Acceso abierto

An overview of technologies and devices against COVID-19 pandemic diffusion: virus detection and monitoring solutions


Cite

Figure 1:

Sandwich immunoassay mechanism of a GMR biosensor forming a capture antibody–target antigen–detection antibody–MNP complex (Wu et al., 2020).
Sandwich immunoassay mechanism of a GMR biosensor forming a capture antibody–target antigen–detection antibody–MNP complex (Wu et al., 2020).

Figure 2:

Picture of the GMR-based hand-held device (a), and top view of the electronic section with highlighted the main components (b) (Wu et al., 2017, 2020).
Picture of the GMR-based hand-held device (a), and top view of the electronic section with highlighted the main components (b) (Wu et al., 2017, 2020).

Figure 3:

Picture of GMR-based portable device reported by the researchers from Stanford University (Choi et al., 2016).
Picture of GMR-based portable device reported by the researchers from Stanford University (Choi et al., 2016).

Figure 4:

Test-strip design and setup (Orlov et al., 2016).
Test-strip design and setup (Orlov et al., 2016).

Figure 5:

Schematic representation of SARS-CoV-2 detection using the electrochemical biosensor. (a) Prepare the premix A and B; (b) Process of electrochemical detection using a smartphone (Zhao et al., 2021).
Schematic representation of SARS-CoV-2 detection using the electrochemical biosensor. (a) Prepare the premix A and B; (b) Process of electrochemical detection using a smartphone (Zhao et al., 2021).

Figure 6:

Schematic of Co-functionalized TiO2 nanotube (Co-TNT)-based sensing platform for detecting SARS-CoV-2 (Vadlamani et al., 2020).
Schematic of Co-functionalized TiO2 nanotube (Co-TNT)-based sensing platform for detecting SARS-CoV-2 (Vadlamani et al., 2020).

Figure 7:

Scanning electron microscopy (SEM) micrographs of (a) TiO2 nanotubes (TNTs) post-annealing. Inset shows sidewalls of TNTs, (b) Co-functionalized TNTs showing the Co (OH)2 precipitate, (c) EDS map of Co confirming its uniform distribution, and (d) EDS spectra confirming the presence of Co (Vadlamani et al., 2020).
Scanning electron microscopy (SEM) micrographs of (a) TiO2 nanotubes (TNTs) post-annealing. Inset shows sidewalls of TNTs, (b) Co-functionalized TNTs showing the Co (OH)2 precipitate, (c) EDS map of Co confirming its uniform distribution, and (d) EDS spectra confirming the presence of Co (Vadlamani et al., 2020).

Figure 8:

Schematic diagram of COVID-19 FET-based biosensor operation (Seo et al., 2020).
Schematic diagram of COVID-19 FET-based biosensor operation (Seo et al., 2020).

Figure 9:

Graphical representation of the working operation of the eCovSens device using SPCE electrode, including COVID-19 antibody (Mahari et al., 2020).
Graphical representation of the working operation of the eCovSens device using SPCE electrode, including COVID-19 antibody (Mahari et al., 2020).

Figure 10:

DhITACT-TR chip for robust detection of target pathogen in a single-step injection of RNA extract (Samson et al., 2020).
DhITACT-TR chip for robust detection of target pathogen in a single-step injection of RNA extract (Samson et al., 2020).

Figure 11:

The surface plasmon polariton (SPP) can only be excited at specific wave vectors and decays evanescently from the surface. The momentum-matching condition leads to the SPP resonance and only exists at certain incident angles (Li et al., 2015).
The surface plasmon polariton (SPP) can only be excited at specific wave vectors and decays evanescently from the surface. The momentum-matching condition leads to the SPP resonance and only exists at certain incident angles (Li et al., 2015).

Figure 12:

Different technologies versus the COVID-19 (Chamola et al., 2020).
Different technologies versus the COVID-19 (Chamola et al., 2020).

Figure 13:

Representation of IoT-based framework for early identification and monitoring of new cases of COVID-19 virus infections (Otoom et al., 2020).
Representation of IoT-based framework for early identification and monitoring of new cases of COVID-19 virus infections (Otoom et al., 2020).

Figure 14:

Scheme of the proposed framework to predict COVID-19 (Maghded et al., 2020).
Scheme of the proposed framework to predict COVID-19 (Maghded et al., 2020).

Figure 15:

Cloud computing for the proposed framework (Maghded et al., 2020).
Cloud computing for the proposed framework (Maghded et al., 2020).

Figure 16:

User registration & results of the test (Maghded et al., 2020).
User registration & results of the test (Maghded et al., 2020).

Figure 17:

iFever (a), Tempdrop (b), iSense (c), Ran’s Night (d), and smart thermometers.
iFever (a), Tempdrop (b), iSense (c), Ran’s Night (d), and smart thermometers.

Figure 18:

Smart Helmet captures temperature by the thermal optical camera (Triaxtec, 2019).
Smart Helmet captures temperature by the thermal optical camera (Triaxtec, 2019).

Figure 19:

Smart glasses temperature capturing (Mohammed et al., 2020).
Smart glasses temperature capturing (Mohammed et al., 2020).

Figure 20:

Thermal imaging drone (Hitconsultant, 2019).
Thermal imaging drone (Hitconsultant, 2019).

Figure 21:

Autonomous swab test robots (South Korean Institute of Machinery and Material, 2019).
Autonomous swab test robots (South Korean Institute of Machinery and Material, 2019).

Figure 22:

The configuration of the headset’s microphone for the respiration rate and breathing detection, (a) configuration of the heart rate, temperature, and respiration rate detection using NTC thermistor, microphone, and PPG sensor, (b) (Stojanović et al., 2020).
The configuration of the headset’s microphone for the respiration rate and breathing detection, (a) configuration of the heart rate, temperature, and respiration rate detection using NTC thermistor, microphone, and PPG sensor, (b) (Stojanović et al., 2020).

Figure 23:

Block diagram of the Arduino based interface for processing vital signs (Stojanović et al., 2020).
Block diagram of the Arduino based interface for processing vital signs (Stojanović et al., 2020).

Figure 24:

The system architecture of the IoT-Q-Band system (Singh et al., 2020).
The system architecture of the IoT-Q-Band system (Singh et al., 2020).

Figure 25:

Data flow diagram of the IoT-Q-Band system (Singh et al., 2020).
Data flow diagram of the IoT-Q-Band system (Singh et al., 2020).

Figure 26:

Mobile application screens of the IoT-Q-Band system showing the cases: (a) when the band is connected, and the subject is within 50 meters of registered quarantine Geo-location, and (b) when the wearable tampered, and the patient is outside the 50 meters of the registered quarantine Geo-location (Singh et al., 2020).
Mobile application screens of the IoT-Q-Band system showing the cases: (a) when the band is connected, and the subject is within 50 meters of registered quarantine Geo-location, and (b) when the wearable tampered, and the patient is outside the 50 meters of the registered quarantine Geo-location (Singh et al., 2020).

Figure 27:

Representation of filter testing setup and the working principle for self-sterilization of the filter (Stanford et al., 2019).
Representation of filter testing setup and the working principle for self-sterilization of the filter (Stanford et al., 2019).

Figure 28:

Example of the Guardian G-Volt mask application (Dezeen, 2019).
Example of the Guardian G-Volt mask application (Dezeen, 2019).

Figure 29:

BX100 Philips Biosensor (Philips, 2019): front view of the device (a), and its application on a patient (b), the graphical scheme of the health monitoring system (c).
BX100 Philips Biosensor (Philips, 2019): front view of the device (a), and its application on a patient (b), the graphical scheme of the health monitoring system (c).

Advantages and disadvantages of different magnetic nano-sensors technologies (Wu et al., 2020).

PlatformAdvantagesDisadvantages
GMRHigh sensitivityMultiple washing steps usually required, thus needing well-trained technicians, but can be wash-free, which reduces the sensitivity
Availability of a portable deviceTime-consuming
Mass production capabilityHigh cost per test; nanofabrication of GMR biosensors required
MTJHigh sensitivityMultiple washing steps usually required, thus needing well-trained technicians, but can be wash-free, which reduces the sensitivity
Mass production capabilityHigh noise; large distance from the MNP to the sensor surface
Hard-to-acquire linear response
Complicated fabrication process
Time-consuming
High cost per test; nanofabrication of MTJ biosensors required
MPS, surface-basedHigh sensitivityMultiple washing steps usually required, thus needing well-trained technicians, but can be wash-free, which reduces the sensitivity
Low cost per testTime-consuming
Availability of a portable device
MPS, volume-basedOne-step wash-free detection allowedMedium sensitivity
Immunoassays that can be hand-held by non-technicians
Low cost per test
Availability of a portable device
NMRAvailability of a portable deviceMultiple washing steps usually required, thus needing well-trained technicians, but can be wash-free, which reduces the sensitivity
Time-consuming
Medium sensitivity

A full list of extracted features (Sun et al., 2020).

CategoryModalityFeaturesExtraction
MobilitySmartphone locationHomestayThe time spent within 200m radius of home location (determined using DBSCAN)
Maximum traveled distance from homeThe maximum distance traveled from home location
Smartphone BluetoothMaximum number of nearby devicesThe maximum number of Bluetooth-enabled nearby devices
Fitbit step countStep countDaily total of Fitbit step count
Functional measuresFitbit sleepSleep durationDaily total duration of sleep categories (light, deep, and rem)
BedtimeThe first sleep category of the night
Fitbit heart rateAverage heart rateThe daily average heart rate
Phone usageSmartphone user interactionUnlock durationThe total duration of phone in the unlocked state
Smartphone usage eventSocial app use durationThe total duration spent on social apps (Google Play categories of Social, Communication, and Dating)

Comparison between the scientific works reported in the second section, in terms of the detection technology, target species, LOD, detection time, application scenario and scalability.

Scientific workDetection mechanismTarget speciesLODDetection timeApplication scenarioScalability
Wu et al. (2020)GMRH1N1 virus H3N2 virus15 ng/mL 125 TCID50/ml10 minVirus screeningLow
Orlov et al. (2016)MPSBoNT A, B and E0.22, 0.11, 0.32 ng/mL25 minFood qualityMedium
Zhang et al. (2013)MPSssDNA400 pM10 secDNA analysisMedium
Lei et al. (2015)NMRCuSO40.2 µM1 mincell isolation, cell culture, DNA amplificationMedium
Zhao et al. (2021)electrochemicalSARS-CoV-2 virus200 copies/mL10 secVirus screeningHigh
Vadlamani et al. (2020)electrochemicalSARS-CoV-2 virus14 nM30 secVirus screeningHigh
Chin et al. (2017)electrochemicalJEV virus5–20 ng/mL20 minVirus screeningHigh
Seo et al. (2020)FET-basedSARS-CoV-2 virus1.7 fM20 secVirus screeningHigh
Moitra et al. (2020)LSPRSARS-CoV-20.18 ng/µL10 minVirus screeningLow
eISSN:
1178-5608
Idioma:
Inglés
Calendario de la edición:
Volume Open
Temas de la revista:
Engineering, Introductions and Overviews, other