Next Article in Journal
Electrochemical Detection of Different Foodborne Bacteria for Point-of-Care Applications
Previous Article in Journal
Using a Smartphone-Based Colorimetric Device with Molecularly Imprinted Polymer for the Quantification of Tartrazine in Soda Drinks
Previous Article in Special Issue
Staphylococcus aureus Detection in Milk Using a Thickness Shear Mode Acoustic Aptasensor with an Antifouling Probe Linker
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antibody Phage Display Technology for Sensor-Based Virus Detection: Current Status and Future Prospects

Institute of Biochemistry and Physiology of Plants and Microorganisms, Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), 13 Prospect Entuziastov, Saratov 410049, Russia
*
Author to whom correspondence should be addressed.
Biosensors 2023, 13(6), 640; https://doi.org/10.3390/bios13060640
Submission received: 26 April 2023 / Revised: 31 May 2023 / Accepted: 8 June 2023 / Published: 9 June 2023

Abstract

:
Viruses are widespread in the environment, and many of them are major pathogens of serious plant, animal, and human diseases. The risk of pathogenicity, together with the capacity for constant mutation, emphasizes the need for measures to rapidly detect viruses. The need for highly sensitive bioanalytical methods to diagnose and monitor socially significant viral diseases has increased in the past few years. This is due, on the one hand, to the increased incidence of viral diseases in general (including the unprecedented spread of a new coronavirus infection, SARS-CoV-2), and, on the other hand, to the need to overcome the limitations of modern biomedical diagnostic methods. Phage display technology antibodies as nano-bio-engineered macromolecules can be used for sensor-based virus detection. This review analyzes the commonly used virus detection methods and approaches and shows the prospects for the use of antibodies prepared by phage display technology as sensing elements for sensor-based virus detection.

1. Introduction

The concept of a virus often raises public concern about the spread of the new coronavirus infection, dengue fever, avian influenza virus, hepatitis, Ebola fever, acquired immunodeficiency syndrome (AIDS), and other deadly diseases [1,2]. However, despite their enormous harmfulness, many viral and virus-like particles have practical applications in bio(nano)medical technologies. In particular, they can be used in targeted drug delivery, in the transfer of genetic material [3,4], in noninvasive imaging for the early detection and treatment of human diseases [5], as biorecognition elements in biosensor systems [6,7], and in the production of vaccines and antimicrobials [8]. Among other things, viral particles can be used in bioelectronics to form certain functional surfaces and materials [4,9], owing to their ability to genetically adapt to any changes from the outside. For instance, icosahedral viral nanoparticles such as tobacco mosaic virus and bacteriophage M13 have been used as nanotubes for batteries and nanowires, while rod-shaped viral nanoparticles such as cowpea mosaic virus and cowpea chlorotic mottle virus have been considered for use in biomedicine [5,10,11]. Additionally, it has been confirmed that archaeal Sulfolobus islandicus rod-shaped virus 2 can be used in bioconjugation chemistry [12,13,14]. Such a useful side of viruses is an object of close research attention, allowing new horizons for their possible applications.
Viruses and noninfectious virus-like particles exhibit the characteristics of ideal building blocks with perfect symmetry and with uniformity of size and shape, which is due to the precise assembly of hundreds of molecules into highly organized scaffolds [15]. Of particular interest is the use of viral particles in the phage display preparation of recombinant antibodies. Phage display is based on the expression of foreign peptides or proteins on the surface of phage particles as part of a chimeric envelope protein [16,17,18]. George P. Smith created this technology in the middle of the 1980s after demonstrating that a foreign protein could be expressed on the surface of bacteriophage M13 (filamentous bacteriophage). He combined the pIII minor coat protein of M13 with the gene encoding the EcoRI restriction endonuclease fragment in a single translation frame [19,20,21,22]. British biochemist Sir Gregory Winter employed phage display to display antigen-binding immunoglobulin fragments on the surface of bacteriophage fd in the 1990s [23]. A fresh combinatorial strategy for preparing recombinant antibodies was thus developed.
The goal of antibody phage display is to create phage particles that show antibodies or their fragments on the membrane with a high degree of specificity to the target antigen or high-affinity phage antibodies. The three main steps of this technology are as follows:
-
Making bacteriophages or choosing them from libraries of available phages;
-
Biopanning (affinity-based enrichment of phage libraries);
-
Phage-displayed antibodies go through a number of affinity selection procedures before being utilized as specific biosensor receptors [24].
Since the beginning of the new millennium, viruses have considerably worsened the global epidemiological situation. Examples include the spread of severe acute respiratory syndrome (SARS) in 2002–2004, the A/H1N1 swine flu pandemic in 2009, and the Ebola outbreak in West Africa in 2014 [25]. In 2019, the world was shaken by the coronavirus pandemic (COVID-19), which has reached unprecedented proportions and has made substantial adjustments to everyone’s habitual way of life [26,27]. According to the World Health Organization (WHO), 763,740,140 cases of COVID-19 infection and 6,908,554 deaths have been confirmed in 230 countries and territories worldwide (as of 20 April 2023) [28]. In addition, complications in the form of unregulated immunological responses, metabolic dysfunction, and multiple organ failure have been reported in patients with acute COVID-19 [29].
On 20 May 2022, WHO released a full report on global health statistics for 2020, the first year of the COVID-19 pandemic that resulted in 4.5 million excess deaths. The statistics show that the pandemic affected the health care system worldwide, in some cases severely limiting access to life-saving resources. The pandemic has significantly slowed the global progress in both life and healthy life expectancy that was achieved in the first 20 years of the century. The report highlights that the global community has been unprepared to recognize the central role of primary health care and to fully fund key elements of health care, which has largely slowed the effectiveness of the response to the spread of the SARS-CoV-2 virus [30].
On 12 August 2022, the United States Center for Disease Control and Prevention reported two new human cases of the H3N2 influenza virus circulating in swine. The outbreaks of influenza A subtype H3N2 among pigs in the United States indicate that the risk of human infection with this virus and its further spread may now be higher than usual. Since the monkeypox outbreak began and as of 3 January, 21,094 confirmed cases have been reported in 29 countries. Five deaths were recorded during the spread of monkeypox: three in Spain, one in Belgium, and one in the Czech Republic [31].
With increased population mobility and international travel, viruses spread rapidly around the world, causing outbreaks of infectious diseases. The emergence of a new virus on a pandemic scale threatens the health and lives of 8 billion people. A major reason for the high prevalence of viral diseases is the lack of effective methods of their detection, including preventive detection in the absence of visible disease symptoms. Therefore, there is a growing need to develop highly sensitive and selective virus detection methods that would be suitable for a wide range of applications, including disease diagnosis, pharmaceutical research, agriculture, and preventive measures.
We describe the advantages of phage display technology in the production of virus-specific antibodies, and we review what is known about virus detection with phage antibodies used in standard and sensor-based methods.

2. Methods of Virus Detection

Many methods are used to identify viruses and virus-like particles. The most common traditional virus detection methods are time-consuming, expensive, often difficult to reproduce, and require special instrumentation and qualified personnel [32]. Virus detection methods facilitate the recognition of different virion components and fractions. Such targets may include fragments of the viral genome, overexpressed antigens, enzymes, and so on (Figure 1).
One “gold standard” method for virus detection is the isolation of pathogens from infected cell cultures [33,34]. Virus isolation can be a very reliable method, but it takes several days to several weeks to produce results and requires trained personnel and expensive equipment. Another “gold standard” method is the polymerase chain reaction (PCR), used in the laboratory diagnosis of viral infections [35,36,37,38,39]. By amplifying the target nucleic acid in the presence of intercalating fluorescent dyes such as SYBR Green or SYTO-13, semi-quantitative detection results may be obtained in a few hours. However, PCR requires a precise thermocycling regimen and, as a rule, special equipment.
Yet another “gold standard” method is the enzyme immunoassay (ELISA), which is often used to detect infections caused by human immunodeficiency virus (HIV), dengue virus (DENV), and other viruses [38,40,41]. The detection by ELISA of viral antigens in clinical samples is limited by the insufficient sensitivity of the assay. Despite these disadvantages, PCR and ELISA enable real-time detection of viruses.
In recent years, DNA microarray technology has been used to diagnose various viral infections, particularly acute respiratory infections [42,43,44]. In addition, the lateral flow assay is very popular for virus diagnosis [45]. The use of immunochemical and serological methods is also hampered by the high antigenic diversity of some virus groups. Table 1 compares the methods traditionally used to detect pathogenic viruses [26]. Owing to these limitations, traditional methods do not allow simultaneous and sensitive detection of viruses and virus-like particles in clinical samples. Figure 2 shows the main diagnostic methods for COVID-19 [46].
One of the most promising tools for virus detection is biosensor-based analysis methods. Biosensors consist of a biologically sensitive element (receptor), a transducer, and a working solution (Figure 3).
The variety of biological materials and methods for their immobilization on the transducer surface has led to the development of three main categories of biosensors, depending on the type of bioreceptor [48]:
-
Enzymatic biosensors;
-
Affinity biosensors;
-
Cell/tissue biosensors.
One immunoanalytical method is affinity biosensors, which track the development of an antigen-antibody combination [48,49]. The efficacy of biosensors depends on such factors as the type of immobilization of recognition elements (biomolecules), with preservation of their natural activity; the availability of the recognition element to the corresponding analyte in solution; and the low nonspecific adsorption on a solid carrier. Therefore, high requirements are imposed on the recognition molecules of biosensors. Specifically, the ligand must be highly specific, chemically and physically stable, and economical to produce. For this reason, antibody-based biosensors as sensitive detection elements are the most common ones.
Kumar et al. [50] showed the prospects for the application of electrochemical sensors in virus detection. Amperometric immunosensors, which combine the principles of voltammetry with immunological responses, are highly sensitive in detecting immune interactions. Figure 4 shows the electrochemical techniques used for SARS-CoV-2 detection [50].
Zhang et al. [51] described an electrochemical method for the detection of H5N1 avian influenza virus. The method uses antibodies immobilized on magnetic nanoparticles through a sandwich immunoassay. Through a sandwich immunoassay, antibodies are fixed on magnetic nanoparticles in the procedure. By electrochemically producing protons from water, the magnetic nanoparticles are transformed into an electroactive counterpart of Prussian blue, releasing Fe3+. The reduction of deposited K3Fe(CN)6 and Fe3+ to K4Fe(CN)6 and Fe2+, respectively, in a subsequent electrochemical step at a lower potential, results in a reaction that creates a Prussian blue analog). This method detects a much lower analyte concentration than traditional methods.
Another antibody-based electrochemical device was described by [52] (Figure 5). The sensor system is based on measuring changes in conductivity by immobilization of a monoclonal antibody to SARS-CoV-2 on screen-printed carbon electrodes. The performance of both electrodes was measured by the interaction of the monoclonal antibody with the specific spike protein of SARS-CoV-2. The limit of detection was 120 fM, whereas it was 90 fM with an in-house built biosensor device (eCovSens) in the case of spiked saliva samples. The advantage of eCovSens is that it can detect the SARS-CoV-2 spike antigen within 10–30 s.
Białobrzeska et al. [53] described an immunosensor for the detection of SARS-CoV-2 N protein sequences on the basis of nucleocapsid antibodies against SARS-CoV-2. The antibodies were prepared and purified in the laboratory and were tightly immobilized on different surfaces (diamond/gold/glass carbon). The biosensor allowed rapid detection of the SARS-CoV-2 virus (detection time, less than 10 min), with detection limits ranging from 0.227 ng/mL (glass carbon) to 0.334 ng/mL (diamond) and 0.362 ng/mL (gold).
For all surfaces tested, a wide linear range of analyte concentrations was obtained (4.4 ng/mL–4.4 pg/mL). Figure 6 compares the developed method with the immunoassay, the “gold standard” in virus detection.
Using the detection of bacteriophage T7 as an example, Lesniewski et al. [54] proposed a colorimetric immunosensor based on gold nanoparticles covalently bound to antibodies specific to this bacteriophage. Owing to the formation of immune complexes of phage particles with antibodies and gold nanoparticles, the immunosensor permitted fast, simple, and selective detection of the virus. As a result, one can observe a solution color change from red to purple with the naked eye. The authors conclude that this method can be used to detect almost all viruses and virus-like particles.
While the first electrochemical devices were being developed, optical sensors, including planar waveguide sensors, were also being developed at the same time [55]. Internal and exterior optical fiber sensors can be split into two major categories. Thus, a feature of internal sensors is that the external force that acts on the fiber can change not only the transit time but also the intensity/polarization of the light propagating down the fiber.
The advantages of optical nanobiosensors include (1) high sensitivity along with an ultralow limit of detection; (2) the ability to modify the analysis for a specific sample, including when the signal is visualized directly with the naked eye for the rapid reading of results and subsequent diagnosis; and (3) the relatively low cost of the portable laser and, therefore, the biosensor itself (Figure 7) [26].
An optical immunosensor was developed to detect the NS1 antigen (nonstructural protein 1) as a biomarker of dengue virus in clinical samples obtained early in infection. The principle of operation is based on the detection of the NS1 antigen by immunofluorescence by using fluorescein-5-isothiocyanate (FITC) conjugated to an IgG antibody. The probe was highly reproducible (relative standard deviation, 2%) and sufficiently stable for 21 days at 4 °C, with a detection limit of 15 ng/mL−1 [56].
Guliy et al. [57] reported on the optical sensor-aided immunodetection of transmissible gastroenteritis virus (TGEV). The analysis is based on the measurement of changes in the electro-optical variables of the sensor before and after the addition of TGEV-specific antibodies to the suspension being analyzed. The measuring process can be fully automated, and it is possible to detect TGEV in the presence of foreign viral particles.
Owing to their excellent chemical stability, acoustic biosensors are most frequently made with piezoelectric crystals such as quartz, lithium niobate, and lithium tantalate. An attractive approach to creating a family of sensors characterized by high sensitivity, rapidity of analysis, low cost, and tiny sizes is the excitation of acoustic waves in a piezoelectric material. Some techniques rely on membranes or receptor antibodies placed on a piezoelectric waveguide or the resonator surface [58]. For instance, immunosensors were developed for the selective detection of the herpes virus in human blood [59] and in natural water reservoirs (rivers, sewers, wastewaters) without the need for preprocessing of the analyzed substrate [60]. These immunosensors immobilized the appropriate antiviral antibodies on the surface of a piezoelectric resonator.
For example, bacteriophage M13 was identified in real-time by Tamarin et al. [61], who used elastic Love waves with horizontal shear polarization in a layered medium. First, phage-specific antibodies were permanently fixed on a silicon oxide substrate. The multilayered structure that had developed on the waveguide surface was then examined after an immunoreaction between the bacteriophage and the immobilized antibodies was induced. The formation of the structure led to a change in the velocity and attenuation of the Love wave. The particles bound to the sensor surface antibodies (control for counting the phage titer) were eluted, and the pH of the solution was changed. Plaque-forming units (PFU) were counted by microbiological methods.
An electroacoustic sensor platform was used for the immunodetection of the transmissible gastroenteritis virus of swine (TGEV) [62]. Recently, colorimetric sensors [63] and surface-enhanced Raman spectroscopy (SERS) [64] have been actively used for virus diagnosis.
Significantly, all immunochemical methods, including biosensor-based ones, depend primarily on the quality of the specific antibodies used to make diagnostic systems. Antibody selection targets range from peptides and recombinant proteins to viral or virus-like particles [65]. Virus-specific antibodies can be derived from libraries generated from different animal species, such as macaque [66] and chimpanzee [67] monkeys, llamas [68], mice [69], chickens [70], and humans [71].

3. Phage Display Technology

Historically, the first source of antibodies was sera from immune animals and humans. The antibodies obtained contain a set of immunoglobulins of different classes and subclasses that specifically recognize their antigen. Different serum antibodies recognize several sites (epitopes) of the antigen. Antibody preparation was then aided by hybridoma technology [72], which makes it possible to obtain antibodies that are produced by a single-cell clone, recognize a single epitope, and retain their properties in many hybrid-cell generations. The advantages of monoclonal antibodies include their high specificity and the possibility to prepare large quantities of antibodies with a given specificity. However, the technology for preparing monoclonal antibodies was developed for mouse and rat studies and is difficult to apply in the case of human antibodies, whereas for therapeutic purposes, human immunoglobulins are required. Difficulties also arise when immunization of animals, for whatever reason, is impossible or the lack of immunogenicity of potential antigens cannot be overcome. Finally, monoclonal antibodies cannot be used with traditional and popular immunological methods based on the precipitation reaction.
To resolve these problems, methods for the molecular cloning of antibody gene fragments have been put forward. One such method is antibody phage display, proposed by [23]. The antigen-binding fragments (scFv, Fab) presented on the surface of a filamentous bacteriophage can be selected on an immobilized antigen. The main idea behind the method is to design a combinatorial library in which the variable regions of the light and heavy chains of immunoglobulins are combined randomly and are presented on the surface of the bacteriophage. Every bacteriophage, such as a B lymphocyte, expresses antibodies of certain specificity. If the size of a library is large enough, the repertoire of variable regions will be comparable to that of antibodies in the body. The advantages of the method are as follows:
-
Antibodies can be selected in vitro, and animal immunization can be omitted;
-
No need to use laboratory animals or maintain long-term cultures of eukaryotic cells;
-
The preparation of individual clone producers of miniantibodies takes 10–14 days, against the several months that hybridoma technology takes;
-
Antibodies are relatively easy to prepare, and their cost is low;
-
It is possible to make hybrid molecules with marker proteins (e.g., tag peptide) in a short time;
-
It is possible to prepare antibodies to autoantigens, weakly immunogenic compounds, and toxins;
-
The method can be used in immunotherapy.
Temperate filamentous phages (M13, f1, fd, etc.) are most commonly used in phage displays. Their virion contains circular single-chain DNA, whose genetic organization and sequence are known by many members of this phage group. The genome size is about 6000 nucleotides. The five structural proteins that make up the capsid of a viral particle are organized similarly in all members of the group [23].
Human phage display libraries or libraries from other species are used to make antibodies. These libraries will be utilized for the in vitro selection (biopanning) of the target molecule. The following actions are part of the process of choosing phage library components:
-
The immobilized antigen is incubated with library clones;
-
Particles of the phage that have not yet attached to the antigen are washed away;
-
The bound phage particles are eluted;
-
The selected clones are infected with bacterial cells (E. coli);
-
The affinity clones are amplified and isolated.
The immunogenicity of the antigens, how they are immobilized, and the quantity of biopanning rounds all affect how long it takes to create an antibody. That is why the process can take a few hours or several days. After biopanning, the monoclonal phage antibodies are analyzed by various immunochemical assays, in particular, ELISA. Subsequently, the genes of the selected antibody fragments can be isolated from the phage particles for subsequent cloning and expression. Figure 8 illustrates the antibody generation process [65]. The availability of the antibody library for screening is the primary prerequisite for antibody phage display. Antibody libraries, depending on how they are constructed, fall into four main categories—naïve, immune, semisynthetic, and synthetic.
The following is the general process for creating a combinatorial phage library:
-
The genes of scFv, Fab, or other fragments are cloned from immune or undamaged humans, mice, rabbits, chickens, pigs, dogs, monkeys, sheep, or cows’ B lymphocyte mRNA;
-
These genes, along with the gene coding for the capsid protein (typically p3), are introduced into the phagemid in a single translation frame;
-
Phagemid genes are expressed, and virions are put together in infected E. coli cells by using the generated phagemid repertoire.
Foreign antibody fragments will be exposed as part of the capsid proteins of the virions. Depending on the chosen vector system, this step takes place with or without the use of helper phages.
In this way, a population of bacteriophages is prepared, each exhibiting a specific antigen-binding domain on its surface [73,74]. The application of phage antibodies in bioreceptors is particularly promising. Phage antibodies themselves are faster and less labor-intensive to prepare than those prepared by hybridoma technology. Phage antibodies have a number of benefits over their natural counterparts, including the following:
-
Owing to the Fc region of the intact antibody, the tiny size of the antibody fragments is typically accompanied by reduced nonspecific binding;
-
The biosensor can immobilize phage antibodies more densely;
-
Phage antibodies, as opposed to full-length antibodies, are produced by E. coli cells, which significantly lowers production costs because no specialized equipment is needed for the culture of hybridoma cells [75].

4. Phage Antibodies for Virus Detection/Identification

Phage display technology antibodies as nano-bio-engineered macromolecules can be used for sensor-based virus detection. In order to recognize lethal viruses such as Zika, Ebola, Hendra, Nipah, Hanta, Middle East respiratory disease (MERS), and SARS, phage display has been utilized to create new diagnostic tools [76,77]. In the current situation of the COVID-19 pandemic, researchers actively search for neutralizing antibodies against SARS-CoV-2 for therapeutic use. Because phage display is an important antibody selection method, the prospects for the use of phage display, with special emphasis on its use in the diagnosis and therapy of coronavirus diseases, were shown by Anand et al. [77].
By now, a huge pool of antibodies against various viruses has been obtained by phage display, by using naïve or immune libraries. From naïve antibody gene libraries, antibodies against human pathogenic viruses such as SARS coronavirus, dengue virus, influenza virus, Venezuelan equine encephalitis virus, norovirus, and hepatitis C virus have been developed by using recombinant viral proteins or complete viral particles [78,79]. Other antibodies have been chosen from immune antibody gene libraries that target the influenza virus, HIV, SARS coronavirus, yellow fever virus, and Western equine encephalitis virus [79]. Even semisynthetic libraries have been utilized to prepare antibodies specific to the influenza virus [80]. In the past, libraries from several species have been used to successfully isolate virus-specific antibodies.
By using phage display, potential therapeutic agents are being developed for other human coronaviruses, such as SARS [81] and MERS [82], and for animal coronaviruses, such as infectious bronchitis virus, TGEV, and porcine epidemic diarrhea virus. High-affinity/small-molecule antibodies have been identified for the E, N, and S proteins of SARS-CoV [77]. For example, Ubah and Palliyil [83] described the advantages of phage antibodies and the prospects for their use against various viral targets. Other VHH segments produced via phage display and animal immunization include those that bind to Rotavirus gp6, H5 hemagglutinin to inhibit H5N1 influenza virus replication, and VHH that recognizes the tail of infectious phage in Lactococcus bacteria.
Examples of the use of phage antibodies for virus detection using classical methods are presented in Table 2.
Undoubtedly, phage antibodies are an excellent alternative to classical antibodies as sensing elements in virus diagnostics by biosensors (Table 3). For example, Kim et al. [180] developed a COVID-19 biosensor based on the lateral flow immunoassay (LFIA). They used three rounds of biopanning to generate phage antibodies (from a naïve chicken phage library) with a single-chain variable fragment (scFv) and crystallizable fragment (Fc) combination, which was specific for SARS-CoV-2 nucleocapsid protein. The scFv–Fc antibodies bound specifically and with high affinity to the nucleocapsid protein of SARS-CoV-2 but not to those other coronaviruses. The detection limit for this virus was 2 ng of antigenic protein. The biosensor could selectively detect SARS-CoV-2 virus within 20 min without detecting SARS-CoV or MERS-CoV virus. In addition, the assay was made more accurate with a portable LFIA reader. When the LFIA biosensor was mounted onto a portable reader, the obtained image was analyzed automatically with LED and CMOS sensors [180].
A sandwiched phage-based enzyme-linked chemiluminescence immunoassay was created by Liu et al. [195] by using the discovered phage expressing a particular peptide as a bifunctional probe. The probe was capable of recognizing the SARS-CoV-2 S1 antigen and amplifying the signal. The method is useful for the detection of SARS-CoV-2 pseudovirus in saliva.
After three rounds of biopanning, Yang et al.’s [199] screening of a phage library for affinity to the SARS-CoV-2 spike protein produced five SARS-CoV-2-specific phage miniantibodies. The resulting antibodies were used to develop a rapid and cost-effective test for the detection of the SARS-CoV-2 virus. With the aid of a handy fluorometer, phage antibodies were successfully transformed into fluorescent immunosensors that allowed quick detection of the SARS-CoV-2 viral antigen [196]. From a combinatorial library, high-affinity synthetic antibodies were isolated by phage display and tagged with gold nanoparticles [197]. These antibodies were successful in identifying the SARS-CoV-2 spike-protein receptor-binding domain and the glycoprotein released by the Ebola virus.
A human combinatorial scFv antibody library was used to retrieve a panel of recombinant single-chain antibodies (scFvs) against structural proteins of tomato spotted wilt virus via phage display [200]. Using phage display to create recombinant antibodies, Dong et al. [201] investigated the specificity of clone binding to the CS protein and hemagglutinin of the H5N1 avian influenza virus. It has been shown that the antibodies identified only epitopes distinct from those seen in other influenza subtypes, enabling quick identification of the H5N1 virus. In order to quickly and accurately detect the H5N1 virus, these recombinant Fab fragments can be employed as a receptor component of sensor systems.
To distinguish avian influenza virus (H5N1) from seasonal influenza viruses (H1N1 and H3N2) (whose nucleoproteins are 90–94% similar in amino acid sequence), Yu et al. [202] developed a phage display-based methodology for obtaining antibodies as affinity reagents against closely related influenza virus nucleoprotein subtypes.
The HBsAg of the hepatitis B virus was demonstrated by ELISA to have a high affinity for the generated phage scFvs, which allowed them to bind to the antigen on the membrane of the infected cells. One of the clones was capable of being internalized into HepG2.2.15 cells that were HBsAg positive, according to indirect fluorescent labeling examination. The ability to internalize phage antibodies to deliver medications to cells that are infected with the hepatitis B virus may be highly promising [120].
By using phage display, methods have been developed to generate specific single-chain variable fragments (scFvs) against dengue fever virus [203] and infectious bursal disease virus (IBDV) [204]. The prepared miniantibodies were used in ELISA to detect viruses in the sera of infected people.
Phage antibody-based ELISA versions have been created for the detection of human disease-causing viruses such as HIV, herpes simplex virus, poliovirus, polyomavirus, rabies virus, respiratory syncytial virus, rotavirus, Sin Nombre virus, Usutu virus, West Nile virus, yellow fever virus, Zika virus, human metapneumovirus, Japanese encephalitis virus, MERS, norovirus, paramyxovirus, chikungunya virus, Ebola virus, enterovirus 71, Epstein-Barr virus, hantavirus, human cytomegalovirus, Hendra virus, Nipah virus, and hepatitis A, C, and E viruses [65].
A useful natural resource for investigating polyvalent interactions with particular antibodies is bacterial viruses. Such research is very useful in the real world and has been used to create improved virus detection methods. For example, Guliy et al. [198] showed the possibility of bacteriophage FAI-Sp59b detection in the presence of foreign viral particles.
The possibility of using phage display antibodies in veterinary medicine was demonstrated in [74]. The great selection potential of phage display antibodies allowed mouse scFvs to recognize the avian influenza virus with excellent sensitivity and specificity [140]. It should be noted that the IBDV virus seriously harms the chicken industry. Owing to the diversity of IBDV strains, it has long been challenging to distinguish between the very virulent classical variation of IBDV (vvIBDV) and the vaccine strain. A single-chain variable fragment that effectively identified the highly conformational epitope of the VP2 protein of vvIBDV was obtained by Sapats et al. [205]. These phage antibodies are perfect for diagnostic techniques such as ELISA since they successfully distinguish vvIBDV from other strains [205,206]. Phage antibodies and an immunoassay were used in related research to find duck hepatitis A virus (DHAV) [99].
One of the most destructive viral illnesses in the dairy and meat sectors is foot-and-mouth disease (FMD). It is known that there are seven FMD serotypes: O, A, C, Asia1, SAT1, SAT2, and SAT3. Therefore, a quick and precise way of differentiating between vaccinated and infected animals is essential to the effectiveness of diagnosis and vaccination. Recombinant chicken scFvs against the FMD 3ABC protein have been produced and demonstrated to be effective for separating infected from vaccine-exposed animals via phage display [110,207]. Additionally, mouse scFvs directed against the VP2 protein of the FMD virus showed a diagnostic capability for a number of serotypes of this virus [113]. For the serological detection of bovine immunodeficiency virus in cattle, recombinant mouse scFv antibodies were utilized in an ELISA with competitive inhibition [87].
The promise of phage antibodies as an experimental tool has been demonstrated to visualize [159] and diagnose [161,208] porcine epidemic diarrhea virus, to diagnose canine parvovirus type 2 (CPV2) [91], and to detect Western and Venezuelan equine encephalitis viruses [189,209].
Possibilities have been shown to detect plant viruses (broad bean mottle virus, grapevine leafroll–associated virus 3, cucumber mosaic cucumovirus, plum pox virus, and grapevine virus B) and zoonotic viruses (Australian bat lyssavirus, bovine viral diarrhea virus, bluetongue virus, classical swine fever virus, canine parvovirus, hematopoietic necrosis virus, ectromelia virus, porcine circovirus, Newcastle disease virus, simian immunodeficiency virus, porcine reproductive and respiratory syndrome virus, transmissible gastroenteritis virus, swine influenza virus, white spot syndrome virus, and vaccinia virus) with phage antibodies and ELISA [65,74].
Norovirus, Maedi-visna, SARS-CoV, Sin Nombre, and duck hepatitis A viruses have been identified with the help of dot-blot immunoassay and phage antibodies [65,74]. Phage antibodies are employed in immunochromatographic assay techniques to diagnose infections brought on by the avian influenza virus, SARS-CoV-2, and dengue virus [65,74].
Note that surface plasmon resonance (SPR) sensors for the detection of chemical and biological species are often the “gold standard” [210,211,212]. SPR biosensors are also used for virus detection. Phage antibodies were used as part of the SPR biosensor for the detection of norovirus [154], porcine circovirus [159], cowpea mosaic virus [213], SARS-CoV-2 [214], etc.
As can be observed from the data displayed, biosensors enable a large decrease in the analysis time owing to the relative simplicity of the procedures, are quite sensitive, and require little pretreatment of the material under examination. The instrumental implementation of these methods should ensure high accuracy of measurements, and in turn, measurements should be made automatically by moderately skilled personnel. When biosensors are designed for the detection of viruses and virus-like particles, the following criteria should be taken into account: property of being all-purpose, ability to be used with various objects, rapidity of detection, specificity, sensitivity, portability, simplicity, ability to use small sample volumes, and low cost [215].

5. Discussion and Prospects

The selection of antibody fragments from phage display libraries thus offers the potential to access very large numbers of molecules with different binding specificities quickly and cheaply, avoiding the need for animal immunizations. The scFv can be genetically engineered to produce tailored constructs as described above. Such phage antibodies can be coated directly onto metal, plastic, silica or carbon surfaces by simple adsorption, which, as discussed above, is adequate for biosensor-based assays.
Viruses are pervasive life forms that need a host to reproduce. From bacteria to plants and animals to viruses, a wide variety of organisms may become infected. The genome, which is represented by double- or single-stranded DNA or RNA, and the capsid are the two primary structural parts of viruses. A small number of proteins encoded by the viral genome are present in numerous copies to create the extremely symmetrical capsid [216]. Methods for virus detection are employed in contemporary virological research for a variety of applications. They can be used in epidemiology to track and manage pandemic outbreaks of illnesses caused by viruses, including swine flu, SARS-CoV-2, and many others. Diagnostic virus detection methods are central in controlling the spread of viruses and help contain viral infections. Despite its drawbacks, the use of PCR to identify particular viral genomic sequences while an infection is still active is still regarded as the best virus detection technique. However, sensor technologies using phage antibodies as sensitive elements are very promising because antibodies generated by phage display are an excellent alternative to classical antibodies. Since its development in 1985, phage display has been a crucial and successful molecular biology technique that has remained essential for the scientific community. A phage library may include millions, or perhaps billions, of distinct and distinctive mapping peptide ligands because a huge number of nucleotide fragments can be cloned into the phage genome. The epitope mapping and antigen presentation on the bacteriophage surface, which constitute the basis for phage display, have been exploited in affinity selection-based biopanning to screen for potential novel vaccine candidates [77].
Phage display is a powerful tool for target ligand selection owing to its being simple, highly effective, rapid, and cheap. Efficient biopanning selection leads to the isolation of ligands with unique, specific, and desired functional characteristics. Phage antibody display offers multiple platforms for the use of antigen-binding; however, the diversity and stability of the library still need to be improved.
In the past few years, the need for highly sensitive bioanalytical methods to diagnose and monitor socially significant diseases has increased. This is due, on the one hand, to the growth of the disease in general, including the unprecedented spread of the new coronavirus infection, SARS-CoV-2, and, on the other hand, to the need to overcome the limitations of current biomedical diagnostic methods. For example, because of puncture invasiveness, a tissue biopsy cannot always be performed, and the results of a single biopsy often cannot provide sufficient information in real-time to diagnose the disease. PCR is currently one of the most popular tools for the rapid detection of viral infections. Nucleic acid-based virus detection usually provides high sensitivity but may require trained personnel and be time-consuming and expensive. The use of isothermal amplification systems may reduce equipment costs, making these systems indispensable when high-performance and rapid cycling testing are required. Alternatives to PCR are immunoassays, which offer reliability and cost-effectiveness. In addition, some immunoassays can be modified with lateral flow technology, which greatly speeds up the generation of results. However, immunoassays are usually inferior in sensitivity to PCR. Along with these methods, the use of next-generation sequencing can provide promising results. In addition, the ability to sequence a large number of viral genomes will provide researchers with expanded information about them and will help in tracing infections.
Characteristics such as high throughput, ease of use, and short running time play an important part, especially in viral outbreaks, in addition to the general requirement for the accuracy, validity, and specificity of viral assays. This strategy enables simple, cheap, rapid, and sensitive detection of specific pathogens, which shows great potential in virus analysis in situ.
The growing advances in phage display and the use of antibodies in the diagnosis of viral diseases suggest this technology will be improved further. This progress will gradually drive innovation to understand the mechanisms involved in infection and to design post-exposure therapies for SARS-CoV-2 and other viral infections.

Author Contributions

O.I.G.: Conceptualization and design, Supervision, Formal Analysis, Writing—Original Draft, Writing—Review and Editing. S.S.E.: Writing—Review and Editing. V.A.K.: Review and Editing. L.A.D.: Project Administration, Formal Analysis and Design, Writing—Original Draft, Writing—Review and Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Ministry of Science and Higher Education of the Russian Federation as a state assignment for the Saratov Scientific Centre of the Russian Academy of Sciences.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. van Rijn, P.; Schirhagl, R. Viruses, artificial viruses and virus-based structures for biomedical applications. Adv. Healthc. Mater. 2016, 5, 1386–1400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Zaidi, S.A. An overview of bio-inspired intelligent imprinted polymers for virus determination. Biosensors 2021, 11, 89. [Google Scholar] [CrossRef] [PubMed]
  3. Ma, Y.; Nolte, R.J.; Cornelissen, J.J. Virus-based nanocarriers for drug delivery. Adv. Drug Deliv. Rev. 2012, 64, 811–825. [Google Scholar] [CrossRef] [PubMed]
  4. Wu, Y.; Yang, H.; Shin, H.J. Viruses as self-assembled nanocontainers for encapsulation of functional cargoes. Korean J. Chem. Eng. 2013, 30, 1359–1367. [Google Scholar] [CrossRef]
  5. Manchester, M.; Singh, P. Virus-based nanoparticles (VNPs): Platform technologies for diagnostic imaging. Adv. Drug Deliv. Rev. 2006, 58, 1505–1522. [Google Scholar] [CrossRef]
  6. Steinmetz, N.F.; Mertens, M.E.; Taurog, R.E.; Johnson, J.E.; Commandeur, U.; Fischer, R.; Manchester, M. Potato virus X as a novel platform for potential biomedical applications. Nano Lett. 2010, 10, 305–312. [Google Scholar] [CrossRef] [Green Version]
  7. Lee, J.H.; Kim, B.C.; Oh, B.K.; Choi, J.W. Rapid and sensitive determination of HIV-1 virus based on surface enhanced Raman spectroscopy. J. Biomed. Nanotechnol. 2015, 11, 2223–2230. [Google Scholar] [CrossRef]
  8. Rappuoli, R.; De Gregorio, E.; Del Giudice, G.; Phogat, S.; Pecetta, S.; Pizza, M.; Hanon, E. Vaccinology in the post-COVID-19 era. Proc. Natl. Acad. Sci. USA 2021, 118, e2020368118. [Google Scholar] [CrossRef]
  9. Daniel, M.C.; Tsvetkova, I.B.; Quinkert, Z.T.; Murali, A.; De, M.; Rotello, V.M.; Kao, C.C.; Dragnea, B. Role of surface charge density in nanoparticle-templated assembly of bromovirus protein cages. ACS Nano 2010, 4, 3853–3860. [Google Scholar] [CrossRef] [Green Version]
  10. Steinmetz, N.F.; Lin, T.; Lomonossoff, G.P.; Johnson, J.E. Structure-based engineering of an icosahedral virus for nanomedicine and nanotechnology. In Viruses and Nanotechnology. Current Topics in Microbiology and Immunology; Manchester, M., Steinmetz, N.F., Eds.; Springer: Berlin/Heidelberg, Germany, 2009; Volume 327, pp. 23–58. [Google Scholar] [CrossRef]
  11. Young, M.; Willits, D.; Uchida, M.; Douglas, T. Plant viruses as biotemplates for materials and their use in nanotechnology. Annu. Rev. Phytopathol. 2008, 46, 361–384. [Google Scholar] [CrossRef]
  12. Nam, K.T.; Kim, D.W.; Yoo, P.J.; Chiang, C.Y.; Meethong, N.; Hammond, P.T.; Chiang, Y.M.; Belcher, A.M. Virus-enabled synthesis and assembly of nanowires for lithium ion battery electrodes. Science 2006, 312, 885–888. [Google Scholar] [CrossRef] [Green Version]
  13. Tseng, R.J.; Tsai, C.; Ma, L.; Ouyang, J.; Ozkan, C.S.; Yang, Y. Digital memory device based on tobacco mosaic virus conjugated with nanoparticles. Nat. Nanotechnol. 2006, 1, 72–77. [Google Scholar] [CrossRef]
  14. Steinmetz, N.F.; Bize, A.; Findlay, K.C.; Lomonossoff, G.P.; Manchester, M.; Evans, D.J.; Prangishvili, D. Site-specific and spatially controlled addressability of a new viral nanobuilding block: Sulfolobus islandicus rod-shaped virus 2. Adv. Funct. Mater. 2008, 18, 3478–3486. [Google Scholar] [CrossRef]
  15. Rong, J.; Niu, Z.; Andrew Lee, L.; Wang, Q. Self-assembly of viral particles. Curr. Opin. Colloid Interface Sci. 2011, 16, 441–450. [Google Scholar] [CrossRef]
  16. Willats, W.G. Phage display: Practicalities and prospects. Plant Mol. Biol. 2002, 50, 837–854. [Google Scholar] [CrossRef]
  17. Wang, L.F.; Yu, M. Epitope identification and discovery using phage display libraries: Applications in vaccine development and diagnostics. Curr. Drug Targets 2004, 5, 1–15. [Google Scholar] [CrossRef]
  18. Kuzmicheva, G.A.; Belyavskaya, V.A. Peptide phage display in biotechnology and biomedicine. Biomed. Khim. 2016, 62, 481–495. [Google Scholar] [CrossRef] [Green Version]
  19. Smith, G.P. Filamentous fusion phage: Novel expression vectors that display cloned antigens on the virion surface. Science 1985, 228, 1315–1317. [Google Scholar] [CrossRef]
  20. Williams, D.D.; Benedek, O.; Turnbough, C.L., Jr. Species-specific peptide ligands for the detection of Bacillus anthracis spores. Appl. Environ. Microbiol. 2003, 69, 6288–6293. [Google Scholar] [CrossRef] [Green Version]
  21. Paoli, G.C.; Chen, C.-Y.; Brewster, J.D. Single-chain Fv antibody with specificity for Listeria monocytogenes. J. Immunol. Methods 2004, 289, 147–155. [Google Scholar] [CrossRef]
  22. Nanduri, V.; Bhunia, A.K.; Tu, S.-I.; Paoli, G.C.; Brewster, J.D. SPR biosensor for the detection of L. monocytogenes using phage-displayed antibody. Biosens. Bioelectron. 2007, 23, 248–252. [Google Scholar] [CrossRef] [PubMed]
  23. McCafferty, J.; Griffiths, A.D.; Winter, G.; Chiswell, D.J. Phage antibodies: Filamentous phage displaying antibody variable domains. Nature 1990, 348, 552–554. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, P.; Yu, G.; Wei, J.; Liao, X.; Zhang, Y.; Ren, Y.; Zhang, C.; Wang, Y.; Zhang, D.; Wang, J.; et al. A single thiolated-phage displayed nanobody-based biosensor for label-free detection of foodborne pathogen. J. Hazard. Mater. 2023, 443, 130157. [Google Scholar] [CrossRef]
  25. Krejcova, L.; Michalek, P.; Rodrigo, M.M.; Heger, Z.; Krizkova, S.; Vaculovicova, M.; Hynek, D.; Adam, V.; Kizek, R. Nanoscale virus biosensors: State of the art. Nanobiosens. Dis. Diagn. 2015, 4, 47–66. [Google Scholar] [CrossRef] [Green Version]
  26. Song, M.; Yang, M.; Hao, J. Pathogenic virus detection by optical nanobiosensors. Cell Rep. Phys. Sci. 2021, 2, 100288. [Google Scholar] [CrossRef]
  27. Aghamirza Moghim Aliabadi, H.; Eivazzadeh-Keihan, R.; Beig Parikhani, A.; Fattahi Mehraban, S.; Maleki, A.; Fereshteh, S.; Bazaz, M.; Zolriasatein, A.; Bozorgnia, B.; Rahmati, S.; et al. COVID-19: A systematic review and update on prevention, diagnosis, and treatment. MedComm 2022, 3, e115. [Google Scholar] [CrossRef]
  28. WHO Coronavirus (COVID-19) Dashboard. 2023. Available online: https://covid19.who.int/ (accessed on 20 April 2023).
  29. Ammulu, M.A.; Nelluri, K.D.D.; Vemuri, P.K.; Mallampati, S.C.; Poda, S. COVID 19: A new insight into organ failure and complications caused by novel SARS-CoV-2 virus and discussion on the role of nanotechnology in detection, treatment and prevention of the disease. Curr. Trends Biotechnol. Pharm. 2022, 16, 577–602. [Google Scholar]
  30. World Health Statistics. 2022. Available online: https://www.who.int/news/item/20-05-2022-world-health-statistics-2022 (accessed on 20 April 2023).
  31. European Centre for Disease Prevention and Control. Communicable Disease Threats Report, 2–8 January 2023, Week 1. Available online: https://www.ecdc.europa.eu/en/publications-data/communicable-disease-threats-report-2-8-january-2023-week-1 (accessed on 20 April 2023).
  32. Internet Resource. Available online: https://www.bmglabtech.com/en/blog/virus-detection-methods/ (accessed on 7 April 2023).
  33. Hsiung, G.D. Diagnostic virology: From animals to automation. Yale J. Biol. Med. 1984, 57, 727–733. [Google Scholar]
  34. Leland, D.S.; Ginocchio, C.C. Role of cell culture for virus detection in the age of technology. Clin. Microbiol. Rev. 2007, 20, 49–78. [Google Scholar] [CrossRef] [Green Version]
  35. Coiras, M.T.; Aguilar, J.C.; García, M.L.; Casas, I.; Pérez-Breña, P. Simultaneous detection of fourteen respiratory viruses in clinical specimens by two multiplex reverse transcription nested-PCR assays. J. Med. Virol. 2004, 72, 484–495. [Google Scholar] [CrossRef]
  36. Syrmis, M.W.; Whiley, D.M.; Thomas, M.; Mackay, I.M.; Williamson, J.; Siebert, D.J.; Nissen, M.D.; Sloots, T.P. A sensitive, specific, and cost-effective multiplex reverse transcriptase-PCR assay for the detection of seven common respiratory viruses in respiratory samples. J. Mol. Diagn. 2004, 6, 125–131. [Google Scholar] [CrossRef] [Green Version]
  37. Templeton, K.E.; Scheltinga, S.A.; Beersma, M.F.C.; Kroes, A.C.M.; Claas, E.C.J. Rapid and sensitive method using multiplex real-time PCR for diagnosis of infections by influenza a and influenza B viruses, respiratory syncytial virus, and parainfluenza viruses 1, 2, 3, and 4. J. Clin. Microbiol. 2004, 42, 1564–1569. [Google Scholar] [CrossRef] [Green Version]
  38. Samson, R.; Navale, G.R.; Dharne, M.S. Biosensors: Frontiers in rapid detection of COVID-19. 3 Biotech 2020, 10, 385. [Google Scholar] [CrossRef]
  39. Zhou, H.; Liu, D.; Ma, L.; Ma, T.; Xu, T.; Ren, L.; Li, L.; Xu, S. A SARS-CoV-2 reference standard quantified by multiple digital PCR platforms for quality assessment of molecular tests. Anal. Chem. 2021, 93, 715–721. [Google Scholar] [CrossRef]
  40. Mehra, B.; Bhattar, S.; Bhalla, P.; Rawat, D. Rapid tests versus ELISA for screening of HIV infection: Our experience from a voluntary counselling and testing facility of a tertiary care centre in North India. ISRN AIDS 2014, 2014, 296840. [Google Scholar] [CrossRef]
  41. Centers for Disease Control and Prevention. Dengue. 2019. Available online: https://www.cdc.gov/dengue/healthcare-providers/diagnosis.html (accessed on 20 April 2023).
  42. Lin, B.; Blaney, K.M.; Malanoski, A.P.; Ligler, A.G.; Schnur, J.M.; Metzgar, D.; Russell, K.L.; Stenger, D.A. Using a resequencing microarray as a multiple respiratory pathogen detection assay. J. Clin. Microbiol. 2007, 45, 443–452. [Google Scholar] [CrossRef] [Green Version]
  43. Mehlmann, M.; Bonner, A.B.; Williams, J.V.; Dankbar, D.M.; Moore, C.L.; Kuchta, R.D.; Podsiad, A.B.; Tamerius, J.D.; Dawson, E.D.; Rowlen, K.L. Comparison of the MChip to viral culture, reverse transcription-PCR, and the QuickVue influenza A+B test for rapid diagnosis of influenza. J. Clin. Microbiol. 2007, 45, 1234–1237. [Google Scholar] [CrossRef] [Green Version]
  44. Huguenin, A.; Moutte, L.; Renois, F.; Leveque, N.; Talmud, D.; Abely, M.; Nguyen, Y.; Carrat, F.; Andreoletti, L. Broad respiratory virus detection in infants hospitalized for bronchiolitis by use of a multiplex RT-PCR DNA microarray system. J. Med. Virol. 2012, 84, 979–985. [Google Scholar] [CrossRef]
  45. Ince, B.; Sezgintürk, M.K. Lateral flow assays for viruses diagnosis: Up-to-date technology and future prospects. Trends Analyt. Chem. 2022, 157, 116725. [Google Scholar] [CrossRef]
  46. Rahman, M.M. Progress in electrochemical biosensing of SARS-CoV-2 virus for COVID-19 management. Chemosensors 2022, 10, 287. [Google Scholar] [CrossRef]
  47. Purohit, B.; Vernekar, P.R.; Shetti, N.P.; Chandra, P. Biosensor nanoengineering: Design, operation, and implementation for biomolecular analysis. Sens. Int. 2020, 1, 100040. [Google Scholar] [CrossRef]
  48. Ribeiro, B.V.; Cordeiro, T.A.R.; Oliveira e Freitas, G.R.; Ferreira, L.F.; Franco, D.L. Biosensors for the detection of respiratory viruses: A review. Talanta Open 2020, 2, 100007. [Google Scholar] [CrossRef] [PubMed]
  49. Monteil, S.; Casson, A.J.; Jones, S.T. Electronic and electrochemical viral detection for point-of-care use: A systematic review. PLoS ONE 2021, 16, e0258002. [Google Scholar] [CrossRef] [PubMed]
  50. Kumar, N.; Shetti, N.P.; Jagannath, S.; Aminabhavi, T.M. Electrochemical sensors for the detection of SARS-CoV-2 virus. Chem. Eng. J. 2022, 430, 132966. [Google Scholar] [CrossRef] [PubMed]
  51. Zhang, Q.; Li, L.; Qiao, Z.; Lei, C.; Fu, Y.; Xie, Q.; Yao, S.; Li, Y.; Ying, Y. Electrochemical conversion of Fe3O4 magnetic nanoparticles to electroactive Prussian blue analogues for self-sacrificial label biosensing of avian influenza virus H5N1. Anal. Chem. 2017, 89, 12145–12151. [Google Scholar] [CrossRef]
  52. Mahari, S.; Roberts, A.; Shahdeo, D.; Gandhi, S. eCovSens-ultrasensitive novel in-house built printed circuit board based electrochemical device for rapid detection of nCovid-19 antigen, a spike protein domain I of SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  53. Białobrzeska, W.; Ficek, M.; Dec, B.; Osella, S.; Trzaskowski, B.; Jaramillo-Botero, A.; Pierpaoli, M.; Rycewicz, M.; Dashkevich, Y.; Łęga, T.; et al. Performance of electrochemical immunoassays for clinical diagnostics of SARS-CoV-2 based on selective nucleocapsid N protein detection: Boron-doped diamond, gold and glassy carbon evaluation. Biosens. Bioelectron. 2022, 209, 114222. [Google Scholar] [CrossRef]
  54. Lesniewski, A.; Los, M.; Jonsson-Niedziółka, M.; Krajewska, A.; Szot, K.; Los, J.M.; Niedziolka-Jonsson, J. Antibody modified gold nanoparticles for fast and selective, colorimetric T7 bacteriophage detection. Bioconjug. Chem. 2014, 25, 644–648. [Google Scholar] [CrossRef]
  55. Evtugyn, G.; Porfireva, A.; Stepanova, V.; Kutyreva, M.; Gataulina, A.; Ulakhovich, N.; Evtugyn, V.; Hianik, T. Impedimetric aptasensor for ochratoxin A determination based on Au nanoparticles stabilized with hyper-branched polymer. Sensors 2013, 13, 16129–16145. [Google Scholar] [CrossRef]
  56. Darwish, N.T.; Sekaran, S.D.; Alias, Y.; Khor, S.M. Immunofluorescence-based biosensor for the determination of dengue virus NS1 in clinical samples. J. Pharm. Biomed. Anal. 2018, 149, 591–602. [Google Scholar] [CrossRef]
  57. Guliy, O.I.; Kanevskiy, M.V.; Fomin, A.S.; Staroverov, S.A.; Bunin, V.D. Progress in the use of an electro-optical sensor for virus detection. Opt. Commun. 2020, 465, 125605. [Google Scholar] [CrossRef]
  58. Ballantine, D.S.; White, R.M.; Martin, S.J.; Ricco, A.J.; Zellers, E.T.; Frye, G.C.; Wohltjen, H. Acoustic Wave Sensors: Theory, Design, and Physico-Chemical Applications, 1st ed.; Academic Press: San Diego, CA, USA, 1997. [Google Scholar]
  59. König, B.; Grätzel, M. A novel immunosensor for herpes viruses. Anal. Chem. 1994, 66, 341–344. [Google Scholar] [CrossRef]
  60. Bisoffi, M.; Hjelle, B.; Brown, D.C.; Branch, D.W.; Edwards, T.L.; Brozik, S.M.; Bondu-Hawkins, V.S.; Larson, R.S. Detection of viral bioagents using a shear horizontal surface acoustic wave biosensor. Biosens. Bioelectron. 2008, 23, 1397–1403. [Google Scholar] [CrossRef]
  61. Tamarin, O.; Comeau, S.; Déjous, C.; Moynet, D.; Rebière, D.; Bezian, J.; Pistré, J. Real time device for biosensing: Design of a bacteriophage model using love acoustic waves. Biosens. Bioelectron. 2003, 18, 755–763. [Google Scholar] [CrossRef]
  62. Guliy, O.; Zaitsev, B.; Teplykh, A.; Balashov, S.; Fomin, A.; Staroverov, S.; Borodina, I. Acoustical slot mode sensor for the rapid coronaviruses detection. Sensors 2021, 21, 1822. [Google Scholar] [CrossRef]
  63. Tessaro, L.; Aquino, A.; Panzenhagen, P.; Joshi, N.; Conte-Junior, C.A. A systematic review of the advancement on colorimetric nanobiosensors for SARS-CoV-2 detection. J. Pharm. Biomed. Anal. 2023, 222, 115087. [Google Scholar] [CrossRef]
  64. Tadesse, L.F.; Safir, F.; Ho, C.-S.; Hasbach, X.; Khuri-Yakub, B.P.; Jeffrey, S.S.; Saleh, A.A.E.; Dionne, J. Toward rapid infectious disease diagnosis with advances in surface-enhanced Raman spectroscopy. J. Chem. Phys. 2020, 152, 240902. [Google Scholar] [CrossRef]
  65. Roth, K.D.R.; Wenzel, E.V.; Ruschig, M.; Steinke, S.; Langreder, N.; Heine, P.A.; Schneider, K.-T.; Ballmann, R.; Fühner, V.; Kuhn, P.; et al. Developing recombinant antibodies by phage display against infectious diseases and toxins for diagnostics and therapy. Front. Cell. Infect. Microbiol. 2021, 11, 697876. [Google Scholar] [CrossRef]
  66. Rülker, T.; Voß, L.; Thullier, P.; O’ Brien, L.M.; Pelat, T.; Perkins, S.D.; Langermann, C.; Schirrmann, T.; Dübel, S.; Marschall, H.-J.; et al. Isolation and characterisation of a human-like antibody fragment (scFv) that inactivates VEEV in vitro and in vivo. PLoS ONE 2012, 7, e37242. [Google Scholar] [CrossRef] [Green Version]
  67. Goncalvez, A.P.; Chien, C.-H.; Tubthong, K.; Gorshkova, I.; Roll, C.; Donau, O.; Schuck, P.; Yoksan, S.; Wang, S.-D.; Purcell, R.H.; et al. Humanized monoclonal antibodies derived from chimpanzee Fabs protect against Japanese encephalitis virus in vitro and in vivo. J. Virol. 2008, 82, 7009–7021. [Google Scholar] [CrossRef] [Green Version]
  68. Fatima, A.; Wang, H.; Kang, K.; Xia, L.; Wang, Y.; Ye, W.; Wang, J.; Wang, X. Development of VHH antibodies against dengue virus type 2 NS1 and comparison with monoclonal antibodies for use in immunological diagnosis. PLoS ONE 2014, 9, e95263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Liu, S.; Moayeri, M.; Leppla, S.H. Anthrax lethal and edema toxins in anthrax pathogenesis. Trends Microbiol. 2014, 22, 317–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. van Wyngaardt, W.; Malatji, T.; Mashau, C.; Fehrsen, J.; Jordaan, F.; Miltiadou, D.; du Plessis, D.H. A large semi-synthetic single-chain Fv phage display library based on chicken immunoglobulin genes. BMC Biotechnol. 2004, 4, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Tikunova, N.; Dubrovskaya, V.; Morozova, V.; Yun, T.; Khlusevich, Y.; Bormotov, N.; Laman, A.; Brovko, F.; Shvalov, A.; Belanov, E. The neutralizing human recombinant antibodies to pathogenic Orthopoxviruses derived from a phage display immune library. Virus Res. 2012, 163, 141–150. [Google Scholar] [CrossRef] [PubMed]
  72. Köhler, G.; Milstein, C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975, 256, 495–497. [Google Scholar] [CrossRef]
  73. Charlton, K.A.; Moyle, S.; Porter, A.J.; Harris, W.J. Analysis of the diversity of a sheep antibody repertoire as revealed from a bacteriophage display library. J. Immunol. 2000, 164, 6221–6229. [Google Scholar] [CrossRef] [Green Version]
  74. Bashir, S.; Paeshuyse, J. Construction of antibody phage libraries and their application in veterinary immunovirology. Antibodies 2020, 9, 21. [Google Scholar] [CrossRef]
  75. Peltomaa, R.; Benito-Peña, E.; Barderas, R.; Moreno-Bondi, M.C. Phage display in the quest for new selective recognition elements for biosensors. ACS Omega 2019, 4, 11569–11580. [Google Scholar] [CrossRef]
  76. Kuhn, P.; Fühner, V.; Unkauf, T.; Moreira, G.M.S.G.; Frenzel, A.; Miethe, S.; Hust, M. Recombinant antibodies for diagnostics and therapy against pathogens and toxins generated by phage display. Proteom. Clin. Appl. 2016, 10, 922–948. [Google Scholar] [CrossRef]
  77. Anand, T.; Virmani, N.; Bera, B.C.; Vaid, R.K.; Vashisth, M.; Bardajatya, P.; Kumar, A.; Tripathi, B.N. Phage display technique as a tool for diagnosis and antibody selection for coronaviruses. Curr. Microbiol. 2021, 78, 1124–1134. [Google Scholar] [CrossRef]
  78. Velappan, N.; Martinez, J.S.; Valero, R.; Chasteen, L.; Ponce, L.; Bondu-Hawkins, V.; Kelly, C.; Pavlik, P.; Hjelle, B.; Bradbury, A.R.M. Selection and characterization of scFv antibodies against the Sin Nombre hantavirus nucleocapsid protein. J. Immunol. Methods 2007, 321, 60–69. [Google Scholar] [CrossRef]
  79. Unkauf, T.; Miethe, S.; Fühner, V.; Schirrmann, T.; Frenzel, A.; Hust, M. Generation of recombinant antibodies against toxins and viruses by phage display for diagnostics and therapy. In Protein Targeting Compounds. Advances in Experimental Medicine and Biology; Böldicke, T., Ed.; Springer: Cham, Switzerland, 2016; Volume 917, pp. 55–76. [Google Scholar] [CrossRef]
  80. Avnir, Y.; Tallarico, A.S.; Zhu, Q.; Bennett, A.S.; Connelly, G.; Sheehan, J.; Sui, J.; Fahmy, A.; Huang, C.-Y.; Cadwell, G.; et al. Molecular signatures of hemagglutinin stem-directed heterosubtypic human neutralizing antibodies against influenza A viruses. PLoS Pathog. 2014, 10, e1004103. [Google Scholar] [CrossRef]
  81. Walter, J.D.; Hutter, C.A.J.; Zimmermann, I.; Wyss, M.; Egloff, P.; Sorgenfrei, M.; Hürlimann, L.M.; Gonda, I.; Meier, G.; Remm, S.; et al. Sybodies targeting the SARS-CoV-2 receptor-binding domain. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  82. He, L.; Li, J.; Ren, S.; Sun, S.; Guo, Y.; Qiu, H.; Liao, Y.; Ji, K.; Fan, R.; Zhao, G.; et al. Construction and identification of nanobody phage display library targeting Middle East respiratory syndrome coronavirus. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2017, 33, 1662–1668. (In Chinese) [Google Scholar]
  83. Ubah, O.; Palliyil, S. Monoclonal antibodies and antibody like fragments derived from immunised phage display libraries. In Recombinant Antibodies for Infectious Diseases. Advances in Experimental Medicine and Biology; Lim, T., Ed.; Springer: Cham, Switzerland, 2017; Volume 1053, pp. 99–117. [Google Scholar] [CrossRef]
  84. Weir, D.L.; Coggins, S.A.; Vu, B.K.; Coertse, J.; Yan, L.; Smith, I.L.; Laing, E.D.; Markotter, W.; Broder, C.C.; Schaefer, B.C. Isolation and Characterization of Cross-Reactive Human Monoclonal Antibodies That Potently Neutralize Australian Bat Lyssavirus Variants and Other Phylogroup 1 Lyssaviruses. Viruses 2021, 13, 391. [Google Scholar] [CrossRef]
  85. Gong, X.; Zhu, M.; Li, G.; Lu, X.; Wan, Y. Specific determination of influenza H7N2 virus based on biotinylated single-domain antibody from a phage displayed library. Anal. Biochem. 2016, 500, 66–72. [Google Scholar] [CrossRef] [PubMed]
  86. Ten Haaf, A.; Kohl, J.; Pscherer, S.; Hamann, H.P.; Eskens, H.U.; Bastian, M.; Gattenlöhner, S.; Tur, M.K. Development of a monoclonal sandwich ELISA for direct detection of bluetongue virus 8 in infected animals. J. Virol. Methods 2017, 243, 172–176. [Google Scholar] [CrossRef]
  87. Bhatia, S.; Gangil, R.; Gupta, D.S.; Sood, R.; Pradhan, H.K.; Dubey, S.C. Single-chain fragment variable antibody against the capsid protein of bovine immunodeficiency virus and its use in ELISA. J. Virol. Methods 2010, 167, 68–73. [Google Scholar] [CrossRef]
  88. Li, T.; Huang, M.; Xiao, H.; Zhang, G.; Ding, J.; Wu, P.; Zhang, H.; Sheng, J.; Chen, C. Selection and characterization of specific nanobody against bovine virus diarrhea virus (BVDV) E2 protein. PLoS ONE 2017, 12, e0178469. [Google Scholar] [CrossRef] [Green Version]
  89. Duan, H.; Ma, Z.; Xu, L.; Zhang, A.; Li, Z.; Xiao, S. A Novel Intracellularly Expressed NS5B-Specific Nanobody Suppresses Bovine Viral Diarrhea Virus Replication. Vet. Microbiol. 2020, 240, 108449. [Google Scholar] [CrossRef]
  90. Ghannam, A.; Kumari, S.; Muyldermans, S.; Abbady, A.Q. Camelid nanobodies with high affinity for broad bean mottle virus: A possible promising tool to immunomodulate plant resistance against viruses. Plant Mol. Biol. 2015, 87, 355–369. [Google Scholar] [CrossRef] [PubMed]
  91. Braganza, A.; Wallace, K.; Pell, L.; Parrish, C.R.; Siegel, D.L.; Mason, N.J. Generation and validation of canine single chain variable fragment phage display libraries. Vet. Immunol. Immunopathol. 2011, 139, 27–40. [Google Scholar] [CrossRef] [PubMed]
  92. Ge, S.; Xu, L.; Li, B.; Zhong, F.; Liu, X.; Zhang, X. Canine parvovirus is diagnosed and neutralized by chicken IgY-scFv generated against the virus capsid protein. Vet. Res. 2020, 51, 110. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, J.L.; Shriver-Lake, L.C.; Zabetakis, D.; Anderson, G.P.; Goldman, E.R. Selection and characterization of protective anti-chikungunya virus single domain antibodies. Mol. Immunol. 2019, 105, 190–197. [Google Scholar] [CrossRef] [PubMed]
  94. Ziegler, A.; Torrance, L.; Macintosh, S.M.; Cowan, G.H.; Mayo Cucumber, M.A. Mosaic cucumovirus antibodies from a synthetic phage display library. Virology 1995, 214, 235–238. [Google Scholar] [CrossRef] [Green Version]
  95. Moreland, N.J.; Tay, M.Y.F.; Lim, E.; Paradkar, P.N.; Doan, D.N.P.; Yau, Y.H.; Geifman Shochat, S.; Vasudevan, S.G. high affinity human antibody fragments to dengue virus nonstructural protein 3. PLoS Negl. Trop. Dis. 2010, 4, e881. [Google Scholar] [CrossRef]
  96. Shriver-Lake, L.C.; Liu, J.L.; Zabetakis, D.; Sugiharto, V.A.; Lee, C.R.; Defang, G.N.; Wu, S.L.; Anderson, G.P.; Goldman, E.R. Selection and characterization of anti-dengue NS1 single domain antibodies. Sci. Rep. 2018, 8, 18086. [Google Scholar] [CrossRef] [Green Version]
  97. Tunghirun, C.; Narkthong, V.; Chaicumpa, W.; Chimnaronk, S. Interference of dengue replication by blocking the access of 3′ SL RNA to the viral RNA-dependent RNA polymerase. Antivir. Res. 2020, 182, 104921. [Google Scholar] [CrossRef]
  98. Wang, Y.; Cui, P.; Zhu, S.; Meng, T.; Hao, F.; Zhu, G.; Zuo, W. Preparation of single-chain antibody against VP3 protein of duck hepatitis virus type 1 by phage display technology. J. Virol. Methods 2018, 257, 73–78. [Google Scholar] [CrossRef]
  99. Xue, W.; Zhao, Q.; Li, P.; Zhang, R.; Lan, J.; Wang, J.; Yang, X.; Xie, Z.; Jiang, S. Identification and characterization of a novel nanobody against duck hepatitis A virus type 1. Virology 2019, 528, 101–109. [Google Scholar] [CrossRef]
  100. Shingarova, L.N.; Tikunova, N.V.; Iun, T.E.; Chepurnov, A.A.; Aliev, T.K.; Batanova, T.A.; Boldyreva, E.F.; Nekrasova, O.V.; Toporova, V.A.; Panina, A.A.; et al. Recombinant full-size human antibody to Ebola virus. Bioorg. Khim. 2007, 33, 598–605. [Google Scholar] [CrossRef]
  101. Goodchild, S.A.; Dooley, H.; Schoepp, R.J.; Flajnik, M.; Lonsdale, S.G. Isolation and characterisation of ebolavirus-specific recombinant antibody fragments from murine and shark immune libraries. Mol. Immunol. 2011, 48, 2027–2037. [Google Scholar] [CrossRef]
  102. Flego, M.; Frau, A.; Accardi, L.; Mallano, A.; Ascione, A.; Gellini, M.; Fanunza, E.; Vella, S.; Di Bonito, P.; Tramontano, E. Intracellular human antibody fragments recognizing the VP35 protein of Zaire Ebola filovirus inhibit the protein activity. BMC Biotechnol. 2019, 19, 64. [Google Scholar] [CrossRef]
  103. Froude, J.W.; Herbert, A.S.; Pelat, T.; Miethe, S.; Zak, S.E.; Brannan, J.M.; Bakken, R.R.; Steiner, A.R.; Yin, G.; Hallam, T.J.; et al. Post-exposure protection in mice against sudan virus by a two antibody cocktail. Viruses 2018, 10, 286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Herbert, A.S.; Froude, J.W.; Ortiz, R.A.; Kuehne, A.I.; Dorosky, D.E.; Bakken, R.R.; Zak, S.E.; Josleyn, N.M.; Musiychuk, K.; Jones, R.M.; et al. Development of an antibody cocktail for treatment of Sudan virus infection. Proc. Natl. Acad. Sci. USA 2020, 117, 3768–3778. [Google Scholar] [CrossRef]
  105. Khlusevich, Y.; Matveev, A.; Baykov, I.; Bulychev, L.; Bormotov, N.; Ilyichev, I.; Shevelev, G.; Morozova, V.; Pyshnyi, D.; Tikunova, N. Phage display antibodies against ectromelia virus that neutralize variola virus: Selection and implementation for p35 neutralizing epitope mapping. Antivir. Res. 2018, 152, 18–25. [Google Scholar] [CrossRef]
  106. Chen, Z.; Ren, X.; Yang, J.; Dong, J.; Xue, Y.; Sun, L.; Zhu, Y.; Jin, Q. An elaborate landscape of the human antibody repertoire against enterovirus 71 infection is revealed by phage display screening and deep sequencing. mAbs 2017, 9, 342–349. [Google Scholar] [CrossRef] [Green Version]
  107. Thanongsaksrikul, J.; Srimanote, P.; Tongtawe, P.; Glab-Ampai, K.; Malik, A.A.; Supasorn, O.; Chiawwit, P.; Poovorawan, Y.; Chaicumpa, W. Identification and production of mouse scFv to specific epitope of enterovirus-71 virion protein-2 (VP2). Arch. Virol. 2018, 163, 1141–1152. [Google Scholar] [CrossRef]
  108. Phanthong, S.; Densumite, J.; Seesuay, W.; Thanongsaksrikul, J.; Teimoori, S.; Sookrung, N.; Poovorawan, Y.; Onvimala, N.; Guntapong, R.; Pattanapanyasat, K.; et al. Human Antibodies to VP4 Inhibit Replication of Enteroviruses Across Subgenotypes and Serotypes, and Enhance Host Innate Immunity. Front. Microbiol. 2020, 11, 562768. [Google Scholar] [CrossRef]
  109. Zhang, D.; Mao, Y.; Cao, Q.; Xiong, L.; Wen, J.; Chen, R.; Zhu, J. Generation and Characterization of a Novel Recombinant Antibody Against LMP1-TES1 of Epstein-Barr Virus Isolated by Phage Display. Viruses 2013, 5, 1131–1142. [Google Scholar] [CrossRef] [Green Version]
  110. Foord, A.J.; Muller, J.D.; Yu, M.; Wang, L.-F.; Heine, H.G. Production and application of recombinant antibodies to foot-and-mouth disease virus non-structural protein 3ABC. J. Immunol. Methods 2007, 321, 142–151. [Google Scholar] [CrossRef] [PubMed]
  111. Wang, D.; Yang, S.; Yin, S.; Shang, Y.; Du, P.; Guo, J.; He, J.; Cai, J.; Liu, X. Characterization of single-domain antibodies against Foot and Mouth Disease Virus (FMDV) serotype O from a camelid and imaging of FMDV in baby hamster kidney-21 cells with single-domain antibody-quantum dots probes. BMC Vet. Res. 2015, 11, 120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Harmsen, M.M.; Seago, J.; Perez, E.; Charleston, B.; Eblé, P.L.; Dekker, A. Isolation of single-domain antibody fragments that preferentially detect intact (146s) particles of foot-and-mouth disease virus for use in vaccine quality control. Front. Immunol. 2017, 8, 3389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Salem, R.; El-Kholy, A.A.; Ibrahim, M. Eight novel single chain antibody fragments recognising VP2 of foot-and-mouth disease virus serotypes A, O, and SAT 2. Virology 2019, 533, 145–154. [Google Scholar] [CrossRef]
  114. Orecchia, M.; Nölke, G.; Saldarelli, P.; Dell’Orco, M.; Uhde-Holzem, K.; Sack, M.; Martelli, G.; Fischer, R.; Schillberg, S. Generation and characterization of a recombinant antibody fragment that binds to the coat protein of grapevine leafroll-associated virus 3. Arch. Virol. 2008, 153, 1075–1084. [Google Scholar] [CrossRef] [PubMed]
  115. Saldarelli, P.; Keller, H.; Dell’Orco, M.; Schots, A.; Elicio, V.; Minafra, A. Isolation of recombinant antibodies (Scfvs) to grapevine virus B. J. Virol. Methods 2005, 124, 191–195. [Google Scholar] [CrossRef]
  116. Koch, J.; Liang, M.; Queitsch, I.; Kraus, A.A.; Bautz, E.K.F. Human recombinant neutralizing antibodies against hantaan virus G2 protein. Virology 2003, 308, 64–73. [Google Scholar] [CrossRef] [Green Version]
  117. Pereira, S.S.; Moreira-Dill, L.S.; Morais, M.S.S.; Prado, N.D.R.; Barros, M.L.; Koishi, A.C.; Mazarrotto, G.A.; Gonçalves, G.M.; Zuliani, J.P.; Calderon, L.A.; et al. Novel camelid antibody fragments targeting recombinant nucleoprotein of araucaria hantavirus: A prototype for an early diagnosis of hantavirus pulmonary syndrome. PLoS ONE 2014, 9, e108067. [Google Scholar] [CrossRef]
  118. Zhu, Z.; Bossart, K.N.; Bishop, K.A.; Crameri, G.; Dimitrov, A.S.; McEachern, J.A.; Feng, Y.; Middleton, D.; Wang, L.F.; Broder, C.C.; et al. Exceptionally potent cross-reactive neutralization of Nipah and Hendra viruses by a human monoclonal antibody. J. Infect. Dis. 2008, 197, 846–853. [Google Scholar] [CrossRef]
  119. Schofield, D.J.; Satterfield, W.; Emerson, S.U.; Purcell, R.H. Four chimpanzee monoclonal antibodies isolated by phage display neutralize hepatitis a virus. Virology 2002, 292, 127–136. [Google Scholar] [CrossRef] [Green Version]
  120. Wen, W.-H.; Qin, W.-J.; Gao, H.; Zhao, J.; Jia, L.-T.; Liao, Q.-H.; Meng, Y.-L.; Jin, B.-Q.; Yao, L.-B.; Chen, S.-Y.; et al. An hepatitis B virus surface antigen specific single chain of variable fragment derived from a natural immune antigen binding fragment phage display library is specifically internalized by HepG2.2.15 cells. J. Viral Hepat. 2007, 14, 512–519. [Google Scholar] [CrossRef]
  121. Barban, V.; Fraysse-Corgier, S.; Paranhos-Baccala, G.; Petit, M.; Manin, C.; Berard, Y.; Prince, A.M.; Mandrand, B.; Meulien, P. Identification of a human epitope in hepatitis C virus (HCV) core protein using a molecularly cloned antibody repertoire from a non-symptomatic, anti-HCV-positive patient. J. Gen. Virol. 2000, 81 Pt 2, 461–469. [Google Scholar] [CrossRef]
  122. Burioni, R.; Bugli, F.; Mancini, N.; Rosa, D.; Di Campli, C.; Moroncini, G.; Manzin, A.; Abrignani, S.; Varaldo, P.E.; Clementi, M.; et al. Nonneutralizing human antibody fragments against hepatitis C virus E2 glycoprotein modulate neutralization of binding activity of human recombinant Fabs. Virology 2001, 288, 29–35. [Google Scholar] [CrossRef] [Green Version]
  123. Jittavisutthikul, S.; Seesuay, W.; Thanongsaksrikul, J.; Thueng-In, K.; Srimanote, P.; Werner, R.G.; Chaicumpa, W. Human transbodies to HCV NS3/4A protease inhibit viral replication and restore host innate immunity. Front. Immunol. 2016, 7, 318. [Google Scholar] [CrossRef] [Green Version]
  124. Glab-Ampai, K.; Chulanetra, M.; Malik, A.A.; Juntadech, T.; Thanongsaksrikul, J.; Srimanote, P.; Thueng-In, K.; Sookrung, N.; Tongtawe, P.; Chaicumpa, W. Human single chain-transbodies that bound to domain-I of non-structural protein 5A (NS5A) of hepatitis C virus. Sci. Rep. 2017, 7, 15042. [Google Scholar] [CrossRef] [Green Version]
  125. Schofield, D.J.; Glamann, J.; Emerson, S.U.; Purcell, R.H. Identification by phage display and characterization of two neutralizing chimpanzee monoclonal antibodies to the hepatitis E virus capsid protein. J. Virol. 2000, 74, 5548–5555. [Google Scholar] [CrossRef] [Green Version]
  126. Sanna, P.P.; Williamson, R.A.; De Logu, A.; Bloom, F.E.; Burton, D.R. Directed selection of recombinant human monoclonal antibodies to herpes simplex virus glycoproteins from phage display libraries. Proc. Natl. Acad. Sci. USA 1995, 92, 6439–6443. [Google Scholar] [CrossRef] [Green Version]
  127. Nejatollahi, F.; Hodgetts, S.J.; Vallely, P.J.; Burnie, J.P. Neutralising human recombinant antibodies to human cytomegalovirus glycoproteins gB and gH. FEMS Immunol. Med. Microbiol. 2002, 34, 237–244. [Google Scholar] [CrossRef]
  128. Moazen, B.; Ebrahimi, E.; Nejatollahi, F. Single chain antibodies against Gp55 of human cytomegalovirus (HCMV) for prophylaxis and treatment of HCMV Infections. Jundishapur J. Microbiol. 2016, 9, e16241. [Google Scholar] [CrossRef] [Green Version]
  129. Gustchina, E.; Louis, J.M.; Lam, S.N.; Bewley, C.A.; Clore, G.M. A monoclonal Fab derived from a human nonimmune phage library reveals a new epitope on gp41 and neutralizes diverse human immunodeficiency virus type 1 strains. J. Virol. 2007, 81, 12946–12953. [Google Scholar] [CrossRef] [Green Version]
  130. da Silva, F.A.; Li, M.; Rato, S.; Maia, S.; Malhó, R.; Warren, K.; Harrich, D.; Craigie, R.; Barbas, C., 3rd; Goncalves, J. Recombinant rabbit single-chain antibodies bind to the catalytic and C-terminal domains of HIV-1 integrase protein and strongly inhibit HIV-1 replication. Biotechnol. Appl. Biochem. 2012, 59, 353–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Mohammadzadeh, S.; Rajabibazl, M.; Fourozandeh, M.; Rasaee, M.J.; Rahbarizadeh, F.; Mohammadi, M. Production of recombinant scFv Against P24 of human immunodeficiency virus type 1 by phage display technology. Monoclon. Antibodies Immunodiagn. Immunother. 2014, 33, 28–33. [Google Scholar] [CrossRef] [PubMed]
  132. Koch, K.; Kalusche, S.; Torres, J.L.; Stanfield, R.L.; Danquah, W.; Khazanehdari, K.; von Briesen, H.; Geertsma, E.R.; Wilson, I.A.; Wernery, U.; et al. Selection of nanobodies with broad neutralizing potential against primary HIV-1 strains using soluble subtype C gp140 envelope trimers. Sci. Rep. 2017, 7, 8390. [Google Scholar] [CrossRef] [PubMed]
  133. Kumar, S.; Kumar, R.; Khan, L.; Makhdoomi, M.A.; Thiruvengadam, R.; Mohata, M.; Agarwal, M.; Lodha, R.; Kabra, S.K.; Sinha, S.; et al. CD4-binding site directed cross-neutralizing scFv monoclonals from HIV-1 subtype C infected Indian children. Front. Immunol. 2017, 8, 1568. [Google Scholar] [CrossRef] [Green Version]
  134. Sun, Y.; Qiao, Y.; Zhu, Y.; Chong, H.; He, Y. Identification of a novel HIV-1-neutralizing antibody from a CRF07_BC-infected chinese donor. Oncotarget 2017, 8, 63047–63063. [Google Scholar] [CrossRef] [Green Version]
  135. Williams, J.V.; Chen, Z.; Cseke, G.; Wright, D.W.; Keefer, C.J.; Tollefson, S.J.; Hessell, A.; Podsiad, A.; Shepherd, B.E.; Sanna, P.P.; et al. A recombinant human monoclonal antibody to human metapneumovirus fusion protein that neutralizes virus in vitro and is effective therapeutically in vivo. J. Virol. 2007, 81, 8315–8324. [Google Scholar] [CrossRef] [Green Version]
  136. Rajput, R.; Sharma, G.; Rawat, V.; Gautam, A.; Kumar, B.; Pattnaik, B.; Pradhan, H.K.; Khanna, M. Diagnostic potential of recombinant scFv antibodies generated against hemagglutinin protein of influenza A virus. Front. Immunol. 2015, 6, 440. [Google Scholar] [CrossRef] [Green Version]
  137. Wyrzucki, A.; Dreyfus, C.; Kohler, I.; Steck, M.; Wilson, I.A.; Hangartner, L. Alternative recognition of the conserved stem epitope in influenza a virus hemagglutinin by a VH3-30-encoded heterosubtypic antibody. J. Virol. 2014, 88, 7083–7092. [Google Scholar] [CrossRef] [Green Version]
  138. Vashisht, S.; Verma, S.; Salunke, D.M. Cross-clade antibody reactivity may attenuate the ability of influenza virus to evade the immune response. Mol. Immunol. 2019, 114, 149–161. [Google Scholar] [CrossRef]
  139. Yodsheewan, R.; Maneewatch, S.; Srimanote, P.; Thueng-In, K.; Songserm, T.; Dong-Din-On, F.; Bangphoomi, K.; Sookrung, N.; Choowongkomon, K.; Chaicumpa, W. Human monoclonal ScFv specific to NS1 protein inhibits replication of influenza viruses across types and subtypes. Antivir. Res. 2013, 100, 226–237. [Google Scholar] [CrossRef]
  140. Sengupta, D.; Shaikh, A.; Bhatia, S.; Pateriya, A.K.; Khandia, R.; Sood, R.; Prakash, A.; Pattnaik, B.; Pradhan, H.K. Development of single-chain Fv against the nucleoprotein of type A influenza virus and its use in ELISA. J. Virol. Methods 2014, 208, 129–137. [Google Scholar] [CrossRef]
  141. Wu, J.; Zeng, X.Q.; Zhang, H.B.; Ni, H.Z.; Pei, L.; Zou, L.R.; Liang, L.J.; Zhang, X.; Lin, J.Y.; Ke, C.W. Novel phage display-derived H5N1-specific scFvs with potential use in rapid avian flu diagnosis. J. Microbiol. Biotechnol. 2014, 24, 704–713. [Google Scholar] [CrossRef] [Green Version]
  142. Zhu, M.; Gong, X.; Hu, Y.; Ou, W.; Wan, Y. Streptavidin-biotin-based directional double nanobody sandwich ELISA for clinical rapid and sensitive detection of influenza H5N1. J. Transl. Med. 2014, 12, 352. [Google Scholar] [CrossRef] [Green Version]
  143. Velappan, N.; Micheva-Viteva, S.; Adikari, S.H.; Waldo, G.S.; Lillo, A.M.; Bradbury, A.R.M. Selection and verification of antibodies against the cytoplasmic domain of M2 of influenza, a transmembrane protein. mAbs 2020, 12, 1843754. [Google Scholar] [CrossRef]
  144. Hirano, D.; Ohshima, N.; Kubota-Koketsu, R.; Yamasaki, A.; Kurosawa, G.; Okuno, Y.; Yoshida, S.; Kurosawa, Y. Three types of broadly reacting antibodies against influenza B viruses induced by vaccination with seasonal influenza viruses. J. Immunol. Res. 2018, 8, 7251793. [Google Scholar] [CrossRef] [Green Version]
  145. Ramage, W.; Gaiotto, T.; Ball, C.; Risley, P.; Carnell, G.W.; Temperton, N.; Cheung, C.Y.; Engelhardt, O.G.; Hufton, S.E. Cross-reactive and lineage-specific single domain antibodies against influenza b hemagglutinin. Antibodies 2019, 8, 14. [Google Scholar] [CrossRef] [Green Version]
  146. Arakawa, M.; Yamashiro, T.; Uechi, G.; Tadano, M.; Nishizono, A. Construction of human Fab (Gamma1/Kappa) library and identification of human monoclonal Fab possessing neutralizing potency against Japanese encephalitis virus. Microbiol. Immunol. 2007, 51, 617–625. [Google Scholar] [CrossRef] [Green Version]
  147. Sherwood, L.J.; Osborn, L.E.; Carrion, R., Jr.; Patterson, J.L.; Hayhurst, A. Rapid assembly of sensitive antigen-capture assays for Marburg virus, using in vitro selection of llama single-domain antibodies, at biosafety level 4. J. Infect. Dis. 2007, 196 (Suppl. S2), S213–S219. [Google Scholar] [CrossRef] [Green Version]
  148. Froude, J.W.; Pelat, T.; Miethe, S.; Zak, S.E.; Wec, A.Z.; Chandran, K.; Brannan, J.M.; Bakken, R.R.; Hust, M.; Thullier, P.; et al. Generation and characterization of protective antibodies to Marburg virus. mAbs 2017, 9, 696–703. [Google Scholar] [CrossRef] [Green Version]
  149. Amatya, P.; Wagner, N.; Chen, G.; Luthra, P.; Shi, L.; Borek, D.; Pavlenco, A.; Rohrs, H.; Basler, C.F.; Sidhu, S.S.; et al. Inhibition of marburg virus RNA synthesis by a synthetic anti-VP35 antibody. ACS Infect. Dis. 2019, 5, 1385–1396. [Google Scholar] [CrossRef]
  150. Kim, Y.; Lee, H.; Park, K.; Park, S.; Lim, J.H.; So, M.K.; Woo, H.M.; Ko, H.; Lee, J.M.; Lim, S.H.; et al. Selection and characterization of monoclonal antibodies targeting middle east respiratory syndrome coronavirus through a human synthetic Fab phage display library panning. Antibodies 2019, 8, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Lim, C.C.; Woo, P.C.Y.; Lim, T.S. Development of a phage display panning strategy utilizing crude antigens: Isolation of MERS-CoV nucleoprotein human antibodies. Sci. Rep. 2019, 9, 6088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Huang, W.; Samanta, M.; Crawford, S.E.; Estes, M.K.; Neill, F.H.; Atmar, R.L.; Palzkill, T. Identification of human single-chain antibodies with broad reactivity for noroviruses. Protein Eng. Des. Sel. 2014, 27, 339–349. [Google Scholar] [CrossRef] [PubMed]
  153. Garaicoechea, L.; Aguilar, A.; Parra, G.I.; Bok, M.; Sosnovtsev, S.V.; Canziani, G.; Green, K.Y.; Bok, K.; Parreño, V. Llama nanoantibodies with therapeutic potential against human norovirus diarrhea. PLoS ONE 2015, 10, e0133665. [Google Scholar] [CrossRef] [PubMed]
  154. Hurwitz, A.M.; Huang, W.; Kou, B.; Estes, M.K.; Atmar, R.L.; Palzkill, T. Identification and characterization of single-chain antibodies that specifically bind GI noroviruses. PLoS ONE 2017, 12, e0170162. [Google Scholar] [CrossRef] [Green Version]
  155. Welch, B.D.; Paduch, M.; Leser, G.P.; Bergman, Z.; Kors, C.A.; Paterson, R.G.; Jardetzky, T.S.; Kossiakoff, A.A.; Lamb, R.A. Probing the functions of the paramyxovirus glycoproteins F and HN with a panel of synthetic antibodies. J. Virol. 2014, 88, 11713–11725. [Google Scholar] [CrossRef] [Green Version]
  156. Hust, M.; Maiss, E.; Jacobsen, H.-J.; Reinard, T. The production of a genus-specific recombinant antibody (scFv) using a recombinant potyvirus protease. J. Virol. Methods 2002, 106, 225–233. [Google Scholar] [CrossRef]
  157. Chen, Z.; Chumakov, K.; Dragunsky, E.; Kouiavskaia, D.; Makiya, M.; Neverov, A.; Rezapkin, G.; Sebrell, A.; Purcell, R. Chimpanzee-human monoclonal antibodies for treatment of chronic poliovirus excretors and emergency postexposure prophylaxis. J. Virol. 2011, 85, 4354–4362. [Google Scholar] [CrossRef] [Green Version]
  158. Chen, G.; Gorelik, L.; Simon, K.J.; Pavlenco, A.; Cheung, A.; Brickelmaier, M.; Chen, L.; Jin, P.; Weinreb, P.H.; Sidhu, S.S. Synthetic antibodies and peptides recognizing progressive multifocal leukoencephalopathy-specific point mutations in Polyomavirus JC capsid viral protein 1. mAbs 2015, 7, 681–692. [Google Scholar] [CrossRef]
  159. Yang, S.; Shang, Y.; Yin, S.; Wang, D.; Cai, J.; Gong, Z.; Serge, M.; Liu, X. A phage-displayed single domain antibody fused to alkaline phosphatase for detection of porcine circovirus type 2. J. Virol. Methods 2015, 213, 84–92. [Google Scholar] [CrossRef]
  160. Mu, Y.; Jia, C.; Zheng, X.; Zhu, H.; Zhang, X.; Xu, H.; Liu, B.; Zhao, Q.; Zhou, E.M. A nanobody-horseradish peroxidase fusion protein-based competitive ELISA for rapid detection of antibodies against porcine circovirus type 2. J. Nanobiotechnol. 2021, 19, 34. [Google Scholar] [CrossRef] [PubMed]
  161. Yang, S.; Li, L.; Yin, S.; Shang, Y.; Khan, M.U.Z.; He, X.; Yuan, L.; Gao, X.; Liu, X.; Cai, J. Single-domain antibodies as promising experimental tools in imaging and isolation of porcine epidemic diarrhea virus. Appl. Microbiol. Biotechnol. 2018, 102, 8931–8942. [Google Scholar] [CrossRef] [PubMed]
  162. Bao, F.; Wang, L.; Zhao, X.; Lu, T.; Na, A.M.; Wang, X.; Cao, J.; Du, Y. Preparation and characterization of a single-domain antibody specific for the porcine epidemic diarrhea virus spike protein. AMB Express 2019, 9, 104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Ma, Z.; Wang, T.; Li, Z.; Guo, X.; Tian, Y.; Li, Y.; Xiao, S. A novel biotinylated nanobody-based blocking ELISA for the rapid and sensitive clinical detection of porcine epidemic diarrhea virus. J. Nanobiotechnol. 2019, 17, 96. [Google Scholar] [CrossRef] [Green Version]
  164. Zhang, F.; Chen, Y.; Ke, Y.; Zhang, L.; Zhang, B.; Yang, L.; Zhu, J. Single chain fragment variable (scFv) antibodies targeting the spike protein of porcine epidemic diarrhea virus provide protection against viral infection in piglets. Viruses 2019, 11, 58. [Google Scholar] [CrossRef] [Green Version]
  165. Liu, H.; Wang, Y.; Duan, H.; Zhang, A.; Liang, C.; Gao, J.; Zhang, C.; Huang, B.; Li, Q.; Li, N.; et al. An intracellularly expressed Nsp9-specific nanobody in MARC-145 cells inhibits porcine reproductive and respiratory syndrome virus replication. Vet. Microbiol. 2015, 181, 252–260. [Google Scholar] [CrossRef]
  166. Liu, H.; Liang, C.; Duan, H.; Zhang, X.; Wang, X.; Xiao, S.; Zhou, E.M. Intracellularly expressed nanobodies against non-structural protein 4 of porcine reproductive and respiratory syndrome virus inhibit virus replication. Biotechnol. Lett. 2016, 38, 1081–1088. [Google Scholar] [CrossRef]
  167. Sun, L.; Chen, Z.; Yu, L.; Wei, J.; Li, C.; Jin, J.; Shen, X.; Lv, X.; Tang, Q.; Li, D.; et al. Generation and characterization of neutralizing human recombinant antibodies against antigenic site II of rabies virus glycoprotein. Appl. Microbiol. Biotechnol. 2012, 96, 357–366. [Google Scholar] [CrossRef]
  168. Boruah, B.M.; Liu, D.; Ye, D.; Gu, T.J.; Jiang, C.L.; Qu, M.; Wright, E.; Wang, W.; He, W.; Liu, C.; et al. Single domain antibody multimers confer protection against rabies infection. PLoS ONE 2013, 8, e71383. [Google Scholar] [CrossRef]
  169. Chen, Z.; Zhang, L.; Tang, A.; Callahan, C.; Pristatsky, P.; Swoyer, R.; Cejas, P.; Nahas, D.; Galli, J.; Cosmi, S.; et al. Discovery and characterization of phage display-derived human monoclonal antibodies against RSV F glycoprotein. PLoS ONE 2016, 11, e0156798. [Google Scholar] [CrossRef] [Green Version]
  170. Rodrıguez-Dıaz, J.; Monedero, V.; Perez-Martınez, G.; Buesa, J. Single-chain variable fragment (scFv) antibodies against rotavirus NSP4 enterotoxin generated by phage display. J. Virol. Methods 2004, 121, 231–238. [Google Scholar] [CrossRef]
  171. Monedero, V.; Rodrıguez-Dıaz, J.; Viana, R.; Buesa, J.; Perez-Martınez, G. Selection of single-chain antibodies against the vp8* subunit of rotavirus VP4 outer capsid protein and their expression in Lactobacillus casei. Appl. Environ. Microbiol. 2004, 70, 6936–6939. [Google Scholar] [CrossRef] [Green Version]
  172. Bracken, C.J.; Lim, S.A.; Solomon, P.; Rettko, N.J.; Nguyen, D.P.; Zha, B.S.; Schaefer, K.; Byrnes, J.R.; Zhou, J.; Lui, I.; et al. QCRG Structural Biology Consortium; Zhou XX, Leung KK, Wells JA. Bi-paratopic and multivalent VH domains block ACE2 binding and neutralize SARS-CoV-2. Nat. Chem. Biol. 2021, 17, 113–121. [Google Scholar] [CrossRef]
  173. Bertoglio, F.; Fühner, V.; Ruschig, M.; Heine, P.A.; Abasi, L.; Klünemann, T.; Rand, U.; Meier, D.; Langreder, N.; Steinke, S.; et al. A SARS-CoV-2 neutralizing antibody selected from COVID-19 patients by phage display is binding to the ACE2-RBD interface and is tolerant to most known recently emerging RBD mutations. bioRxiv 2020. [Google Scholar] [CrossRef]
  174. Ku, Z.; Xie, X.; Davidson, E.; Ye, X.; Su, H.; Menachery, V.D.; Li, Y.; Yuan, Z.; Zhang, X.; Muruato, A.E.; et al. Molecular determinants and mechanism for antibody cocktail preventing SARS-CoV-2 escape. Nat. Commun. 2021, 12, 469. [Google Scholar] [CrossRef]
  175. Lim, S.A.; Gramespacher, J.A.; Pance, K.; Rettko, N.J.; Solomon, P.; Jin, J.; Lui, I.; Elledge, S.K.; Liu, J.; Bracken, C.J.; et al. Bispecific VH/Fab antibodies targeting neutralizing and non-neutralizing Spike epitopes demonstrate enhanced potency against SARS-CoV-2. mAbs 2021, 13, 1893426. [Google Scholar] [CrossRef]
  176. Ma, H.; Zeng, W.; Meng, X.; Huang, X.; Yang, Y.; Zhao, D.; Zhou, P.; Wang, X.; Zhao, C.; Sun, Y.; et al. Potent Neutralization of SARS-CoV-2 by hetero-bivalent alpaca nanobodies targeting the spike receptor-binding domain. J. Virol. 2021, 95, e02438-20. [Google Scholar] [CrossRef]
  177. Gai, J.; Ma, L.; Li, G.; Zhu, M.; Qiao, P.; Li, X.; Zhang, H.; Zhang, Y.; Chen, Y.; Ji, W.; et al. A potent neutralizing nanobody against SARS-CoV-2 with inhaled delivery potential. MedComm 2021, 2, 101–113. [Google Scholar] [CrossRef]
  178. Slezak, T.; Kossiakoff, A.A. Engineered ultra-high affinity synthetic antibodies for SARS-CoV-2 neutralization and detection. J. Mol. Biol. 2021, 433, 166956. [Google Scholar] [CrossRef]
  179. Zhou, D.; Chan, J.F.; Zhou, B.; Zhou, R.; Li, S.; Shan, S.; Liu, L.; Zhang, A.J.; Chen, S.J.; Chan, C.C.; et al. Robust SARS-CoV-2 infection in nasal turbinates after treatment with systemic neutralizing antibodies. Cell Host Microbe 2021, 29, 551–563.e5. [Google Scholar] [CrossRef]
  180. Kim, H.-Y.; Lee, J.-H.; Kim, M.J.; Park, S.C.; Choi, M.; Lee, W.; Ku, K.B.; Kim, B.T.; Changkyun Park, E.; Kim, H.G.; et al. Development of a SARS-CoV-2-specific biosensor for antigen detection using scFv-Fc fusion proteins. Biosens. Bioelectron. 2021, 175, 112868. [Google Scholar] [CrossRef] [PubMed]
  181. Ita, S.; Agostinho, M.R.; Sullivan, K.; Yub Han, S.; Akleh, R.; Johnson, W.E.; Fofana, I.B.F. Analysis of SIVmac Envelope-Specific Antibodies Selected Through Phage Display. AIDS Res. Hum. Retrovir. 2017, 33, 869–879. [Google Scholar] [CrossRef] [PubMed]
  182. Du, T.; Zhu, G.; Wu, X.; Fang, J.; Zhou, E.-M. Biotinylated single domain antibody-based blocking ELISA for detection of antibodies against swine influenza virus. Int. J. Nanomed. 2019, 14, 9337–9349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Zhang, F.; Chen, Y.; Yang, L.; Zhu, J. Construction and characterization of porcine single-chain fragment variable antibodies that neutralize transmissible gastroenteritis virus in vitro. Arch. Virol. 2019, 164, 983–994. [Google Scholar] [CrossRef] [PubMed]
  184. Schoenenwald, A.K.J.; Gwee, C.P.; Stiasny, K.; Hermann, M.; Vasudevan, S.G.; Skern, T. Development and characterization of specific antiusutu virus chicken-derived single chain variable fragment antibodies. Protein Sci. Publ. Protein Soc. 2020, 29, 2175–2188. [Google Scholar] [CrossRef]
  185. Shin, Y.W.; Chang, K.-H.; Hong, G.-W.; Yeo, S.-G.; Jee, Y.; Kim, J.-H.; Oh, M.-d.; Cho, D.-H.; Kim, S.-H. Selection of vaccinia virus-neutralizing antibody from a phage display human-antibody library. J. Microbiol. Biotechnol. 2019, 29, 651–657. [Google Scholar] [CrossRef] [Green Version]
  186. Yuan, L.; Zhang, X.; Xiao, N.; Dai, L.; Chen, W.; Jia, C.; Zhao, R.; Hemmingsen, S.M.; Dai, H. Identification of a WSSV neutralizing scFv antibody by phage display technology and in vitro screening. Dis. Aquat. Organ. 2006, 72, 93–99. [Google Scholar] [CrossRef] [Green Version]
  187. Duan, T.; Ferguson, M.; Yuan, L.; Xu, F.; Li, G. Human monoclonal Fab antibodies against west nile virus and its neutralizing activity analyzed in vitro and in vivo. J. Antivir. Antiretrovir. 2009, 1, 36–42. [Google Scholar] [CrossRef] [Green Version]
  188. Rizzo, S.; Imperato, P.; Mora-Cárdenas, E.; Konstantinidou, S.; Marcello, A.; Sblattero, D. Selection and characterization of highly specific recombinant antibodies against west nile virus E protein. J. Biotechnol. 2020, 311, 35–43. [Google Scholar] [CrossRef]
  189. Long, M.C.; Jager, S.; Mah, D.C.; Jebailey, L.; Mah, M.A.; Masri, S.A.; Nagata, L.P. Construction and characterization of a novel recombinant single-chain variable fragment antibody against Western equine encephalitis virus. Hybridoma 2000, 19, 1–13. [Google Scholar] [CrossRef]
  190. Burke, C.W.; Froude, J.W.; Miethe, S.; Hülseweh, B.; Hust, M.; Glass, P.J. Human-like neutralizing antibodies protect mice from aerosol exposure with western equine encephalitis virus. Viruses 2018, 10, 147. [Google Scholar] [CrossRef] [Green Version]
  191. Liu, J.L.; Shriver-Lake, L.C.; Zabetakis, D.; Goldman, E.R.; Anderson, G.P. Selection of single-domain antibodies towards western equine encephalitis virus. Antibodies 2018, 7, 44. [Google Scholar] [CrossRef] [Green Version]
  192. Lu, X.; Xiao, H.; Li, S.; Pang, X.; Song, J.; Liu, S.; Cheng, H.; Li, Y.; Wang, X.; Huang, C.; et al. Double lock of a human neutralizing and protective monoclonal antibody targeting the yellow fever virus envelope. Cell Rep. 2019, 26, 438–446.e5. [Google Scholar] [CrossRef] [Green Version]
  193. Mwale, P.F.; Lee, C.H.; Lin, L.T.; Leu, S.J.; Huang, Y.J.; Chiang, L.C.; Mao, Y.C.; Yang, Y.Y. Expression, purification, and characterization of anti-Zika virus envelope protein: Polyclonal and chicken-derived single chain variable fragment antibodies. Int. J. Mol. Sci. 2020, 21, 492. [Google Scholar] [CrossRef] [Green Version]
  194. Delfin-Riela, T.; Rossotti, M.; Alvez-Rosado, R.; Leizagoyen, C.; González-Sapienza, G. Highly sensitive detection of Zika virus nonstructural protein 1 in serum samples by a two-site nanobody ELISA. Biomolecules 2020, 10, 1652. [Google Scholar] [CrossRef]
  195. Liu, J.; Ma, P.; Yu, H.; Wang, M.; Yin, P.; Pang, S.; Jiao, Y.; Dong, T.; Liu, A. Discovery of a phage peptide specifically binding to the SARS-CoV-2 spike S1 protein for the sensitive phage-based enzyme-linked chemiluminescence immunoassay of the SARS-CoV-2 antigen. Anal. Chem. 2022, 94, 11591–11599. [Google Scholar] [CrossRef]
  196. Li, H.; Zhu, B.; Li, B.; Chen, L.; Ning, X.; Dong, H.; Liang, J.; Yang, X.; Dong, J.; Ueda, H. Isolation of a human SARS-CoV-2 neutralizing antibody from a synthetic phage library and its conversion to fluorescent biosensors. Sci. Rep. 2022, 12, 15496. [Google Scholar] [CrossRef]
  197. Chen, X.; Kang, S.; Ikbal, M.A.; Zhao, Z.; Pan, Y.; Zuo, J.; Gu, L.; Wang, C. Synthetic nanobody-functionalized nanoparticles for accelerated development of rapid, accessible detection of viral antigens. Biosens. Bioelectron. 2022, 202, 113971. [Google Scholar] [CrossRef]
  198. Guliy, O.I.; Zaitsev, B.D.; Borodina, I.A.; Shikhabudinov, A.M.; Staroverov, S.A.; Dykman, L.A.; Fomin, A.S. Electro-acoustic sensor for the real-time identification of the bacteriophages. Talanta 2018, 178, 743–750. [Google Scholar] [CrossRef]
  199. Yang, F.; Liu, L.; Neuenschwander, P.F.; Idell, S.; Vankayalapati, R.; Jain, K.G.; Du, K.; Ji, H.; Yi, G. Phage display-derived peptide for the specific binding of SARS-CoV-2. ACS Omega 2021, 7, 3203–3211. [Google Scholar] [CrossRef]
  200. Griep, R.A.; Prins, M.; van Twisk, C.; Keller, H.J.; Kerschbaumer, R.J.; Kormelink, R.; Goldbach, R.W.; Schots, A. Application of phage display in selecting Tomato spotted wilt virus-specific single-chain antibodies (scFvs) for sensitive diagnosis in ELISA. Phytopathology 2000, 90, 183–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Dong, J.; Sakurai, A.; Nomura, N.; Park, E.Y.; Shibasaki, F.; Ueda, H. Isolation of recombinant phage antibodies targeting the hemagglutinin cleavage site of highly pathogenic avian influenza virus. PLoS ONE 2013, 8, e61158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Yu, C.-M.; Chen, I.-C.; Tung, C.-P.; Peng, H.-P.; Jian, J.-W.; Chiu, Y.-K.; Tsou, Y.-L.; Chen, H.-S.; Huang, Y.-J.; Hsiao, W.W.-W.; et al. A panel of anti-influenza virus nucleoprotein antibodies selected from phage-displayed synthetic antibody libraries with rapid diagnostic capability to distinguish diverse influenza virus subtypes. Sci. Rep. 2020, 10, 13318. [Google Scholar] [CrossRef] [PubMed]
  203. Cabezas, S.; Rojas, G.; Pavon, A.; Bernardo, L.; Castellanos, Y.; Alvarez, M.; Pupo, M.; Guillen, G.; Guzman, M.G. Phage-displayed antibody fragments recognizing dengue 3 and dengue 4 viruses as tools for viral serotyping in sera from infected individuals. Arch. Virol. 2009, 154, 1035–1045. [Google Scholar] [CrossRef]
  204. Pashaki, A.S.; Safarnejad, M.R.; Safdar, A.H.A.; Safarpour, H.; Tabatabaie, M. Production of a phage-displayed single chain variable fragment antibody against infectious bursal disease virus. Trop. J. Pharm. Res. 2017, 16, 2801–2809. [Google Scholar] [CrossRef] [Green Version]
  205. Sapats, S.; Gould, G.; Trinidad, L.; Parede, L.H.; David, C.; Ignjatovic, J. An ELISA for detection of infectious bursal disease virus and differentiation of very virulent strains based on single chain recombinant chicken antibodies. Avian Pathol. 2005, 34, 449–455. [Google Scholar] [CrossRef] [Green Version]
  206. Sapats, S.I.; Trinidad, L.; Gould, G.; Heine, H.G.; van den Berg, T.P.; Eterradossi, N.; Jackwood, D.; Parede, L.; Toquin, D.; Ignjatovic, J. Chicken recombinant antibodies specific for very virulent infectious bursal disease virus. Arch. Virol. 2006, 151, 1551–1566. [Google Scholar] [CrossRef]
  207. Sharma, G.K.; Mahajan, S.; Matura, R.; Subramaniam, S.; Mohapatra, J.K.; Pattnaik, B. Production and characterization of single-chain antibody (scFv) against 3ABC non-structural protein in Escherichia coli for sero-diagnosis of Foot and Mouth Disease virus. Biologicals 2014, 42, 339–345. [Google Scholar] [CrossRef]
  208. Chen, S.; Li, S.; Sun, H.; Li, Y.; Ji, S.; Song, K.; Zhang, L.; Luo, Y.; Sun, Y.; Ma, J.; et al. Expression and characterization of a recombinant porcinized antibody against the E2 protein of classical swine fever virus. Appl. Microbiol. Biotechnol. 2018, 102, 961–970. [Google Scholar] [CrossRef]
  209. Duggan, J.M.; Coates, D.M.; Ulaeto, D.O. Isolation of single-chain antibody fragments against Venezuelan equine encephalomyelitis virus from two different immune sources. Viral Immunol. 2001, 14, 263–273. [Google Scholar] [CrossRef]
  210. Homola, J. Surface plasmon resonance sensors for detection of chemical and biological species. Chem. Rev. 2008, 108, 462–493. [Google Scholar] [CrossRef]
  211. Howes, P.D.; Rana, S.; Stevens, M.M. Plasmonic nanomaterials for biodiagnostics. Chem. Soc. Rev. 2014, 43, 3835–3853. [Google Scholar] [CrossRef]
  212. Walper, S.A.; Aragonés, G.L.; Sapsford, K.E.; Brown, C.W., III; Rowland, C.E.; Breger, J.C.; Medintz, I.L. Detecting biothreat agents: From current diagnostics to developing sensor technologies. ACS Sens. 2018, 3, 1894–2024. [Google Scholar] [CrossRef] [Green Version]
  213. Torrance, L.; Ziegler, A.; Pittman, H.; Paterson, M.; Toth, R.; Eggleston, I. Oriented immobilisation of engineered single-chain antibodies to develop biosensors for virus detection. J. Virol. Methods 2006, 134, 164–170. [Google Scholar] [CrossRef]
  214. Kim, J.W.; Cho, A.H.; Shin, H.G.; Jang, S.H.; Cho, S.Y.; Lee, Y.R.; Lee, S. Development and characterization of phage display-derived monoclonal antibodies to the S2 domain of spike proteins of wild-type SARS-CoV-2 and multiple variants. Viruses 2023, 15, 174. [Google Scholar] [CrossRef]
  215. Soleymani, L.; Li, F. Mechanistic challenges and advantages of biosensor miniaturization into the nanoscale. ACS Sens. 2017, 2, 458–467. [Google Scholar] [CrossRef]
  216. Domingo, E. Virus as Populations: Composition, Complexity, Dynamics, and Biological Implications, 1st ed.; Academic Press: San Diego, CA, USA, 2015. [Google Scholar]
Figure 1. Viral lifecycle in a eukaryotic cell (A), virus detection methods (B), and the overall scheme of antibody phage display technology for sensor-based virus detection (C) [32] with modification.
Figure 1. Viral lifecycle in a eukaryotic cell (A), virus detection methods (B), and the overall scheme of antibody phage display technology for sensor-based virus detection (C) [32] with modification.
Biosensors 13 00640 g001
Figure 2. Methods used to diagnose COVID-19. (a) Molecular testing based on nucleic acid amplification assays, (b) lateral flow immunoassay, and (c) biosensors [46].
Figure 2. Methods used to diagnose COVID-19. (a) Molecular testing based on nucleic acid amplification assays, (b) lateral flow immunoassay, and (c) biosensors [46].
Biosensors 13 00640 g002
Figure 3. General design scheme of biosensors [47].
Figure 3. General design scheme of biosensors [47].
Biosensors 13 00640 g003
Figure 4. Electrochemical techniques used for SARS-CoV-2 detection [50].
Figure 4. Electrochemical techniques used for SARS-CoV-2 detection [50].
Biosensors 13 00640 g004
Figure 5. (A) Fabricated electrochemical eCovSens device; and (B) fabrication of an SPCE electrode, in which nCovid-19 antibody (Ab) is immobilized onto the transducer of the SPCE. The transducer detects changes in the electrical signal resulting from the antigen–antibody (Ag–Ab) interaction [52].
Figure 5. (A) Fabricated electrochemical eCovSens device; and (B) fabrication of an SPCE electrode, in which nCovid-19 antibody (Ab) is immobilized onto the transducer of the SPCE. The transducer detects changes in the electrical signal resulting from the antigen–antibody (Ag–Ab) interaction [52].
Biosensors 13 00640 g005
Figure 6. Clinical diagnostics with gold immunoassays. (A) Schematic comparison of the conventional qRT−PCR detection methods and the point-of-care ultrafast immunoassay. (B) Impedimetric spectra for the gold sensor were acquired when it was incubated with a mixture of positive samples (saliva swabs containing SARS-CoV-2) and negative samples (saliva swabs missing SARS-CoV-2 but carrying other pathogens (three samples) or from healthy individuals (two samples). (C) Evaluation of the tested samples’ Ct and ΔRct values. (D) Testing the antibody cross-reactivity with various upper respiratory tract viruses, including human rhinovirus, influenza A virus, and respiratory syncytial virus [53].
Figure 6. Clinical diagnostics with gold immunoassays. (A) Schematic comparison of the conventional qRT−PCR detection methods and the point-of-care ultrafast immunoassay. (B) Impedimetric spectra for the gold sensor were acquired when it was incubated with a mixture of positive samples (saliva swabs containing SARS-CoV-2) and negative samples (saliva swabs missing SARS-CoV-2 but carrying other pathogens (three samples) or from healthy individuals (two samples). (C) Evaluation of the tested samples’ Ct and ΔRct values. (D) Testing the antibody cross-reactivity with various upper respiratory tract viruses, including human rhinovirus, influenza A virus, and respiratory syncytial virus [53].
Biosensors 13 00640 g006
Figure 7. Schematic diagram of optical nanobiosensors used to detect pathogenic viruses. These biosensors are designed on the basis of quantum dots, upconversion nanoparticles, noble metal nanoparticles, 2D nanoprobes based on fluorescent organic molecules, and chemiluminescence immunoassay [26].
Figure 7. Schematic diagram of optical nanobiosensors used to detect pathogenic viruses. These biosensors are designed on the basis of quantum dots, upconversion nanoparticles, noble metal nanoparticles, 2D nanoprobes based on fluorescent organic molecules, and chemiluminescence immunoassay [26].
Biosensors 13 00640 g007
Figure 8. Scheme for antibody preparation [65].
Figure 8. Scheme for antibody preparation [65].
Biosensors 13 00640 g008
Table 1. Comparison of methods traditionally used to detect pathogenic viruses [26].
Table 1. Comparison of methods traditionally used to detect pathogenic viruses [26].
TechniquePrincipleTimeAdvantagesDisadvantage
PCRnucleic acidhours
-
well established
-
small number of samples
-
ease to contamination
-
time-consuming
ELISAviral proteinhourshighly specific
-
low sensitivity
-
high-quality sample preparation
Cell culturinginfectivity assaydays to weeks
-
suitable for virus sub-typing recovery of novel and divergent strains
-
inexpensive
-
contamination problems
-
time-consuming and labor intensive
-
unavailable for immediate patient care
Electron microscopyviral particleshoursrapid method
-
well-trained personnel
-
low specificity
Computed tomographychest scanninghoursgood basis for clinical diagnosis and treatment
-
technical expertise
-
centralized facilities
Table 2. Examples of using recombinant antibodies for virus detection.
Table 2. Examples of using recombinant antibodies for virus detection.
VirusPhage Ab TargetType of Phage LibraryFormat of Phage AbsApplication of Phage AbsReference
Australian bat lyssavirus (ABLV)glycoprotein GnaïveFab, IgGELISA, in vitro neutralization[84]
Avian influenza virus
H7N2 (AIV)
complete virusimmuneVHHELISA[85]
Bluetongue virus (BTV)complete virussemi-syntheticscFv, scFv-FcELISA[86]
Bovine immunodeficiency virus (BIV)capsid (CA) proteinsemi-syntheticscFvELISA, WB[87]
Bovine viral diarrhea virus (BVDV)envelope 2 (E2) proteinimmuneVHHELISA, qRT-PCR,
in vitro neutralization
[88]
nonstructural protein 5
(NS5B)
immuneVHHELISA[89]
Broad bean mottle virus (BBMV)complete virusimmuneVHHELISA, in vitro neutralization[90]
Canine parvovirus type 2 (CPV2)complete virussemi-syntheticscFvELISA[91]
virus-like particles (VLP)immunescFvELISA, ICA, virus suppression assay[92]
Chikungunya virus (CHIKV)VLPimmuneVHHELISA, MagPlex,
in vitro neutralization
[93]
Cucumber mosaic virus (CMV)complete virussemi-syntheticscFvELISA, WB[94]
Dengue virus (DENV)non-structural protein 3 (NS3)naïveFabELISA, in vitro neutralization[95]
nonstructural protein 1 (NS1)immuneVHHMagPlex assay[96]
RNA-dependent RNA polymerase (RdRp) in the viral non-structural protein 5 (NS5)naïvescFvELISA, WB, in vitro inhibition[97]
Duck hepatitis A virus (DHAV)VP3 proteinimmunescFvELISA, in vivo neutralization[98]
VP1 proteinimmuneVHHELISA, IF, dot-blot[99]
Ebola virusnucleoproteinsyntheticscFv, IgGELISA, WB[100]
viable Zaire ebolavirus, viral matrix protein VP40, viral nucleoprotein (NP)immunescFv, IgNAR VELISA, WB[101]
multifunctional viral protein (VP35)semi-syntheticscFvELISA, WB[102]
glycoprotein (GP)immunescFv, scFv-FcELISA, in vitro neutralization,
in vivo protection
[103,104]
Ectromelia virus (ECTV)epitope p35immunescFvELISA, in vitro neutralization, WB[105]
Enterovirus 71 (EV71)inactivated EV71 virionsimmuneFabELISA, WB, in vitro neutralization[106]
virion protein 2 (VP2)naïvescFvELISA, WB[107]
internal capsid protein (VP4)naïvescFvELISA, WB, IF, in vitro neutralization[108]
Epstein–Barr virus (EBV)latent membrane protein 1 (LMP1)naïveFabELISA, WB, IF, FACS,
in vitro inhibition
[109]
Foot-and-mouth disease virus (FMDV)recombinant non-structural protein (NSP) 3ABCimmunescFvELISA, WB[110]
VLPimmuneVHHELISA, IF[111]
intact (146S) FMDVimmuneVHHELISA[112]
VP2 capsid proteinimmunescFvELISA[113]
Grapevine leafroll-associated virus 3 (GLRaV-3)coat proteinimmunescFvELISA[114]
Grapevine virus B (GVB)virus particlessemi-syntheticscFvELISA[115]
Hantavirusesvirus particlesimmuneFabELISA, IF, WB[116]
nucleoproteinimmuneVHHELISA, WB[117]
Hendra virus (NiV) and Nipah virus (HeV)attachment envelope glycoprotein GnaïveFab, IgGELISA, immunoprecipitation, WB, in vitro neutralization, IF, in vivo neutralization[118]
Hepatitis A (HAV)HAV capsidimmuneFab, IgGELISA, in vitro neutralization[119]
Hepatitis B virus (HBV)hepatitis B virus surface antigen (HBsAg)immuneFab, scFvELISA, IF[120]
Hepatitis C virus (HCV)core proteinimmuneFabELISA[121]
E2 glycoproteinimmuneFabELISA, in vitro neutralization[122]
non-structural protein-3/4A (NS3/4A)naïvescFvELISA, IF, in vitro neutralization[123]
non-structural protein-5A (NS5A)naïvescFvELISA, WB, IF, in vitro neutralization[124]
Hepatitis E virus (HEV)ORF2 proteinimmuneFabELISA, WB, in vitro neutralization[125]
Herpes simplex virus (HSV-1, HSV-2)glycoproteins gD and gBimmuneFabELISA, immuno precipitation, WB, in vitro neutralization[126]
Human cytomegalovirus (HCMV)gycoprotein B (gB) and H (gH)immunescFvELISA, in vitro neutralization[127]
glycoprotein 55 (gp55)immunescFvELISA, in vitro neutralization[128]
Human immunodeficiency viruses (HIV)transmembrane glycoprotein gp41syntheticFabWB, in vitro neutralization[129]
integrase
(IN) protein
immunescFvELISA, WB, IF, in vitro neutralization[130]
p24immunescFvELISA[131]
envelope glycoprotein gp140immuneVHHELISA, in vitro neutralization[132]
CD4bs region of subtype CimmunescFvELISA, in vitro neutralization[133]
Envelope glycoprotein gp120immuneFabELISA, in vitro neutralization, IF[134]
Human metapneumovirus (HMPV)F proteinimmuneFabELISA, IF, in vitro neutralization, in
vivo protection
[135]
Influenza AH1N1hemagglutinin protein (HA)immunescFvELISA[136]
H2N2hemagglutinin protein HA (stem region)immuneFabELISA, in vitro neutralization[137]
H3N2HA protein and its variantssemi-syntheticscFvELISA[138]
non-structural protein-1 (NS1)naïvescFvELISA, WB, in vitro neutralization, IF[139]
nucleoprotein (NP)immunescFvELISA, WB, in vitro inhibition[140]
hemagglutinin protein (HA)semi-syntheticscFvELISA[141]
complete inactivated virusimmuneVHHELISA[142]
Influenza AM2 protein (cytoplasmatic domain)naïvescFv, scFv-FcWB, IHC[143]
Influenza Bwhole virusimmuneFabELISA, WB, IF, in vitro neutralization[144]
hemagglutinin protein (HA)immuneVHHELISA[145]
Japanese encephalitis virus (JEV)purified virionimmuneFabELISA, immunoprecipitation, in vitro neutralization[146]
domains I, II, III of envelope proteinimmuneFab, IgGELISA, WB, immunoprecipitation, in vitro neutralization, in vivo protection[67]
Marburg virusnucleoproteinimmunesdAbsELISA[147]
transmembrane glycoprotein (GP)immunescFv, scFv-FcWB, in vitro neutralization, in vivo protection[148]
VP35 proteinsyntheticFabELISA[149]
Middle East respiratory syndrome-related coronavirus (MERS-CoV)S2 subunit of the MERS-CoV spike protein (MERS-S2P)syntheticFab, IgGELISA, IF, in vitro neutralization, ACCEL ELISA™[150]
nucleoprotein (NP)naïvescFvELISA[151]
NorovirusVLPsemi-syntheticscFvELISA, WB[152]
VLP with major capsid protein VP1 and a minor structural protein VP2immuneVHHELISA, WB, in vitro inhibition, IF[153]
P-domain of the GI.1 (VLP) major capsid proteinsemi-syntheticscFvELISA, dot-blot, surface plasmon resonance (SPR)[154]
Paramyxovirusglycoproteins F (fusion protein) and HN (attachment protein)syntheticFab, sAbELISA, in vitro neutralization, immunoprecipitation[155]
Plum pox virus (PPV)NIa proteasesemi-syntheticscFvWB, dot-blot[156]
Polioviruscapsid proteins VP1 and VP3immuneFab, IgGELISA, in vitro neutralization, in vivo protection[157]
Polyomavirusmajor capsid viral protein 1 (VP1)syntheticFab, IgGELISA[158]
Porcine circovirus type-2 (PCV2)complete virusimmunesdAbsWestern blot, ELISA, and SPR[159]
cap proteinimmuneVHHELISA[160]
Porcine epidemic diarrhea virus (PEDV)membrane protein of PEDVimmunesdAb fragments (sdAb-Mc19/29/30/37)ELISA[161]
S1 domain of spike
protein
immuneVHHELISA, in vitro neutralization[162]
nucleocapsid (N) proteinimmuneVHHELISA[163]
S1 region of the spike proteinimmunescFvELISA, IF, in vivo protection[164]
Porcine reproductive and respiratory syndrome virus (PRRSV)non-structural protein 9 (Nsp9)immuneVHHELISA, IF, immunoprecipitation[165]
non-structural protein 4 (Nsp4)immuneVHHELISA, immunoprecipitation[166]
Rabies lyssavirusglycoprotein (antigenic site II)immuneFab, IgGELISA, IF, in vitro neutralization[167]
glycoprotein GnaïveVHHELISA, in vitro neutralization, in vivo protection[168]
Respiratory syncytial virus (RSV)glycoprotein FsyntheticFab, IgGELISA, in vitro neutralization[169]
Rotavirusnon-structural protein Nsp4semi-syntheticscFvELISA, WB[170]
VP8* fraction of rotaviral VP4 outer capsidsemi-syntheticscFvELISA, WB, in vitro inhibition[171]
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)S protein (RBD)syntheticVHELISA, in vitro neutralization[172]
immunescFv, scFv-Fc, IgGELISA,
in vitro inhibition,
in vitro neutralization,
in vivo protection
[173]
naïvescFvELISA, in vitro neutralization,
in vitro inhibition,
in vivo protection
[174]
syntheticFab, biospecific Fab+VHin vitro neutralization[175]
immuneVHH, VHH-FcELISA,
in vitro neutralization
[176]
RBDimmuneVHHELISA, in vitro inhibition, in vitro neutralization[177]
semi-syntheticFabELISA, in vitro neutralization[178]
immuneFab, IgGELISA, in vitro neutralization assay, in vivo protection[179]
nucleocapsid protein (NP)immunescFv, scFv-FcELISA, WB, dot-blot, lateral flow strip assay[180]
Sin Nombre orthohantavirus (SNV)Sin Nombre Virus nucleocapsid protein (SNV-N)naïvescFvELISA, WB, dot-blot[78]
Simian immunodeficiency virus (SIV)trimeric (gp140) and monomeric (gp120) forms of the SIVmac239 envelope glycoproteinimmunescFv, scFv-FcELISA, WB, in vitro neutralization[181]
Swine influenza virus (SIV)SIV nucleoprotein (SIV-NP)immuneVHHELISA, WB[182]
Transmissible gastroenteritis virus (TGEV)whole virusimmunescFvELISA, in vitro neutralization,
IF, WB
[183]
Usutu virus (USUV)domain III (DIII) of the USUV E proteinimmunescFvELISA, WB, in vitro neutralization[184]
Vaccinia virus (VACV)virus particlesimmunescFv, IgGinhibition ELISA,
in vitro
neutralization
[185]
Venezuelan equine encephalitis viruses (VEEV)viral E1 envelope proteinimmunescFv, scFv-FcELISA, WB, in vitro
neutralization,
in vivo protection
[66]
White spot syndrome virus (WSSV)virus particlesimmunescFvELISA, in vitro neutralization[186]
West Nile virus (WNV)domain IIII of WNV envelope (E) proteinimmuneFabELISA, IF, in vitro neutralization,
in vivo protection
[187]
envelope (E)proteinnaïvescFv, scFv-FcELISA, WB, in vitro neutralization[188]
Western equine encephalitis viruses (WEEV)virusimmunescFvEnzyme-linked immunosorbent assays (ELISAs)[189]
virus particlesimmunescFv, scFv-Fcin vitro neutralization, in vivo protection[190]
E2/E3E2 envelope proteinsimmuneVHHELISA (MagPlex assay)[191]
Yellow fever virus (YFV)domain II of envelope proteinimmunescFv, IgGin vitro neutralization,
in vivo protection
[192]
Zika virus (ZIKV)envelope (E) proteinimmunescFvELISA, WB, FACS, in vitro inhibition[193]
nonstructural protein 1 (NS1)immuneVHHELISA[194]
Table 3. Examples of biosensor systems using recombinant antibodies as a receptor element for detecting viruses.
Table 3. Examples of biosensor systems using recombinant antibodies as a receptor element for detecting viruses.
Type of Sensory SystemPhage Antibody FormatVirusTargetDetection LimitAnalysis TimeReference
Biosensor based on the lateral flow immunoassay (LFIA)scFv–FcSARS-CoV-2nucleocapsid protein (NP)2 ng of antigenic protein
2.5 × 104 pfu cultured virus
20 min[180]
Sandwiched phage-based enzyme-linked chemiluminescence immunoassay (ELCLIA)n.i.SARS-CoV-2S1 domain of the spike glycoprotein78 pg/mL of antigenic proteinn.i.[195]
Quench-based fluorescent immunosensor
(Quenchbodies)
Fab, IgSARS-CoV-2S1 domain of the spike glycoprotein0.1 nM of S protein trimer 105 copies/mL of virus2 min[196]
Nanobody-functionalized nanoparticles for rapid, electronic detection (Nano2RED)n.i.SARS-CoV-2spike protein receptor binding domain (RBD)40 pg/mL
1.3 pM
5–20 min[197]
Ebola virussecreted glycoprotein (sGP)10 pg/mL
0.13 pM
Electro-acoustic sensorIgbacteriophage
FAl-SR65
whole virus particles106
phages/mL
5 min[198]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guliy, O.I.; Evstigneeva, S.S.; Khanadeev, V.A.; Dykman, L.A. Antibody Phage Display Technology for Sensor-Based Virus Detection: Current Status and Future Prospects. Biosensors 2023, 13, 640. https://doi.org/10.3390/bios13060640

AMA Style

Guliy OI, Evstigneeva SS, Khanadeev VA, Dykman LA. Antibody Phage Display Technology for Sensor-Based Virus Detection: Current Status and Future Prospects. Biosensors. 2023; 13(6):640. https://doi.org/10.3390/bios13060640

Chicago/Turabian Style

Guliy, Olga I., Stella S. Evstigneeva, Vitaly A. Khanadeev, and Lev A. Dykman. 2023. "Antibody Phage Display Technology for Sensor-Based Virus Detection: Current Status and Future Prospects" Biosensors 13, no. 6: 640. https://doi.org/10.3390/bios13060640

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop