman HK, Martin GR (2005) Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol 15: 378–386. doi: 10.1016/j.semcancer.2005.05.004
![]() |
[52] |
Moll S, Ebeling M, Weibel F, et al. (2013) Epithelial cells as active player in fibrosis: findings from an in vitro model. PLoS One 8: e56575. doi: 10.1371/journal.pone.0056575
![]() |
[53] |
Mu X, Zheng W, Xiao L, et al. (2013) Engineering a 3D vascular network in hydrogel for mimicking a nephron. Lab Chip 13: 1612–1618. doi: 10.1039/c3lc41342j
![]() |
[54] | King SM, Higgins JW, Nino CR, et al. (2017) 3D Proximal Tubule Tissues Recapitulate Key Aspects of Renal Physiology to Enable Nephrotoxicity Testing. Front Physiol 8: 123. |
[55] | Nguyen DG, King SM, Presnell SC (2016) Engineered Renal Tissues, Arrays Thereof, and Methods of Making the Same. |
[56] |
Matak D, Brodaczewska KK, Lipiec M, et al. (2017) Colony, hanging drop, and methylcellulose three dimensional hypoxic growth optimization of renal cell carcinoma cell lines. Cytotechnology 69: 565–578. doi: 10.1007/s10616-016-0063-2
![]() |
[57] |
Astashkina AI, Mann BK, Prestwich GD, et al. (2012) A 3-D organoid kidney culture model engineered for high-throughput nephrotoxicity assays. Biomaterials 33: 4700–4711. doi: 10.1016/j.biomaterials.2012.02.063
![]() |
[58] |
Astashkina AI, Mann BK, Prestwich GD, et al. (2012) Comparing predictive drug nephrotoxicity biomarkers in kidney 3-D primary organoid culture and immortalized cell lines. Biomaterials 33: 4712–4721. doi: 10.1016/j.biomaterials.2012.03.001
![]() |
[59] |
Ozbolat IT, Hospodiuk M (2016) Current advances and future perspectives in extrusion-based bioprinting. Biomaterials 76: 321–343. doi: 10.1016/j.biomaterials.2015.10.076
![]() |
[60] | Alconcel SNS, Baas AS, Maynard HD (2017) FDA-approved poly(ethylene glycol)–protein conjugate drugs. Polymer Chemistry 2: 1442–1448. |
[61] |
Holzl K, Lin S, Tytgat L, et al. (2016) Bioink properties before, during and after 3D bioprinting. Biofabrication 8: 032002. doi: 10.1088/1758-5090/8/3/032002
![]() |
[62] |
Hospodiuk M, Dey M, Sosnoski D, et al. (2017) The bioink: A comprehensive review on bioprintable materials. Biotechnol Adv 35: 217–239. doi: 10.1016/j.biotechadv.2016.12.006
![]() |
[63] |
Nguyen KT, West JL (2002) Photopolymerizable hydrogels for tissue engineering applications. Biomaterials 23: 4307–4314. doi: 10.1016/S0142-9612(02)00175-8
![]() |
[64] |
Arcaute K, Mann BK, Wicker RB (2006) Stereolithography of three-dimensional bioactive poly(ethylene glycol) constructs with encapsulated cells. Ann Biomed Eng 34: 1429–1441. doi: 10.1007/s10439-006-9156-y
![]() |
[65] |
Hockaday LA, Kang KH, Colangelo NW, et al. (2012) Rapid 3D printing of anatomically accurate and mechanically heterogeneous aortic valve hydrogel scaffolds. Biofabrication 4: 035005. doi: 10.1088/1758-5082/4/3/035005
![]() |
[66] |
He Y, Tuck CJ, Prina E, et al. (2017) A new photocrosslinkable polycaprolactone-based ink for three-dimensional inkjet printing. J Biomed Mater Res B Appl Biomater 105: 1645–1657. doi: 10.1002/jbm.b.33699
![]() |
[67] |
Hribar KC, Soman P, Warner J, et al. (2014) Light-assisted direct-write of 3D functional biomaterials. Lab Chip 14: 268–275. doi: 10.1039/C3LC50634G
![]() |
[68] |
Li WW, Li H, Liu ZF, et al. (2009) Determination of residual acrylamide in medical polyacrylamide hydrogel by high performance liquid chromatography tandem mass spectroscopy. Biomed Environ Sci 22: 28–31. doi: 10.1016/S0895-3988(09)60018-0
![]() |
[69] |
Rana D, Ramalingam M (2017) Enhanced proliferation of human bone marrow derived mesenchymal stem cells on tough hydrogel substrates. Mater Sci Eng C Mater Biol Appl 76: 1057–1065. doi: 10.1016/j.msec.2017.03.202
![]() |
[70] | Hron P, Slechtova J, Smetana K, et al. (1997) Silicone rubber-hydrogel composites as polymeric biomaterials. IX. Composites containing powdery polyacrylamide hydrogel. Biomaterials 18: 1069–1073. |
[71] | Xi TF, Fan CX, Feng XM, et al. (2006) Cytotoxicity and altered c-myc gene expression by medical polyacrylamide hydrogel. J Biomed Mater Res A 78: 283–290. |
[72] |
Xi L, Wang T, Zhao F, et al. (2014) Evaluation of an injectable thermosensitive hydrogel as drug delivery implant for ocular glaucoma surgery. PLoS One 9: e100632. doi: 10.1371/journal.pone.0100632
![]() |
[73] |
Dumortier G, Grossiord JL, Agnely F, et al. (2006) A review of poloxamer 407 pharmaceutical and pharmacological characteristics. Pharm Res 23: 2709–2728. doi: 10.1007/s11095-006-9104-4
![]() |
[74] |
Wu W, DeConinck A, Lewis JA (2011) Omnidirectional printing of 3D microvascular networks. Adv Mater 23: H178–83. doi: 10.1002/adma.201004625
![]() |
[75] | Ozbolat IT (2017) 3D Bioprinting: Fundamentals, principles and Applications, London: Academy Press. |
[76] |
Gómez-Guillén MC, Giménez B, López-Caballero ME, et al. (2011) Functional and bioactive properties of collagen and gelatin from alternative sources: A review. Food Hydrocolloids 25: 1813–1827. doi: 10.1016/j.foodhyd.2011.02.007
![]() |
[77] |
Kuijpers AJ, van Wachem PB, van Luyn MJ, et al. (2000) In vivo compatibility and degradation of crosslinked gelatin gels incorporated in knitted Dacron. J Biomed Mater Res 51: 136–145. doi: 10.1002/(SICI)1097-4636(200007)51:1<136::AID-JBM18>3.0.CO;2-W
![]() |
[78] |
Kolesky DB, Homan KA, Skylar-Scott MA, et al. (2016) Three-dimensional bioprinting of thick vascularized tissues. Proc Natl Acad Sci U S A 113: 3179–3184. doi: 10.1073/pnas.1521342113
![]() |
[79] |
McManus MC, Boland ED, Koo HP, et al. (2006) Mechanical properties of electrospun fibrinogen structures. Acta Biomater 2: 19–28. doi: 10.1016/j.actbio.2005.09.008
![]() |
[80] |
Cui X, Boland T (2009) Human microvasculature fabrication using thermal inkjet printing technology. Biomater 30: 6221–6227. doi: 10.1016/j.biomaterials.2009.07.056
![]() |
[81] |
Fussenegger M, Meinhart J, Hobling W, et al. (2003) Stabilized autologous fibrin-chondrocyte constructs for cartilage repair in vivo. Ann Plast Surg 51: 493–498. doi: 10.1097/01.sap.0000067726.32731.E1
![]() |
[82] |
Achilli M, Mantovani D (2010) Tailoring Mechanical Properties of Collagen-Based Scaffolds for Vascular Tissue Engineering: The Effects of pH, Temperature and Ionic Strength on Gelation. Polymers 2: 664–680. doi: 10.3390/polym2040664
![]() |
[83] |
Park JY, Choi JC, Shim JH, et al. (2014) A comparative study on collagen type I and hyaluronic acid dependent cell behavior for osteochondral tissue bioprinting. Biofabrication 6: 035004. doi: 10.1088/1758-5082/6/3/035004
![]() |
[84] |
Lee KY, Moone DJ (2012) Alginate: Properties and biomedical applications. Progress in Polymer Science 37: 106–126. doi: 10.1016/j.progpolymsci.2011.06.003
![]() |
[85] |
Cohen DL, Lo W, Tsavaris A, et al. (2011) Increased mixing improves hydrogel homogeneity and quality of three-dimensional printed constructs. Tissue Eng Part C Methods 17: 239–248. doi: 10.1089/ten.tec.2010.0093
![]() |
[86] |
Gudapati H, Dey M, Ozbolat I (2016) A comprehensive review on droplet-based bioprinting: Past, present and future. Biomaterials 102: 20–42. doi: 10.1016/j.biomaterials.2016.06.012
![]() |
[87] | Yan J, Huang Y, Chrisey DB (2013) Laser-assisted printing of alginate long tubes and annular constructs. Biofabrication 5: 015002. |
[88] |
Nguyen DG, Funk J, Robbins JB, et al. (2016) Bioprinted 3D Primary Liver Tissues Allow Assessment of Organ-Level Response to Clinical Drug Induced Toxicity In Vitro. PLoS One 11: e0158674. doi: 10.1371/journal.pone.0158674
![]() |
[89] |
Thirumala S, Gimble JM, Devireddy RV (2013) Methylcellulose based thermally reversible hydrogel system for tissue engineering applications. Cells 2: 460–475. doi: 10.3390/cells2030460
![]() |
[90] |
Lott JR, McAllister JW, Arvidson SA, et al. (2013) Fibrillar structure of methylcellulose hydrogels. Biomacromolecules 14: 2484–2488. doi: 10.1021/bm400694r
![]() |
[91] | Oxlund H, Andreassen TT (1980) The roles of hyaluronic acid, collagen and elastin in the mechanical properties of connective tissues. J Anat 131: 611–620. |
[92] |
Luo Y, Kirker KR, Prestwich GD (2000) Cross-linked hyaluronic acid hydrogel films: new biomaterials for drug delivery. J Control Release 69: 169–184. doi: 10.1016/S0168-3659(00)00300-X
![]() |
[93] |
Jeon O, Song SJ, Lee K, et al. (2007) Mechanical properties and degradation behaviors of hyaluronic acid hydrogels cross-linked at various cross-linking densities. Carbohydrate Polymers 70: 251–257. doi: 10.1016/j.carbpol.2007.04.002
![]() |
[94] |
Gruene M, Pflaum M, Hess C, et al. (2011) Laser printing of three-dimensional multicellular arrays for studies of cell-cell and cell-environment interactions. Tissue Eng Part C Methods 17: 973–982. doi: 10.1089/ten.tec.2011.0185
![]() |
[95] |
Chuah JKC, Zink D (2017) Stem cell-derived kidney cells and organoids: Recent breakthroughs and emerging applications. Biotechnol Adv 35: 150–167. doi: 10.1016/j.biotechadv.2016.12.001
![]() |
[96] |
Reint G, Rak-Raszewska A, Vainio SJ (2017) Kidney development and perspectives for organ engineering. Cell Tissue Res 369: 171–183. doi: 10.1007/s00441-017-2616-x
![]() |
[97] |
Ozbolat I, Khoda A (2014) Design of a New Parametric Path Plan for Additive Manufacturing of Hollow Porous Structures With Functionally Graded Materials. ASME J Comput Inf Sci Eng 14: 041005. doi: 10.1115/1.4028418
![]() |
[98] | Kamisuki S, Hagata T, Tezuka C, et al. (1998) A low power, small, electrostatically-driven commercial inkjet head: Proceedings MEMS 98. IEEE. Eleventh Annual International Workshop on Micro Electro Mechanical Systems. An Investigation of Micro Structures, Sensors, Actuators, Machines and Systems 1998 Jan 25–29, Heidelberg, Germany, pp. 63–68. |
[99] |
Gasperini L, Maniglio D, Motta A, et al. (2015) An electrohydrodynamic bioprinter for alginate hydrogels containing living cells. Tissue Eng Part C Methods 21: 123–132. doi: 10.1089/ten.tec.2014.0149
![]() |
[100] | Moghadam H, Samimi M, Samimi A, et al. (2010) Electrospray modeling of highly viscous and non-Newtonian liquids. Journal of Applied Polymer Science 118: 1288–1296. |
[101] |
Demirci U, Montesano G (2007) Single cell epitaxy by acoustic picolitre droplets. Lab Chip 7: 1139–1145. doi: 10.1039/b704965j
![]() |
[102] |
Khalil S, Nam J, Sun W (2005) Multi-nozzle deposition for construction of 3D biopolymer tissue scaffolds. Rapid Prototyp J 11: 9–17. doi: 10.1108/13552540510573347
![]() |
[103] |
Bajaj P, Schweller RM, Khademhosseini A, et al. (2014) 3D biofabrication strategies for tissue engineering and regenerative medicine. Annu Rev Biomed Eng 16: 247–276. doi: 10.1146/annurev-bioeng-071813-105155
![]() |
[104] |
Bhattacharjee N, Urrios A, Kang S, et al. (2016) The upcoming 3D-printing revolution in microfluidics. Lab Chip 16: 1720–1742. doi: 10.1039/C6LC00163G
![]() |
[105] |
Ovsianikov A, Muhleder S, Torgersen J, et al. (2014) Laser photofabrication of cell-containing hydrogel constructs. Langmuir 30: 3787–3794. doi: 10.1021/la402346z
![]() |
[106] |
Turunen S, Joki T, Hiltunen ML, et al. (2017) Direct Laser Writing of Tubular Microtowers for 3D Culture of Human Pluripotent Stem Cell-Derived Neuronal Cells. ACS Appl Mater Interfaces 9: 25717–25730. doi: 10.1021/acsami.7b05536
![]() |
[107] |
Odde DJ, Renn MJ (2000) Laser-guided direct writing of living cells. Biotechnol Bioeng 67: 312–318. doi: 10.1002/(SICI)1097-0290(20000205)67:3<312::AID-BIT7>3.0.CO;2-F
![]() |
[108] | Delaporte P, Alloncle A (2016) Laser-induced forward transfer: A high resolution additive manufacturing technology. Optics & Laser Technology 78: 33–41. |
[109] | Yan Y, Li S, Zhang R, et al. (2009) Rapid prototyping and manufacturing technology: principles, representative techniques, applications and development trends. Tsinghua Science & Technology 14: 1–12. |
[110] |
Zein I, Hutmacher DW, Tan KC, et al. (2002) Fused deposition modeling of novel scaffold architectures for tissue engineering applications. Biomaterials 23: 1169–1185. doi: 10.1016/S0142-9612(01)00232-0
![]() |
[111] | Boland T, Mironov V, Gutowska A, et al. (2003) Cell and organ printing 2: fusion of cell aggregates in three-dimensional gels. Anat Rec A Discov Mol Cell Evol Biol 272: 497–502. |
[112] | Bakhshinejad A, D'Souza R (2015) A brief comparison between available bio-printing methods. IEEE Great Lakes Biomedical Conference (GLBC), 2015 May 14–17, Milwaukee, WI, pp. 1–3. |
[113] | Wilson WC, Boland T (2003) Cell and organ printing 1: protein and cell printers. Anat Rec A Discov Mol Cell Evol Biol 272: 491–496. |
[114] |
Klebe RJ (1988) Cytoscribing: a method for micropositioning cells and the construction of two- and three-dimensional synthetic tissues. Exp Cell Res 179: 362–373. doi: 10.1016/0014-4827(88)90275-3
![]() |
[115] |
Boland T, Tao X, Damon B, et al. (2007) Drop-on-demand printing of cells and materials for designer tissue constructs. Mater Sci Eng C 27: 372–376. doi: 10.1016/j.msec.2006.05.047
![]() |
[116] |
Derby B (2008) Bioprinting: Inkjet printing protein and hybrid cell-containing materials and structures. J Mater Chem 18: 5717–5721. doi: 10.1039/b807560c
![]() |
[117] |
Choi YJ, Yi HG, Kim SW, et al. (2017) 3D Cell Printed Tissue Analogues: A New Platform for Theranostics. Theranostics 7: 3118–3137. doi: 10.7150/thno.19396
![]() |
[118] |
Dhariwala B, Hunt E, Boland T (2004) Rapid prototyping of tissue-engineering constructs, using photopolymerizable hydrogels and stereolithography. Tissue Eng 10: 1316–1322. doi: 10.1089/ten.2004.10.1316
![]() |
[119] |
Odde DJ, Renn MJ (1999) Laser-guided direct writing for applications in biotechnology. Trends Biotechnol 17: 385–389. doi: 10.1016/S0167-7799(99)01355-4
![]() |
[120] |
Ringeisen BR, Kim H, Barron JA, et al. (2004) Laser printing of pluripotent embryonal carcinoma cells. Tissue Eng 10: 483–491. doi: 10.1089/107632704323061843
![]() |
[121] |
Ovsianikov A, Schlie S, Ngezahayo A, et al. (2007) Two-photon polymerization technique for microfabrication of CAD-designed 3D scaffolds from commercially available photosensitive materials. J Tissue Eng Regen Med 1: 443–449. doi: 10.1002/term.57
![]() |
[122] |
Guillemot F, Souquet A, Catros S, et al. (2010) High-throughput laser printing of cells and biomaterials for tissue engineering. Acta Biomater 6: 2494–2500. doi: 10.1016/j.actbio.2009.09.029
![]() |
[123] |
Gittard SD, Narayan RJ (2010) Laser direct writing of micro- and nano-scale medical devices. Expert Rev Med Devices 7: 343–356. doi: 10.1586/erd.10.14
![]() |
[124] |
Jang J, Yi H, Cho D (2016) 3D Printed Tissue Models: Present and Future. ACS Biomater Sci Eng 2: 1722–1731. doi: 10.1021/acsbiomaterials.6b00129
![]() |
[125] |
Ozbolat IT, Yu Y (2013) Bioprinting toward organ fabrication: challenges and future trends. IEEE Trans Biomed Eng 60: 691–699. doi: 10.1109/TBME.2013.2243912
![]() |
[126] |
Mironov V, Visconti RP, Kasyanov V, et al. (2009) Organ printing: tissue spheroids as building blocks. Biomaterials 30: 2164–2174. doi: 10.1016/j.biomaterials.2008.12.084
![]() |
[127] |
Jakab K, Norotte C, Marga F, et al. (2010) Tissue engineering by self-assembly and bio-printing of living cells. Biofabrication 2: 022001. doi: 10.1088/1758-5082/2/2/022001
![]() |
[128] |
Chung JHY, Naficy S, Yue Z, et al. (2013) Bio-ink properties and printability for extrusion printing living cells. Biomater Sci 1: 763–773. doi: 10.1039/c3bm00012e
![]() |
[129] |
Levato R, Visser J, Planell JA, et al. (2014) Biofabrication of tissue constructs by 3D bioprinting of cell-laden microcarriers. Biofabrication 6: 035020. doi: 10.1088/1758-5082/6/3/035020
![]() |
[130] |
Pati F, Ha DH, Jang J, et al. (2015) Biomimetic 3D tissue printing for soft tissue regeneration. Biomaterials 62: 164–175. doi: 10.1016/j.biomaterials.2015.05.043
![]() |
[131] |
Kang HW, Lee SJ, Ko IK, et al. (2016) A 3D bioprinting system to produce human-scale tissue constructs with structural integrity. Nat Biotechnol 34: 312–319. doi: 10.1038/nbt.3413
![]() |
[132] |
Rouwkema J, Rivron NC, van Blitterswijk CA (2008) Vascularization in tissue engineering. Trends Biotechnol 26: 434–441. doi: 10.1016/j.tibtech.2008.04.009
![]() |
[133] |
Rouwkema J, Khademhosseini A (2016) Vascularization and Angiogenesis in Tissue Engineering: Beyond Creating Static Networks. Trends Biotechnol 34: 733–745. doi: 10.1016/j.tibtech.2016.03.002
![]() |
[134] |
Lovett M, Lee K, Edwards A, et al. (2009) Vascularization strategies for tissue engineering. Tissue Eng Part B Rev 15: 353–370. doi: 10.1089/ten.teb.2009.0085
![]() |
[135] | Costa-Almeida R, Granja PL, Soares R, et al. (2014) Cellular strategies to promote vascularisation in tissue engineering applications. Eur Cell Mater 28: 51–66; discussion 66–67. |
[136] |
Novosel EC, Kleinhans C, Kluger PJ (2011) Vascularization is the key challenge in tissue engineering. Adv Drug Deliv Rev 63: 300–311. doi: 10.1016/j.addr.2011.03.004
![]() |
[137] |
Zhang C, Hou J, Zheng S, et al. (2011) Vascularized atrial tissue patch cardiomyoplasty with omentopexy improves cardiac performance after myocardial infarction. Ann Thorac Surg 92: 1435–1442. doi: 10.1016/j.athoracsur.2011.05.054
![]() |
[138] |
Hammerman MR (2002) Xenotransplantation of developing kidneys. Am J Physiol Renal Physiol 283: F601–606. doi: 10.1152/ajprenal.00126.2002
![]() |
[139] |
Halt KJ, Parssinen HE, Junttila SM, et al. (2016) CD146(+) cells are essential for kidney vasculature development. Kidney Int 90: 311–324. doi: 10.1016/j.kint.2016.02.021
![]() |
[140] | Robert B, St John PL, Abrahamson DR (1998) Direct visualization of renal vascular morphogenesis in Flk1 heterozygous mutant mice. Am J Physiol 275: F164–172. |
[141] |
Gao X, Chen X, Taglienti M, et al. (2005) Angioblast-mesenchyme induction of early kidney development is mediated by Wt1 and Vegfa. Development 132: 5437–5449. doi: 10.1242/dev.02095
![]() |
[142] | Robert B, St John PL, Hyink DP, et al. (1996) Evidence that embryonic kidney cells expressing flk-1 are intrinsic, vasculogenic angioblasts. Am J Physiol 271: F744–753. |
[143] | Hyink DP, Tucker DC, St John PL, et al. (1996) Endogenous origin of glomerular endothelial and mesangial cells in grafts of embryonic kidneys. Am J Physiol 270: F886–899. |
[144] |
Loughna S, Hardman P, Landels E, et al. (1997) A molecular and genetic analysis of renalglomerular capillary development. Angiogenesis 1: 84–101. doi: 10.1023/A:1018357116559
![]() |
[145] | Rymer CC, Sims-Lucas S (2015) In utero intra-cardiac tomato-lectin injections on mouse embryos to gauge renal blood flow. J Vis Exp 2015: 52398 . |
[146] |
Serluca FC, Drummond IA, Fishman MC (2002) Endothelial signaling in kidney morphogenesis: a role for hemodynamic forces. Curr Biol 12: 492–497. doi: 10.1016/S0960-9822(02)00694-2
![]() |
[147] |
Bersini S, Moretti M (2015) 3D functional and perfusable microvascular networks for organotypic microfluidic models. J Mater Sci Mater Med 26: 180. doi: 10.1007/s10856-015-5520-5
![]() |
[148] |
Song JW, Bazou D, Munn LL (2015) Microfluidic model of angiogenic sprouting. Methods Mol Biol 1214: 243–254. doi: 10.1007/978-1-4939-1462-3_15
![]() |
[149] |
Jeon JS, Bersini S, Gilardi M, et al. (2015) Human 3D vascularized organotypic microfluidic assays to study breast cancer cell extravasation. Proc Natl Acad Sci U S A 112: 214–219. doi: 10.1073/pnas.1417115112
![]() |
[150] |
Miller JS, Stevens KR, Yang MT, et al. (2012) Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nat Mater 11: 768–774. doi: 10.1038/nmat3357
![]() |
[151] |
Kolesky DB, Truby RL, Gladman AS, et al. (2014) 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs. Adv Mater 26: 3124–3130. doi: 10.1002/adma.201305506
![]() |
[152] |
Huling J, Ko IK, Atala A, et al. (2016) Fabrication of biomimetic vascular scaffolds for 3D tissue constructs using vascular corrosion casts. Acta Biomater 32: 190–197. doi: 10.1016/j.actbio.2016.01.005
![]() |
[153] | Atala A (2004) Tissue engineering for the replacement of organ function in the genitourinary system. Am J Transplant 4 Suppl 6: 58–73. |
[154] | Cui T, Terlecki R, Atala A (2014) Tissue engineering in urethral reconstruction. Arch Esp Urol 67: 29–34. |
[155] |
Atala A (2011) Tissue engineering of human bladder. Br Med Bull 97: 81–104. doi: 10.1093/bmb/ldr003
![]() |
[156] |
de Kemp V, de Graaf P, Fledderus JO, et al. (2015) Tissue engineering for human urethral reconstruction: systematic review of recent literature. PLoS One 10: e0118653. doi: 10.1371/journal.pone.0118653
![]() |
[157] | Hirashima T, Hoshuyama M, Adachi T (2017) In vitro tubulogenesis of Madin-Darby canine kidney (MDCK) spheroids occurs depending on constituent cell number and scaffold gel concentration. J Theor Biol 435: 110–115. |
[158] |
Zhang K, Fu Q, Yoo J, et al. (2017) 3D bioprinting of urethra with PCL/PLCL blend and dual autologous cells in fibrin hydrogel: An in vitro evaluation of biomimetic mechanical property and cell growth environment. Acta Biomater 50: 154–164. doi: 10.1016/j.actbio.2016.12.008
![]() |
[159] |
Mironov V, Kasyanov V, Drake C, et al. (2008) Organ printing: promises and challenges. Regen Med 3: 93–103. doi: 10.2217/17460751.3.1.93
![]() |
[160] |
Visconti RP, Kasyanov V, Gentile C, et al. (2010) Towards organ printing: engineering an intra-organ branched vascular tree. Expert Opin Biol Ther 10: 409–420. doi: 10.1517/14712590903563352
![]() |
[161] |
Kelm JM, Lorber V, Snedeker JG, et al. (2010) A novel concept for scaffold-free vessel tissue engineering: self-assembly of microtissue building blocks. J Biotechnol 148: 46–55. doi: 10.1016/j.jbiotec.2010.03.002
![]() |
[162] |
Itoh M, Nakayama K, Noguchi R, et al. (2015) Scaffold-Free Tubular Tissues Created by a Bio-3D Printer Undergo Remodeling and Endothelialization when Implanted in Rat Aortae. PLoS One 10: e0136681. doi: 10.1371/journal.pone.0136681
![]() |
[163] |
Norotte C, Marga FS, Niklason LE, et al. (2009) Scaffold-free vascular tissue engineering using bioprinting. Biomaterials 30: 5910–5917. doi: 10.1016/j.biomaterials.2009.06.034
![]() |
[164] |
Gentile C, Fleming PA, Mironov V, et al. (2008) VEGF-mediated fusion in the generation of uniluminal vascular spheroids. Dev Dyn 237: 2918–2925. doi: 10.1002/dvdy.21720
![]() |
[165] |
Fleming PA, Argraves WS, Gentile C, et al. (2010) Fusion of uniluminal vascular spheroids: a model for assembly of blood vessels. Dev Dyn 239: 398–406. doi: 10.1002/dvdy.22161
![]() |
[166] |
Davis GE, Bayless KJ, Mavila A (2002) Molecular basis of endothelial cell morphogenesis in three-dimensional extracellular matrices. Anat Rec 268: 252–275. doi: 10.1002/ar.10159
![]() |
[167] |
Kamei M, Saunders WB, Bayless KJ, et al. (2006) Endothelial tubes assemble from intracellular vacuoles in vivo. Nature 442: 453–456. doi: 10.1038/nature04923
![]() |
[168] | Nunes SS, Krishnan L, Gerard CS, et al. (2010) Angiogenic potential of microvessel fragments is independent of the tissue of origin and can be influenced by the cellular composition of the implants. Microcirculation 17: 557–567. |
[169] |
Laschke MW, Menger MD (2015) Adipose tissue-derived microvascular fragments: natural vascularization units for regenerative medicine. Trends Biotechnol 33: 442–448. doi: 10.1016/j.tibtech.2015.06.001
![]() |
[170] |
Alajati A, Laib AM, Weber H, et al. (2008) Spheroid-based engineering of a human vasculature in mice. Nat Methods 5: 439–445. doi: 10.1038/nmeth.1198
![]() |
[171] |
Klein D (2018) iPSCs-based generation of vascular cells: reprogramming approaches and applications. Cell Mol Life Sci 75: 1411–1433. doi: 10.1007/s00018-017-2730-7
![]() |
[172] | Pazhayattil GS, Shirali AC (2014) Drug-induced impairment of renal function. Int J Nephrol Renovasc Dis 7: 457–468. |
[173] |
Perneger TV, Whelton PK, Klag MJ (1994) Risk of kidney failure associated with the use of acetaminophen, aspirin, and nonsteroidal antiinflammatory drugs. N Engl J Med 331: 1675–1679. doi: 10.1056/NEJM199412223312502
![]() |
[174] | Kataria A, Trasande L, Trachtman H (2015) The effects of environmental chemicals on renal function. Nat Rev Nephrol 11: 610–625. |
[175] |
Gutierrez OM (2013) Sodium- and phosphorus-based food additives: persistent but surmountable hurdles in the management of nutrition in chronic kidney disease. Adv Chronic Kidney Dis 20: 150–156. doi: 10.1053/j.ackd.2012.10.008
![]() |
[176] |
Jha V, Rathi M (2008) Natural medicines causing acute kidney injury. Semin Nephrol 28: 416–428. doi: 10.1016/j.semnephrol.2008.04.010
![]() |
[177] |
Isnard Bagnis C, Deray G, Baumelou A, et al. (2004) Herbs and the kidney. Am J Kidney Dis 44: 1–11. doi: 10.1016/S0272-6386(04)01098-4
![]() |
[178] |
Gintant G, Sager PT, Stockbridge N (2016) Evolution of strategies to improve preclinical cardiac safety testing. Nat Rev Drug Discov 15: 457–471. doi: 10.1038/nrd.2015.34
![]() |
[179] |
Mehta RL, Pascual MT, Soroko S, et al. (2004) Spectrum of acute renal failure in the intensive care unit: the PICARD experience. Kidney Int 66: 1613–1621. doi: 10.1111/j.1523-1755.2004.00927.x
![]() |
[180] |
Uchino S, Kellum JA, Bellomo R, et al. (2005) Acute renal failure in critically ill patients: a multinational, multicenter study. JAMA 294: 813–818. doi: 10.1001/jama.294.7.813
![]() |
[181] |
Chu X, Bleasby K, Evers R (2013) Species differences in drug transporters and implications for translating preclinical findings to humans. Expert Opin Drug Metab Toxicol 9: 237–252. doi: 10.1517/17425255.2013.741589
![]() |
[182] |
Greek R, Menache A (2013) Systematic reviews of animal models: methodology versus epistemology. Int J Med Sci 10: 206–221. doi: 10.7150/ijms.5529
![]() |
[183] |
Olson H, Betton G, Robinson D, et al. (2000) Concordance of the toxicity of pharmaceuticals in humans and in animals. Regul Toxicol Pharmacol 32: 56–67. doi: 10.1006/rtph.2000.1399
![]() |
[184] |
Huang HC, Chang YJ, Chen WC, et al. (2013) Enhancement of renal epithelial cell functions through microfluidic-based coculture with adipose-derived stem cells. Tissue Eng Part A 19: 2024–2034. doi: 10.1089/ten.tea.2012.0605
![]() |
[185] |
Jang KJ, Cho HS, Kang DH, et al. (2011) Fluid-shear-stress-induced translocation of aquaporin-2 and reorganization of actin cytoskeleton in renal tubular epithelial cells. Integr Biol (Camb) 3: 134–141. doi: 10.1039/C0IB00018C
![]() |
[186] |
Sciancalepore AG, Sallustio F, Girardo S, et al. (2014) A bioartificial renal tubule device embedding human renal stem/progenitor cells. PLoS One 9: e87496. doi: 10.1371/journal.pone.0087496
![]() |
[187] |
Sochol RD, Gupta NR, Bonventre JV (2016) A Role for 3D Printing in Kidney-on-a-Chip Platforms. Curr Transplant Rep 3: 82–92. doi: 10.1007/s40472-016-0085-x
![]() |
[188] |
Lee J, Kim S (2018) Kidney-on-a-Chip: a New Technology for Predicting Drug Efficacy, Interactions, and Drug-induced Nephrotoxicity. Curr Drug Metab 19: 577–583. doi: 10.2174/1389200219666180309101844
![]() |
[189] |
Wilmer MJ, Ng CP, Lanz HL, et al. (2016) Kidney-on-a-Chip Technology for Drug-Induced Nephrotoxicity Screening. Trends Biotechnol 34: 156–170. doi: 10.1016/j.tibtech.2015.11.001
![]() |
[190] |
Xie HG, Wang SK, Cao CC, et al. (2013) Qualified kidney biomarkers and their potential significance in drug safety evaluation and prediction. Pharmacol Ther 137: 100–107. doi: 10.1016/j.pharmthera.2012.09.004
![]() |
[191] |
Cruz NM, Freedman BS (2018) CRISPR Gene Editing in the Kidney. Am J Kidney Dis 71: 874–883. doi: 10.1053/j.ajkd.2018.02.347
![]() |
[192] |
Mundel P (2017) Podocytes and the quest for precision medicines for kidney diseases. Pflugers Arch 469: 1029–1037. doi: 10.1007/s00424-017-2015-x
![]() |
[193] |
Jansson K, Nguyen AN, Magenheimer BS, et al. (2012) Endogenous concentrations of ouabain act as a cofactor to stimulate fluid secretion and cyst growth of in vitro ADPKD models via cAMP and EGFR-Src-MEK pathways. Am J Physiol Renal Physiol 303: F982–90. doi: 10.1152/ajprenal.00677.2011
![]() |
[194] |
Freedman BS, Lam AQ, Sundsbak JL, et al. (2013) Reduced ciliary polycystin-2 in induced pluripotent stem cells from polycystic kidney disease patients with PKD1 mutations. J Am Soc Nephrol 24: 1571–1586. doi: 10.1681/ASN.2012111089
![]() |
[195] |
Cruz NM, Song X, Czerniecki SM, et al. (2017) Organoid cystogenesis reveals a critical role of microenvironment in human polycystic kidney disease. Nat Mater 16: 1112–1119. doi: 10.1038/nmat4994
![]() |
[196] |
Wang L, Tao T, Su W, et al. (2017) A disease model of diabetic nephropathy in a glomerulus-on-a-chip microdevice. Lab Chip 17: 1749–1760. doi: 10.1039/C7LC00134G
![]() |
[197] |
Waters JP, Richards YC, Skepper JN, et al. (2017) A 3D tri-culture system reveals that activin receptor-like kinase 5 and connective tissue growth factor drive human glomerulosclerosis. J Pathol 243: 390–400. doi: 10.1002/path.4960
![]() |
[198] | Freedman BS (2015) Modeling Kidney Disease with iPS Cells. Biomark Insights 10: 153–169. |
[199] |
Clevers H (2016) Modeling Development and Disease with Organoids. Cell 165: 1586–1597. doi: 10.1016/j.cell.2016.05.082
![]() |
[200] | Garreta E, Montserrat N, Belmonte JCI (2018) Kidney organoids for disease modeling. Oncotarget 9: 12552–12553. |
[201] |
Kim YK, Refaeli I, Brooks CR, et al. (2017) Gene-Edited Human Kidney Organoids Reveal Mechanisms of Disease in Podocyte Development. Stem Cells 35: 2366–2378. doi: 10.1002/stem.2707
![]() |
[202] | Little MH, McMahon AP (2012) Mammalian kidney development: principles, progress, and projections. Cold Spring Harb Perspect Biol 4: a008300. |
[203] |
Yokote S, Matsunari H, Iwai S, et al. (2015) Urine excretion strategy for stem cell-generated embryonic kidneys. Proc Natl Acad Sci U S A 112: 12980–12985. doi: 10.1073/pnas.1507803112
![]() |
1. | Bruna Machado, Katharine Hodel, Valdir Barbosa-Júnior, Milena Soares, Roberto Badaró, The Main Molecular and Serological Methods for Diagnosing COVID-19: An Overview Based on the Literature, 2020, 13, 1999-4915, 40, 10.3390/v13010040 | |
2. | Özgecan Erdem, Esma Derin, Kutay Sagdic, Eylul Gulsen Yilmaz, Fatih Inci, Smart materials-integrated sensor technologies for COVID-19 diagnosis, 2021, 2522-5731, 10.1007/s42247-020-00150-w | |
3. | Zhong Yao, Luka Drecun, Farzaneh Aboualizadeh, Sun Jin Kim, Zhijie Li, Heidi Wood, Emelissa J. Valcourt, Kathy Manguiat, Simon Plenderleith, Lily Yip, Xinliu Li, Zoe Zhong, Feng Yun Yue, Tatiana Closas, Jamie Snider, Jelena Tomic, Steven J. Drews, Michael A. Drebot, Allison McGeer, Mario Ostrowski, Samira Mubareka, James M. Rini, Shawn Owen, Igor Stagljar, A homogeneous split-luciferase assay for rapid and sensitive detection of anti-SARS CoV-2 antibodies, 2021, 12, 2041-1723, 10.1038/s41467-021-22102-6 | |
4. | Boris G. Andryukov, Natalya N. Besednova, Tatyana A. Kuznetsova, Ludmila N. Fedyanina, Laboratory-Based Resources for COVID-19 Diagnostics: Traditional Tools and Novel Technologies. A Perspective of Personalized Medicine, 2021, 11, 2075-4426, 42, 10.3390/jpm11010042 | |
5. | Boris Georgievich Andryukov, I. N. Lyapun, COVID-19 diagnostic laboratory strategies: modern technologies and development trends (review of literature), 2020, 65, 2412-1320, 757, 10.18821/0869-2084-2020-65-12-757-766 | |
6. | Felix Weihs, Alisha Anderson, Stephen Trowell, Karine Caron, Resonance Energy Transfer-Based Biosensors for Point-of-Need Diagnosis—Progress and Perspectives, 2021, 21, 1424-8220, 660, 10.3390/s21020660 | |
7. | S. Ilbeigi, R. Dehdari Vais, N. Sattarahmady, Photo-genosensor for Trichomonas vaginalis based on gold nanoparticles-genomic DNA, 2021, 15721000, 102290, 10.1016/j.pdpdt.2021.102290 | |
8. | Riccarda Antiochia, Paper-Based Biosensors: Frontiers in Point-of-Care Detection of COVID-19 Disease, 2021, 11, 2079-6374, 110, 10.3390/bios11040110 | |
9. | Lucilene Dornelles Mello, Potential contribution of ELISA and LFI Assays to assessment of the oxidative stress condition based on 8-oxodG biomarker, 2021, 00032697, 114215, 10.1016/j.ab.2021.114215 | |
10. | Mina A. Nessiem, Harry Coppock, Mostafa M. Mohamed, Björn W. Schuller, 2023, 9780323917940, 255, 10.1016/B978-0-323-91794-0.00016-0 | |
11. | Susraba Chatterjee, Sumi Mukhopadhyay, Recent advances of lateral flow immunoassay components as “point of need”, 2022, 43, 1532-1819, 579, 10.1080/15321819.2022.2122063 | |
12. | Solen Monteil, Alexander J. Casson, Samuel T. Jones, Vivek Maheshwari, Electronic and electrochemical viral detection for point-of-care use: A systematic review, 2021, 16, 1932-6203, e0258002, 10.1371/journal.pone.0258002 | |
13. | Drishti V Lohiya, Swanand S Pathak, Role of Technology in Detection of COVID-19, 2022, 2168-8184, 10.7759/cureus.29138 | |
14. | Neda Rafat, Lee Brewer, Nabojeet Das, Dhruti J. Trivedi, Balazs K. Kaszala, Aniruddh Sarkar, Inexpensive High-Throughput Multiplexed Biomarker Detection Using Enzymatic Metallization with Cellphone-Based Computer Vision, 2023, 8, 2379-3694, 534, 10.1021/acssensors.2c01429 | |
15. | Su-Hua Yang, Hao-Yu Zhang, Chih-Chia Huang, Yi-Yan Tsai, Shun-Ming Liao, Red Zn2SiO4:Eu3+ and Mg2TiO4:Mn4+ nanophosphors for on-site rapid optical detections: Synthesis and characterization, 2021, 127, 0947-8396, 10.1007/s00339-021-04733-0 | |
16. | Robert S. Matson, 2023, Chapter 11, 978-1-0716-2902-4, 141, 10.1007/978-1-0716-2903-1_11 | |
17. | Daniel W. Bradbury, Jasmine T. Trinh, Milo J. Ryan, Cassandra M. Cantu, Jiakun Lu, Frances D. Nicklen, Yushen Du, Ren Sun, Benjamin M. Wu, Daniel T. Kamei, On-demand nanozyme signal enhancement at the push of a button for the improved detection of SARS-CoV-2 nucleocapsid protein in serum, 2021, 146, 0003-2654, 7386, 10.1039/D1AN01350E | |
18. | Marta Wanat, Mary Logan, Jennifer A Hirst, Charles Vicary, Joseph J Lee, Rafael Perera, Irene Tracey, Gordon Duff, Peter Tufano, Thomas Fanshawe, Lazaro Mwandigha, Brian D Nicholson, Sarah Tonkin-Crine, Richard Hobbs, Perceptions on undertaking regular asymptomatic self-testing for COVID-19 using lateral flow tests: a qualitative study of university students and staff, 2021, 11, 2044-6055, e053850, 10.1136/bmjopen-2021-053850 | |
19. | Genna E. Davies, Christopher R. Thornton, Development of a Monoclonal Antibody and a Serodiagnostic Lateral-Flow Device Specific to Rhizopus arrhizus (Syn. R. oryzae), the Principal Global Agent of Mucormycosis in Humans, 2022, 8, 2309-608X, 756, 10.3390/jof8070756 | |
20. | Busiswa Dyan, Palesa Pamela Seele, Amanda Skepu, Phumlane Selby Mdluli, Salerwe Mosebi, Nicole Remaliah Samantha Sibuyi, A Review of the Nucleic Acid-Based Lateral Flow Assay for Detection of Breast Cancer from Circulating Biomarkers at a Point-of-Care in Low Income Countries, 2022, 12, 2075-4418, 1973, 10.3390/diagnostics12081973 | |
21. | Wesley Wei-Wen Hsiao, Trong-Nghia Le, Dinh Minh Pham, Hui-Hsin Ko, Huan-Cheng Chang, Cheng-Chung Lee, Neha Sharma, Cheng-Kang Lee, Wei-Hung Chiang, Recent Advances in Novel Lateral Flow Technologies for Detection of COVID-19, 2021, 11, 2079-6374, 295, 10.3390/bios11090295 | |
22. | Jung Soo Park, Seokjoon Kim, Jinjoo Han, Jung Ho Kim, Ki Soo Park, Equipment-free, salt-mediated immobilization of nucleic acids for nucleic acid lateral flow assays, 2022, 351, 09254005, 130975, 10.1016/j.snb.2021.130975 | |
23. | Anel I. Saviñon-Flores, Fernanda Saviñon-Flores, G. Trejo, Erika Méndez, Ştefan Ţălu, Miguel A. González-Fuentes, Alia Méndez-Albores, A review of cardiac troponin I detection by surface enhanced Raman spectroscopy: Under the spotlight of point-of-care testing, 2022, 10, 2296-2646, 10.3389/fchem.2022.1017305 | |
24. | Alexander Biby, Xiaochuan Wang, Xinliang Liu, Olivia Roberson, Allya Henry, Xiaohu Xia, Rapid testing for coronavirus disease 2019 (COVID-19), 2022, 12, 2159-6859, 12, 10.1557/s43579-021-00146-5 | |
25. | Catia Delmiglio, David W. Waite, Sonia T. Lilly, Juncong Yan, Candace E. Elliott, Julie Pattemore, Paul L. Guy, Jeremy R. Thompson, New Virus Diagnostic Approaches to Ensuring the Ongoing Plant Biosecurity of Aotearoa New Zealand, 2023, 15, 1999-4915, 418, 10.3390/v15020418 | |
26. | Yuen Yung Hui, Yi‐Xiu Tang, Terumitsu Azuma, Hsin‐Hung Lin, Fang‐Zhen Liao, Qing‐Ying Chen, Jen‐Hwa Kuo, Yuh‐Lin Wang, Huan‐Cheng Chang, Design and implementation of a low‐cost portable reader for thermometric lateral flow immunoassay, 2022, 69, 0009-4536, 1356, 10.1002/jccs.202200124 | |
27. | Ruihua Tang, Nur Alam, Min Li, Mingyue Xie, Yonghao Ni, Dissolvable sugar barriers to enhance the sensitivity of nitrocellulose membrane lateral flow assay for COVID-19 nucleic acid, 2021, 268, 01448617, 118259, 10.1016/j.carbpol.2021.118259 | |
28. | Nathan K. Khosla, Jake M. Lesinski, Monika Colombo, Léonard Bezinge, Andrew J. deMello, Daniel A. Richards, Simplifying the complex: accessible microfluidic solutions for contemporary processes within in vitro diagnostics, 2022, 22, 1473-0197, 3340, 10.1039/D2LC00609J | |
29. | Lokman Liv, Hilal Kayabay, An Electrochemical Biosensing Platform for the SARS‐CoV‐2 Spike Antibody Detection Based on the Functionalised SARS‐CoV‐2 Spike Antigen Modified Electrode, 2022, 7, 2365-6549, 10.1002/slct.202200256 | |
30. | Jacob L. Binsley, Stefano Pagliara, Feodor Y. Ogrin, Numerical investigation of flexible Purcell-like integrated microfluidic pumps, 2022, 132, 0021-8979, 164701, 10.1063/5.0109263 | |
31. | Matheus Bernardes Torres Fogaça, Arun K. Bhunia, Leonardo Lopes-Luz, Eduardo Pimenta Ribeiro Pontes de Almeida, José Daniel Gonçalves Vieira, Samira Bührer-Sékula, Antibody- and nucleic acid–based lateral flow immunoassay for Listeria monocytogenes detection, 2021, 413, 1618-2642, 4161, 10.1007/s00216-021-03402-8 | |
32. | Mansoreh Abdolhosseini, Farshid Zandsalimi, Fahimeh Salasar Moghaddam, Gholamreza Tavoosidana, A review on colorimetric assays for DNA virus detection, 2022, 301, 01660934, 114461, 10.1016/j.jviromet.2022.114461 | |
33. | Maria‐Ana Huergo, Francis Schuknecht, Jinhua Zhang, Theobald Lohmüller, Plasmonic Nanoagents in Biophysics and Biomedicine, 2022, 10, 2195-1071, 2200572, 10.1002/adom.202200572 | |
34. | Simran Kaur, Niharika Gupta, Bansi D. Malhotra, Recent developments in wearable & non-wearable point-of-care biosensors for cortisol detection, 2023, 1473-7159, 1, 10.1080/14737159.2023.2184260 | |
35. | Lokman Liv, Melisa Yener, Gizem Çoban, Şevval Arzu Can, Electrochemical biosensing platform based on hydrogen bonding for detection of the SARS-CoV-2 spike antibody, 2022, 414, 1618-2642, 1313, 10.1007/s00216-021-03752-3 | |
36. | Noor Jamaludeen, Christian Beyer, Ulrike Billing, Katrin Vogel, Monika Brunner-Weinzierl, Myra Spiliopoulou, Potential of Point-of-Care and At-Home Assessment of Immune Status via Rapid Cytokine Detection and Questionnaire-Based Anamnesis, 2021, 21, 1424-8220, 4960, 10.3390/s21154960 | |
37. | Juan Carlos Gómez de la Torre Pretell, Miguel Hueda-Zavaleta, José Alonso Cáceres-DelAguila, Claudia Barletta-Carrillo, Cesar Copaja-Corzo, Maria del Pilar Suarez Poccorpachi, María Soledad Vega Delgado, Gloria Maria Magdalena Levano Sanchez, Vicente A. Benites-Zapata, Clinical Characteristics Associated with Detected Respiratory Microorganism Employing Multiplex Nested PCR in Patients with Presumptive COVID-19 but Negative Molecular Results in Lima, Peru, 2022, 7, 2414-6366, 340, 10.3390/tropicalmed7110340 | |
38. | Panagiotis G. Georgiou, Collette S. Guy, Muhammad Hasan, Ashfaq Ahmad, Sarah-Jane Richards, Alexander N. Baker, Neer V. Thakkar, Marc Walker, Sarojini Pandey, Neil R. Anderson, Dimitris Grammatopoulos, Matthew I. Gibson, Plasmonic Detection of SARS-CoV-2 Spike Protein with Polymer-Stabilized Glycosylated Gold Nanorods, 2022, 11, 2161-1653, 317, 10.1021/acsmacrolett.1c00716 | |
39. | Lokman Liv, Electrochemical immunosensor platform based on gold-clusters, cysteamine and glutaraldehyde modified electrode for diagnosing COVID-19, 2021, 168, 0026265X, 106445, 10.1016/j.microc.2021.106445 | |
40. | Ahmed Y. El-Moghazy, Noha Amaly, Gang Sun, Nitin Nitin, Development and clinical evaluation of commercial glucose meter coupled with nanofiber based immuno-platform for self-diagnosis of SARS-CoV-2 in saliva, 2023, 253, 00399140, 124117, 10.1016/j.talanta.2022.124117 | |
41. | Wenjie Jiang, Wangquan Ji, Yu Zhang, Yaqi Xie, Shuaiyin Chen, Yuefei Jin, Guangcai Duan, An Update on Detection Technologies for SARS-CoV-2 Variants of Concern, 2022, 14, 1999-4915, 2324, 10.3390/v14112324 | |
42. | J. Michael Janda, 2022, 9780323903035, 19, 10.1016/B978-0-12-818731-9.00085-9 | |
43. | Rebeka Rudolf, Vojkan Lazić, Peter Majerič, Andrej Ivanič, Gregor Kravanja, Karlo T. Raić, 2022, Chapter 1, 978-3-030-98745-9, 1, 10.1007/978-3-030-98746-6_1 | |
44. | Nayeli Shantal Castrejón-Jiménez, Blanca Estela García-Pérez, Nydia Edith Reyes-Rodríguez, Vicente Vega-Sánchez, Víctor Manuel Martínez-Juárez, Juan Carlos Hernández-González, Challenges in the Detection of SARS-CoV-2: Evolution of the Lateral Flow Immunoassay as a Valuable Tool for Viral Diagnosis, 2022, 12, 2079-6374, 728, 10.3390/bios12090728 | |
45. | Jennifer A Hirst, Mary Logan, Thomas R Fanshawe, Lazaro Mwandigha, Marta Wanat, Charles Vicary, Rafael Perera, Sarah Tonkin-Crine, Joseph Jonathan Lee, Irene Tracey, Gordon Duff, Peter Tufano, Marya Besharov, Lionel Tarassenko, Brian D Nicholson, F D Richard Hobbs, Feasibility and Acceptability of Community Coronavirus Disease 2019 Testing Strategies (FACTS) in a University Setting, 2021, 8, 2328-8957, 10.1093/ofid/ofab495 | |
46. | Ernst Emmanuel Etienne, Bharath Babu Nunna, Niladri Talukder, Yudong Wang, Eon Soo Lee, COVID-19 Biomarkers and Advanced Sensing Technologies for Point-of-Care (POC) Diagnosis, 2021, 8, 2306-5354, 98, 10.3390/bioengineering8070098 | |
47. | Andreea-Cristina Mirica, Dana Stan, Ioana-Cristina Chelcea, Carmen Marinela Mihailescu, Augustin Ofiteru, Lorena-Andreea Bocancia-Mateescu, Latest Trends in Lateral Flow Immunoassay (LFIA) Detection Labels and Conjugation Process, 2022, 10, 2296-4185, 10.3389/fbioe.2022.922772 | |
48. | Navaporn Sritong, Marina Sala de Medeiros, Laud Anthony Basing, Jacqueline C. Linnes, Promise and perils of paper-based point-of-care nucleic acid detection for endemic and pandemic pathogens, 2023, 23, 1473-0197, 888, 10.1039/D2LC00554A | |
49. | Kriangsak Faikhruea, Ilada Choopara, Naraporn Somboonna, Wanchai Assavalapsakul, Byeang Hyean Kim, Tirayut Vilaivan, Enhancing Peptide Nucleic Acid–Nanomaterial Interaction and Performance Improvement of Peptide Nucleic Acid-Based Nucleic Acid Detection by Using Electrostatic Effects, 2022, 5, 2576-6422, 789, 10.1021/acsabm.1c01177 | |
50. | Gourav Bhattacharya, Sam J. Fishlock, Shahzad Hussain, Sudipta Choudhury, Annan Xiang, Baljinder Kandola, Anurag Pritam, Navneet Soin, Susanta Sinha Roy, James A. McLaughlin, Disposable Paper-Based Biosensors: Optimizing the Electrochemical Properties of Laser-Induced Graphene, 2022, 14, 1944-8244, 31109, 10.1021/acsami.2c06350 | |
51. | Lokman Liv, Aysu Baş, Discriminative electrochemical biosensing of wildtype and omicron variant of SARS-CoV-2 nucleocapsid protein with single platform, 2022, 657, 00032697, 114898, 10.1016/j.ab.2022.114898 | |
52. | Jongwon Park, Lateral Flow Immunoassay Reader Technologies for Quantitative Point-of-Care Testing, 2022, 22, 1424-8220, 7398, 10.3390/s22197398 | |
53. | Mulya Supianto, Hye Jin Lee, Recent research trends in fluorescent reporters‐based lateral flow immunoassay for protein biomarkers specific to acute myocardial infarction , 2022, 43, 1229-5949, 4, 10.1002/bkcs.12430 | |
54. | Phuoc Loc Truong, Yiming Yin, Daeho Lee, Seung Hwan Ko, Advancement in COVID‐19 detection using nanomaterial‐based biosensors, 2023, 3, 2766-2098, 20210232, 10.1002/EXP.20210232 | |
55. | Haseeb Ahsan, Monoplex and multiplex immunoassays: approval, advancements, and alternatives, 2021, 31, 1618-565X, 333, 10.1007/s00580-021-03302-4 | |
56. | Cristina Tortolini, Antonio Angeloni, Riccarda Antiochia, A Comparative Study of Voltammetric vs Impedimetric Immunosensor for Rapid SARS‐CoV‐2 Detection at the Point‐of‐care, 2022, 1040-0397, 10.1002/elan.202200349 | |
57. | Nursel Olgaç, Yücel Şahin, Lokman Liv, Development and characterisation of cysteine-based gold electrodes for the electrochemical biosensing of the SARS-CoV-2 spike antigen, 2022, 147, 0003-2654, 4462, 10.1039/D2AN01225A | |
58. | Rowa Y. Alhabbab, Lateral Flow Immunoassays for Detecting Viral Infectious Antigens and Antibodies, 2022, 13, 2072-666X, 1901, 10.3390/mi13111901 | |
59. | Hervé Boutal, Christian Moguet, Lilas Pommiès, Stéphanie Simon, Thierry Naas, Hervé Volland, The Revolution of Lateral Flow Assay in the Field of AMR Detection, 2022, 12, 2075-4418, 1744, 10.3390/diagnostics12071744 | |
60. | Navid Rajil, Shahriar Esmaeili, Benjamin W. Neuman, Reed Nessler, Hung-Jen Wu, Zhenhuan Yi, Robert W. Brick, Alexei V. Sokolov, Philip R. Hemmer, Marlan O. Scully, Quantum optical immunoassay: upconversion nanoparticle-based neutralizing assay for COVID-19, 2022, 12, 2045-2322, 10.1038/s41598-021-03978-2 | |
61. | Isabelle C. Samper, Ana Sánchez-Cano, Wisarut Khamcharoen, Ilhoon Jang, Weena Siangproh, Eva Baldrich, Brian J. Geiss, David S. Dandy, Charles S. Henry, Electrochemical Capillary-Flow Immunoassay for Detecting Anti-SARS-CoV-2 Nucleocapsid Protein Antibodies at the Point of Care, 2021, 6, 2379-3694, 4067, 10.1021/acssensors.1c01527 | |
62. | José R. Botella, Point-of-Care DNA Amplification for Disease Diagnosis and Management, 2022, 60, 0066-4286, 1, 10.1146/annurev-phyto-021621-115027 | |
63. | Diana I. Meira, Ana I. Barbosa, Joel Borges, Rui L. Reis, Vitor M. Correlo, Filipe Vaz, Recent advances in nanomaterial-based optical biosensors for food safety applications: Ochratoxin-A detection, as case study, 2023, 1040-8398, 1, 10.1080/10408398.2023.2168248 | |
64. | Poorya Sadeghi, Hessamaddin Sohrabi, Maryam Hejazi, Ali Jahanban-Esfahlan, Behzad Baradaran, Maryam Tohidast, Mir Reza Majidi, Ahad Mokhtarzadeh, Seyed Mohammad Tavangar, Miguel de la Guardia, Lateral flow assays (LFA) as an alternative medical diagnosis method for detection of virus species: The intertwine of nanotechnology with sensing strategies, 2021, 145, 01659936, 116460, 10.1016/j.trac.2021.116460 | |
65. | Wen-Yeh Hsieh, Cheng-Han Lin, Tzu-Ching Lin, Chao-Hsu Lin, Hui-Fang Chang, Chin-Hung Tsai, Hsi-Tien Wu, Chih-Sheng Lin, Development and Efficacy of Lateral Flow Point-of-Care Testing Devices for Rapid and Mass COVID-19 Diagnosis by the Detections of SARS-CoV-2 Antigen and Anti-SARS-CoV-2 Antibodies, 2021, 11, 2075-4418, 1760, 10.3390/diagnostics11101760 | |
66. | Lokman Liv, Gizem Çoban, Nuri Nakiboğlu, Tanıl Kocagöz, A rapid, ultrasensitive voltammetric biosensor for determining SARS-CoV-2 spike protein in real samples, 2021, 192, 09565663, 113497, 10.1016/j.bios.2021.113497 | |
67. | Terumitsu Azuma, Yuen Yung Hui, Oliver Y. Chen, Yuh-Lin Wang, Huan-Cheng Chang, Thermometric lateral flow immunoassay with colored latex beads as reporters for COVID-19 testing, 2022, 12, 2045-2322, 10.1038/s41598-022-07963-1 | |
68. | Wei Zhao, Yang Kim, Randall G. Cameron, A novel multiplex lateral flow assay for rapid assessment of pectin structural/functional properties, 2022, 133, 0268005X, 107988, 10.1016/j.foodhyd.2022.107988 | |
69. | Monika Chhillar, Deepak kukkar, Preeti Kukkar, Ki-Hyun Kim, Nanoparticle-antibody conjugate-based immunoassays for detection of CKD-associated biomarkers, 2023, 158, 01659936, 116857, 10.1016/j.trac.2022.116857 | |
70. | Ashwin Krishnamoorthy, Subashini Chandrapalan, Gohar JalayeriNia, Yaqza Hussain, Ayman Bannaga, Ian Io Lei, Ramesh Arasaradnam, Influence of seasonal and operator variations on diagnostic accuracy of lateral flow devices during the COVID-19 pandemic: a systematic review and meta-analysis, 2023, 23, 1470-2118, 144, 10.7861/clinmed.2022-0319 | |
71. | Magnus Philipp, Lisa Müller, Marcel Andrée, Kai P. Hussnaetter, Heiner Schaal, Michael Feldbrügge, Kerstin Schipper, Efficient virus detection utilizing chitin-immobilized nanobodies synthesized in Ustilago maydis, 2023, 01681656, 10.1016/j.jbiotec.2023.03.005 | |
72. | Siddarth Arumugam, Jiawei Ma, Uzay Macar, Guangxing Han, Kathrine McAulay, Darrell Ingram, Alex Ying, Harshit Harpaldas Chellani, Terry Chern, Kenta Reilly, David A. M. Colburn, Robert Stanciu, Craig Duffy, Ashley Williams, Thomas Grys, Shih-Fu Chang, Samuel K. Sia, Rapidly adaptable automated interpretation of point-of-care COVID-19 diagnostics, 2023, 3, 2730-664X, 10.1038/s43856-023-00312-x | |
73. | André Shamsabadi, Tabasom Haghighi, Sara Carvalho, Leah C. Frenette, Molly M. Stevens, The Nanozyme Revolution: Enhancing the Performance of Medical Biosensing Platforms, 2024, 36, 0935-9648, 10.1002/adma.202300184 | |
74. | Suchanat Boonkaew, Katarzyna Szot-Karpińska, Joanna Niedziółka-Jönsson, Ario de Marco, Martin Jönsson-Niedziółka, NFC Smartphone-Based Electrochemical Microfluidic Device Integrated with Nanobody Recognition for C-Reactive Protein, 2024, 9, 2379-3694, 3066, 10.1021/acssensors.4c00249 | |
75. | Masato Matsuda, Kosuke Itoh, Takahiro Sugai, Yoshiki Hoshiyama, Toshiaki Kikuchi, Shuji Terai, Improving diagnostic performance of coronavirus disease 2019 rapid antigen testing through computer-based feedback training using open-source experimental psychology software, 2024, 30, 1341321X, 292, 10.1016/j.jiac.2023.10.019 | |
76. | Ying Zhao, Jingwei Sang, Yusheng Fu, Jiuchuan Guo, Jinhong Guo, Magnetic nanoprobe-enabled lateral flow assays: recent advances, 2023, 148, 0003-2654, 3418, 10.1039/D3AN00044C | |
77. | Christopher R. Thornton, Genna E. Davies, Laura Dougherty, Development of a monoclonal antibody and a lateral-flow device for the rapid detection of a Mucorales-specific biomarker, 2023, 13, 2235-2988, 10.3389/fcimb.2023.1305662 | |
78. | Yeonjeong Ha, Exploiting the Potential of Magnetic Nanoparticles for Rapid Diagnosis Tests (RDTs): Nanoparticle-Antibody Conjugates and Color Development Strategies, 2023, 13, 2075-4418, 3033, 10.3390/diagnostics13193033 | |
79. | Qingwen Sun, Qihong Ning, Tangan Li, Qixia Jiang, Shaoqing Feng, Ning Tang, Daxiang Cui, Kan Wang, Immunochromatographic enhancement strategy for SARS-CoV-2 detection based on nanotechnology, 2023, 15, 2040-3364, 15092, 10.1039/D3NR02396F | |
80. | Mahmoud El-Maghrabey, Galal Magdy, Heba M. Hashem, Mohamed N. Amin, Abdelaziz Elgaml, Aya Saad Radwan, Magda Ahmed El-Sherbeny, Rania El-Shaheny, Comprehending COVID-19 diagnostic tests and greenness assessment of its reported detection methods, 2023, 169, 01659936, 117379, 10.1016/j.trac.2023.117379 | |
81. | Yiming Zhang, Zijun Fang, Yusheng Fu, Yi Wu, Jiuchuan Guo, Jinhong Guo, Diangeng Li, Jingshan Duan, Long afterglow nanoprobes labeled image enhancement using deep learning in rapid and sensitive lateral flow immunoassay, 2024, 379, 09244247, 115956, 10.1016/j.sna.2024.115956 | |
82. | Min Jung Kim, Izzati Haizan, Min Ju Ahn, Dong-Hyeok Park, Jin-Ha Choi, Recent Advances in Lateral Flow Assays for Viral Protein Detection with Nanomaterial-Based Optical Sensors, 2024, 14, 2079-6374, 197, 10.3390/bios14040197 | |
83. | Lourdes AN. Julius, Sarai M. Torres Delgado, Rohit Mishra, Nigel Kent, Eadaoin Carthy, Jan G. Korvink, Dario Mager, Jens Ducrée, David J. Kinahan, Programmable fluidic networks on centrifugal microfluidic discs, 2024, 1288, 00032670, 342159, 10.1016/j.aca.2023.342159 | |
84. | Hanhao Zhang, Neda Rafat, Josiah Rudge, Sai Preetham Peddireddy, Yoo Na Kim, Taaseen Khan, Aniruddh Sarkar, High throughput electronic detection of biomarkers using enzymatically amplified metallization on nanostructured surfaces, 2024, 1759-9660, 10.1039/D4AY01657B | |
85. | Nana Lyu, Amin Hassanzadeh-Barforoushi, Laura M. Rey Gomez, Wei Zhang, Yuling Wang, SERS biosensors for liquid biopsy towards cancer diagnosis by detection of various circulating biomarkers: current progress and perspectives, 2024, 11, 2196-5404, 10.1186/s40580-024-00428-3 | |
86. | Martin Bartosik, Ludmila Moranova, Nasim Izadi, Johana Strmiskova, Ravery Sebuyoya, Jitka Holcakova, Roman Hrstka, Advanced technologies towards improved HPV diagnostics, 2024, 96, 0146-6615, 10.1002/jmv.29409 | |
87. | Xushuo Zhang, Sam Fishlock, Peter Sharpe, James McLaughlin, Alistair H. Kean, Nikhil Bhalla, 2023, Cystatin C for chronic kidney disease monitoring via the application of gold core-shells in lateral flow immunoassays, 9781510668454, 6, 10.1117/12.2690967 | |
88. | Zhao-Yu Lu, Yang-Hsiang Chan, The importance of antibody orientation for enhancing sensitivity and selectivity in lateral flow immunoassays, 2024, 3, 2635-0998, 1613, 10.1039/D4SD00206G | |
89. | Shazreen Shaharuddin, Nik Mohd Afizan Nik Abd Rahman, Mas Jaffri Masarudin, Mohammedarfat N Alamassi, The importance of hypoxia sensors in detecting HIF-1 biomarker at high altitude, 2023, 7, 25764500, 132, 10.15406/aaoaj.2023.07.00182 | |
90. | Hannah Littlecott, Clare Herd, John O'Rourke, Lina Toncon Chaparro, Matt Keeling, G. James Rubin, Elizabeth Fearon, Effectiveness of testing, contact tracing and isolation interventions among the general population on reducing transmission of SARS-CoV-2: a systematic review, 2023, 381, 1364-503X, 10.1098/rsta.2023.0131 | |
91. | Po‐Chun Huang, Ying Zhou, Erin B. Porter, Ravindra G. Saxena, Andrea Gomez, Matthew Ykema, Naomi L. Senehi, Dongjoo Lee, Chia‐Ping Tseng, Pedro J. Alvarez, Yizhi J. Tao, Yilin Li, Rafael Verduzco, Organic Electrochemical Transistors functionalized with Protein Minibinders for Sensitive and Specific Detection of SARS‐CoV‐2, 2023, 10, 2196-7350, 10.1002/admi.202202409 | |
92. | Yedi Herdiana, Ferry Ferdiansyah Sofian, Shaharum Shamsuddin, Taofik Rusdiana, Towards halal pharmaceutical: Exploring alternatives to animal-based ingredients, 2024, 10, 24058440, e23624, 10.1016/j.heliyon.2023.e23624 | |
93. | Ankit Tiwari, Divyanshu Singh, Dharmendra Kumar, Vikas Chandra, Naveen Kumar Vishvakarma, Dhananjay Shukla, Harit Jha, Rajat Pratap Singh, 2024, Chapter 15, 978-981-97-4722-1, 389, 10.1007/978-981-97-4723-8_15 | |
94. | Sijie Liu, Rui Shu, Mingrui Zhang, Cong Zhao, Kexin Wang, Jiayi Zhang, Jing Sun, Leina Dou, Daohong Zhang, Jianlong Wang, Goat anti-mouse immunoglobulin as “crosslinker” assisted signal tracer assemble with intensive antibody utilization efficiency for sensitive paper-based strip nanobiosensors, 2024, 258, 01418130, 128923, 10.1016/j.ijbiomac.2023.128923 | |
95. | Sijie Liu, Chenyang Sun, Xiyue Zhang, Rui Shu, Jiayi Zhang, Biao Wang, Kexin Wang, Leina Dou, Lunjie Huang, Qingyu Yang, Jianlong Wang, Advances in enhancement-type signal tracers and analysis strategies driven Lateral flow immunoassay for guaranteeing the agri-food safety, 2025, 268, 09565663, 116920, 10.1016/j.bios.2024.116920 | |
96. | Zhigang Cao, Li Yi, Xiangnan Liu, Jinyuan Shang, Yuening Cheng, Erkai Feng, Xingyu Liu, Yuping Fan, Xiaoliang Hu, Wenlong Cai, Feng Cong, Shipeng Cheng, Rapid lateral flow immunoassay for fluorescence detection of canine distemper virus (CDV), 2024, 11, 2297-1769, 10.3389/fvets.2024.1413420 | |
97. | N. Sathishkumar, Bhushan J. Toley, Direct comparison of colorimetric signal amplification techniques in lateral flow immunoassays, 2024, 16, 1759-9660, 7200, 10.1039/D4AY01416B | |
98. | Balamurugan Shanmugaraj, Perawat Jirarojwattana, Waranyoo Phoolcharoen, Molecular Farming Strategy for the Rapid Production of Protein-Based Reagents for Use in Infectious Disease Diagnostics, 2023, 89, 0032-0943, 1010, 10.1055/a-2076-2034 | |
99. | Naresh Mandal, Raja Mitra, Bidhan Pramanick, C-MEMS-derived glassy carbon electrochemical biosensors for rapid detection of SARS-CoV-2 spike protein, 2023, 9, 2055-7434, 10.1038/s41378-023-00601-4 | |
100. | Anuj Kumar, Pralay Maiti, Paper-based sustainable biosensors, 2024, 5, 2633-5409, 3563, 10.1039/D3MA01019H | |
101. | Miroslav Pohanka, Current trends in digital camera-based bioassays for point-of-care tests, 2024, 552, 00098981, 117677, 10.1016/j.cca.2023.117677 | |
102. | Anna Toldrà, Georgios Chondrogiannis, Mahiar M. Hamedi, A 3D paper microfluidic device for enzyme‐linked assays: Application to DNA analysis, 2023, 18, 1860-6768, 10.1002/biot.202300143 | |
103. | Kumaravel Vealan, Narcisse Joseph, Sharizah Alimat, Anandi S. Karumbati, Karuppiah Thilakavathy, Lateral flow assay: a promising rapid point-of-care testing tool for infections and non-communicable diseases, 2023, 17, 1875-855X, 250, 10.2478/abm-2023-0068 | |
104. | Fanjin Wang, Anthony Harker, Mohan Edirisinghe, Maryam Parhizkar, Micro‐ and Nanomanufacturing for Biomedical Applications and Nanomedicine: A Perspective, 2023, 3, 2688-4046, 10.1002/smsc.202300039 | |
105. | Tao Wang, Chuanjiang Ran, Xinyue He, Shengzhou Li, Hongguang Xiang, Yan Shen, Jue Wang, Hongxia Wei, Effects on molecular interactions of hollow gold nanoparticles and antibody for sensitizing P24 antigen determination, 2024, 14, 2046-2069, 30154, 10.1039/D4RA05277C | |
106. | Rafaela S. Andre, Rafaela C. Sanfelice, Mardoqueu M. da Costa, Luiza A. Mercante, Daniel S. Correa, Adriana Pavinatto, 2024, 9780443153808, 345, 10.1016/B978-0-443-15380-8.00012-6 | |
107. | Abozar Ghorbani, Sajad Astaraki, Mahsa Rostami, Arezoo Pakdel, Unleashing the power of colloidal gold immunochromatographic assays for plant virus diagnostics, 2024, 12, 22150161, 102498, 10.1016/j.mex.2023.102498 | |
108. | Zhicheng Jin, Wonjun Yim, Maurice Retout, Emily Housel, Wenbin Zhong, Jiajing Zhou, Michael S. Strano, Jesse V. Jokerst, Colorimetric sensing for translational applications: from colorants to mechanisms, 2024, 53, 0306-0012, 7681, 10.1039/D4CS00328D | |
109. | Sandra E. Rodriguez‐Cruz, Evaluating the sensitivity, stability, and cross‐reactivity of commercial fentanyl immunoassay test strips, 2023, 68, 0022-1198, 1555, 10.1111/1556-4029.15332 | |
110. | Jack Hassall, Carmen Coxon, Vishal C. Patel, Simon D. Goldenberg, Chrysi Sergaki, Limitations of current techniques in clinical antimicrobial resistance diagnosis: examples and future prospects, 2024, 2, 2731-8745, 10.1038/s44259-024-00033-8 | |
111. | Norberto A. Guzman, Daniel E. Guzman, Timothy Blanc, Advancements in portable instruments based on affinity-capture-migration and affinity-capture-separation for use in clinical testing and life science applications, 2023, 1704, 00219673, 464109, 10.1016/j.chroma.2023.464109 | |
112. | Jaehi Kim, Min-Sup Shin, Jonghyun Shin, Hyung-Mo Kim, Xuan-Hung Pham, Seung-min Park, Dong-Eun Kim, Young Jun Kim, Bong-Hyun Jun, Recent Trends in Lateral Flow Immunoassays with Optical Nanoparticles, 2023, 24, 1422-0067, 9600, 10.3390/ijms24119600 | |
113. | Simone Cavalera, Alessandro Gelli, Fabio Di Nardo, Thea Serra, Valentina Testa, Stefano Bertinetti, Laura Ozella, Claudio Forte, Claudio Baggiani, Laura Anfossi, Improving the sensitivity and the cost-effectiveness of a competitive visual lateral flow immunoassay through sequential designs of experiments, 2025, 208, 0026265X, 112450, 10.1016/j.microc.2024.112450 | |
114. | Viola Papini, Angelo Meloni, Susanna Pecchia, Development of a Duplex PCR-NALFIA Assay for the Simultaneous Detection of Macrophomina phaseolina and Verticillium dahliae Causal Agents of Crown and Root Rot of Strawberry, 2025, 15, 2077-0472, 160, 10.3390/agriculture15020160 | |
115. | Sarah Courdier, Alexandre Bouchet, Maxime Karlen, Julien Boucher, Valérie D’Acremont, David Vernez, Stephane Goutte, The direct emissions related to Global Warming Potential of different types of diagnostic tests at different phases of the COVID pandemic: A climate-focused life-cycle assessment, 2025, 4, 2767-3200, e0000561, 10.1371/journal.pclm.0000561 | |
116. | Dania Al Ismail, Edgar I. Campos-Madueno, Valentina Donà, Andrea Endimiani, Hypervirulent Klebsiella pneumoniae (hvKp): Overview, Epidemiology, and Laboratory Detection, 2025, 10, 2469-2964, 80, 10.20411/pai.v10i1.777 | |
117. | Wei Xue, Tao Sheng, Pan Jia, Guangxiao Zhang, Yangyang Chang, Meng Liu, 2025, 9780443133565, 1, 10.1016/B978-0-443-13356-5.00006-9 | |
118. | Po-Yu Chu, Po-Shuan Huang, Chih-Yu Chen, Kun-Yu Tsai, Shu-Ying Chiu, Le-Wei Fan, Yu-Chen Cheng, Chi-Jui Lin, Chia-Hsun Hsieh, Min-Hsien Wu, Development of a point-of-care testing (POCT)-use paper-based device for recombinase polymerase amplification (RPA)-based bioassays-: Demonstration of the detection of Neisseria gonorrhoeae, 2025, 26660539, 100307, 10.1016/j.snr.2025.100307 | |
119. | Alexander Spreinat, Willfried Kunz, Christian H. Maack, Carola Wilczek, Britta Nestler, Andrea Ernst, Fluid propagation and protein adsorption patterns in porous nitrocellulose membranes for lateral flow assays, 2025, 37, 1070-6631, 10.1063/5.0257343 | |
120. | Supriya Atta, Yuanhao Zhao, Sebastian Sanchez, Sabina V. Yampolsky, Tuan Vo-Dinh, Plasmonics-Enhanced Dual-Modal Colorimetric and Photothermal Lateral Flow Immunoassay Using Gold Nanocages, 2025, 0003-2700, 10.1021/acs.analchem.4c05384 | |
121. | Pakapreud Khumwan, Stephan Ruttloff, Johannes Götz, Dieter Nees, Conor O’Sullivan, Alvaro Conde, Mirko Lohse, Christian Wolf, Nastasia Okulova, Janine Brommert, Richard Benauer, Ingo Katzmayr, Nikolaus Ladenhauf, Wilfried Weigel, Maciej Skolimowski, Max Sonnleitner, Martin Smolka, Anja Haase, Barbara Stadlober, Jan Hesse, Translation of COVID-19 Serology Test on Foil-Based Lateral Flow Chips: A Journey from Injection Molding to Scalable Roll-to-Roll Nanoimprint Lithography, 2025, 15, 2079-6374, 229, 10.3390/bios15040229 | |
122. | Thiciany Blener Lopes, Fabiana Fioravante Coelho, Tárcio Peixoto Roca, Jéssica Karoline Augusta Oliveira, Valérian Delagarde, Ségolène Brichler, Diana Paola Gómez Mendoza, Juan Miguel Villalobos Salcedo, Deusilene Souza Vieira, Frédéric Le Gal, Ricardo Tostes Gazzinelli, Ana Paula Fernandes, Elitza S. Theel, A universal point-of-care immunochromatographic test for the serodiagnosis of hepatitis D, 2025, 0095-1137, 10.1128/jcm.01999-24 | |
123. | Georgios Goumas, Efthymia N. Vlachothanasi, Evangelos C. Fradelos, Dimitra S. Mouliou, Biosensors, Artificial Intelligence Biosensors, False Results and Novel Future Perspectives, 2025, 15, 2075-4418, 1037, 10.3390/diagnostics15081037 | |
124. | Amadeo Sena-Torralba, Yulieth D. Banguera-Ordoñez, Javier Carrascosa, Ángel Maquieira, Sergi Morais, Portable electrophoretic lateral flow biosensing for ultra-sensitive human lactate dehydrogenase detection in serum samples, 2025, 282, 09565663, 117504, 10.1016/j.bios.2025.117504 | |
125. | Yi‐Xiu Tang, Yuen Yung Hui, An‐Jie Liu, Wesley W.‐W. Hsiao, Huan‐Cheng Chang, Quantitative laser‐scanning lateral flow immunoassay of luteinizing hormone with a handheld analyzer, 2025, 0009-4536, 10.1002/jccs.70024 | |
126. | Chuanjiang Ran, Qingjie Zhang, Zhenglin Long, Yuhao Li, Xinyue He, Shengzhou Li, Chen Chen, Cong Cheng, Ying Xu, Wenli Zhang, Wei Guo, Yan Shen, An in-situ gold growth self-catalytic signal amplification system enhanced hollow gold immunochromatography for ultrasensitive detection of p24 antigen in HIV infection, 2025, 515, 13858947, 163350, 10.1016/j.cej.2025.163350 | |
127. | Julia Pedreira-Rincón, Lourdes Rivas, Joan Comenge, Vasso Skouridou, Daniel Camprubí-Ferrer, Jose Muñoz, Ciara K. O'Sullivan, Alejandro Chamorro-Garcia, Claudio Parolo, A comprehensive review of competitive lateral flow assays over the past decade, 2025, 1473-0197, 10.1039/D4LC01075B | |
128. | Hong-Ngoc Tran-Doan, Huu-Dang Tran, Anh-Kiet Lam, Khon Huynh, 2025, Chapter 17, 978-3-031-90193-5, 223, 10.1007/978-3-031-90194-2_17 | |
129. | Hajar Samadian, Ferial Ghaemi, 2025, 9780443338717, 167, 10.1016/B978-0-443-33871-7.00010-6 | |
130. | Celia Fuentes-Chust, Claudio Parolo, Andrew Piper, Arben Merkoçi, Stability matters: evaluating the long-term performance of AuNP–DNA conjugates in lateral flow assays through varied conjugation methods and storage buffers, 2025, 2040-3364, 10.1039/D4NR05328A |
Infectious agent | Detectable marker of infectious contamination (target analyte) | Format of test systems LFIA | Refs |
Dengue virus | Dengue non-structural protein 1 (NS1) | Magneto-enzyme | [51],[52] |
Zika virus | Zika virus nonstructural protein 1 (NS1) | Smartphone-based fluorescent | [53] |
Chikungunya virus | SD Bioline, IgM OnSite, IgM |
Chromatographic Chromatographic |
[54] |
Yellow fever (YF) virus | YF non-structural protein 1 (NS1) | Chromatographic | [55] |
Ebola virus | Antigen Ebola virus ReEBOV VP40 | Magneto-enzyme | [56] |
Dengue virus | Ig G/ IgM | Multiplex | [42] |
Yellow fever (YF) virus | Ig G/ IgM | Multiplex | [42] |
Ebola virus | Ig G/ IgM | Multiplex | [42] |
Human immunodeficiency virus (HIV) | Anti-HIV IgG | Multiplex | [41] |
Hepatitis C virus (HCV) | Anti-HCV IgG | Multiplex | [41] |
Hepatitis B virus (HBV) | Hepatitis B e-antigen (HBeAg) | Monoplex | [57] |
Human Immunodeficiency Virus (HIV-1) | HIV-1 p24 antigen | Monoplex | [58],[59] |
Foot-and-mouth disease virus | Antigen detection for all 7 serotypes for types O, A, C and Asia1 | Multiplex | [60] |
Respiratory viruses | Human adenovirus, influenza A H1N1 virus | Magnetic SERS-based LFIA (FeO @ Ag) | [61] |
Newcastle disease virus (NDV) | Amplification (RPA)-nucleic acid lateral flow (NALF) immunoassay | Multiplex RPA-NALF | [62] |
Infectious bronchitis virus (IBV) | Amplification (RPA)-nucleic acid lateral flow (NALF) immunoassay | Multiplex RPA-NALF | [62] |
Human Polyomavirus BK (BKV) | DNA BKV | Monoplex sandwich-type | [63] |
Bordetella pertussis | Anti-toxin pertussis IgG | Fluorescent Eu-nanoparticle reporters | [64] |
Staphylococcus aureus | Staphylococcal enterotoxin B | SERS-based lateral flow immunoassay | [65] |
Streptococcus pyogenes, Group A | S. pyogenes (A) | SERS-based lateral flow immunoassay | [66] |
Yersinia pestis | F1 capsular antigen | Monoplex | [67] |
Escherichia coli O157: H7 | E. coli O157: H7 | Sandwich models | [68] |
Listeria monocytogenes | L. monocytogenes | SERS-based lateral flow immunoassay | [69] |
Salmonella typhimurium | S. typhimurium | SERS-based lateral flow immunoassay | [69] |
Helicobacter pylori (HpSA-test) | Antigen H. pylori HpSA | Monoplex | [70] |
Advantage | Refs | Limitations | Refs |
|
[3],[27],[40] |
|
[27],[29],[40],[62] |
|
[23],[32],[40] |
|
[23],[28],[32],[47] |
|
[12],[31],[37] |
|
[10],[29],[37],[42] |
|
[3],[18],[28],[32] |
|
[3],[13],[32],[54] |
|
[4],[10],[13] |
|
[4],[28],[51],[55] |
|
[13],[24],[26] |
|
[13],[33],[52],[59] |
|
[12],[25],[33],[35] |
|
[12],[23],[42],[58] |
|
[23],[28],[37],[38] |
|
[23],[28],[47],[62] |
Detection platforms principle | Company/Country | Detector Model | Mode of Measurements | Company website |
Optical | Axxin/Australia | AXXIN AX-2X | Colorimetry, fluorimetry | axxin.com |
Bio-AMD/United Kingdom | Digital Strip Reader | Colorimetry | bioamd.com | |
BioAssay Works/United States | Cube-Reader | Colorimetry | bioassayworks.com | |
Hamamatsu/Japan | Immunochromato-Reader C11787 | Colorimetry, fluorimetry | hamamatsu.com | |
Magnetic Labels (Magneto-enzyme) | Magna BioSciences/United States | MICT® Bench-Top System | Open System | magnabiosciences.com |
Magnasense Technologies/Finland | Magnasense's Magnetometric Reader | Open System | magnasense.com | |
VWR International/United States | FoodChek™ MICT System | Closed System | vwr.com |
Detecting Device and Construction | Target Analytes | Range of Concentration Measured | Refs |
Dual LFIA with iPhone 5s | Salmonella enteritidis | 20–107 CFU/mL | [7] |
E. coli O157:H7 | 34–107 CFU/mL | ||
UC-LFS platform | Brain natriuretic peptide | 5–100 pg/mL | [31] |
Suppression of tumorigenicity 2 | 1–25 ng/mL | ||
Smartphone's ambient-light-sensor-based reader (SPALS-reader) | |||
Cadmium ion | 0.16–50 ng/mL | [35] | |
Clenbuterol | 0.046–1 ng/mL | ||
Porcine epidemic diarrhea virus | 0.055–20 µg/mL | ||
Electrochemical detection | Human chorionic gonadotrophin (HCG) | 25–50 mIU HCG in serum | [82] |
Magneto-enzyme | Dengue virus non-structural protein 1 (NS1) | 0, 1–0,25 ng/ml | [51] |
Magnetic SERS (Raman scattering-based LFIA) | Human adenovirus, | from 50 PFU/mL | [61] |
Influenza A H1N1 virus | from 10 PFU/mL | ||
Raman scattering-based LFIA (SERS-LFIA) | Streptococcus pyogenes, Group A | 0,2–100 KOE/mL | [66] |
Listeria monocytogenes | 102–107 KOE/mL | [69] |
Country-developers and producer companies | Sensitivity/ specificity of the test-systems (%) | Description of the test system | Biosubstrates used for diagnosis, analysis time | Links |
US / China, Cellex Inc. | 93,8/95,6 | IgM/IgG is detected by SARS-CoV-2 protein nucelocapside | Serum, plasma or whole blood (K2-EDTA, sodium citrate), 20 min | [113] |
US, ChemBio | 92,7 (IgM) и 95,9 (IgG)/99,0 (IgM и IgG) | IgM/IgG is detected by SARS-CoV-2 protein nucelocapside | Finger or vein whole blood, serum and plasma (lithium heparin, K2-EDTA), 15 min | [114] |
US, Autobio Diagnostics Co. Ltd. (+ Hardy Diagnostics) | 95,7 (IgM) и 99,0 (IgG)/99,0 (IgM и IgG) | IgM/IgG is detected by SARS-CoV-2 antigens | Finger or vein whole blood, serum and plasma (heparin, K2-EDTA), 15 min | [115] |
US / China, Healgen Scientific LLC | 96,7 (IgG), 86,7 (IgM), 96,7 comb./98,0 (IgG), 99,0 (IgM), 97,0 comb. | IgM/IgG is detected by SARS-CoV-2 antigens | Finger or vein whole blood, serum and plasma (heparin, K2-EDTA), 10 min | [116] |
China, Hangzhou Biotest Biotech Co., Ltd | 92.5 (IgM), 91.56 (IgG)/98.1 (IgM), 99.52 (IgG) | IgM/IgG is detected by SARS-CoV-2 recombinant spike protein receptor binding domain | Finger or vein whole blood, serum and plasma (heparin, K2-EDTA), up to 20 min | [117] |
China, Biohit Healthcare (Heifei) Co. Ltd. | 33.0 (IgM, days 1–7), 56.6 (IgG days 8–14), 83.0 (IgM days 8–14), 96.2 (IgG days 15+), 97.7 (IgM days 15 +)/99.5 (IgM), 100.0 (IgG) | IgM/IgG is detected antibodies by SARS-CoV-2 recombinant N-protein antigen and mouse anti human IgM/IgG antibody | Samples for human serum, plasma or whole blood (heparin, K2-EDTA, and sodium citrate), 15 min | [118] |
China, Hangzhou Laihe Biotech Co., Ltd | 100,0 (IgM, 0–6 days), 85.7 (IgM, 7–14 days), 76,0 (IgG, 7–14 days), 99.25 (IgM, 14+ days), 98.5 (IgG, 14+ days) / 99.43 | IgM/IgG is detected by SARS-CoV-2 antibodies. The target antigen is the S1 region of the spike protein. | Samples for human serum, plasma or whole blood (heparin, K2-EDTA, and sodium citrate), 15 min | [119] |
US / China, Aytu Biosciences / Orient Gene Biotech | 87.9 (IgM) & 97.2 (IgG)/100,0 for IgG and IgM | IgM/IgG is detected antibodies by SARS-CoV-2 antigen. | Samples for human serum, plasma or whole blood, 10 min | [120] |
Note: * - according to the Center for Health Security at J. Hopkins University
Infectious agent | Detectable marker of infectious contamination (target analyte) | Format of test systems LFIA | Refs |
Dengue virus | Dengue non-structural protein 1 (NS1) | Magneto-enzyme | [51],[52] |
Zika virus | Zika virus nonstructural protein 1 (NS1) | Smartphone-based fluorescent | [53] |
Chikungunya virus | SD Bioline, IgM OnSite, IgM |
Chromatographic Chromatographic |
[54] |
Yellow fever (YF) virus | YF non-structural protein 1 (NS1) | Chromatographic | [55] |
Ebola virus | Antigen Ebola virus ReEBOV VP40 | Magneto-enzyme | [56] |
Dengue virus | Ig G/ IgM | Multiplex | [42] |
Yellow fever (YF) virus | Ig G/ IgM | Multiplex | [42] |
Ebola virus | Ig G/ IgM | Multiplex | [42] |
Human immunodeficiency virus (HIV) | Anti-HIV IgG | Multiplex | [41] |
Hepatitis C virus (HCV) | Anti-HCV IgG | Multiplex | [41] |
Hepatitis B virus (HBV) | Hepatitis B e-antigen (HBeAg) | Monoplex | [57] |
Human Immunodeficiency Virus (HIV-1) | HIV-1 p24 antigen | Monoplex | [58],[59] |
Foot-and-mouth disease virus | Antigen detection for all 7 serotypes for types O, A, C and Asia1 | Multiplex | [60] |
Respiratory viruses | Human adenovirus, influenza A H1N1 virus | Magnetic SERS-based LFIA (FeO @ Ag) | [61] |
Newcastle disease virus (NDV) | Amplification (RPA)-nucleic acid lateral flow (NALF) immunoassay | Multiplex RPA-NALF | [62] |
Infectious bronchitis virus (IBV) | Amplification (RPA)-nucleic acid lateral flow (NALF) immunoassay | Multiplex RPA-NALF | [62] |
Human Polyomavirus BK (BKV) | DNA BKV | Monoplex sandwich-type | [63] |
Bordetella pertussis | Anti-toxin pertussis IgG | Fluorescent Eu-nanoparticle reporters | [64] |
Staphylococcus aureus | Staphylococcal enterotoxin B | SERS-based lateral flow immunoassay | [65] |
Streptococcus pyogenes, Group A | S. pyogenes (A) | SERS-based lateral flow immunoassay | [66] |
Yersinia pestis | F1 capsular antigen | Monoplex | [67] |
Escherichia coli O157: H7 | E. coli O157: H7 | Sandwich models | [68] |
Listeria monocytogenes | L. monocytogenes | SERS-based lateral flow immunoassay | [69] |
Salmonella typhimurium | S. typhimurium | SERS-based lateral flow immunoassay | [69] |
Helicobacter pylori (HpSA-test) | Antigen H. pylori HpSA | Monoplex | [70] |
Advantage | Refs | Limitations | Refs |
|
[3],[27],[40] |
|
[27],[29],[40],[62] |
|
[23],[32],[40] |
|
[23],[28],[32],[47] |
|
[12],[31],[37] |
|
[10],[29],[37],[42] |
|
[3],[18],[28],[32] |
|
[3],[13],[32],[54] |
|
[4],[10],[13] |
|
[4],[28],[51],[55] |
|
[13],[24],[26] |
|
[13],[33],[52],[59] |
|
[12],[25],[33],[35] |
|
[12],[23],[42],[58] |
|
[23],[28],[37],[38] |
|
[23],[28],[47],[62] |
Detection platforms principle | Company/Country | Detector Model | Mode of Measurements | Company website |
Optical | Axxin/Australia | AXXIN AX-2X | Colorimetry, fluorimetry | axxin.com |
Bio-AMD/United Kingdom | Digital Strip Reader | Colorimetry | bioamd.com | |
BioAssay Works/United States | Cube-Reader | Colorimetry | bioassayworks.com | |
Hamamatsu/Japan | Immunochromato-Reader C11787 | Colorimetry, fluorimetry | hamamatsu.com | |
Magnetic Labels (Magneto-enzyme) | Magna BioSciences/United States | MICT® Bench-Top System | Open System | magnabiosciences.com |
Magnasense Technologies/Finland | Magnasense's Magnetometric Reader | Open System | magnasense.com | |
VWR International/United States | FoodChek™ MICT System | Closed System | vwr.com |
Detecting Device and Construction | Target Analytes | Range of Concentration Measured | Refs |
Dual LFIA with iPhone 5s | Salmonella enteritidis | 20–107 CFU/mL | [7] |
E. coli O157:H7 | 34–107 CFU/mL | ||
UC-LFS platform | Brain natriuretic peptide | 5–100 pg/mL | [31] |
Suppression of tumorigenicity 2 | 1–25 ng/mL | ||
Smartphone's ambient-light-sensor-based reader (SPALS-reader) | |||
Cadmium ion | 0.16–50 ng/mL | [35] | |
Clenbuterol | 0.046–1 ng/mL | ||
Porcine epidemic diarrhea virus | 0.055–20 µg/mL | ||
Electrochemical detection | Human chorionic gonadotrophin (HCG) | 25–50 mIU HCG in serum | [82] |
Magneto-enzyme | Dengue virus non-structural protein 1 (NS1) | 0, 1–0,25 ng/ml | [51] |
Magnetic SERS (Raman scattering-based LFIA) | Human adenovirus, | from 50 PFU/mL | [61] |
Influenza A H1N1 virus | from 10 PFU/mL | ||
Raman scattering-based LFIA (SERS-LFIA) | Streptococcus pyogenes, Group A | 0,2–100 KOE/mL | [66] |
Listeria monocytogenes | 102–107 KOE/mL | [69] |
Country-developers and producer companies | Sensitivity/ specificity of the test-systems (%) | Description of the test system | Biosubstrates used for diagnosis, analysis time | Links |
US / China, Cellex Inc. | 93,8/95,6 | IgM/IgG is detected by SARS-CoV-2 protein nucelocapside | Serum, plasma or whole blood (K2-EDTA, sodium citrate), 20 min | [113] |
US, ChemBio | 92,7 (IgM) и 95,9 (IgG)/99,0 (IgM и IgG) | IgM/IgG is detected by SARS-CoV-2 protein nucelocapside | Finger or vein whole blood, serum and plasma (lithium heparin, K2-EDTA), 15 min | [114] |
US, Autobio Diagnostics Co. Ltd. (+ Hardy Diagnostics) | 95,7 (IgM) и 99,0 (IgG)/99,0 (IgM и IgG) | IgM/IgG is detected by SARS-CoV-2 antigens | Finger or vein whole blood, serum and plasma (heparin, K2-EDTA), 15 min | [115] |
US / China, Healgen Scientific LLC | 96,7 (IgG), 86,7 (IgM), 96,7 comb./98,0 (IgG), 99,0 (IgM), 97,0 comb. | IgM/IgG is detected by SARS-CoV-2 antigens | Finger or vein whole blood, serum and plasma (heparin, K2-EDTA), 10 min | [116] |
China, Hangzhou Biotest Biotech Co., Ltd | 92.5 (IgM), 91.56 (IgG)/98.1 (IgM), 99.52 (IgG) | IgM/IgG is detected by SARS-CoV-2 recombinant spike protein receptor binding domain | Finger or vein whole blood, serum and plasma (heparin, K2-EDTA), up to 20 min | [117] |
China, Biohit Healthcare (Heifei) Co. Ltd. | 33.0 (IgM, days 1–7), 56.6 (IgG days 8–14), 83.0 (IgM days 8–14), 96.2 (IgG days 15+), 97.7 (IgM days 15 +)/99.5 (IgM), 100.0 (IgG) | IgM/IgG is detected antibodies by SARS-CoV-2 recombinant N-protein antigen and mouse anti human IgM/IgG antibody | Samples for human serum, plasma or whole blood (heparin, K2-EDTA, and sodium citrate), 15 min | [118] |
China, Hangzhou Laihe Biotech Co., Ltd | 100,0 (IgM, 0–6 days), 85.7 (IgM, 7–14 days), 76,0 (IgG, 7–14 days), 99.25 (IgM, 14+ days), 98.5 (IgG, 14+ days) / 99.43 | IgM/IgG is detected by SARS-CoV-2 antibodies. The target antigen is the S1 region of the spike protein. | Samples for human serum, plasma or whole blood (heparin, K2-EDTA, and sodium citrate), 15 min | [119] |
US / China, Aytu Biosciences / Orient Gene Biotech | 87.9 (IgM) & 97.2 (IgG)/100,0 for IgG and IgM | IgM/IgG is detected antibodies by SARS-CoV-2 antigen. | Samples for human serum, plasma or whole blood, 10 min | [120] |