Next Article in Journal
A Score-Based Approach to 18F-FDG PET Images as a Tool to Describe Metabolic Predictors of Myocardial Doxorubicin Susceptibility
Next Article in Special Issue
Utility of Two-Dimensional Difference Gel Electrophoresis in Diagnosis of Multiple Sclerosis
Previous Article in Journal
Prospective Comparison of F-18 Choline PET/CT Scan Versus Axial MRI for Detecting Bone Metastasis in Biochemically Relapsed Prostate Cancer Patients
Previous Article in Special Issue
Comparison of Direct Sequencing, Real-Time PCR-High Resolution Melt (PCR-HRM) and PCR-Restriction Fragment Length Polymorphism (PCR-RFLP) Analysis for Genotyping of Common Thiopurine Intolerant Variant Alleles NUDT15 c.415C>T and TPMT c.719A>G (TPMT*3C)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Single Domain Antibodies as New Biomarker Detectors

1
Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia
2
Bacterial Pathogenesis and Scabies Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
3
Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kelantan 16150, Malaysia
4
Department of Drug Resistance and Diagnostics, Australian Army Malaria Institute, Brisbane 4051, Australia
5
Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia
6
Clinical Tropical Medicine Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia
*
Author to whom correspondence should be addressed.
Diagnostics 2017, 7(4), 52; https://doi.org/10.3390/diagnostics7040052
Submission received: 23 August 2017 / Revised: 7 September 2017 / Accepted: 8 September 2017 / Published: 17 October 2017
(This article belongs to the Collection Biomarkers in Medicine)

Abstract

:
Biomarkers are defined as indicators of biological processes, pathogenic processes, or pharmacological responses to a therapeutic intervention. Biomarkers have been widely used for early detection, prediction of response after treatment, and for monitoring the progression of diseases. Antibodies represent promising tools for recognition of biomarkers, and are widely deployed as analytical tools in clinical settings. For immunodiagnostics, antibodies are now exploited as binders for antigens of interest across a range of platforms. More recently, the discovery of antibody surface display and combinatorial chemistry techniques has allowed the exploration of new binders from a range of animals, for instance variable domains of new antigen receptors (VNAR) from shark and variable heavy chain domains (VHH) or nanobodies from camelids. These single domain antibodies (sdAbs) have some advantages over conventional murine immunoglobulin owing to the lack of a light chain, making them the smallest natural biomarker binders thus far identified. In this review, we will discuss several biomarkers used as a means to validate diseases progress. The potential functionality of modern singe domain antigen binders derived from phylogenetically early animals as new biomarker detectors for current diagnostic and research platforms development will be described.

1. Introduction

Early and accurate diagnosis of disease is important for providing appropriate treatment to individuals with most human diseases. Clinical diagnosis remains a mainstay method for clinical care in many settings, such as among febrile patients in endemic areas [1]. However, the overlap of clinical symptoms of many diseases makes misdiagnosis likely and frequent, thereby impeding treatment decisions and epidemiologic information [2].
Microbiological methods represent definitive diagnostic method for various infectious diseases in laboratory settings [3]. Samples tested are predominantly blood, serum, stool and urine that can be collected from both outpatients and hospitalized individuals. Despite high specificity, the requirement of maintaining specific temperature during transportation of clinical specimens to laboratory for processing is problematic, especially in less well-resourced settings [4,5]. Microscopy, when possible, is one of the least expensive methods for laboratory diagnosis. During the Malaria Eradication era, slide-based diagnosis assisted the successful elimination of malaria in many countries [6]. However, obstacles such as time consuming nature of this test, the need for highly trained and experienced staff, and system maintenance have limited the utility of this method for diagnosis of malaria [7,8].
Biomarkers are defined as indicators of biological processes, pathogenic processes, or pharmacological responses to a therapeutic intervention [9]. These include the presence of specific antigens, changes in enzyme activities, and unique changes in DNA and proteins, for example, carcinoembryonic antigen (CEA) in colorectal cancer [10], chitinase enzyme activity in Alzheimer’s disease [11], and circulating tumor DNA in breast cancer [12]. Identification of unique biomarkers of diseases is important for early screening, diagnosis, and monitoring of disease progression. The introduction of high-throughput screening methods, particularly through to genomics, transcriptomics and proteomics, has expanded the knowledge base and potential for diagnosis using biomarkers [13,14]. Methods of directly detecting DNA and RNA have enhanced the precision with which biomarkers of diseases are identified [15]. However, unlike nucleic acid detection methods, post-translation modifications of proteins can complicate the detection of clinically-relevant biomarkers [16].
Antibodies are the primary tool used as recognition reagents to detect biomarkers, and are widely used as analytical tools in clinical settings. For immunodiagnostics, antibodies are now exploited as binders for antigen of interest in a range of platforms, including the enzyme-linked immunosorbant assay (ELISA), immunoblotting (Western Blotting), immunocytochemistry, and the immunoprecipitation assay [17,18]. With the help of sophisticated instruments, antibodies are also used as tools for protein microarrays, flow cytometric analysis, and immunoaffinity analysis [19,20,21]. Therefore, monoclonal antibodies are widely used as a main source of antibodies in rapid diagnostic tests for detecting various infectious diseases such as malaria, dengue, HIV and so on. Antibodies can be isolated from a range of sources, with plasma and serum being the most common sources in laboratory practice [22]. Furthermore, alternative sources, such as chicken egg yolk, have been used to produce functional antibodies against certain biomarkers [23]. Antibodies have also been used as therapeutic agents and animal models such as mice, rabbits, horses and guinea pig are critical to the development of therapeutic antibodies [24].
Small-molecule antibodies are postulated to have better solubility capability [25,26]. Nowadays, the shrinking of intact immunoglobulin into smaller antibody fragments such as scFv, Fv, Fab and dual or tetra-domain fragments has become possible via molecular engineering approaches (Figure S1) [27,28]. An advantage of small antibody fragments is their ease of genetic manipulation due to their smaller size, ease of expression in bacterial systems, and low lot-to-lot variation, and in scaled-up production [29,30,31]. Moreover, the desired antibody fragment repertoire can also be developed from any animal immunoglobulin with an appropriate set of specific primers [32,33].
The introduction of antibody surface display and combinatorial chemistry techniques has greatly allowed the exploration of new binders from various organisms. More recently, natural single domain antibodies (sdAbs) that functioned well in vivo were discovered from a few ancient vertebrates, including VNAR from shark, and VHH from camelids. These groups of minimized antibodies offer the potential of providing advantages over conventional antibodies such as IgG and IgM derived from higher animals [34,35]. The unusual antibodies derived from these groups of animal have been reported to provide promising specificity and sensitivity for their target antigens [36,37,38]. In addition to possessing the capability of better tissue penetration, the peculiar structure of these sdAbs naturally renders them to have better thermostability, enabling them withstand the harsh environment [39,40].

2. Classes of Diagnostic Tests that Target Biomarkers

As described above, conventional diagnostic methods such as microscopy remain problematic in many settings. Thus, there is a significant need for new diagnostic techniques that are capable of displaying high specificity and sensitivity [41]. In recent decades, molecular-based diagnostic tools have become well developed, generating new strategies for diagnosis. Examples include polymerase chain reaction (PCR) [42], loop-mediated isothermal amplification (LAMP) [43], mass spectrometry (MS) [44], fluorescence-activated cell sorting (FACS) [45], and single molecule array (SIMOA) technology [46]. In spite of some advantages related to their extremely good specificity, the implementation of these methods is hampered by specific limitations such as their time-consuming nature, cost, need for electricity supply, and lack of experienced technicians, especially in less well-resourced settings. These issues have been extensively reviewed [47,48].
To address these shortcomings, immune-based assays would be ideal tools for biomarker detection in clinical diagnostic settings [49]. As the gold standard for measuring protein content in human body fluids such as blood, the enzyme-linked immunosorbent assay (ELISA) remains the predominant method for detection of antigens of interest (biomarkers) [50]. Furthermore, the invention of new point-of-care systems, such as rapid diagnostic tests (RDTs), has been a major contributor to clinical case management [51,52] patients undergoing self-monitoring at home; for instance, blood glucose reading for diabetics [53]. Many high-throughput antibody-based screening methods have been integrated across various diagnostic platforms to enable rapid identification of biomarkers that may aid individual treatment and monitoring of responses to therapy. Current multiplexed immunoassays are based on multi-marker strategies, in which high-affinity capture ligands (antibodies or proteins/peptides) are immobilized in parallel assays [54,55]. Several commercial multiplexed immunoassay platforms are available on this emerging market, including the Luminex bead-based platform [56], Meso Scale Discovery’s Multi Array Technology [57], and protein array platforms from Whatman [58].

2.1. Non-Infectious Diseases and Non-Diseases

Non-infectious diseases, commonly known as non-communicable diseases (NCDs), are the leading cause of morbidity and mortality worldwide. According to the WHO, about 40 million people suffering from cancers, cardiovascular diseases and stroke, chronic respiratory diseases, and diabetes are killed each year in the world [59]. Unlike infectious diseases, NCDs affect people from across the whole world. Of chronic diseases, cancers have been the highest cause of mortality, followed by diabetes and cardiovascular disease [60,61]. In addition, other conditions, such as pregnancy and envenomation, are common cases. Prompt screening for these could render a proper plan, and also the selection of appropriate anti-sera for those who are envenomated. Hence, the development of simple, cost-effective, and low-complexity point-of-care (POC) devices represents an important goal in global health.

2.1.1. Diagnostics for Cancer

Despite substantial investments and progress made in therapy, cancer remains as a major threat to human life across the globe. In the past 5 years, more than 12 million new cases and 7.6 million deaths have been caused by various cancers [62]. The incidence of cancer is projected to continuously increase to 26.8 million new cases and 17.1 million deaths annually by the year 2030 [63]. The most common cancers are prostate, lung, breast, colorectal, liver, stomach and cervical cancers, with lung cancer and breast cancer resulting in the highest mortality among men and women, respectively. The success rate of therapy for cancers can be significantly improved if they are diagnosed early [64].
Antibodies to target biomarkers are widely used, for detecting prostate specific antigen (PSA), specifically expressed in prostate cancer [65], carcinogenic-embryonic antigen (CEA) in colorectal cancer [66], CA15-3 antigen and her-2/neu are proteins associated with breast cancer [67,68], CA19-9 in gastrointestinal cancer [69], while CA125 is a biomarker for ovarian cancer diagnosis [70]. ELISA tests are the current gold standard for identifying cancer biomarkers due to their high sensitivity, specificity, and ability to quantify target antigens [71,72]. The monoclonal antibody (mAb) within NMP22® BladderChek® Test (Inverness Medical Innovations Inc., North America) for example, is an FDA-approved lateral flow immunochromatographic test designed to detect nuclear matrix proteins (NMPs) for bladder cancer. The sensitivity and specificity of this test has been reported to be up to 95% using just 4 drops of urine, with results available in 30 min [73].
Another approach to cancer biomarker detection is by coupling antibodies to electrode-based devices [74]. These systems enable sample profiling on a large scale [75]. An automated diagnostic device with high sensitivity and specificity was developed by Kashani-Sabet et al. to distinguish benign nevi from melanoma via integration of gene expression data and tissue array profiling [76]. Using such tissue proteomics approaches, several potential biomarkers have been identified from melanoma, such as actin-related protein 2/3 complex, subunit 2 (ARPC2), fibronectin 1 (FN1), and regulator of G protein signaling 1 (RGS1) [76].

2.1.2. Diagnostics for Pregnancy

Diagnosis of pregnancy using test kits is one of the most successful immunoassays. Today, many different brands are commercially available worldwide [77,78,79]. In the past, several pregnancy-specific materials were targeted as candidate biomarkers. These include Schwangerschafts protein 1 (SP1) [80], placental protein 14 (PP14) [81] and early pregnancy factor (EPF) [82,83]. In recent years, however, the hormone human chorionic gonadotropin (hCG) has become the most widely detected biomarker for early detection of pregnancy due to its abundance and it being amenable to immunoassay design [84,85,86]. β-hCG is a dimeric glycoprotein with a size of 46 kDa that is synthesized by the trophoblastic tissue in placenta [87], with the amount increasingly secreted after implantation [88]. The first urine pregnancy test using monoclonal antibodies to detect β-hCG based on simple immunological principles was introduced over 30 years ago [89]. Owing to their cost effectiveness, and convenience, pregnancy diagnostic test kits are very widely used by both the general public and healthcare professionals for early detection of pregnancy [90,91].
Assays using traditional polyclonal antibodies have suffered from the drawback of insufficient specificity [92]. Thus, monoclonal antibody (mAb)-based pregnancy tests predominate, and offer nearly 100% sensitivity and specificity for detection of β-hCG at a threshold concentration of 25 mili-IU/mL in urine [93,94]. For example, the Clearview hCG (Unipath, UK) test is able to detect pregnancy on the day of a missed period with just three drops of urine loading to the sample window, within five minutes with accuracy greater than 99% [95]. Most pregnancy test kits presently are very robust because membranes and antibodies are engineered to be protected by a plastic housing, and are sealed to prevent damage from humidity. In addition, when stored at room temperature, the shelf-life for all pregnancy test kits can be up to 2 to 3 years [96].

2.1.3. Diagnostics for Envenomation

Venoms are biological toxins secreted by certain kinds of poisonous animals, such as snakes, scorpions, spiders, hornets and wasps, which usually use them for attacking their prey by targeting the victim’s lymphatic system through biting or “injection” using a special “weapon” or so-called sting [97,98,99]. Snakebite envenomation remains a life-threatening medical emergency worldwide [100]. To prevent inappropriate treatment, which can result in allergy, paralysis or even death of the victims, the establishment of rapid, reliable and specific detection of envenomation is necessary. A diagnostic test is therefore essential to improving emergency management of envenomation to facilitate the provision of appropriate antivenom therapy [101].
Techniques for detection of different snake venoms have been extensively reviewed. Antigen capturing ELISA has been claimed to be the best method for detection of snake venom [102,103]. The first commercial diagnostic kit for detection of snake venoms was introduced by the Commonwealth Serum Laboratories (CSL) in Australia in 1991 [104]. This kit was specially designed to detect the venoms of the five most dangerous snakes in Australia and Papua New Guinea, including Australian Tiger snake, Brown snake, Black snake, Death adder and Taipan [105,106]. The CSL snake venom diagnostic kit (SVDK) promises to provide a rapid, easy-to-use, low-cost and long-shelf-life test. As snake venoms are complex mixtures, a polyclonal antibody is used in each well in SVDK to distinguish particular types of snake venom [107]. The SVDK has been widely evaluated in both humans and animals in many clinical studies, and has shown high sensitivity and specificity [107,108,109]. This kit has been reported to be able to detect 2.5 ng/mL venom in less than 20 min, and has a specificity of 100%. Due to high stability of immunoreagents and low cross reactivity, these characteristics have made SVDK widely used by health workers in tropical countries [107].
Several alternative diagnostic kits have also been developed to identify the venoms of the four common snakes from Asia [110,111,112]. Unlike SVDK, whole blood can directly be used as sample in the AB-microELISA kit. The sensitivity of this assay for detection of venom has been shown to be 10 ng/mL. In addition, 600 μL of whole blood is sufficient for this kit, and results are available within 30 min [113]. Apart from whole blood, serum, urine blister fluids and bite site swabs can also be used as assay samples. However, further studies are ongoing to validate the prototype of AB-microELISA kit for field use [110].

2.2. Infectious Diseases

Despite decades of advances, infectious diseases continue to represent leading causes of morbidity and mortality throughout the world [114,115,116]. Millions of people are living under threat of a diversity of diseases caused by bacteria (e.g., Staphylococcus aureus and Salmonella typhi), viruses (Human Immunodefiency Virus and Hepatitis C virus), fungi (Candidasis, Coccidioides and Pneumocystis) and parasites (malaria and helminths). Infections lead to disability, death and social and economic disruption [117,118]. According to WHO statistics, approximately 15 million people are killed by such diseases in developing countries annually [119]. Moreover, sexually transmitted infections such as syphilis [120], and tropical parasitic infections such as schitosomiasis also cause enormous morbidity [121]. Due to the diversity of environmental conditions in varying developing countries, a need has arisen for the development of simple, accurate, and stable diagnostic tools. The deployment of highly sensitive and specific diagnostic tests is also needed to counteract the spreading of drug resistance of infectious diseases [122].

2.2.1. Diagnostics for Viral Disease

Since the first case of AIDS was reported in early 1980s, HIV/AIDS has caused 1.7 million HIV-related deaths in 2011 alone and estimated about 34 million people were living with HIV worldwide in 2010 [123]. AIDS, referring to acquired immunodeficiency syndrome, is a dangerous infectious disease that eventually causes death without treatment. The etiological agent of AIDS is known as human immunodeficiency virus (HIV), a retrovirus with a single-stranded RNA (ssRNA) [124]. However, many persons who are infected with HIV are not aware of the infection until late in the course of disease.
To increase access to early treatment and prevention, rapid HIV tests play an integral role in HIV prevention activities in both clinical and non-clinical settings [125,126]. ELISA assays are extensively used to screen for the appearance of specific antibodies. To perform a typical indirect assay, the serum collected from a patient is incubated to detect a response to an HIV target antigen; for example, p24, gp24 or gp120. A positive antibody response is then detected by an enzyme-labeled anti-human antibody or an enzyme-labeled antigen [127,128].
Instead of using blood or serum as a sample, HIV infection can also be diagnosed by detecting the presence of anti-HIV antibodies in a patient’s saliva or urine samples [129,130]. For example, the OraSure® assay is a specific salivary test that is designed to determine anti-HIV IgG antibodies from saliva [131]. However, HIV diagnostic tests based on urine and saliva are still not comparable to blood samples in terms of detailed information and the specific characteristics of the HIV subtype responsible for the infection [132].
With the aid of rapid tests, screening tests can be conducted in hard-to-reach patient populations. Unlike high-throughput EIA screening tests, RDTs can perform well for HIV diagnosis even when the volume of samples is low [133]. With the administration of combination anti-retroviral therapies, the transmission rate of HIV can be reduced from over 25% to less than 2% for those infected pregnant patients [134]. Recently, the US FDA has approved four types rapid tests for screening HIV-1 infection, including Murex® Single Use Diagnostic System HIV-1 Test (Murex Diagnostics, Inc., USA), OralQuick® Rapid HIV-1 Antibody Test (OraSure Technologies, Inc., USA), Reveal® Rapid HIV-1 Antibody Test (MedMira Laboratories Inc., Canada), and UniGold Recombigen® HIV (Trinity Biotech PLC, Ireland) [135].
However, false-negative results can occur in individuals who are in the acute phase of infection [136]. Diagnostic tests that directly detect the HIV p24 antigen in serum samples have been shown to be superior to antibody-based detection tests for early infection [137]. For instance, the commercial Vidas Duo assay (bioMerieux Inc., March-L’Etoile) is a fourth-generation ELISA. This assay possesses the advantage by decreasing the diagnostic window to an average of 7 days. In addition to detecting anti-HIV antibodies, this kit targets the HIV p24 antigen that is present in the blood of an HIV infected individual in the early phase [138].

2.2.2. Diagnostics for Bacterial Disease

Typhoid fever is a serious systemic illness caused by the Salmonella enterica serotype Typhi. This disease represents the most common cause of community-acquired bacteremia in developing countries [139]. The annual global incidence of typhoid fever is over 21.5 million cases, resulting in more than 200,000 deaths [140]. The emergence of multidrug resistance S. typhi has complicated treatment [141]. Therefore, rapid and accurate diagnosis is essential to provide early antimicrobial treatment, for preventions of mortality cases, and for the control of disease transmission [142].
In the past, the Widal test was the most widely used serologic test for detection of host antibodies. This agglutination test targeted typhoid antisera such as lipopolysaccharide (O) and flagellar (H), and Vi antigens of S. typhi [143,144]. However, tests was limited by false-positive results due to cross-reaction of the antigenic determinants with non-typhoid Salmonella or other tropical diseases such as malaria and dengue [145,146]. False-negative reactions may also occur if the blood sample is collected too early from the infected individuals [147]. The Widal test is not a satisfactory test for diagnosing typhoid fever in endemic areas [148]. Recently, a new generation of rapid serologic tests has been developed, for example Linear Cromotest® (Linear Chemicals, Barcelona, Spain). This test aims to detect host IgM and IgG antibodies which are specific to O and H antigens of S. typhi. The highest reported specificity (50%) and sensitivity (95.2%) limited the accuracy of diagnosis of typhoid fever at two sub-Saharan African sites [149].
TUBEX® (IDL Biotech AB, Bromma, Sweden) is semiquantitative colorometric rapid test that use polystyrene particle agglutination to detect anti-O9 IgM antibodies specific for group D of S. typhi. This kit enables rapid diagnosis of typhoid fever patients, with only 3 min per test at room temperature [149]. The TUBEX® test kit is designed for detection of antibodies in the patient’s serum by inhibiting the binding between an indicator antibody-bound particle and a magnetic antigen-bound particle [150]. Thus, an acute stage of S. typhi infections can be indicated by elevated levels of anti-O9 IgM antibodies in combination with typical clinical symptoms of typhoid fever [151,152]. However, the TUBEX® colorimetric reaction may be subject to false-positive results due to hemolyzed samples in individuals with recent S. enteritidis infection [150]. Despite the promising test performance, the requirement for additional laboratory equipment has limited this test kit being used in resource-limited endemic regions [149].
Another rapid test for diagnosing S. typhi of typhoid fever is Typhidot® (Malaysian Biodiagnostic Research, Bangi, Malaysia). This is a dot EIA test that detects either host IgM or IgG antibodies against the S. typhi antigen [153]. The target antigen used in the assay is neither an O nor H antigen, but a 50 kDa outer membrane protein (OMP) antigen of S. typhi. This recombinant protein is coated on a nitrocellulose strip for detection of the antigen-antibody complex by employing an anti-human antibody conjugated peroxidise and a chromogenic substrate [154]. Due to host immune response, the 50 kDa OMP is a good antigen to identify S. typhi specific antibodies in the sera of individuals with typhoid [155,156]. Since the IgG antibody can persist in the host for more than 2 years, the detection of IgG antibodies can lead to false positive results by confusing between acute or convalescent cases [157]. An upgraded version of Typhidot-M® has been introduced. It activates antibodies to allow accessibility of OMP antigens to the specific IgM. With this approach, detection of acute typhoid infection can be obtained within 3 h [158]. Both Typhidot® and Typhidot-M® are simple, fast, specific, sensitive, and economical dot diagnostic assays for providing early detection of S. typhi infections. Evaluation studies on the Typhidot® and Typhidot-M® tests in clinical settings showed that these tests performed better than the Widal test and conventional gold standard culture methods [158]. Both Typhidot® and TUBEX® kit have reportedly given good performance for diagnosis of typhoid fever in small cohorts of hospitalized patients [159,160].

2.2.3. Diagnostics for Parasitic Diseases

Malaria remains a severe parasitic disease leading to high morbidity and mortality in tropics [161]. This protozoan parasitic disease is transmitted by female Anopheles mosquitoes. According to the World Malaria Report 2012, it was estimated that about 219 million cases of malaria, causing 660,000 deaths, occurred throughout the world in 2010. Africa is the most prevalent region, where up to 90% of all malaria deaths occur [161]. Four species of Plasmodium parasites are well-known as causative agents for human malaria, namely P. falciparum, P. malariae, P. ovale, and P. vivax. However, the simian species P. knowlesi has been recently identified as a new species that can cause malaria infection in humans [162]. This mainly occurs in Malaysian Borneo [163] and other South East Asian countries [164,165]. Of these species, P. falciparum is the most pathogenic, accounting for the majority of febrile illness and death [166]. Therefore, it is crucial to understand the important parameters in the transmission of the disease, and develop effective diagnostic strategies for its prevention and control.
Malaria rapid diagnostic tests (RDTs) using antigen capture technology were developed in the early 1990s, and have led to much-improved access to diagnostic tests for malaria. Such devices are intended to provide simple, swift, accurate and reliable diagnosis of malaria in areas where microscopic diagnosis is not applicable [167,168]. Other advantages of RDTs are that they do not require complex methodologies, intensive training, and electricity supply, thus representing promising diagnostic tools in remote areas [169,170].
A variety of antigens have been investigated as candidate targets for malaria RDTs. Plasmodium falciparum histidine rich protein 2 (PfHRP2), Plasmodium lactate dehydrogenase (pLDH), and parasite fructose 1,6-biphosphate aldolase (Aldolase) are predominantly used as biomarkers in malaria RDTs. PfHRP2 is water soluble protein that is specific to P. falciparum, and is produced by parasite 2 h after invasion of red blood cell [171,172]. The molecular weight of the secreted PfHRP2 varies from 60 to 105 kDa [173]. pLDH is a soluble glycoprotein enzyme produced by the asexual and sexual stages of parasites [174]. Different isomers of pLDH can be identified in all human malaria species [175]. Aldolase is an enzyme of the parasite glycolytic pathway that is also synthesized by all human malaria species [176]. Although three types of tissue-specific aldolase isoenzymes can be found in all higher vertebrates, P. falciparum and P. vivax possess only one aldolase isoenzyme, which is also similar to that possessed by Giardia lamblia and Trypanosoma brucei [177].
Nowadays, malaria RDTs have been developed into a range of test formats, including dipstick, strip, card, pad, well, or cassette devices [175]. PfHRP2-detecting tests were the first type of RDT to become available specifically for P. falciparum detection (ParaSight-F® and ICT®), where mAb against PfHRP2 were used as signal and capture antibodies [178,179]. It was followed soon after by pLDH, and Aldolase detection tests, such as OptiMAL® which are able to detect all four human Plasmodium species (pan-malaria) [180,181,182]. Polyclonal antibodies have been used as capture antibody in the qualitative and quantitative immunoassay test for targeting pLDH [183]. Meanwhile, monoclonal antibodies against parasite Aldolase that are pan-specific have been used in a combined “P. f/pan” immunochromatographic test to detect non-P. falciparum spp., along with PfHRP2 [184]. Recently, the increased demand for RDTs has resulted in more than 200 malaria RDT products from 60 manufacturers currently being available in the global market [185].

3. Currently Available Antibody Binders for Detection of Biomarkers

Antibodies can be classified into three different categories: polyclonal antibodies, monoclonal antibodies, and recombinant antibodies [186]. Polyclonal antibodies (Polyclonal Abs) are heterogeneous antibody mixtures that are derived from multiple plasma cell lines. Owing to their complexity, polyclonal antibodies have excellent properties for recognizing complex antigens carrying numerous epitopes [187]. A monoclonal antibody (mAb) is a homogeneous antibody generated from a single B lymphocyte clone. Antibodies produced in mAb format have extremely high specificity against a single epitope on antigens [188]. Recombinant antibodies or antibody fragments (rAbs) are antibodies generated using molecular techniques in laboratory. They are aimed at improving the sensitivity, selectivity, stability and immobilization properties in diagnostic applications, for example, in biosensors [189].
In making the decision to use or generate polyclonal, monoclonal or recombinant antibodies, several factors should be considered, including commercial availability, animals to raise, types of applications, time length and cost [186]. A comparison of parameters for producing different source of antibodies is shown in Table 1.

3.1. Monoclonal Antibodies

The first description of mAb production was by Nobel Prize winner Kohler and Milstein in 1975 [190]. The fusion technique between splenic B cells and myeloma cells, termed the hydridoma technique has revolutionized immunology. The production of mAbs is not influenced by the animal sources used, thus, giving mAbs a better homogeneity and consistency in scaled-up production [191]. mAb technology has been widely applied in biomedical research and the pharmaceutical industry.
Unlike polyclonal Abs, the monospecificity of mAb enables targeting a single epitope. This permits a range of applications, including targeting members of a protein family and evaluating changes in molecular conformation and protein-protein interactions. However, the functionality and sensitivity of mAbs can be reduced by small changes in the structure of antigen determining regions, or even by minor changes in pH or salt concentration. One advantage is that mAbs can be produced at greater concentration and much higher purity than polyclonal Abs [191]. Disadvantages of mAb can be overcome using combinations of multiple mAbs specific to desired antigens. However, this pooling method can be difficult, costly, and time consuming [191]. Nowadays, the mass production of mAbs through the ascites method has been largely replaced by in vitro technology such as bioreactors due to the constraint of needing use of mice as host animals [192,193,194,195,196].

3.2. Limitations of Conventional Monoclonal Antibodies

As bivalent antibodies, IgG represents the most abundant immunoglobulin proteins (approximately 85%) found in all mammalian serum (Figure 1) [197]. Due to their ability to confer high affinity and retention times, monospecific IgGs are the preferred reagents in biomedical research, as well as in therapeutic and diagnostic applications. However, several practical drawbacks are apparent for diagnostic reagents based on conventional IgG antibodies. The complex architecture and large molecular size (~150 kDa) may result in weak bindings, when small sizes or small amounts of protein antigens are not easily recognized by the concave surfaces of CDR loops [198,199].
More importantly, there is a concern in the application of RDTs in tropical countries regarding their shelf life, because some of these antibodies are susceptible to degradation by excessive temperatures (>40 °C), or by storage for extended periods under conditions of high humidity [200]. To overcome humidity, most RDT devices are now protected in a hermetically sealed plastic packet, or a desiccant is used to ensure that the test strips remain dry [201]. However, it is still a challenge to protect RDTs from high ambient temperatures, resulting in reduced performance of RDTs, especially in tropical countries, where temperatures regularly rise to 45 °C. Hence, for most RDT devices a storage temperature between 4 to 30 °C is recommended. However, this condition is difficult to meet, especially in endemic areas where refrigerated storage systems may not be available [202,203].
To address these problems, initial attempts to generate single domain antibody fragments by separating expression of individual VH or VL units was introduced Ward and co-workers [204]. However, this approach reportedly resulted in solubility problems in aqueous solvents, higher cost, a more time-consuming process, and the requirement of sophisticated protein engineering approaches [205]. Moreover, its failure to recognize selected mAbs on conserved epitopes of specific antigens due to unbound reactivities mediated by the Fc region hinder its utility for diagnostic applications [206,207].
With the emergence of DNA engineering, surface display has been widely used to discover new antibody fragments for the purposes of diagnostic and therapeutic application. As a consequence, a range of different types of new antibodies has been investigated, aiming to overcome the limitations presented in the conventional antibodies.

4. Phage Display Technology for New Biomarker Binder Discovery

Screening phage display libraries is a powerful tool for identifying specific binders from libraries that contain a large diversity of targets [33,208]. Library construction is achieved by splicing a repertoire of genes (genotype) that encodes the peptide into a gene that encodes a capsid structural protein (phenotype). The “displayed” peptides are included in the capsid layer on the phage surface. Ideally, these proteins should not interfere with the phage structure [209].
Recombinant DNA technology has enabled phage library construction whereby billions of variant peptides and proteins are able to be presented on the surface of the phage [210,211]. From this diversity library, binders specific to proteins of interest with high affinity can be selected by biopanning. This technique facilitates understanding of protein–ligand interactions [212], antigen-antibody interactions [213,214], and permits improvement of the affinity of proteins to their binding partner [215,216]. For instance, phage display antibody libraries with diversities as high as 1010 can be established using display technology [217,218]. Recently, surface display technology has expanded to include ribosome display [219], yeast surface display [220], and mammalian cell display [221]. Such technologies have enabled the exploration of new antibodies that may not otherwise have been discovered, from humans and animals including shark, camel, llama, and lamprey [222,223,224,225].
Antibody phage display libraries have been used extensively for isolation of high-affinity specific binders against unique antigens from different targets [226,227,228,229,230]. Three types of antibody libraries are typically constructed: naïve, synthetic, and immunized libraries [231]. A naïve antibody library refers to the repertoire of antibody genes derived from non-immunized donors. Synthetic antibody libraries are constructed using synthesized V-gene fragments with randomized CDRs, whereas immunized libraries are based on a host immunized with the target antigen of the disease [232]. The principle of the phage display is represented in Figure 2, indicating the workflows of library construction, biopanning, and clone screening prior to protein expression and purification for functional assays.

5. Natural Single Domain Antibodies

The evolution of immunoglobulins from invertebrates began ~550 million years ago [233]. With the emergence of antibody surface display technology, interest has increased in new binders from less commonly used animals, including VHH from camelids, and VNAR from sharks. The unusual antibodies derived from these groups of animals have been reported to provide promising specificity and sensitivity for target antigens [36,37,38]. The availability of new binders derived from lower vertebrates is now discussed.

5.1. VHH Heavy Chain Domain in Camelids

As in all mammals, members of the camelid family produce immunoglobulin G which comprises two heavy chains and two light chains fused with disulfide bonds. However, unlike the VH and VL domains in conventional antibodies, a unique subclass of immunoglobulin containing only a heavy chain domain and lacking a light chain was found in the circulatory system of camelids. Owing to its peculiar structure, this antibody has been configured as “heavy chain only” antibodies (HCAbs) [234]. The capacity of camelid HCAbs to retain the reversibility and binding activity after heat denaturation has enabled new applications where transient heating may occur [235,236,237].
HCAbs are slightly different from IgG, in that they also include both a constant (Fc) and variable domain. The isolated variable domain region of camelids HCAbs is known as VHH (variable domain of the heavy chain of HCAbs) or Nanobody® (Nb; Ablynx) [238]. Similar to the products of protein engineering the VH domain from other mammals, the N-terminal of VHH is naturally utilized as a binding surface to interact with the target antigen [234]. The molecular weight of VHH is 15 kDa, ten times lower than that of an intact conventional antibody (Figure 1). It was thereby considered the smallest possible antibody fragment, and has attracted the interest of many scientists [239,240,241].
The major advantage of the VHH antibody is its greater solubility compared to classical VH [205]. This is due to the hydrophilic amino acid substitution present in the framework 2 region. Meanwhile, the single coding exon of less than 450 base pairs facilitates genetic engineering of VHH fragments [240,242]. In addition, on account of its smaller antigen binding surface area, the unique CDR3 region enables the heavy domain of camelids to penetrate into antigen cleft regions that are not easily recognized by conventional antibodies [243,244]. From a phylogenetic prospect, it is conceivably possible to produce humanized VHH [245], a process that may be “easier” than the complicated manipulation required to “humanize” murine or other more distant species to reduce an alloresponse, such as the human antimouse antibody (HAMA) response [246]. Furthermore, due to their high intrinsic domain stability, camelid VHH is now under investigation as a probe for diagnostics [247,248]. The diagnostic potential of camelid VHH as a probe in immunodetection systems offers possibilities for improving the diagnosis of infection [249], cancers [250], and caffeine contaminants in the food and beverage industries [251,252].

5.2. VNAR Heavy Chain Domain in Sharks

A class of naturally occurring single variable domain antibodies was discovered in the serum of elasmobranch cartilaginous fish during the early 1990s [253,254,255]. These natural functional repertoires were termed immunoglobulin new antigen receptors (IgNARs). IgNARs are an unconventional and unique class of proteins found in sharks, including nurse sharks (Ginglymostoma cirratum) [256], wobbegong sharks (Orectolobus maculatus) [257], smooth dogfish (Mustelus canis) [258], banded hound sharks (Triakis scyllium) [259], and horn sharks (Heterodontus francisci) [260]. Investigations has revealed that IgNARs function as antibody and immune response mediators in sharks. However, unlike camelid VHH domains, the IgNAR V region is more similar to light chain and T-cell receptor variable regions than to other VH regions [254,261].
Several desirable biological properties of IgNAR V-domains have been identified, and their potential as alternative antigen binders explored [257,258,262]. The natural habitat of sharks has resulted in them evolving an extraordinarily stable immune system such that the functionality of antibodies can be retained in a harsh environment [263]. Electron microscopic studies have indicated that the intact IgNAR exists as a disulfide-bonded homodimer that consists of a polyprotein with one variable domain (VNAR) and five constant domains (CNAR) (Figure 1) [264].
Similar to the camelid VHH, the VNAR has only a heavy chain domain. However, the cross-species conservation of amino acid sequences with human VH is extremely low in VNAR (~25%), whereas it is more than 80% homologous to classical VH scaffolds in camelid VHH [255,265]. It is hypothesized that IgNARs lack many residues that exist in conventional antibodies; these are replaced by other hydrophilic residues. The greatly truncated CDR2 region, herein defined as an HV2 region, has created a signature hallmark for shark VNAR. Due to this unusual structure, the single variable heavy domain proteins of shark IgNARs are currently the smallest antibody fragments observed in the animal kingdom, having a size of only 12 kDa. Yet, in combination with the peculiar feature of a long CDR3 region, these VNAR domains thought to more readily penetrate cleft regions of antigens, thereby increasing the opportunity to target small target epitopes that may not be accessible to conventional IgG [266].
In terms of heat-stability, VNAR also possesses refolding properties as found in camelid VHH. The ability to retain fully functional binding-specific activity after exposure to temperatures of up to 95 °C may make VNAR ideally suited to protein array and diagnostic applications where transient heating may occur as part of the protein immobilization process [258,267]. It is partly due to the presence of cysteine residues in these single domain antibodies, making an extraordinary structure conformation [268].
VNAR domains are more easily produced as recombinant proteins than conventional antibodies. Additionally, due to hydrophilic residues present within VNAR surfaces, high yields of expressed proteins associated with high solubility are achievable, and are thus easy to express in prokaryotic systems [257]. Therefore, the potential utility of VNAR as an alternative binder for clinical applications is now being investigated in a variety of research areas for diagnostic and therapeutic purposes.

6. Use of Different Binders for Specific Applications

To date, mammals remain the main source of intact antibodies for targeting diseases. However, with the aid of DNA technology, a number of new antibodies have been engineered as smaller single domain fragments to improve of immunoassays, immunosensors, and imaging probes in various applications. As described recently, the discovery of natural single heavy domain antibodies from camelids VHH, shark VNAR, and lamprey VLRs offer some advantages over conventional antibody fragments. This range of natural antibodies is expected to open applications such as enzyme inhibitors and intrabodies, and as detection units in biosensors or immunodiagnostics. In the following section, the deployment of different binders in specific diagnostic applications will be reviewed.

6.1. Applications of Camelids VHH Domain or Nanobodies®

To monitor infections, single domain antibodies naturally derived from camelids (nanobodies) may enable superior detection of species-specific antigens to classical monoclonal antibodies in immunodiagnostic tests. Trypanosome infection causes African sleeping sickness and Chagas disease. Both are severe parasitic diseases caused by protozoa of the genus Trypanosoma. Sleeping sickness disease is mainly reported in rural Africa. The antigenic variation strategy adopted by this parasite represents a major barrier to the immune system to eliminate it. Consequently, it is difficult for specific mAbs to detect genus-specific antigens [269]. By adopting an in vitro selection method, novel nanobody clones that showed specificity to T. evansi at a species level and genus-specific reactivity against various Trypanosoma species were isolated. Due to their small sizes, nanobodies were shown to be capable of penetrating into the conserved epitopes of antigens that are inaccessible to classical mAbs [270].
Cysticercosis is a serious tissue infection caused by larval cysts of the pork tapeworm that is prevalent in many low-income countries [271]. Monoclonal antibodies that are currently deployed in sandwich ELISAs are mainly genus-specific against Taenia sp., but poorly specific at a species level to identify Taenia solium, the major Taenia species threatening human health [272,273]. To circumvent such limitations, an in vitro selection of nanobodies from immunized dromedaries was developed to recognize a specific marker on T. solium. After in vitro selection, the nanobodies showed no cross-reactivity against other livestock Taenia species, while having a very specific response to a specific 14 kDa glycoprotein (Ts14) in T. solium. Therefore, nanobodies showed potential as an alternative to genus-species mAb for developing unambiguous ELISA tests for human cysticerosis [249]. Apart from being used as diagnostic reagents for infectious diseases, nanobodies have been identified as alternative binders to analyze the compositions of substances in the food and beverage industries. Due to their excellent thermal stability, nanobodies showed superior performance to classical mouse mAbs in ELISA at measuring caffeine concentration in hot and cold beverages [252].
Camelid sdAbs have recently been applied in ELISA methods to detect a wide range of small molecules, including explosive materials (trinitroluene or TNT) [274], agents of bioterrorism (Botulinum A neurotoxin) [235], toxins (ricin, cholera, staphylococcal enterotoxin B) [275], scorpion toxin [276], and viruses (HIV, rotavirus, Vaccinia, and Marburg) [277,278,279]. Owing to the combination of several favorable properties, camelid nanobodies have also been employed in some sophisticated devices to diagnose diseases. In miniature device development, the advanced features of highly stable and unique conformational structures of nanobodies have permitted overcoming many problems faced by traditional whole antibodies and scFv fragments such as cross-reactivity and nanoparticle agglutination. The development of biosensors coupled with nanobodies (nanoconjugates system) has enabled significant improvement in the performance of a device at identifying harmful bacteria (Staphylococcus aureus) at down to a nanometer scale within 10 min [280].
Nevertheless, mAbs remain common binding agents for identifying and tracing tumor-associated proteins for noninvasive in vivo imaging. However, due to their limitations, particularly their large size (150 kDa) and their Fc regions, mAbs penetrate poorly into solid tumors [281]. The emergence of native nanobodies offers the possibility of resolving such problems, and thereby promises the development of probes for diagnosing tumor markers such as EGF receptors [282]. This will enable cancer staging predictions in the blood circulation such as prostate-specific antigen [283]. In view of therapeutic potential, Argen-X (www.argenx.com) has recently developed SIMPLE AntibodyTM (chimeric humanized IgGs) that derived from llama VH and VL domains for the treatment of severe autoimmune diseases and cancer. More applications using camelids VHH targeting specified antigens from various diseases is summarized in Table 2.

6.2. Applications of Shark VNAR Domain

Evidence that IgNAR is part of the shark adaptive immune response was demonstrated in work where increasing levels of hen egg lysozyme (HEL) specific IgNAR developed in shark sera after 4–5 months immunization [224]. The peculiar structure of the shark IgNAR variable domain renders it amenable to the creation of synthetic peptide mimetics to target specific epitopes that are inaccessible to conventional antibodies [264]. Therefore, VNAR may be suitable as new molecular reagents for research and diagnostic applications, and for immunotherapeutic applications.
Apical membrane antigen-1 (AMA1) is a highly polymorphic 83 kDa merozoite surface protein that is essential for erythrocyte invasion by malaria parasites [312]. A VNAR isolated from a wobbegong shark showed high binding affinity to P. falciparum AMA1 through its unique CDR3 region after undergoing affinity maturation [313]. The binding specificity of a monovalent VNAR clone to P. falciparum AMA1 was comparable with commercially available binding reagents, derived from conventional polyclonal sera, monoclonal antibodies, small fragments (Fab, scFv) and peptides [314]. Foley and co-workers demonstrated that the heat stability of purified recombinant VNAR was superior to that of conventional mAbs by targeting immobilized P. falciparum AMA1 in various format at 45 °C, and the refolding property of VNAR was retained when the temperature increased to 80 °C. The excellent stability property at extreme pH and resistance to proteolytic cleavage was further evidenced by incubating VNAR with homogenized murine stomach tissues under in vivo conditions [267]. Based on these properties, it was proposed that VNAR domains have potential for development as alternate binders for malaria diagnostics platforms.
Human periodontal disease is an advanced gingivitis caused by the bacterial pathogen Porhyromonas gingivalis [315]. Late treatment often leads to dental loss due to the accumulation of lysine gingipain (KgP). KgP is a high molecular weight polyprotease produced by P. gingivalis [316]. This bacterial toxin is responsible for destruction of dental tissue of host by suppressing the secretion of specific lytic enzymes from immune system [317]. Nuttall and co-workers (2002) identified two distinct clones specific to KgP from a wobbegong shark VNAR phage display library. The high stability and binding affinity towards P. gingivalis KgP indicated the potential for VNAR as a valuable source of single domain binding reagents [318].
In recent studies, shark VNAR domains have been reported to detect markers from viral diseases at a greater sensitivity. Ebolavirus hemorrhagic fever (EVHF) is a highly lethal disease caused by Bundibugyo virus (BDBV), Sudan virus (SUDV), Tai Forest virus (TAFV), and Zaire Ebolavirus (ZEBOV) [319,320,321]. Shark VNAR and murine scFv phage display libraries have been generated against specified markers on Zaire Ebolavirus. The results indicated that the sensitivity and thermal stability of shark VNAR against viral nucleoprotein (NP) was superior to murine mAb and scFv in this class [262].
As in the case with camelids nanobodies, highly diversified shark VNAR libraries have also been used to detect different kind of toxins, including staphylococcal enterotoxin B (SEB), ricin, botulinum toxin A (BoNT/A) complex toxoid [322], and cholera toxin (CT) [258]. In addition to identifying markers from non-infectious diseases, the intrabody of VNAR has been reported to recognize immunosilent targets in humans, for example the 70 kDa translocase of outer membrane (Tom70) [323]. Owing to the findings of negligible cross-reactivity with other unspecified antigens, and superior heat stability, shark VNAR domains may be potent source of thermal sdAbs over conventional antibodies in diagnostic and biotherapeutic applications. The applications of recombinant shark VNAR against specified antigens from various diseases is summarized in Table 3.

7. Conclusions

Diagnosis by biomarker detection has become a new trend in a wide range of diagnostics, as it could be beneficial for personalized therapy. Conventional antibodies such as IgG and IgM derived from mammals are commonly used as antigen binders in immunoassays for identification of particular disease. With the emergence of genetic engineering, the production of monoclonal antibodies is undoubtedly overcoming many shortcomings presented in polyclonal sera. In order to achieve greater binding efficacy, molecular scientists have continually explored new binders with smaller size and better durability. Natural small-molecule single domain antibodies (sdAbs), functional but not structurally related to the conventional antibodies, have recently been discovered in some ancient animals. They are known as VHH or Nanobodies® from camelids, VNAR from sharks, and, recently, variable-like lymphocytes (VLRs) from lamprey fish. Unlike mammal antibodies, these sdAbs are only composed of heavy protein chains, thereby making them the smallest antibodies thus far. Although most remain at the stage of proof of concept, the advantages of natural sdAbs including better solubility, tissue penetration, stability towards heat and enzymes, and comparatively low production costs offer the posibility of advances in finding new binders for use in research, diagnostic and clinical.

Supplementary Materials

The following are available online at www.mdpi.com/2075-4418/07/4/52/s1.

Acknowledgments

This work is fully sponsored by Malaysian Government and University Sains Malaysia, including Malaysian Ministry of Higher Education the Higher Institutions Centre of Excellence Program (Grant no.: 311/CIPPM/4401005), RUI Grant no.: RU(1001/CIPPM/811296), and USM Short Term (Grant no.: 304/CIPPM/6313191).

Author Contributions

All authors made contribution to this study. Chiuan Herng Leow, Chiuan Yee Leow and Candy Chuah conceived the antibody and biomarker discovery section. Katja Fischer, Qin Cheng and James McCarthy coordinated and led the manuscript writing. All authors have read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hendriksen, I.C.; Ferro, J.; Montoya, P.; Chhaganlal, K.D.; Seni, A.; Gomes, E.; Silamut, K.; Lee, S.J.; Lucas, M.; Chotivanich, K.; et al. Diagnosis, clinical presentation, and in-hospital mortality of severe malaria in HIV-coinfected children and adults in Mozambique. Clin. Infect. Dis. 2012, 55, 1144–1153. [Google Scholar] [CrossRef] [PubMed]
  2. Murphy, J.F. Reducing misdiagnosis in clinical practice. Ir. Med. J. 2009, 102, 68. [Google Scholar] [PubMed]
  3. Zegarra Montes, L.Z.; Sanchez Mejia, A.A.; Loza Munarriz, C.A.; Gutierrez, E.C. Semen and urine culture in the diagnosis of chronic bacterial prostatitis. Int. Braz. J. Urol. 2008, 34, 30–37; discussion 38–40. [Google Scholar] [CrossRef] [PubMed]
  4. Olson, W.C.; Smolkin, M.E.; Farris, E.M.; Fink, R.J.; Czarkowski, A.R.; Fink, J.H.; Chianese-Bullock, K.A.; Slingluff, C.L., Jr. Shipping blood to a central laboratory in multicenter clinical trials: Effect of ambient temperature on specimen temperature, and effects of temperature on mononuclear cell yield, viability and immunologic function. J. Transl. Med. 2011, 9, 26. [Google Scholar] [CrossRef] [PubMed]
  5. Wilson, M.L. General principles of specimen collection and transport. Clin. Infect. Dis. 1996, 22, 766–777. [Google Scholar] [CrossRef] [PubMed]
  6. Wongsrichanalai, C.; Barcus, M.J.; Muth, S.; Sutamihardja, A.; Wernsdorfer, W.H. A review of malaria diagnostic tools: Microscopy and rapid diagnostic test (RDT). Am. J. Trop. Med. Hyg. 2007, 77, 119–127. [Google Scholar] [PubMed]
  7. Thomson, S.; Lohmann, R.C.; Crawford, L.; Dubash, R.; Richardson, H. External quality assessment in the examination of blood films for malarial parasites within Ontario, Canada. Arch. Pathol. Lab. Med. 2000, 124, 57–60. [Google Scholar] [PubMed]
  8. Johnston, S.P.; Pieniazek, N.J.; Xayavong, M.V.; Slemenda, S.B.; Wilkins, P.P.; da Silva, A.J. PCR as a confirmatory technique for laboratory diagnosis of malaria. J. Clin. Microbiol. 2006, 44, 1087–1089. [Google Scholar] [CrossRef] [PubMed]
  9. Hulka, B.S. Overview of biological markers. In Biological Markers in Epidemiology; Oxford University Press: New York, NY, USA, 1990; pp. 3–15. [Google Scholar]
  10. Tiernan, J.P.; Perry, S.L.; Verghese, E.T.; West, N.P.; Yeluri, S.; Jayne, D.G.; Hughes, T.A. Carcinoembryonic antigen is the preferred biomarker for in vivo colorectal cancer targeting. Br. J. Cancer 2013, 108, 662–667. [Google Scholar] [CrossRef] [PubMed]
  11. Watabe-Rudolph, M.; Song, Z.; Lausser, L.; Schnack, C.; Begus-Nahrmann, Y.; Scheithauer, M.O.; Rettinger, G.; Otto, M.; Tumani, H.; Thal, D.R.; et al. Chitinase enzyme activity in CSF is a powerful biomarker of Alzheimer disease. Neurology 2012, 78, 569–577. [Google Scholar] [CrossRef] [PubMed]
  12. Dawson, S.J.; Tsui, D.W.; Murtaza, M.; Biggs, H.; Rueda, O.M.; Chin, S.F.; Dunning, M.J.; Gale, D.; Forshew, T.; Mahler-Araujo, B.; et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef] [PubMed]
  13. Niedbala, R.S.; Mauck, C.; Harrison, P.; Doncel, G.F. Biomarker discovery: Validation and decision-making in product development. Sex. Transm. Dis. 2009, 36, S76–S80. [Google Scholar] [CrossRef] [PubMed]
  14. Rai, A.J.; Zhang, Z.; Rosenzweig, J.; Shih Ie, M.; Pham, T.; Fung, E.T.; Sokoll, L.J.; Chan, D.W. Proteomic approaches to tumor marker discovery. Arch. Pathol. Lab. Med. 2002, 126, 1518–1526. [Google Scholar] [PubMed]
  15. Lopez, M.M.; Bertolini, E.; Olmos, A.; Caruso, P.; Gorris, M.T.; Llop, P.; Penyalver, R.; Cambra, M. Innovative tools for detection of plant pathogenic viruses and bacteria. Int. Microbiol. 2003, 6, 233–243. [Google Scholar] [CrossRef] [PubMed]
  16. Kerner, J.; Lee, K.; Tandler, B.; Hoppel, C.L. VDAC proteomics: Post-translation modifications. Biochim. Biophys. Acta 2012, 1818, 1520–1525. [Google Scholar] [CrossRef] [PubMed]
  17. Demerdash, Z.A.; Diab, T.M.; Aly, I.R.; Mohamed, S.H.; Mahmoud, F.S.; Zoheiry, M.K.; Mansour, W.A.; Attia, M.E.; El-Bassiouny, A.E. Diagnostic efficacy of monoclonal antibody based sandwich enzyme linked immunosorbent assay (ELISA) for detection of Fasciola gigantica excretory/secretory antigens in both serum and stool. Parasites Vectors 2011, 4, 176. [Google Scholar] [CrossRef] [PubMed]
  18. Fujiwara, K.; Yoshizaki, Y.; Shin, M.; Miyazaki, T.; Saita, T.; Nagata, S. Immunocytochemistry for vancomycin using a monoclonal antibody that reveals accumulation of the drug in rat kidney and liver. Antimicrob. Agents Chemother. 2012, 56, 5883–5891. [Google Scholar] [CrossRef] [PubMed]
  19. Zhou, J.; Belov, L.; Solomon, M.J.; Chan, C.; Clarke, S.J.; Christopherson, R.I. Colorectal cancer cell surface protein profiling using an antibody microarray and fluorescence multiplexing. J. Vis. Exp. 2011. [Google Scholar] [CrossRef] [PubMed]
  20. Raponi, S.; de Propris, M.S.; Intoppa, S.; Milani, M.L.; Vitale, A.; Elia, L.; Perbellini, O.; Pizzolo, G.; Foa, R.; Guarini, A. Flow cytometric study of potential target antigens (CD19, CD20, CD22, CD33) for antibody-based immunotherapy in acute lymphoblastic leukemia: Analysis of 552 cases. Leuk. Lymphoma 2011, 52, 1098–1107. [Google Scholar] [CrossRef] [PubMed]
  21. Zandian, M.; Jungbauer, A. An immunoaffinity column with a monoclonal antibody as ligand for human follicle stimulating hormone. J. Sep. Sci. 2009, 32, 1585–1591. [Google Scholar] [CrossRef] [PubMed]
  22. Vora, A.J.; Nuttall, P.; James, V. Screening plasma HAV antibody sources. Lancet 1991, 338, 62. [Google Scholar] [CrossRef]
  23. Spillner, E.; Braren, I.; Greunke, K.; Seismann, H.; Blank, S.; du Plessis, D. Avian IgY antibodies and their recombinant equivalents in research, diagnostics and therapy. Biologicals 2012, 40, 313–322. [Google Scholar] [CrossRef] [PubMed]
  24. Hanly, W.C.; Artwohl, J.E.; Bennett, B.T. Review of Polyclonal Antibody Production Procedures in Mammals and Poultry. ILAR J. 1995, 37, 93–118. [Google Scholar] [CrossRef] [PubMed]
  25. Gaciarz, A.; Veijola, J.; Uchida, Y.; Saaranen, M.J.; Wang, C.; Horkko, S.; Ruddock, L.W. Systematic screening of soluble expression of antibody fragments in the cytoplasm of E. coli. Microb. Cell Fact. 2016, 15, 22. [Google Scholar] [CrossRef] [PubMed]
  26. Zielonka, S.; Empting, M.; Grzeschik, J.; Konning, D.; Barelle, C.J.; Kolmar, H. Structural insights and biomedical potential of IgNAR scaffolds from sharks. MAbs 2015, 7, 15–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Saerens, D.; Ghassabeh, G.H.; Muyldermans, S. Single-domain antibodies as building blocks for novel therapeutics. Curr. Opin. Pharmacol. 2008, 8, 600–608. [Google Scholar] [CrossRef] [PubMed]
  28. O’Kennedy, R.; Roben, P. Antibody engineering: An overview. Essays Biochem. 1991, 26, 59–75. [Google Scholar] [PubMed]
  29. Ahmad, Z.A.; Yeap, S.K.; Ali, A.M.; Ho, W.Y.; Alitheen, N.B.; Hamid, M. scFv antibody: Principles and clinical application. Clin. Dev. Immunol. 2012, 2012, 980250. [Google Scholar] [CrossRef] [PubMed]
  30. Tu, B.; Ziemann, R.N.; Tieman, B.C.; Hawksworth, D.J.; Tyner, J.; Scheffel, J.; Pinkus, M.S.; Brophy, S.E.; Werneke, J.M.; Gutierrez, R.; et al. Generation and characterization of chimeric antibodies against NS3, NS4, NS5, and core antigens of hepatitis C virus. Clin. Vaccine Immunol. 2010, 17, 1040–1047. [Google Scholar] [CrossRef] [PubMed]
  31. Karu, A.E.; Bell, C.W.; Chin, T.E. Recombinant Antibody Technology. ILAR J. 1995, 37, 132–141. [Google Scholar] [CrossRef]
  32. Azzazy, H.M.; Highsmith, W.E., Jr. Phage display technology: Clinical applications and recent innovations. Clin. Biochem. 2002, 35, 425–445. [Google Scholar] [CrossRef]
  33. Hoogenboom, H.R. Selecting and screening recombinant antibody libraries. Nat. Biotechnol. 2005, 23, 1105–1116. [Google Scholar] [CrossRef] [PubMed]
  34. Dick, H.M. Single domain antibodies. Br. Med. J. 1990, 300, 959–960. [Google Scholar] [CrossRef]
  35. Diaz, M.; Stanfield, R.L.; Greenberg, A.S.; Flajnik, M.F. Structural analysis, selection, and ontogeny of the shark new antigen receptor (IgNAR): Identification of a new locus preferentially expressed in early development. Immunogenetics 2002, 54, 501–512. [Google Scholar] [CrossRef] [PubMed]
  36. Stijlemans, B.; Caljon, G.; Natesan, S.K.; Saerens, D.; Conrath, K.; Perez-Morga, D.; Skepper, J.N.; Nikolaou, A.; Brys, L.; Pays, E.; et al. High affinity nanobodies against the Trypanosome brucei VSG are potent trypanolytic agents that block endocytosis. PLoS Pathog. 2011, 7, e1002072. [Google Scholar] [CrossRef] [PubMed]
  37. Shao, C.Y.; Secombes, C.J.; Porter, A.J. Rapid isolation of IgNAR variable single-domain antibody fragments from a shark synthetic library. Mol. Immunol. 2007, 44, 656–665. [Google Scholar] [CrossRef] [PubMed]
  38. Herrin, B.R.; Alder, M.N.; Roux, K.H.; Sina, C.; Ehrhardt, G.R.; Boydston, J.A.; Turnbough, C.L., Jr.; Cooper, M.D. Structure and specificity of lamprey monoclonal antibodies. Proc. Natl. Acad. Sci. USA 2008, 105, 2040–2045. [Google Scholar] [CrossRef] [PubMed]
  39. Akazawa-Ogawa, Y.; Takashima, M.; Lee, Y.H.; Ikegami, T.; Goto, Y.; Uegaki, K.; Hagihara, Y. Heat-induced irreversible denaturation of the camelid single domain VHH antibody is governed by chemical modifications. J. Biol. Chem. 2014, 289, 15666–15679. [Google Scholar] [CrossRef] [PubMed]
  40. Kovaleva, M.; Ferguson, L.; Steven, J.; Porter, A.; Barelle, C. Shark variable new antigen receptor biologics—A novel technology platform for therapeutic drug development. Expert Opin. Biol. Ther. 2014, 14, 1527–1539. [Google Scholar] [CrossRef] [PubMed]
  41. Tucker, J.D.; Bu, J.; Brown, L.B.; Yin, Y.P.; Chen, X.S.; Cohen, M.S. Accelerating worldwide syphilis screening through rapid testing: A systematic review. Lancet Infect. Dis. 2010, 10, 381–386. [Google Scholar] [CrossRef]
  42. Suppiah, J.; Thimma, J.S.; Cheah, S.H.; Vadivelu, J. Development and evaluation of polymerase chain reaction assay to detect Burkholderia genus and to differentiate the species in clinical specimens. FEMS Microbiol. Lett. 2010, 306, 9–14. [Google Scholar] [CrossRef] [PubMed]
  43. Saetiew, C.; Limpaiboon, T.; Jearanaikoon, P.; Daduang, S.; Pientong, C.; Kerdsin, A.; Daduang, J. Rapid detection of the most common high-risk human papillomaviruses by loop-mediated isothermal amplification. J. Virol. Methods 2011, 178, 22–30. [Google Scholar] [CrossRef] [PubMed]
  44. Tessitore, A.; Gaggiano, A.; Cicciarelli, G.; Verzella, D.; Capece, D.; Fischietti, M.; Zazzeroni, F.; Alesse, E. Serum biomarkers identification by mass spectrometry in high-mortality tumors. Int. J. Proteom. 2013, 2013, 125858. [Google Scholar] [CrossRef] [PubMed]
  45. Balmer, O.; Tostado, C. New fluorescence markers to distinguish co-infecting Trypanosoma brucei strains in experimental multiple infections. Acta Trop. 2006, 97, 94–101. [Google Scholar] [CrossRef] [PubMed]
  46. Petersen, E.R.B.; Olsen, D.A.; Christensen, H.; Hansen, S.B.; Christensen, C.; Brandslund, I. Rhodopsin in plasma from patients with diabetic retinopathy—Development and validation of digital ELISA by Single Molecule Array (Simoa) technology. J. Immunol. Methods 2017, 446, 60–69. [Google Scholar] [CrossRef] [PubMed]
  47. Ndao, M. Diagnosis of parasitic diseases: Old and new approaches. Interdiscip. Perspect. Infect. Dis. 2009, 2009, 278246. [Google Scholar] [CrossRef] [PubMed]
  48. Tangpukdee, N.; Duangdee, C.; Wilairatana, P.; Krudsood, S. Malaria diagnosis: A brief review. Korean J. Parasitol. 2009, 47, 93–102. [Google Scholar] [CrossRef] [PubMed]
  49. Esquivel-Velazquez, M.; Ostoa-Saloma, P.; Morales-Montor, J.; Hernandez-Bello, R.; Larralde, C. Immunodiagnosis of neurocysticercosis: Ways to focus on the challenge. J. Biomed. Biotechnol. 2011, 2011, 516042. [Google Scholar] [CrossRef] [PubMed]
  50. Hang, V.T.; Nguyet, N.M.; Trung, D.T.; Tricou, V.; Yoksan, S.; Dung, N.M.; van Ngoc, T.; Hien, T.T.; Farrar, J.; Wills, B.; et al. Diagnostic accuracy of NS1 ELISA and lateral flow rapid tests for dengue sensitivity, specificity and relationship to viraemia and antibody responses. PLoS Negl. Trop. Dis. 2009, 3, e360. [Google Scholar] [CrossRef] [PubMed]
  51. Houze, S.; Boutron, I.; Marmorat, A.; Dalichampt, M.; Choquet, C.; Poilane, I.; Godineau, N.; le Guern, A.S.; Thellier, M.; Broutier, H.; et al. Performance of rapid diagnostic tests for imported malaria in clinical practice: Results of a national multicenter study. PLoS ONE 2013, 8, e75486. [Google Scholar] [CrossRef] [PubMed]
  52. Masanja, M.I.; McMorrow, M.; Kahigwa, E.; Kachur, S.P.; McElroy, P.D. Health workers’ use of malaria rapid diagnostic tests (RDTs) to guide clinical decision making in rural dispensaries, Tanzania. Am. J. Trop. Med. Hyg. 2010, 83, 1238–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ng, W.Y.; Tiong, C.C.; Jacob, E. Maltose interference-free test strips for blood glucose testing at point-of-care: A laboratory performance evaluation. Diabetes Technol. Ther. 2010, 12, 889–893. [Google Scholar] [CrossRef] [PubMed]
  54. Lafleur, L.; Stevens, D.; McKenzie, K.; Ramachandran, S.; Spicar-Mihalic, P.; Singhal, M.; Arjyal, A.; Osborn, J.; Kauffman, P.; Yager, P.; et al. Progress toward multiplexed sample-to-result detection in low resource settings using microfluidic immunoassay cards. Lab. Chip 2012, 12, 1119–1127. [Google Scholar] [CrossRef] [PubMed]
  55. Sheppard, C.L.; Guiver, M.; Hartley, J.; Harrison, T.G.; George, R.C. Use of a multiplexed immunoassay for detection of serotype-specific Streptococcus pneumoniae antigen in pleural fluid and cerebrospinal fluid specimens. J. Med. Microbiol. 2011, 60, 1879–1881. [Google Scholar] [CrossRef] [PubMed]
  56. Taniuchi, M.; Verweij, J.J.; Noor, Z.; Sobuz, S.U.; Lieshout, L.; Petri, W.A., Jr.; Haque, R.; Houpt, E.R. High throughput multiplex PCR and probe-based detection with Luminex beads for seven intestinal parasites. Am. J. Trop. Med. Hyg. 2011, 84, 332–337. [Google Scholar] [CrossRef] [PubMed]
  57. Van Doorn, H.R.; Kinh, N.; Tuan, H.M.; Tuan, T.A.; Minh, N.N.; Bryant, J.E.; Hang, V.; Uyen le, T.T.; Thinh le, Q.; Anh, T.; et al. Clinical validation of a point-of-care multiplexed in vitro immunoassay using monoclonal antibodies (the MSD influenza test) in four hospitals in Vietnam. J. Clin. Microbiol. 2012, 50, 1621–1625. [Google Scholar] [CrossRef] [PubMed]
  58. Balboni, I.; Limb, C.; Tenenbaum, J.D.; Utz, P.J. Evaluation of microarray surfaces and arraying parameters for autoantibody profiling. Proteomics 2008, 8, 3443–3449. [Google Scholar] [CrossRef] [PubMed]
  59. World Health Organization (WHO). Noncommunicable Diseases—Fact Sheet Updated June 2017; Media Centre: Geneva, Switzerland, 2017. [Google Scholar]
  60. Collier, A.; Ghosh, S.; McGlynn, B.; Hollins, G. Prostate cancer, androgen deprivation therapy, obesity, the metabolic syndrome, type 2 diabetes, and cardiovascular disease: A review. Am. J. Clin. Oncol. 2012, 35, 504–509. [Google Scholar] [CrossRef] [PubMed]
  61. Van Horn, L. What do cancer, diabetes, and cardiovascular disease have in common? J. Am. Diet. Assoc. 2009, 109, 1329. [Google Scholar] [CrossRef] [PubMed]
  62. World Health Organization (WHO). World Cancer Report 2008; World Health Organization (WHO): Geneva, Switzerland, 2008; p. 512. [Google Scholar]
  63. Ferlay, J.; Bray, F.; Pisani, P.; Parkin, D.M. Cancer, Incidence, Mortality and Prevalence Worldwide IARC CancerBase; Version 2.0; IARC Press: Lyon, France, 2004. [Google Scholar]
  64. Li, X.; Scarfe, A.; King, K.; Fenton, D.; Butts, C.; Winget, M. Timeliness of cancer care from diagnosis to treatment: A comparison between patients with breast, colon, rectal or lung cancer. Int. J. Qual. Health Care 2013, 25, 197–204. [Google Scholar] [CrossRef] [PubMed]
  65. Gretzer, M.B.; Partin, A.W. PSA markers in prostate cancer detection. Urol. Clin. N. Am. 2003, 30, 677–686. [Google Scholar] [CrossRef]
  66. Crawford, N.P.; Colliver, D.W.; Galandiuk, S. Tumor markers and colorectal cancer: Utility in management. J. Surg. Oncol. 2003, 84, 239–248. [Google Scholar] [CrossRef] [PubMed]
  67. Cheung, K.L.; Robertson, F.R. Objective measurement of remission and progression in metastatic breast cancer by the use of serum tumour markers. Minerva Chir. 2003, 58, 297–303. [Google Scholar] [PubMed]
  68. Hanna, W.M.; Kahn, H.J.; Pienkowska, M.; Blondal, J.; Seth, A.; Marks, A. Defining a test for HER-2/neu evaluation in breast cancer in the diagnostic setting. Mod. Pathol. 2001, 14, 677–685. [Google Scholar] [CrossRef] [PubMed]
  69. Trompetas, V.; Panagopoulos, E.; Priovolou-Papaevangelou, M.; Ramantanis, G. Giant benign true cyst of the spleen with high serum level of CA 19-9. Eur. J. Gastroenterol. Hepatol. 2002, 14, 85–88. [Google Scholar] [CrossRef] [PubMed]
  70. Anderiesz, C.; Quinn, M.A. Screening for ovarian cancer. Med. J. Aust. 2003, 178, 655–656. [Google Scholar] [PubMed]
  71. Wayner, E.A.; Quek, S.I.; Ahmad, R.; Ho, M.E.; Loprieno, M.A.; Zhou, Y.; Ellis, W.J.; True, L.D.; Liu, A.Y. Development of an ELISA to detect the secreted prostate cancer biomarker AGR2 in voided urine. Prostate 2012, 72, 1023–1034. [Google Scholar] [CrossRef] [PubMed]
  72. Cheng, H.Y.; Lai, L.J.; Ko, F.H. Rapid and sensitive detection of rare cancer cells by the coupling of immunomagnetic nanoparticle separation with ELISA analysis. Int. J. Nanomed. 2012, 7, 2967–2973. [Google Scholar] [CrossRef] [PubMed]
  73. Tritschler, S.; Scharf, S.; Karl, A.; Tilki, D.; Knuechel, R.; Hartmann, A.; Stief, C.G.; Zaak, D. Validation of the diagnostic value of NMP22 BladderChek test as a marker for bladder cancer by photodynamic diagnosis. Eur. Urol. 2007, 51, 403–407; discussion 407–408. [Google Scholar] [CrossRef] [PubMed]
  74. Yang, M.; Javadi, A.; Li, H.; Gong, S. Ultrasensitive immunosensor for the detection of cancer biomarker based on graphene sheet. Biosens. Bioelectron. 2010, 26, 560–565. [Google Scholar] [CrossRef] [PubMed]
  75. Adams, A.A.; Okagbare, P.I.; Feng, J.; Hupert, M.L.; Patterson, D.; Gottert, J.; McCarley, R.L.; Nikitopoulos, D.; Murphy, M.C.; Soper, S.A. Highly efficient circulating tumor cell isolation from whole blood and label-free enumeration using polymer-based microfluidics with an integrated conductivity sensor. J. Am. Chem. Soc. 2008, 130, 8633–8641. [Google Scholar] [CrossRef] [PubMed]
  76. Kashani-Sabet, M.; Rangel, J.; Torabian, S.; Nosrati, M.; Simko, J.; Jablons, D.M.; Moore, D.H.; Haqq, C.; Miller, J.R., 3rd; Sagebiel, R.W. A multi-marker assay to distinguish malignant melanomas from benign nevi. Proc. Natl. Acad. Sci. USA 2009, 106, 6268–6272. [Google Scholar] [CrossRef] [PubMed]
  77. Moraes, G.S.; Amaral Cristovam, R.; Savaris, R.F. Comparative analysis of the accuracy of urinary hCG tests in vitro. Rev. Assoc. Med. Bras. 2011, 57, 516–522. [Google Scholar] [CrossRef] [PubMed]
  78. Taylor, A.E.; Khoury, R.H.; Crowley, W.F., Jr. A comparison of 13 different immunometric assay kits for gonadotropins: Implications for clinical investigation. J. Clin. Endocrinol. Metab. 1994, 79, 240–247. [Google Scholar] [PubMed]
  79. Coons, S.J. A look at the purchase and use of home pregnancy-test kits. Am. Pharm. 1989, NS29, 46–48. [Google Scholar] [PubMed]
  80. Grudzinskas, J.G.; Lenton, E.A.; Gordon, Y.B.; Kelso, I.M.; Jeffrey, D.; Sobowale, O.; Chard, T. Circulating levels of pregnancy-specific beta1-glycoprotein in early pregnancy. Br. J. Obstet. Gynaecol. 1977, 84, 740–742. [Google Scholar] [CrossRef] [PubMed]
  81. Bohn, H.; Kraus, W.; Winckler, W. New soluble placental tissue proteins: Their isolation, characterization, localization and quantification. Placenta Suppl. 1982, 4, 67–81. [Google Scholar] [PubMed]
  82. Whyte, A.; Heap, R.B. Reproductive immunology. Early pregnancy factor. Nature 1983, 304, 121–122. [Google Scholar] [CrossRef] [PubMed]
  83. Morton, H.; Rolfe, B.; Clunie, G.J. An early pregnancy factor detected in human serum by the rosette inhibition test. Lancet 1977, 1, 394–397. [Google Scholar] [CrossRef]
  84. Snyder, J.A.; Haymond, S.; Parvin, C.A.; Gronowski, A.M.; Grenache, D.G. Diagnostic considerations in the measurement of human chorionic gonadotropin in aging women. Clin. Chem. 2005, 51, 1830–1835. [Google Scholar] [CrossRef] [PubMed]
  85. Matsui, H.; Kihara, M.; Usui, H.; Tate, S.; Mitsuhashi, A.; Shozu, M. Comparison of 2 commercially available human chorionic gonadotropin immunoassays used in the management of gestational trophoblastic neoplasia. J. Reprod. Med. 2009, 54, 631–635. [Google Scholar] [PubMed]
  86. Vaitukaitis, J.L. Development of the home pregnancy test. Ann. N. Y. Acad. Sci. 2004, 1038, 220–222. [Google Scholar] [CrossRef] [PubMed]
  87. Lapthorn, A.J.; Harris, D.C.; Littlejohn, A.; Lustbader, J.W.; Canfield, R.E.; Machin, K.J.; Morgan, F.J.; Isaacs, N.W. Crystal structure of human chorionic gonadotropin. Nature 1994, 369, 455–461. [Google Scholar] [CrossRef] [PubMed]
  88. Alfthan, H.; Schroder, J.; Fraser, R.; Koskimies, A.; Halila, H.; Stenman, U.H. Choriogonadotropin and its beta subunit separated by hydrophobic-interaction chromatography and quantified in serum during pregnancy by time-resolved immunofluorometric assays. Clin. Chem. 1988, 34, 1758–1762. [Google Scholar] [PubMed]
  89. Spona, J.; Dancoine, F.; Lorenz, U.; Valente, D.; Wasley, G.D. Early detection of pregnancy by new beta-hCG monoclonal urine test. Gynecol. Obstet. Investig. 1985, 19, 6–10. [Google Scholar] [CrossRef]
  90. Bastian, L.A.; Nanda, K.; Hasselblad, V.; Simel, D.L. Diagnostic efficiency of home pregnancy test kits. A meta-analysis. Arch. Fam. Med. 1998, 7, 465–469. [Google Scholar] [CrossRef] [PubMed]
  91. Cole, L.A.; Khanlian, S.A. The need for a quantitative urine hCG assay. Clin. Biochem. 2009, 42, 676–683. [Google Scholar] [CrossRef] [PubMed]
  92. De Medeiros, S.F.; Amato, F.; Matthews, C.D.; Norman, R.J. Comparison of specific immunoassays for detection of the β-core human chorionic gonadotrophin fragment in body fluids. J. Endocrinol. 1992, 135, 161–174. [Google Scholar] [CrossRef] [PubMed]
  93. Norman, R.J.; Lowings, C.; Chard, T. Dipstick method for human chorionic gonadotropin suitable for emergency use on whole blood and other fluids. Lancet 1985, 1, 19–20. [Google Scholar] [CrossRef]
  94. Chow, S.N.; Ouyang, P.C.; Chu, C.T.; Lee, C.Y. Rapid and simple immunoassays for measurement of human chorionic gonadotropin using monoclonal antibodies. J. Formos. Med. Assoc. 1990, 89, 792–798. [Google Scholar] [PubMed]
  95. Spyropoulou, I.; Karamalegos, C.; Bolton, V.N. A prospective randomized study comparing the outcome of in vitro fertilization and embryo transfer following culture of human embryos individually or in groups before embryo transfer on day 2. Hum. Reprod. 1999, 14, 76–79. [Google Scholar] [CrossRef] [PubMed]
  96. Holl, K.; Lundin, E.; Kaasila, M.; Grankvist, K.; Afanasyeva, Y.; Hallmans, G.; Lehtinen, M.; Pukkala, E.; Surcel, H.M.; Toniolo, P.; et al. Effect of long-term storage on hormone measurements in samples from pregnant women: The experience of the Finnish Maternity Cohort. Acta Oncol. 2008, 47, 406–412. [Google Scholar] [CrossRef] [PubMed]
  97. Karalliedde, L. Animal toxins. Br. J. Anaesth. 1995, 74, 319–327. [Google Scholar] [CrossRef] [PubMed]
  98. Pollyea, D.A.; George, T.I.; Corless, C.; Gotlib, J. When yellow jackets attack: Recurrent and severe anaphylactic reactions to insect bites and stings. Am. J. Hematol. 2009, 84, 843–846. [Google Scholar] [CrossRef] [PubMed]
  99. Nowatzki, J.; Sene, R.V.; Paludo, K.S.; Rizzo, L.E.; Souza-Fonseca-Guimaraes, F.; Veiga, S.S.; Nader, H.B.; Franco, C.R.; Trindade, E.S. Brown spider (Loxosceles intermedia) venom triggers endothelial cells death by anoikis. Toxicon 2012, 60, 396–405. [Google Scholar] [CrossRef] [PubMed]
  100. Cruz, L.S.; Vargas, R.; Lopes, A.A. Snakebite envenomation and death in the developing world. Ethn. Dis. 2009, 19, S42–S46. [Google Scholar]
  101. Girish, K.S.; Kemparaju, K. Overlooked issues of snakebite management: Time for strategic approach. Curr. Top. Med. Chem. 2011, 11, 2494–2508. [Google Scholar] [CrossRef] [PubMed]
  102. Theakston, R.D.; Lloyd-Jones, M.J.; Reid, H.A. Micro-ELISA for detecting and assaying snake venom and venom-antibody. Lancet 1977, 2, 639–641. [Google Scholar] [CrossRef]
  103. Rial, A.; Morais, V.; Rossi, S.; Massaldi, H. A new ELISA for determination of potency in snake antivenoms. Toxicon 2006, 48, 462–466. [Google Scholar] [CrossRef] [PubMed]
  104. Cox, J.C.; Moisidis, A.V.; Shepherd, J.M.; Drane, D.P.; Jones, S.L. A novel format for a rapid sandwich EIA and its application to the identification of snake venoms. J. Immunol. Methods 1992, 146, 213–218. [Google Scholar] [CrossRef]
  105. Sutherland, S.K. Antivenom use in Australia. Premedication, adverse reactions and the use of venom detection kits. Med. J. Aust. 1992, 157, 734–739. [Google Scholar] [PubMed]
  106. Sutherland, S.K.; Leonard, R.L. Snakebite deaths in Australia 1992–1994 and a management update. Med. J. Aust. 1995, 163, 616–618. [Google Scholar] [PubMed]
  107. Trevett, A.J.; Lalloo, D.G.; Nwokolo, N.C.; Theakston, D.G.; Naraqi, S.; Warrell, D.A. Venom detection kits in the management of snakebite in Central province, Papua New Guinea. Toxicon 1995, 33, 703–705. [Google Scholar] [CrossRef]
  108. Tibballs, J.; Kuruppu, S.; Hodgson, W.C.; Carroll, T.; Hawdon, G.; Sourial, M.; Baker, T.; Winkel, K. Cardiovascular, haematological and neurological effects of the venom of the Papua New Guinean small-eyed snake (Micropechis ikaheka) and their neutralisation with CSL polyvalent and black snake antivenoms. Toxicon 2003, 42, 647–655. [Google Scholar] [CrossRef] [PubMed]
  109. Ong, R.K.; Swindells, K.; Mansfield, C.S. Prospective determination of the specificity of a commercial snake venom detection kit in urine samples from dogs and cats. Aust. Vet. J. 2010, 88, 222–224. [Google Scholar] [CrossRef] [PubMed]
  110. Van Dong, L.; Quyen le, K.; Eng, K.H.; Gopalakrishnakone, P. Immunogenicity of venoms from four common snakes in the South of Vietnam and development of ELISA kit for venom detection. J. Immunol. Methods 2003, 282, 13–31. [Google Scholar] [CrossRef]
  111. Le Dong, V.; Selvanayagam, Z.E.; Gopalakrishnakone, P.; Eng, K.H. A new avidin-biotin optical immunoassay for the detection of β-bungarotoxin and application in diagnosis of experimental snake envenomation. J. Immunol. Methods 2002, 260, 125–136. [Google Scholar] [CrossRef]
  112. Ho, M.; Warrell, M.J.; Warrell, D.A.; Bidwell, D.; Voller, A. A critical reappraisal of the use of enzyme-linked immunosorbent assays in the study of snake bite. Toxicon 1986, 24, 211–221. [Google Scholar] [CrossRef]
  113. Selvanayagam, Z.E.; Gopalakrishnakone, P. Tests for detection of snake venoms, toxins and venom antibodies: Review on recent trends (1987–1997). Toxicon 1999, 37, 565–586. [Google Scholar] [CrossRef]
  114. Bonacini, M.; Lin, H.J.; Hollinger, F.B. Effect of coexisting HIV-1 infection on the diagnosis and evaluation of hepatitis C virus. J. Acquir. Immune Defic. Syndr. 2001, 26, 340–344. [Google Scholar] [CrossRef] [PubMed]
  115. Alvarez-Fernandez, M.; Potel-Alvarellos, C.; Alvarez-Garcia, P.; Otero-Varela, M.I.; Constenla-Carames, L. An outbreak of negative rapid agglutination test meticillin-resistant Staphylococcus aureus. J. Hosp. Infect. 2008, 69, 194–195. [Google Scholar] [CrossRef] [PubMed]
  116. Abate, K. Modern-day malaria: An overview of this lingering threat. Adv. Nurse Pract 2008, 16, 67–68. [Google Scholar] [PubMed]
  117. Bygbjerg, I.C. Double burden of noncommunicable and infectious diseases in developing countries. Science 2012, 337, 1499–1501. [Google Scholar] [CrossRef] [PubMed]
  118. Zaidi, A.K.; Awasthi, S.; deSilva, H.J. Burden of infectious diseases in South Asia. Br. Med. J. 2004, 328, 811–815. [Google Scholar] [CrossRef] [PubMed]
  119. World Health Organization (WHO). World Health Organization Fact. Sheets: Infectious Diseases; World Health Organization (WHO): Geneva, Switzerland, 2010. [Google Scholar]
  120. Pultorak, E.; Wong, W.; Rabins, C.; Mehta, S.D. Economic burden of sexually transmitted infections: Incidence and direct medical cost of Chlamydia, gonorrhea, and syphilis among Illinois adolescents and young adults, 2005–2006. Sex. Transm. Dis. 2009, 36, 629–636. [Google Scholar] [CrossRef] [PubMed]
  121. Mao, S.P.; Shao, B.R. Schistosomiasis control in the people’s Republic of China. Am. J. Trop. Med. Hyg. 1982, 31, 92–99. [Google Scholar] [CrossRef] [PubMed]
  122. Qiu, S.; Xu, X.; Wang, Y.; Yang, G.; Wang, Z.; Wang, H.; Zhang, L.; Liu, N.; Chen, C.; Liu, W.; et al. Emergence of resistance to fluoroquinolones and third-generation cephalosporins in Shigella flexneri subserotype 1c isolates from China. Clin. Microbiol. Infect. 2012, 18, E95–E98. [Google Scholar] [CrossRef] [PubMed]
  123. The Joint United Nations Programme on HIV/AIDS (UNAIDS). Global HIV/AIDS Response Progress Report 2011: Epidemic Update and Health Sector Progress towards Universal Access; World Health Organization, UNAIDS: Geneva, Switzerland, 2011; p. 229. [Google Scholar]
  124. Goldsby, R.A.; Kindt, T.J.; Osborne, B.A. AIDS and Other Immunodeficiencies, 4th ed.; W.H. Freeman and Company: New York, NY, USA, 2000; pp. 467–496. [Google Scholar]
  125. Piwowar-Manning, E.M.; Tustin, N.B.; Sikateyo, P.; Kamwendo, D.; Chipungu, C.; Maharaj, R.; Mushanyu, J.; Richardson, B.A.; Hillier, S.; Brooks Jackson, J. Validation of rapid HIV antibody tests in 5 African countries. J. Int. Assoc. Physicians AIDS Care 2010, 9, 170–172. [Google Scholar] [CrossRef] [PubMed]
  126. Ferreira Junior, O.C.; Ferreira, C.; Riedel, M.; Widolin, M.R.; Barbosa-Junior, A. Evaluation of rapid tests for anti-HIV detection in Brazil. AIDS 2005, 19, S70–S75. [Google Scholar] [CrossRef] [PubMed]
  127. Gurtler, L. Difficulties and strategies of HIV diagnosis. Lancet 1996, 348, 176–179. [Google Scholar] [CrossRef]
  128. Kivuyo, S.L.; Johannessen, A.; Troseid, M.; Kasubi, M.J.; Gundersen, S.G.; Naman, E.; Mushi, D.; Ngowi, B.J.; Mfinanga, G.S.; Bruun, J.N. p24 antigen detection on dried blood spots is a feasible and reliable test for infant HIV infection in rural Tanzania. Int. J. STD AIDS 2011, 22, 719–721. [Google Scholar] [CrossRef] [PubMed]
  129. Connell, J.A.; Parry, J.V.; Mortimer, P.P.; Duncan, J. Novel assay for the detection of immunoglobulin G antihuman immunodeficiency virus in untreated saliva and urine. J. Med. Virol. 1993, 41, 159–164. [Google Scholar] [CrossRef] [PubMed]
  130. Gershy-Damet, G.M.; Koffi, K.; Abouya, L.; Sasson-Morokro, M.; Brattegaard, K.; Parry, J.V.; de Cock, K.M.; Mortimer, P.P. Salivary and urinary diagnosis of human immunodeficiency viruses 1 and 2 infection in Cote d’Ivoire, using two assays. Trans. R. Soc. Trop. Med. Hyg. 1992, 86, 670–671. [Google Scholar] [CrossRef]
  131. Gallo, D.; George, J.R.; Fitchen, J.H.; Goldstein, A.S.; Hindahl, M.S. Evaluation of a system using oral mucosal transudate for HIV-1 antibody screening and confirmatory testing. OraSure HIV Clinical Trials Group. JAMA 1997, 277, 254–258. [Google Scholar] [CrossRef] [PubMed]
  132. Respess, R.A.; Rayfield, M.A.; Dondero, T.J. Laboratory testing and rapid HIV assays: Applications for HIV surveillance in hard-to-reach populations. AIDS 2001, 15, S49–S59. [Google Scholar] [CrossRef] [PubMed]
  133. Larson, B.A.; Schnippel, K.; Ndibongo, B.; Xulu, T.; Brennan, A.; Long, L.; Fox, M.P.; Rosen, S. Rapid point-of-care CD4 testing at mobile HIV testing sites to increase linkage to care: An evaluation of a pilot program in South Africa. J. Acquir. Immune Defic. Syndr. 2012, 61, e13–e17. [Google Scholar] [CrossRef] [PubMed]
  134. Kanshana, S.; Simonds, R.J. National program for preventing mother-child HIV transmission in Thailand: Successful implementation and lessons learned. AIDS 2002, 16, 953–959. [Google Scholar] [CrossRef] [PubMed]
  135. US Food and Drug Administation (USFDA). US Food and Drug Administation Center for Biologics Evaluation and Research. Available online: http://www.fda.gov/cber/products/testkits.htm (accessed on 15 February 2017).
  136. Christiansen, C.B.; Jessen, T.E.; Nielsen, C.; Staun-Olsen, P. False negative anti-HIV-1/HIV-2 ELISAs in acute HIV-2 infection. Vox Sang. 1996, 70, 144–147. [Google Scholar] [CrossRef] [PubMed]
  137. Parpia, Z.A.; Elghanian, R.; Nabatiyan, A.; Hardie, D.R.; Kelso, D.M. p24 antigen rapid test for diagnosis of acute pediatric HIV infection. J. Acquir. Immune Defic. Syndr. 2010, 55, 413–419. [Google Scholar] [CrossRef] [PubMed]
  138. Bourlet, T.; Pretis, C.; Pillet, S.; Lesenechal, M.; Piche, J.; Pozzetto, B. Comparative evaluation of the VIDAS HIV DUO Ultra assay for combined detection of HIV-1 antigen and antibodies to HIV. J. Virol. Methods 2005, 127, 165–167. [Google Scholar] [CrossRef] [PubMed]
  139. Kariuki, S.; Revathi, G.; Kiiru, J.; Mengo, D.M.; Mwituria, J.; Muyodi, J.; Munyalo, A.; Teo, Y.Y.; Holt, K.E.; Kingsley, R.A.; et al. Typhoid in Kenya is associated with a dominant multidrug-resistant Salmonella enterica serovar Typhi haplotype that is also widespread in Southeast Asia. J. Clin. Microbiol. 2010, 48, 2171–2176. [Google Scholar] [CrossRef] [PubMed]
  140. Parry, C.M.; Hien, T.T.; Dougan, G.; White, N.J.; Farrar, J.J. Typhoid fever. N. Engl. J. Med. 2002, 347, 1770–1782. [Google Scholar] [CrossRef] [PubMed]
  141. Pratap, C.B.; Patel, S.K.; Shukla, V.K.; Tripathi, S.K.; Singh, T.B.; Nath, G. Drug resistance in Salmonella enterica serotype Typhi isolated from chronic typhoid carriers. Int. J. Antimicrob. Agents 2012, 40, 279–280. [Google Scholar] [CrossRef] [PubMed]
  142. World Health Organization (WHO). Background Document: The Diagnosis, Treatment and Prevention of Typhoid Fever; WHO Documentary: Geneva, Switzerland, 2003; Volume WHO/V and B/03.07. [Google Scholar]
  143. Pu, S.J.; Huang, H.S. Diagnostic value of a single Widal test. Chin. J. Microbiol. Immunol. 1985, 18, 256–263. [Google Scholar]
  144. Brodie, J. Antibodies and the Aberdeen typhoid outbreak of 1964. I. The Widal reaction. J. Hyg. 1977, 79, 161–180. [Google Scholar] [CrossRef] [PubMed]
  145. Jhaveri, K.N.; Nandwani, S.K.; Mehta, P.K.; Surati, R.R.; Parmar, B.D. False positive modified Widal test in acute malaria. J. Assoc. Physicians India 1995, 43, 754–755. [Google Scholar] [PubMed]
  146. Parry, C.M.; Hoa, N.T.; Diep, T.S.; Wain, J.; Chinh, N.T.; Vinh, H.; Hien, T.T.; White, N.J.; Farrar, J.J. Value of a single-tube widal test in diagnosis of typhoid fever in Vietnam. J. Clin. Microbiol. 1999, 37, 2882–2886. [Google Scholar] [PubMed]
  147. Pandya, M.; Pillai, P.; Deb, M. Rapid diagnosis of typhoid fever by detection of Barber protein and Vi antigen of Salmonella serotype typhi. J. Med. Microbiol. 1995, 43, 185–188. [Google Scholar] [CrossRef] [PubMed]
  148. Gopalakrishnan, V.; Sekhar, W.Y.; Soo, E.H.; Vinsent, R.A.; Devi, S. Typhoid fever in Kuala Lumpur and a comparative evaluation of two commercial diagnostic kits for the detection of antibodies to Salmonella typhi. Singapore Med. J. 2002, 43, 354–358. [Google Scholar] [PubMed]
  149. Keddy, K.H.; Sooka, A.; Letsoalo, M.E.; Hoyland, G.; Chaignat, C.L.; Morrissey, A.B.; Crump, J.A. Sensitivity and specificity of typhoid fever rapid antibody tests for laboratory diagnosis at two sub-Saharan African sites. Bull. World Health Organ. 2011, 89, 640–647. [Google Scholar] [CrossRef] [PubMed]
  150. Oracz, G.; Feleszko, W.; Golicka, D.; Maksymiuk, J.; Klonowska, A.; Szajewska, H. Rapid diagnosis of acute Salmonella gastrointestinal infection. Clin. Infect. Dis. 2003, 36, 112–115. [Google Scholar] [CrossRef] [PubMed]
  151. Feleszko, W.; Maksymiuk, J.; Oracz, G.; Golicka, D.; Szajewska, H. The TUBEX typhoid test detects current Salmonella infections. J. Immunol. Methods 2004, 285, 137–138. [Google Scholar] [CrossRef] [PubMed]
  152. Tam, F.C.; Lim, P.L. The TUBEX typhoid test based on particle-inhibition immunoassay detects IgM but not IgG anti-O9 antibodies. J. Immunol. Methods 2003, 282, 83–91. [Google Scholar] [CrossRef] [PubMed]
  153. Choo, K.E.; Oppenheimer, S.J.; Ismail, A.B.; Ong, K.H. Rapid serodiagnosis of typhoid fever by dot enzyme immunoassay in an endemic area. Clin. Infect. Dis. 1994, 19, 172–176. [Google Scholar] [CrossRef] [PubMed]
  154. Ismail, A.; Kader, Z.S.; Kok-Hai, O. Dot enzyme immunosorbent assay for the serodiagnosis of typhoid fever. Southeast Asian J. Trop. Med. Public Health 1991, 22, 563–566. [Google Scholar] [PubMed]
  155. Ismail, A.; Hai, O.K.; Kader, Z.A. Demonstration of an antigenic protein specific for Salmonella typhi. Biophys. Res. Commun. 1991, 181, 301–305. [Google Scholar] [CrossRef]
  156. Jackson, A.A.; Ismail, A.; Ibrahim, T.A.; Kader, Z.S.; Nawi, N.M. Retrospective review of dot enzyme immunoassay test for typhoid fever in an endemic area. Southeast Asian J. Trop. Med. Public Health 1995, 26, 625–630. [Google Scholar] [PubMed]
  157. Choo, K.E.; Davis, T.M.; Ismail, A.; Ong, K.H. Longevity of antibody responses to a Salmonella typhi-specific outer membrane protein: Interpretation of a dot enzyme immunosorbent assay in an area of high typhoid fever endemicity. Am. J. Trop. Med. Hyg. 1997, 57, 656–659. [Google Scholar] [CrossRef] [PubMed]
  158. Choo, K.E.; Davis, T.M.; Ismail, A.; Tuan Ibrahim, T.A.; Ghazali, W.N. Rapid and reliable serological diagnosis of enteric fever: Comparative sensitivity and specificity of Typhidot and Typhidot-M tests in febrile Malaysian children. Acta Trop. 1999, 72, 175–183. [Google Scholar] [CrossRef]
  159. House, D.; Wain, J.; Ho, V.A.; Diep, T.S.; Chinh, N.T.; Bay, P.V.; Vinh, H.; Duc, M.; Parry, C.M.; Dougan, G.; et al. Serology of typhoid fever in an area of endemicity and its relevance to diagnosis. J. Clin. Microbiol. 2001, 39, 1002–1007. [Google Scholar] [CrossRef] [PubMed]
  160. Olsen, S.J.; Pruckler, J.; Bibb, W.; Nguyen, T.M.; Tran, M.T.; Sivapalasingam, S.; Gupta, A.; Phan, T.P.; Nguyen, T.C.; Nguyen, V.C.; et al. Evaluation of rapid diagnostic tests for typhoid fever. J. Clin. Microbiol. 2004, 42, 1885–1889. [Google Scholar] [CrossRef] [PubMed]
  161. World Health Organization (WHO). World Malaria Report 2012; WHO Press: Geneva, Switzerland, 2012; p. 288. [Google Scholar]
  162. Singh, B.; Kim Sung, L.; Matusop, A.; Radhakrishnan, A.; Shamsul, S.S.; Cox-Singh, J.; Thomas, A.; Conway, D.J. A large focus of naturally acquired Plasmodium knowlesi infections in human beings. Lancet 2004, 363, 1017–1024. [Google Scholar] [CrossRef]
  163. Lee, K.S.; Cox-Singh, J.; Brooke, G.; Matusop, A.; Singh, B. Plasmodium knowlesi from archival blood films: Further evidence that human infections are widely distributed and not newly emergent in Malaysian Borneo. Int. J. Parasitol. 2009, 39, 1125–1128. [Google Scholar] [CrossRef] [PubMed]
  164. Sulistyaningsih, E.; Fitri, L.E.; Loscher, T.; Berens-Riha, N. Diagnostic difficulties with Plasmodium knowlesi infection in humans. Emerg. Infect. Dis. 2010, 16, 1033–1034. [Google Scholar] [CrossRef] [PubMed]
  165. Van den Eede, P.; van, H.N.; van Overmeir, C.; Vythilingam, I.; Duc, T.N.; Hung le, X.; Manh, H.N.; Anne, J.; D’Alessandro, U.; Erhart, A. Human Plasmodium knowlesi infections in young children in central Vietnam. Malar. J. 2009, 8, 249. [Google Scholar] [CrossRef] [PubMed]
  166. Snow, R.W.; Guerra, C.A.; Noor, A.M.; Myint, H.Y.; Hay, S.I. The global distribution of clinical episodes of Plasmodium falciparum malaria. Nature 2005, 434, 214–217. [Google Scholar] [CrossRef] [PubMed]
  167. Bell, D.; Wongsrichanalai, C.; Barnwell, J.W. Ensuring quality and access for malaria diagnosis: How can it be achieved? Nat. Rev. Microbiol. 2006, 4, S7–S20. [Google Scholar] [CrossRef] [PubMed]
  168. Murray, C.K.; Bell, D.; Gasser, R.A.; Wongsrichanalai, C. Rapid diagnostic testing for malaria. Trop. Med. Int. Health 2003, 8, 876–883. [Google Scholar] [CrossRef] [PubMed]
  169. Van den Ende, J.; Jacobs, J.; Bisoffi, Z. Utility of point-of-care malaria rapid diagnostic tests. Am. J. Trop. Med. Hyg. 2010, 83, 207. [Google Scholar] [PubMed]
  170. Bisoffi, Z.; Gobbi, F.; Angheben, A.; van den Ende, J. The role of rapid diagnostic tests in managing malaria. PLoS Med. 2009, 6, e1000063. [Google Scholar] [CrossRef] [PubMed]
  171. Howard, R.J.; Uni, S.; Aikawa, M.; Aley, S.B.; Leech, J.H.; Lew, A.M.; Wellems, T.E.; Rener, J.; Taylor, D.W. Secretion of a malarial histidine-rich protein (Pf HRP II) from Plasmodium falciparum-infected erythrocytes. J. Cell Biol. 1986, 103, 1269–1277. [Google Scholar] [CrossRef] [PubMed]
  172. Rock, E.P.; Marsh, K.; Saul, A.J.; Wellems, T.E.; Taylor, D.W.; Maloy, W.L.; Howard, R.J. Comparative analysis of the Plasmodium falciparum histidine-rich proteins HRP-I, HRP-II and HRP-III in malaria parasites of diverse origin. Parasitology 1987, 95 Pt 2, 209–227. [Google Scholar] [CrossRef] [PubMed]
  173. Parra, M.E.; Evans, C.B.; Taylor, D.W. Identification of Plasmodium falciparum histidine-rich protein 2 in the plasma of humans with malaria. J. Clin. Microbiol. 1991, 29, 1629–1634. [Google Scholar] [PubMed]
  174. Aslan, G.; Ulukanligil, M.; Seyrek, A.; Erel, O. Diagnostic performance characteristics of rapid dipstick test for Plasmodium vivax malaria. Mem. Inst. Oswaldo Cruz 2001, 96, 683–686. [Google Scholar] [CrossRef] [PubMed]
  175. Moody, A. Rapid diagnostic tests for malaria parasites. Clin. Microbiol. Rev. 2002, 15, 66–78. [Google Scholar] [CrossRef] [PubMed]
  176. Meier, B.; Dobeli, H.; Certa, U. Stage-specific expression of aldolase isoenzymes in the rodent malaria parasite Plasmodium berghei. Mol. Biochem. Parasitol. 1992, 52, 15–27. [Google Scholar] [CrossRef]
  177. Cloonan, N.; Fischer, K.; Cheng, Q.; Saul, A. Aldolase genes of Plasmodium species. Mol. Biochem. Parasitol. 2001, 113, 327–330. [Google Scholar] [CrossRef]
  178. Aguilar, R.; Machevo, S.; Menendez, C.; Bardaji, A.; Nhabomba, A.; Alonso, P.L.; Mayor, A. Comparison of placental blood microscopy and the ICT HRP2 rapid diagnostic test to detect placental malaria. Trans. R. Soc. Trop. Med. Hyg. 2012, 106, 573–575. [Google Scholar] [CrossRef] [PubMed]
  179. Forney, J.R.; Magill, A.J.; Wongsrichanalai, C.; Sirichaisinthop, J.; Bautista, C.T.; Heppner, D.G.; Miller, R.S.; Ockenhouse, C.F.; Gubanov, A.; Shafer, R.; et al. Malaria rapid diagnostic devices: Performance characteristics of the ParaSight F device determined in a multisite field study. J. Clin. Microbiol. 2001, 39, 2884–2890. [Google Scholar] [CrossRef] [PubMed]
  180. Mueller, I.; Betuela, I.; Ginny, M.; Reeder, J.C.; Genton, B. The sensitivity of the OptiMAL rapid diagnostic test to the presence of Plasmodium falciparum gametocytes compromises its ability to monitor treatment outcomes in an area of Papua New Guinea in which malaria is endemic. J. Clin. Microbiol. 2007, 45, 627–630. [Google Scholar] [CrossRef] [PubMed]
  181. Soto Tarazona, A.; Solari Zerpa, L.; Mendoza Requena, D.; Llanos-Cuentas, A.; Magill, A. Evaluation of the rapid diagnostic test OptiMAL for diagnosis of malaria due to Plasmodium vivax. Braz. J. Infect. Dis. 2004, 8, 151–155. [Google Scholar] [CrossRef] [PubMed]
  182. Playford, E.G.; Walker, J. Evaluation of the ICT malaria P.f/P.v and the OptiMal rapid diagnostic tests for malaria in febrile returned travellers. J. Clin. Microbiol. 2002, 40, 4166–4171. [Google Scholar] [CrossRef] [PubMed]
  183. Piper, R.; Lebras, J.; Wentworth, L.; Hunt-Cooke, A.; Houze, S.; Chiodini, P.; Makler, M. Immunocapture diagnostic assays for malaria using Plasmodium lactate dehydrogenase (pLDH). Am. J. Trop. Med. Hyg. 1999, 60, 109–118. [Google Scholar] [CrossRef] [PubMed]
  184. Tjitra, E.; Suprianto, S.; Dyer, M.; Currie, B.J.; Anstey, N.M. Field evaluation of the ICT malaria P.f/P.v immunochromatographic test for detection of Plasmodium falciparum and Plasmodium vivax in patients with a presumptive clinical diagnosis of malaria in eastern Indonesia. J. Clin. Microbiol. 1999, 37, 2412–2417. [Google Scholar] [PubMed]
  185. World Health Organization (WHO). List of Known Commercially Available Antigen-Detecting Malaria RDTs. Available online: http://www.wpro.who.int/malaria/sites/rdt/ (accessed on 12 April 2017).
  186. Conroy, P.J.; Hearty, S.; Leonard, P.; O’Kennedy, R.J. Antibody production, design and use for biosensor-based applications. Semin. Cell Dev. Biol. 2009, 20, 10–26. [Google Scholar] [CrossRef] [PubMed]
  187. Newcombe, C.; Newcombe, A.R. Antibody production: Polyclonal-derived biotherapeutics. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2007, 848, 2–7. [Google Scholar] [CrossRef] [PubMed]
  188. Milstein, C. 12th Sir Hans Krebs Lecture. From antibody diversity to monoclonal antibodies. Eur. J. Biochem. 1981, 118, 429–436. [Google Scholar] [CrossRef] [PubMed]
  189. 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] [PubMed]
  190. Kohler, G.; Milstein, C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975, 256, 495–497. [Google Scholar] [CrossRef] [PubMed]
  191. Lipman, N.S.; Jackson, L.R.; Trudel, L.J.; Weis-Garcia, F. Monoclonal versus polyclonal antibodies: Distinguishing characteristics, applications, and information resources. ILAR J. 2005, 46, 258–268. [Google Scholar] [CrossRef] [PubMed]
  192. Yokoyama, W.M. Production of monoclonal antibody supernatant and ascites fluid. Curr. Protoc. Mol. Biol. 2008. [Google Scholar] [CrossRef]
  193. Jackson, L.R.; Trudel, L.J.; Fox, J.G.; Lipman, N.S. Monoclonal antibody production in murine ascites. I. Clinical and pathologic features. Lab. Anim. Sci. 1999, 49, 70–80. [Google Scholar] [PubMed]
  194. Hendriksen, C.F.; de Leeuw, W. Production of monoclonal antibodies by the ascites method in laboratory animals. Res. Immunol. 1998, 149, 535–542. [Google Scholar] [CrossRef]
  195. Li, F.; Vijayasankaran, N.; Shen, A.Y.; Kiss, R.; Amanullah, A. Cell culture processes for monoclonal antibody production. MAbs 2010, 2, 466–479. [Google Scholar] [CrossRef] [PubMed]
  196. Dewar, V.; Voet, P.; Denamur, F.; Smal, J. Industrial implementation of in vitro production of monoclonal antibodies. ILAR J. 2005, 46, 307–313. [Google Scholar] [CrossRef] [PubMed]
  197. Janeway, C.A.; Travers, J.P.; WAlport, M.; Schlomchik, M. Immunology, 5th ed.; Garland Science: New York, NY, USA, 2001. [Google Scholar]
  198. Wesolowski, J.; Alzogaray, V.; Reyelt, J.; Unger, M.; Juarez, K.; Urrutia, M.; Cauerhff, A.; Danquah, W.; Rissiek, B.; Scheuplein, F.; et al. Single domain antibodies: Promising experimental and therapeutic tools in infection and immunity. Med. Microbiol. Immunol. 2009, 198, 157–174. [Google Scholar] [CrossRef] [PubMed]
  199. Braden, B.C.; Souchon, H.; Eisele, J.L.; Bentley, G.A.; Bhat, T.N.; Navaza, J.; Poljak, R.J. Three-dimensional structures of the free and the antigen-complexed Fab from monoclonal anti-lysozyme antibody D44.1. J. Mol. Biol. 1994, 243, 767–781. [Google Scholar] [CrossRef]
  200. Murray, C.K.; Gasser, R.A., Jr.; Magill, A.J.; Miller, R.S. Update on rapid diagnostic testing for malaria. Clin. Microbiol. Rev. 2008, 21, 97–110. [Google Scholar] [CrossRef] [PubMed]
  201. Guire, P.E. Stability issues for protein-based in vitro diagnostic products. IVD Technol. 1999, 5, 50–54. [Google Scholar]
  202. Chiodini, P.L.; Bowers, K.; Jorgensen, P.; Barnwell, J.W.; Grady, K.K.; Luchavez, J.; Moody, A.H.; Cenizal, A.; Bell, D. The heat stability of Plasmodium lactate dehydrogenase-based and histidine-rich protein 2-based malaria rapid diagnostic tests. Trans. R. Soc. Trop. Med. Hyg. 2007, 101, 331–337. [Google Scholar] [CrossRef] [PubMed]
  203. Jorgensen, P.; Chanthap, L.; Rebueno, A.; Tsuyuoka, R.; Bell, D. Malaria rapid diagnostic tests in tropical climates: The need for a cool chain. Am. J. Trop. Med. Hyg. 2006, 74, 750–754. [Google Scholar] [PubMed]
  204. Ward, E.S.; Gussow, D.; Griffiths, A.D.; Jones, P.T.; Winter, G. Binding activities of a repertoire of single immunoglobulin variable domains secreted from E. coli. Nature 1989, 341, 544–546. [Google Scholar] [CrossRef] [PubMed]
  205. Barthelemy, P.A.; Raab, H.; Appleton, B.A.; Bond, C.J.; Wu, P.; Wiesmann, C.; Sidhu, S.S. Comprehensive analysis of the factors contributing to the stability and solubility of autonomous human VH domains. J. Biol. Chem. 2008, 283, 3639–3654. [Google Scholar] [CrossRef] [PubMed]
  206. Huang, L.; Reekmans, G.; Saerens, D.; Friedt, J.M.; Frederix, F.; Francis, L.; Muyldermans, S.; Campitelli, A.; van Hoof, C. Prostate-specific antigen immunosensing based on mixed self-assembled monolayers, camel antibodies and colloidal gold enhanced sandwich assays. Biosens. Bioelectron. 2005, 21, 483–490. [Google Scholar] [CrossRef] [PubMed]
  207. Hudson, P.J.; Souriau, C. Engineered antibodies. Nat. Med. 2003, 9, 129–134. [Google Scholar] [CrossRef] [PubMed]
  208. Jostock, T.; Vanhove, M.; Brepoels, E.; van Gool, R.; Daukandt, M.; Wehnert, A.; Van Hegelsom, R.; Dransfield, D.; Sexton, D.; Devlin, M.; et al. Rapid generation of functional human IgG antibodies derived from Fab-on-phage display libraries. J. Immunol. Methods 2004, 289, 65–80. [Google Scholar] [CrossRef] [PubMed]
  209. 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]
  210. Hoogenboom, H.R. Overview of antibody phage-display technology and its applications. Methods Mol. Biol. 2002, 178, 1–37. [Google Scholar] [PubMed]
  211. Hoogenboom, H.R.; de Bruine, A.P.; Hufton, S.E.; Hoet, R.M.; Arends, J.W.; Roovers, R.C. Antibody phage display technology and its applications. Immunotechnology 1998, 4, 1–20. [Google Scholar] [CrossRef]
  212. Iason, G. The role of plant secondary metabolites in mammalian herbivory: Ecological perspectives. Proc. Nutr. Soc. 2005, 64, 123–131. [Google Scholar] [CrossRef] [PubMed]
  213. Griffiths, A.D.; Malmqvist, M.; Marks, J.D.; Bye, J.M.; Embleton, M.J.; McCafferty, J.; Baier, M.; Holliger, K.P.; Gorick, B.D.; Hughes-Jones, N.C.; et al. Human anti-self antibodies with high specificity from phage display libraries. EMBO J. 1993, 12, 725–734. [Google Scholar] [PubMed]
  214. Winter, G.; Griffiths, A.D.; Hawkins, R.E.; Hoogenboom, H.R. Making antibodies by phage display technology. Annu. Rev. Immunol. 1994, 12, 433–455. [Google Scholar] [CrossRef] [PubMed]
  215. Hill, J.E.; Hemmingsen, S.M.; Goldade, B.G.; Dumonceaux, T.J.; Klassen, J.; Zijlstra, R.T.; Goh, S.H.; van Kessel, A.G. Comparison of ileum microflora of pigs fed corn-, wheat-, or barley-based diets by chaperonin-60 sequencing and quantitative PCR. Appl. Environ. Microbiol. 2005, 71, 867–875. [Google Scholar] [CrossRef] [PubMed]
  216. Neri, D.; Petrul, H.; Roncucci, G. Engineering recombinant antibodies for immunotherapy. Cell Biophys. 1995, 27, 47–61. [Google Scholar] [CrossRef] [PubMed]
  217. Xia, M.S.; Hu, C.H.; Xu, Z.R. Effects of copper-bearing montmorillonite on growth performance, digestive enzyme activities, and intestinal microflora and morphology of male broilers. Poult. Sci. 2004, 83, 1868–1875. [Google Scholar] [CrossRef] [PubMed]
  218. Paul, S.S.; Kamra, D.N.; Sastry, V.R.; Sahu, N.P.; Agarwal, N. Effect of anaerobic fungi on in vitro feed digestion by mixed rumen microflora of buffalo. Reprod. Nutr. Dev. 2004, 44, 313–319. [Google Scholar] [CrossRef] [PubMed]
  219. Hume, M.E.; Poole, T.L.; Pultz, N.J.; Hanrahan, J.A.; Donskey, C.J. Inhibition of vancomycin-resistant enterococcus by continuous-flow cultures of human stool microflora with and without anaerobic gas supplementation. Curr. Microbiol. 2004, 48, 364–367. [Google Scholar] [CrossRef] [PubMed]
  220. Franklin, M.A.; Mathew, A.G.; Vickers, J.R.; Clift, R.A. Characterization of microbial populations and volatile fatty acid concentrations in the jejunum, ileum, and cecum of pigs weaned at 17 vs. 24 days of age. J. Anim. Sci. 2002, 80, 2904–2910. [Google Scholar] [CrossRef] [PubMed]
  221. Engberg, R.M.; Hedemann, M.S.; Jensen, B.B. The influence of grinding and pelleting of feed on the microbial composition and activity in the digestive tract of broiler chickens. Br. Poult. Sci. 2002, 43, 569–579. [Google Scholar] [CrossRef] [PubMed]
  222. Abbady, A.Q.; Al-Mariri, A.; Zarkawi, M.; Al-Assad, A.; Muyldermans, S. Evaluation of a nanobody phage display library constructed from a Brucella-immunised camel. Vet. Immunol. Immunopathol. 2011, 142, 49–56. [Google Scholar] [CrossRef] [PubMed]
  223. Dolk, E.; van der Vaart, M.; Lutje Hulsik, D.; Vriend, G.; de Haard, H.; Spinelli, S.; Cambillau, C.; Frenken, L.; Verrips, T. Isolation of llama antibody fragments for prevention of dandruff by phage display in shampoo. Appl. Environ. Microbiol. 2005, 71, 442–450. [Google Scholar] [CrossRef] [PubMed]
  224. Dooley, H.; Flajnik, M.F.; Porter, A.J. Selection and characterization of naturally occurring single-domain (IgNAR) antibody fragments from immunized sharks by phage display. Mol. Immunol. 2003, 40, 25–33. [Google Scholar] [CrossRef]
  225. Xu, G.; Tasumi, S.; Pancer, Z. Yeast surface display of lamprey variable lymphocyte receptors. Methods Mol. Biol. 2011, 748, 21–33. [Google Scholar] [CrossRef] [PubMed]
  226. Hussack, G.; Arbabi-Ghahroudi, M.; Mackenzie, C.R.; Tanha, J. Isolation and characterization of Clostridium difficile toxin-specific single-domain antibodies. Methods Mol. Biol. 2012, 911, 211–239. [Google Scholar] [CrossRef] [PubMed]
  227. Liu, Y.; Regula, L.K.; Stewart, A.; Lai, J.R. Synthetic Fab fragments that bind the HIV-1 gp41 heptad repeat regions. Biochem. Biophys. Res. Commun. 2011, 413, 611–615. [Google Scholar] [CrossRef] [PubMed]
  228. Shui, X.; Huang, J.; Li, Y.H.; Xie, P.L.; Li, G.C. Construction and selection of human Fab antibody phage display library of liver cancer. Hybridoma 2009, 28, 341–347. [Google Scholar] [CrossRef] [PubMed]
  229. Sowa, K.M.; Cavanagh, D.R.; Creasey, A.M.; Raats, J.; McBride, J.; Sauerwein, R.; Roeffen, W.F.; Arnot, D.E. Isolation of a monoclonal antibody from a malaria patient-derived phage display library recognising the Block 2 region of Plasmodium falciparum merozoite surface protein-1. Mol. Biochem. Parasitol. 2001, 112, 143–147. [Google Scholar] [CrossRef]
  230. Yang, G.H.; Yoon, S.O.; Jang, M.H.; Hong, H.J. Affinity maturation of an anti-hepatitis B virus PreS1 humanized antibody by phage display. J. Microbiol. 2007, 45, 528–533. [Google Scholar] [PubMed]
  231. Carmen, S.; Jermutus, L. Concepts in antibody phage display. Brief Funct. Genomic Proteomic 2002, 1, 189–203. [Google Scholar] [CrossRef] [PubMed]
  232. Brichta, J.; Hnilova, M.; Viskovic, T. Generation of hapten-specific recombinant antibodies: Antibody phage display technology: A review. Vet. Med. 2005, 50, 231–252. [Google Scholar]
  233. Flajnik, M.F. Comparative analyses of immunoglobulin genes: Surprises and portents. Nat. Rev. Immunol. 2002, 2, 688–698. [Google Scholar] [CrossRef] [PubMed]
  234. Hamers-Casterman, C.; Atarhouch, T.; Muyldermans, S.; Robinson, G.; Hamers, C.; Songa, E.B.; Bendahman, N.; Hamers, R. Naturally occurring antibodies devoid of light chains. Nature 1993, 363, 446–448. [Google Scholar] [CrossRef] [PubMed]
  235. Goldman, E.R.; Anderson, G.P.; Conway, J.; Sherwood, L.J.; Fech, M.; Vo, B.; Liu, J.L.; Hayhurst, A. Thermostable llama single domain antibodies for detection of botulinum A neurotoxin complex. Anal. Chem. 2008, 80, 8583–8591. [Google Scholar] [CrossRef] [PubMed]
  236. Perez, J.M.; Renisio, J.G.; Prompers, J.J.; van Platerink, C.J.; Cambillau, C.; Darbon, H.; Frenken, L.G. Thermal unfolding of a llama antibody fragment: A two-state reversible process. Biochemistry 2001, 40, 74–83. [Google Scholar] [CrossRef] [PubMed]
  237. Dumoulin, M.; Conrath, K.; van Meirhaeghe, A.; Meersman, F.; Heremans, K.; Frenken, L.G.; Muyldermans, S.; Wyns, L.; Matagne, A. Single-domain antibody fragments with high conformational stability. Protein Sci. 2002, 11, 500–515. [Google Scholar] [CrossRef] [PubMed]
  238. Harmsen, M.M.; de Haard, H.J. Properties, production, and applications of camelid single-domain antibody fragments. Appl. Microbiol. Biotechnol. 2007, 77, 13–22. [Google Scholar] [CrossRef] [PubMed]
  239. Muyldermans, S.; Atarhouch, T.; Saldanha, J.; Barbosa, J.A.; Hamers, R. Sequence and structure of VH domain from naturally occurring camel heavy chain immunoglobulins lacking light chains. Protein Eng. 1994, 7, 1129–1135. [Google Scholar] [CrossRef] [PubMed]
  240. Muyldermans, S. Single domain camel antibodies: Current status. J. Biotechnol. 2001, 74, 277–302. [Google Scholar] [CrossRef]
  241. Dumoulin, M.; Last, A.M.; Desmyter, A.; Decanniere, K.; Canet, D.; Larsson, G.; Spencer, A.; Archer, D.B.; Sasse, J.; Muyldermans, S.; et al. A camelid antibody fragment inhibits the formation of amyloid fibrils by human lysozyme. Nature 2003, 424, 783–788. [Google Scholar] [CrossRef] [PubMed]
  242. Vance, D.J.; Tremblay, J.M.; Mantis, N.J.; Shoemaker, C.B. Stepwise engineering of heterodimeric single domain camelid VHH antibodies that passively protect mice from ricin toxin. J. Biol. Chem. 2013, 288, 36538–36547. [Google Scholar] [CrossRef] [PubMed]
  243. De Genst, E.; Silence, K.; Decanniere, K.; Conrath, K.; Loris, R.; Kinne, J.; Muyldermans, S.; Wyns, L. Molecular basis for the preferential cleft recognition by dromedary heavy-chain antibodies. Proc. Natl. Acad. Sci. USA 2006, 103, 4586–4591. [Google Scholar] [CrossRef] [PubMed]
  244. Desmyter, A.; Transue, T.R.; Ghahroudi, M.A.; Thi, M.H.; Poortmans, F.; Hamers, R.; Muyldermans, S.; Wyns, L. Crystal structure of a camel single-domain VH antibody fragment in complex with lysozyme. Nat. Struct. Biol. 1996, 3, 803–811. [Google Scholar] [CrossRef] [PubMed]
  245. Vincke, C.; Loris, R.; Saerens, D.; Martinez-Rodriguez, S.; Muyldermans, S.; Conrath, K. General strategy to humanize a camelid single-domain antibody and identification of a universal humanized nanobody scaffold. J. Biol. Chem. 2009, 284, 3273–3284. [Google Scholar] [CrossRef] [PubMed]
  246. Line, B.R.; Breyer, R.J.; McElvany, K.D.; Earle, D.C.; Khazaeli, M.B. Evaluation of human anti-mouse antibody response in normal volunteers following repeated injections of fanolesomab (NeutroSpec), a murine anti-CD15 IgM monoclonal antibody for imaging infection. Nucl. Med. Commun. 2004, 25, 807–811. [Google Scholar] [CrossRef] [PubMed]
  247. Dmitriev, O.Y.; Lutsenko, S.; Muyldermans, S. Nanobodies as Probes for Protein Dynamics in Vitro and in Cells. J. Biol. Chem. 2016, 291, 3767–3775. [Google Scholar] [CrossRef] [PubMed]
  248. Van Brussel, A.S.; Adams, A.; Oliveira, S.; Dorresteijn, B.; El Khattabi, M.; Vermeulen, J.F.; van der Wall, E.; Mali, W.P.; Derksen, P.W.; van Diest, P.J.; et al. Hypoxia-Targeting Fluorescent Nanobodies for Optical Molecular Imaging of Pre-Invasive Breast Cancer. Mol. Imaging Biol. 2016, 18, 535–544. [Google Scholar] [CrossRef] [PubMed]
  249. Deckers, N.; Saerens, D.; Kanobana, K.; Conrath, K.; Victor, B.; Wernery, U.; Vercruysse, J.; Muyldermans, S.; Dorny, P. Nanobodies, a promising tool for species-specific diagnosis of Taenia solium cysticercosis. Int. J. Parasitol. 2009, 39, 625–633. [Google Scholar] [CrossRef] [PubMed]
  250. Roovers, R.C.; Laeremans, T.; Huang, L.; De Taeye, S.; Verkleij, A.J.; Revets, H.; de Haard, H.J.; van Bergen en Henegouwen, P.M. Efficient inhibition of EGFR signaling and of tumour growth by antagonistic anti-EFGR Nanobodies. Cancer Immunol. Immunother. 2007, 56, 303–317. [Google Scholar] [CrossRef] [PubMed]
  251. Franco, E.J.; Sonneson, G.J.; DeLegge, T.J.; Hofstetter, H.; Horn, J.R.; Hofstetter, O. Production and characterization of a genetically engineered anti-caffeine camelid antibody and its use in immunoaffinity chromatography. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2010, 878, 177–186. [Google Scholar] [CrossRef] [PubMed]
  252. Ladenson, R.C.; Crimmins, D.L.; Landt, Y.; Ladenson, J.H. Isolation and characterization of a thermally stable recombinant anti-caffeine heavy-chain antibody fragment. Anal. Chem. 2006, 78, 4501–4508. [Google Scholar] [CrossRef] [PubMed]
  253. Diaz, M.; Greenberg, A.S.; Flajnik, M.F. Somatic hypermutation of the new antigen receptor gene (NAR) in the nurse shark does not generate the repertoire: Possible role in antigen-driven reactions in the absence of germinal centers. Proc. Natl. Acad. Sci. USA 1998, 95, 14343–14348. [Google Scholar] [CrossRef] [PubMed]
  254. Greenberg, A.S.; Avila, D.; Hughes, M.; Hughes, A.; McKinney, E.C.; Flajnik, M.F. A new antigen receptor gene family that undergoes rearrangement and extensive somatic diversification in sharks. Nature 1995, 374, 168–173. [Google Scholar] [CrossRef] [PubMed]
  255. Roux, K.H.; Greenberg, A.S.; Greene, L.; Strelets, L.; Avila, D.; McKinney, E.C.; Flajnik, M.F. Structural analysis of the nurse shark (new) antigen receptor (NAR): Molecular convergence of NAR and unusual mammalian immunoglobulins. Proc. Natl. Acad. Sci. USA 1998, 95, 11804–11809. [Google Scholar] [CrossRef] [PubMed]
  256. Dooley, H.; Stanfield, R.L.; Brady, R.A.; Flajnik, M.F. First molecular and biochemical analysis of in vivo affinity maturation in an ectothermic vertebrate. Proc. Natl. Acad. Sci. USA 2006, 103, 1846–1851. [Google Scholar] [CrossRef] [PubMed]
  257. Nuttall, S.D.; Krishnan, U.V.; Hattarki, M.; De Gori, R.; Irving, R.A.; Hudson, P.J. Isolation of the new antigen receptor from wobbegong sharks, and use as a scaffold for the display of protein loop libraries. Mol. Immunol. 2001, 38, 313–326. [Google Scholar] [CrossRef]
  258. Liu, J.L.; Anderson, G.P.; Delehanty, J.B.; Baumann, R.; Hayhurst, A.; Goldman, E.R. Selection of cholera toxin specific IgNAR single-domain antibodies from a naive shark library. Mol. Immunol. 2007, 44, 1775–1783. [Google Scholar] [CrossRef] [PubMed]
  259. Ohtani, M.; Hikima, J.; Jung, T.S.; Kondo, H.; Hirono, I.; Aoki, T. Construction of an artificially randomized IgNAR phage display library: Screening of variable regions that bind to hen egg white lysozyme. Mar. Biotechnol. 2013, 15, 56–62. [Google Scholar] [CrossRef] [PubMed]
  260. Camacho-Villegas, T.; Mata-Gonzalez, T.; Paniagua-Solis, J.; Sanchez, E.; Licea, A. Human TNF cytokine neutralization with a vNAR from Heterodontus francisci shark: A potential therapeutic use. MAbs 2013, 5, 80–85. [Google Scholar] [CrossRef] [PubMed]
  261. Streltsov, V.A.; Varghese, J.N.; Carmichael, J.A.; Irving, R.A.; Hudson, P.J.; Nuttall, S.D. Structural evidence for evolution of shark Ig new antigen receptor variable domain antibodies from a cell-surface receptor. Proc. Natl. Acad. Sci. USA 2004, 101, 12444–12449. [Google Scholar] [CrossRef] [PubMed]
  262. 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] [PubMed]
  263. Barelle, C.; Gill, D.S.; Charlton, K. Shark novel antigen receptors--the next generation of biologic therapeutics? Adv. Exp. Med. Biol. 2009, 655, 49–62. [Google Scholar] [CrossRef] [PubMed]
  264. Stanfield, R.L.; Dooley, H.; Flajnik, M.F.; Wilson, I.A. Crystal structure of a shark single-domain antibody V region in complex with lysozyme. Science 2004, 305, 1770–1773. [Google Scholar] [CrossRef] [PubMed]
  265. Muyldermans, S.; Cambillau, C.; Wyns, L. Recognition of antigens by single-domain antibody fragments: The superfluous luxury of paired domains. Trends Biochem. Sci. 2001, 26, 230–235. [Google Scholar] [CrossRef]
  266. Streltsov, V.A.; Carmichael, J.A.; Nuttall, S.D. Structure of a shark IgNAR antibody variable domain and modeling of an early-developmental isotype. Protein Sci. 2005, 14, 2901–2909. [Google Scholar] [CrossRef] [PubMed]
  267. Griffiths, K.; Dolezal, O.; Parisi, K.; Angerosa, J.; Dogovski, C.; Barraclough, M.; Sanalla, A.; Casey, J.; González, I.; Perugini, M.; et al. Shark Variable New Antigen Receptor (VNAR) Single Domain Antibody Fragments: Stability and Diagnostic Applications. Antibodies 2013, 2, 66–81. [Google Scholar] [CrossRef]
  268. Flajnik, M.F.; Deschacht, N.; Muyldermans, S. A case of convergence: Why did a simple alternative to canonical antibodies arise in sharks and camels? PLoS Biol. 2011, 9, e1001120. [Google Scholar] [CrossRef] [PubMed]
  269. Pays, E.; Vanhamme, L.; Perez-Morga, D. Antigenic variation in Trypanosoma brucei: Facts, challenges and mysteries. Curr. Opin. Microbiol. 2004, 7, 369–374. [Google Scholar] [CrossRef] [PubMed]
  270. Saerens, D.; Stijlemans, B.; Baral, T.N.; Nguyen Thi, G.T.; Wernery, U.; Magez, S.; de Baetselier, P.; Muyldermans, S.; Conrath, K. Parallel selection of multiple anti-infectome Nanobodies without access to purified antigens. J. Immunol. Methods 2008, 329, 138–150. [Google Scholar] [CrossRef] [PubMed]
  271. Hernandez, M.; Beltran, C.; Garcia, E.; Fragoso, G.; Gevorkian, G.; Fleury, A.; Parkhouse, M.; Harrison, L.; Sotelo, J.; Sciutto, E. Cysticercosis: Towards the design of a diagnostic kit based on synthetic peptides. Immunol. Lett. 2000, 71, 13–17. [Google Scholar] [CrossRef]
  272. Dorny, P.; Brandt, J.; Zoli, A.; Geerts, S. Immunodiagnostic tools for human and porcine cysticercosis. Acta Trop. 2003, 87, 79–86. [Google Scholar] [CrossRef]
  273. Garcia, H.H.; Harrison, L.J.; Parkhouse, R.M.; Montenegro, T.; Martinez, S.M.; Tsang, V.C.; Gilman, R.H. A specific antigen-detection ELISA for the diagnosis of human neurocysticercosis. The Cysticercosis Working Group in Peru. Trans. R. Soc. Trop. Med. Hyg. 1998, 92, 411–414. [Google Scholar] [CrossRef]
  274. Anderson, G.P.; Goldman, E.R. TNT detection using llama antibodies and a two-step competitive fluid array immunoassay. J. Immunol. Methods 2008, 339, 47–54. [Google Scholar] [CrossRef] [PubMed]
  275. Goldman, E.R.; Anderson, G.P.; Liu, J.L.; Delehanty, J.B.; Sherwood, L.J.; Osborn, L.E.; Cummins, L.B.; Hayhurst, A. Facile generation of heat-stable antiviral and antitoxin single domain antibodies from a semisynthetic llama library. Anal. Chem. 2006, 78, 8245–8255. [Google Scholar] [CrossRef] [PubMed]
  276. Hmila, I.; Abdallah, R.B.; Saerens, D.; Benlasfar, Z.; Conrath, K.; Ayeb, M.E.; Muyldermans, S.; Bouhaouala-Zahar, B. VHH, bivalent domains and chimeric Heavy chain-only antibodies with high neutralizing efficacy for scorpion toxin AahI’. Mol. Immunol. 2008, 45, 3847–3856. [Google Scholar] [CrossRef] [PubMed]
  277. Strokappe, N.; Szynol, A.; Aasa-Chapman, M.; Gorlani, A.; Forsman Quigley, A.; Hulsik, D.L.; Chen, L.; Weiss, R.; de Haard, H.; Verrips, T. Llama antibody fragments recognizing various epitopes of the CD4bs neutralize a broad range of HIV-1 subtypes A, B and C. PLoS ONE 2012, 7, e33298. [Google Scholar] [CrossRef] [PubMed]
  278. Vanlandschoot, P.; Stortelers, C.; Beirnaert, E.; Ibanez, L.I.; Schepens, B.; Depla, E.; Saelens, X. Nanobodies(R): New ammunition to battle viruses. Antivir. Res. 2011, 92, 389–407. [Google Scholar] [CrossRef] [PubMed]
  279. Pant, N.; Marcotte, H.; Hermans, P.; Bezemer, S.; Frenken, L.; Johansen, K.; Hammarstrom, L. Lactobacilli producing bispecific llama-derived anti-rotavirus proteins in vivo for rotavirus-induced diarrhea. Future Microbiol. 2011, 6, 583–593. [Google Scholar] [CrossRef] [PubMed]
  280. Ryan, S.; Kell, A.J.; van Faassen, H.; Tay, L.L.; Simard, B.; MacKenzie, R.; Gilbert, M.; Tanha, J. Single-domain antibody-nanoparticles: Promising architectures for increased Staphylococcus aureus detection specificity and sensitivity. Bioconjug.Chem. 2009, 20, 1966–1974. [Google Scholar] [CrossRef] [PubMed]
  281. Kenanova, V.; Wu, A.M. Tailoring antibodies for radionuclide delivery. Expert Opin. Drug Deliv. 2006, 3, 53–70. [Google Scholar] [CrossRef] [PubMed]
  282. Huang, L.; Gainkam, L.O.; Caveliers, V.; Vanhove, C.; Keyaerts, M.; De Baetselier, P.; Bossuyt, A.; Revets, H.; Lahoutte, T. SPECT imaging with 99mTc-labeled EGFR-specific nanobody for in vivo monitoring of EGFR expression. Mol. Imaging Biol. 2008, 10, 167–175. [Google Scholar] [CrossRef] [PubMed]
  283. Pleschberger, M.; Saerens, D.; Weigert, S.; Sleytr, U.B.; Muyldermans, S.; Sara, M.; Egelseer, E.M. An S-layer heavy chain camel antibody fusion protein for generation of a nanopatterned sensing layer to detect the prostate-specific antigen by surface plasmon resonance technology. Bioconjug. Chem. 2004, 15, 664–671. [Google Scholar] [CrossRef] [PubMed]
  284. Pruszynski, M.; Koumarianou, E.; Vaidyanathan, G.; Revets, H.; Devoogdt, N.; Lahoutte, T.; Zalutsky, M.R. Targeting breast carcinoma with radioiodinated anti-HER2 Nanobody. Nucl. Med. Biol. 2013, 40, 52–59. [Google Scholar] [CrossRef] [PubMed]
  285. Vaneycken, I.; Devoogdt, N.; van Gassen, N.; Vincke, C.; Xavier, C.; Wernery, U.; Muyldermans, S.; Lahoutte, T.; Caveliers, V. Preclinical screening of anti-HER2 nanobodies for molecular imaging of breast cancer. FASEB J. 2011, 25, 2433–2446. [Google Scholar] [CrossRef] [PubMed]
  286. Keyaerts, M.; Xavier, C.; Heemskerk, J.; Devoogdt, N.; Everaert, H.; Ackaert, C.; Vanhoeij, M.; Duhoux, F.P.; Gevaert, T.; Simon, P.; et al. Phase I Study of 68Ga-HER2-Nanobody for PET/CT Assessment of HER2 Expression in Breast Carcinoma. J. Nucl. Med. 2016, 57, 27–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Anderson, G.P.; Moreira, S.C.; Charles, P.T.; Medintz, I.L.; Goldman, E.R.; Zeinali, M.; Taitt, C.R. TNT detection using multiplexed liquid array displacement immunoassays. Anal. Chem. 2006, 78, 2279–2285. [Google Scholar] [CrossRef] [PubMed]
  288. Tillib, S.V.; Ivanova, T.I.; Lyssuk, E.Y.; Larin, S.S.; Kibardin, A.V.; Korobko, E.V.; Vikhreva, P.N.; Gnuchev, N.V.; Georgiev, G.P.; Korobko, I.V. Nanoantibodies for detection and blocking of bioactivity of human vascular endothelial growth factor A(165). Biochemistry 2012, 77, 659–665. [Google Scholar] [CrossRef] [PubMed]
  289. Minaeian, S.; Rahbarizadeh, F.; Zarkesh-Esfahani, S.H.; Ahmadvand, D.; Broom, O.J. Neutralization of human papillomavirus by specific nanobodies against major capsid protein L1. J. Microbiol. Biotechnol. 2012, 22, 721–728. [Google Scholar] [CrossRef] [PubMed]
  290. Vosjan, M.J.; Vercammen, J.; Kolkman, J.A.; Stigter-van Walsum, M.; Revets, H.; van Dongen, G.A. Nanobodies targeting the hepatocyte growth factor: Potential new drugs for molecular cancer therapy. Mol. Cancer Ther. 2012, 11, 1017–1025. [Google Scholar] [CrossRef] [PubMed]
  291. Smolarek, D.; Hattab, C.; Hassanzadeh-Ghassabeh, G.; Cochet, S.; Gutierrez, C.; de Brevern, A.G.; Udomsangpetch, R.; Picot, J.; Grodecka, M.; Wasniowska, K.; et al. A recombinant dromedary antibody fragment (VHH or nanobody) directed against human Duffy antigen receptor for chemokines. Cell Mol. Life Sci. 2010, 67, 3371–3387. [Google Scholar] [CrossRef] [PubMed]
  292. Thys, B.; Saerens, D.; Schotte, L.; De Bleeser, G.; Muyldermans, S.; Hassanzadeh-Ghassabeh, G.; Rombaut, B. A simple quantitative affinity capturing assay of poliovirus antigens and subviral particles by single-domain antibodies using magnetic beads. J. Virol. Methods 2011, 173, 300–305. [Google Scholar] [CrossRef] [PubMed]
  293. Thys, B.; Schotte, L.; Muyldermans, S.; Wernery, U.; Hassanzadeh-Ghassabeh, G.; Rombaut, B. in vitro antiviral activity of single domain antibody fragments against poliovirus. Antivir. Res. 2010, 87, 257–264. [Google Scholar] [CrossRef] [PubMed]
  294. Ahmadvand, D.; Rasaee, M.J.; Rahbarizadeh, F.; Kontermann, R.E.; Sheikholislami, F. Cell selection and characterization of a novel human endothelial cell specific nanobody. Mol. Immunol. 2009, 46, 1814–1823. [Google Scholar] [CrossRef] [PubMed]
  295. Ahmadvand, D.; Rasaee, M.J.; Rahbarizadeh, F.; Mohammadi, M. Production and characterization of a high-affinity nanobody against human endoglin. Hybridoma 2008, 27, 353–360. [Google Scholar] [CrossRef] [PubMed]
  296. Abbady, A.Q.; Al-Daoude, A.; Al-Mariri, A.; Zarkawi, M.; Muyldermans, S. Chaperonin GroEL a Brucella immunodominant antigen identified using Nanobody and MALDI-TOF-MS technologies. Vet. Immunol. Immunopathol. 2012, 146, 254–263. [Google Scholar] [CrossRef] [PubMed]
  297. Goldman, E.R.; Anderson, G.P.; Bernstein, R.D.; Swain, M.D. Amplification of immunoassays using phage-displayed single domain antibodies. J. Immunol. Methods 2010, 352, 182–185. [Google Scholar] [CrossRef] [PubMed]
  298. Swain, M.D.; Anderson, G.P.; Zabetakis, D.; Bernstein, R.D.; Liu, J.L.; Sherwood, L.J.; Hayhurst, A.; Goldman, E.R. Llama-derived single-domain antibodies for the detection of botulinum A neurotoxin. Anal. Bioanal. Chem. 2010, 398, 339–348. [Google Scholar] [CrossRef] [PubMed]
  299. Leung, K. 99mTc(CO)3-Anti-carcinoembryonic antigen (CEA) humanized CEA5 graft nanobody. In Molecular Imaging and Contrast Agent Database (MICAD); Bethesda: Rockyville, MD, USA, 2004. [Google Scholar]
  300. Sukhanova, A.; Even-Desrumeaux, K.; Kisserli, A.; Tabary, T.; Reveil, B.; Millot, J.M.; Chames, P.; Baty, D.; Artemyev, M.; Oleinikov, V.; et al. Oriented conjugates of single-domain antibodies and quantum dots: Toward a new generation of ultrasmall diagnostic nanoprobes. Nanomedicine 2012, 8, 516–525. [Google Scholar] [CrossRef] [PubMed]
  301. Vaneycken, I.; Govaert, J.; Vincke, C.; Caveliers, V.; Lahoutte, T.; de Baetselier, P.; Raes, G.; Bossuyt, A.; Muyldermans, S.; Devoogdt, N. in vitro analysis and in vivo tumor targeting of a humanized, grafted nanobody in mice using pinhole SPECT/micro-CT. J. Nucl. Med. 2010, 51, 1099–1106. [Google Scholar] [CrossRef] [PubMed]
  302. Leung, K. 99mTc(CO)3-Anti-vascular cell adhesion molecule-1 nanobody cAbVCAM1-5. In Molecular Imaging and Contrast Agent Database (MICAD); Bethesda: Rockyville, MD, USA, 2004. [Google Scholar]
  303. Leung, K. Microbubbles conjugated with anti-vascular cell adhesion molecule-1 nanobody cAbVCAM1-5. In Molecular Imaging and Contrast Agent Database (MICAD); Bethesda: Rockyville, MD, USA, 2004. [Google Scholar]
  304. Broisat, A.; Hernot, S.; Toczek, J.; De Vos, J.; Riou, L.M.; Martin, S.; Ahmadi, M.; Thielens, N.; Wernery, U.; Caveliers, V.; et al. Nanobodies targeting mouse/human VCAM1 for the nuclear imaging of atherosclerotic lesions. Circ. Res. 2012, 110, 927–937. [Google Scholar] [CrossRef] [PubMed]
  305. Schmitz, K.R.; Bagchi, A.; Roovers, R.C.; van Bergen en Henegouwen, P.M.; Ferguson, K.M. Structural evaluation of EGFR inhibition mechanisms for nanobodies/VHH domains. Structure 2013, 21, 1214–1224. [Google Scholar] [CrossRef] [PubMed]
  306. Roovers, R.C.; Vosjan, M.J.; Laeremans, T.; el Khoulati, R.; de Bruin, R.C.; Ferguson, K.M.; Verkleij, A.J.; van Dongen, G.A.; van Bergen en Henegouwen, P.M. A biparatopic anti-EGFR nanobody efficiently inhibits solid tumour growth. Int. J. Cancer 2011, 129, 2013–2024. [Google Scholar] [CrossRef] [PubMed]
  307. Hmila, I.; Saerens, D.; Ben Abderrazek, R.; Vincke, C.; Abidi, N.; Benlasfar, Z.; Govaert, J.; El Ayeb, M.; Bouhaouala-Zahar, B.; Muyldermans, S. A bispecific nanobody to provide full protection against lethal scorpion envenoming. FASEB J. 2010, 24, 3479–3489. [Google Scholar] [CrossRef] [PubMed]
  308. Altintas, I.; Kok, R.J.; Schiffelers, R.M. Targeting epidermal growth factor receptor in tumors: From conventional monoclonal antibodies via heavy chain-only antibodies to nanobodies. Eur. J. Pharm. Sci. 2012, 45, 399–407. [Google Scholar] [CrossRef] [PubMed]
  309. Chopra, A. [99mTc]Epidermal growth factor receptor-specific nanobody. In Molecular Imaging and Contrast Agent Database (MICAD); Bethesda: Rockyville, MD, USA, 2004. [Google Scholar]
  310. Friedman, M.; Stahl, S. Engineered affinity proteins for tumour-targeting applications. Biotechnol. Appl. Biochem. 2009, 53, 1–29. [Google Scholar] [CrossRef] [PubMed]
  311. Hultberg, A.; Temperton, N.J.; Rosseels, V.; Koenders, M.; Gonzalez-Pajuelo, M.; Schepens, B.; Ibanez, L.I.; Vanlandschoot, P.; Schillemans, J.; Saunders, M.; et al. Llama-derived single domain antibodies to build multivalent, superpotent and broadened neutralizing anti-viral molecules. PLoS ONE 2011, 6, e17665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  312. Narum, D.L.; Thomas, A.W. Differential localization of full-length and processed forms of PF83/AMA-1 an apical membrane antigen of Plasmodium falciparum merozoites. Mol. Biochem. Parasitol. 1994, 67, 59–68. [Google Scholar] [CrossRef]
  313. Nuttall, S.D.; Humberstone, K.S.; Krishnan, U.V.; Carmichael, J.A.; Doughty, L.; Hattarki, M.; Coley, A.M.; Casey, J.L.; Anders, R.F.; Foley, M.; et al. Selection and affinity maturation of IgNAR variable domains targeting Plasmodium falciparum AMA1. Proteins 2004, 55, 187–197. [Google Scholar] [CrossRef] [PubMed]
  314. Henderson, K.A.; Streltsov, V.A.; Coley, A.M.; Dolezal, O.; Hudson, P.J.; Batchelor, A.H.; Gupta, A.; Bai, T.; Murphy, V.J.; Anders, R.F.; et al. Structure of an IgNAR-AMA1 complex: Targeting a conserved hydrophobic cleft broadens malarial strain recognition. Structure 2007, 15, 1452–1466. [Google Scholar] [CrossRef] [PubMed]
  315. Slots, J.; Ting, M. Actinobacillus actinomycetemcomitans and Porphyromonas gingivalis in human periodontal disease: Occurrence and treatment. Periodontol. 2000 1999, 20, 82–121. [Google Scholar] [CrossRef] [PubMed]
  316. Kadowaki, T.; Nakayama, K.; Okamoto, K.; Abe, N.; Baba, A.; Shi, Y.; Ratnayake, D.B.; Yamamoto, K. Porphyromonas gingivalis proteinases as virulence determinants in progression of periodontal diseases. J. Biochem. 2000, 128, 153–159. [Google Scholar] [CrossRef] [PubMed]
  317. Aduse-Opoku, J.; Davies, N.N.; Gallagher, A.; Hashim, A.; Evans, H.E.; Rangarajan, M.; Slaney, J.M.; Curtis, M.A. Generation of lys-gingipain protease activity in Porphyromonas gingivalis W50 is independent of Arg-gingipain protease activities. Microbiology 2000, 146, 1933–1940. [Google Scholar] [CrossRef] [PubMed]
  318. Nuttall, S.D.; Krishnan, U.V.; Doughty, L.; Nathanielsz, A.; Ally, N.; Pike, R.N.; Hudson, P.J.; Kortt, A.A.; Irving, R.A. A naturally occurring NAR variable domain binds the Kgp protease from Porphyromonas gingivalis. FEBS Lett. 2002, 516, 80–86. [Google Scholar] [CrossRef]
  319. Papaneri, A.B.; Wirblich, C.; Cooper, K.; Jahrling, P.B.; Schnell, M.J.; Blaney, J.E. Further characterization of the immune response in mice to inactivated and live rabies vaccines expressing Ebola virus glycoprotein. Vaccine 2012, 30, 6136–6141. [Google Scholar] [CrossRef] [PubMed]
  320. Kondratowicz, A.S.; Maury, W.J. Ebolavirus: A brief review of novel therapeutic targets. Future Microbiol. 2012, 7, 1–4. [Google Scholar] [CrossRef] [PubMed]
  321. Fausther-Bovendo, H.; Mulangu, S.; Sullivan, N.J. Ebolavirus vaccines for humans and apes. Curr. Opin. Virol. 2012, 2, 324–329. [Google Scholar] [CrossRef] [PubMed]
  322. Liu, J.L.; Anderson, G.P.; Goldman, E.R. Isolation of anti-toxin single domain antibodies from a semi-synthetic spiny dogfish shark display library. BMC Biotechnol. 2007, 7, 78. [Google Scholar] [CrossRef] [PubMed]
  323. Nuttall, S.D.; Krishnan, U.V.; Doughty, L.; Pearson, K.; Ryan, M.T.; Hoogenraad, N.J.; Hattarki, M.; Carmichael, J.A.; Irving, R.A.; Hudson, P.J. Isolation and characterization of an IgNAR variable domain specific for the human mitochondrial translocase receptor Tom70. Eur. J. Biochem. 2003, 270, 3543–3554. [Google Scholar] [CrossRef] [PubMed]
  324. Walsh, R.; Nuttall, S.; Revill, P.; Colledge, D.; Cabuang, L.; Soppe, S.; Dolezal, O.; Griffiths, K.; Bartholomeusz, A.; Locarnini, S. Targeting the hepatitis B virus precore antigen with a novel IgNAR single variable domain intrabody. Virology 2011, 411, 132–141. [Google Scholar] [CrossRef] [PubMed]
  325. Streltsov, V.A.; Varghese, J.N.; Masters, C.L.; Nuttall, S.D. Crystal structure of the amyloid-beta p3 fragment provides a model for oligomer formation in Alzheimer’s disease. J. Neurosci. 2011, 31, 1419–1426. [Google Scholar] [CrossRef] [PubMed]
  326. Bojalil, R.; Mata-Gonzalez, M.T.; Sanchez-Munoz, F.; Yee, Y.; Argueta, I.; Bolanos, L.; Amezcua-Guerra, L.M.; Camacho-Villegas, T.A.; Sanchez-Castrejon, E.; Garcia-Ubbelohde, W.J.; et al. Anti-tumor necrosis factor VNAR single domains reduce lethality and regulate underlying inflammatory response in a murine model of endotoxic shock. BMC Immunol. 2013, 14, 17. [Google Scholar] [CrossRef] [PubMed]
  327. Konning, D.; Rhiel, L.; Empting, M.; Grzeschik, J.; Sellmann, C.; Schroter, C.; Zielonka, S.; Dickgiesser, S.; Pirzer, T.; Yanakieva, D.; et al. Semi-synthetic vNAR libraries screened against therapeutic antibodies primarily deliver anti-idiotypic binders. Sci. Rep. 2017, 7, 9676. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the comparison conventional antibody IgG with natural single domain antibodies derived from camelids and sharks. Single V (colored ovals); C domains (grey colored). Each color in V domains VH, VL, or single heavy domain represents the specific source of animals.
Figure 1. Schematic representation of the comparison conventional antibody IgG with natural single domain antibodies derived from camelids and sharks. Single V (colored ovals); C domains (grey colored). Each color in V domains VH, VL, or single heavy domain represents the specific source of animals.
Diagnostics 07 00052 g001
Figure 2. Principle of filamentous bacteriophage M13 phage display using a phagemid vector. Gene encoding for millions of variants of libraries are cloned into a phagemid vector carrying the gene encoding for one of five phage coat proteins (pIII). Large phage libraries can be obtained by transforming E. coli with phagemids and rescue of phages with helper phage. Hence, phages displaying the specific-binding antibodies against immobilized targets can be selected and isolated by several rounds of biopanning. These involve binding, washing, elution, infection and amplification. The eluted bound phages are subsequently screened by ELISA assay and followed by DNA sequencing prior to their protein expression and purification.
Figure 2. Principle of filamentous bacteriophage M13 phage display using a phagemid vector. Gene encoding for millions of variants of libraries are cloned into a phagemid vector carrying the gene encoding for one of five phage coat proteins (pIII). Large phage libraries can be obtained by transforming E. coli with phagemids and rescue of phages with helper phage. Hence, phages displaying the specific-binding antibodies against immobilized targets can be selected and isolated by several rounds of biopanning. These involve binding, washing, elution, infection and amplification. The eluted bound phages are subsequently screened by ELISA assay and followed by DNA sequencing prior to their protein expression and purification.
Diagnostics 07 00052 g002
Table 1. Comparison of parameters for producing monoclonal, polyclonal, and recombinant antibodies.
Table 1. Comparison of parameters for producing monoclonal, polyclonal, and recombinant antibodies.
CharacteristicsMonoclonal AntibodyPolyclonal AntibodiesRecombinant Antibodies
Cost to produce++++++
Difficulty of production++++++
Skills or training++++++
Time scale++++++
Specificity and affinity+++++++
Amounts++++++
Commercial availability++++++
Variability+++++
“+” represents the least, “++” represents the moderate, and “+++” represents the most (Adapted from [186] with permission from publishing journal).
Table 2. The applications of camelids VHH against specified antigens from various diseases.
Table 2. The applications of camelids VHH against specified antigens from various diseases.
Target AntigensDiseasesApplicationsReference
HER2Breast cancerDiagnostic[284,285,286]
TNTExplosiveDiagnostic[274,287]
Ts14 glycoproteinT. solium cysticercosisDiagnostic[249]
LMM, ES, CSE, TSB, LLGPs, VF of T. soliumNeurocysticercosisImmunodiagnosis[249]
VEGF-A165NeoangiogenesisDiagnostic and therapeutic[288]
HPV-16 L1 proteinCervical cancerDiagnostic and therapeutic[289,290]
DARCMalaria (by P. vivax)Diagnostic or therapeutic[291]
Poliovirus type 1 Sabin strain particlesPoliomyelitisDiagnostic and therapeutic[292,293]
CD105Angiogenesis related tumorsDiagnostic and therapeutic[294,295]
HSP-60Brucellosis (Livestock)Diagnostic and vaccine[222,296]
Caffeine carboxylate KLHBeveragesDetection and separation[251,252]
SEBToxinSensor and diagnostic[275]
RicinToxinSensor and diagnostic[275]
BoNT/AToxinSensor and diagnostic[235,297,298]
CEAColon cancerIn vivo imaging[299,300,301]
VCAM1Atherosclerotic plaquesMolecular imaging[302,303,304]
EGFRTumoursDetection and imaging[305,306]
Scorpion AahIIToxinNeutralizing and therapeutic[276,307]
Heat-killed B. melitensis Riv1BrucellosisDiagnostic, therapeutic and vaccination[282,308,309,310]
RSV protein FAcute lower respiratory tractTherapeutic[311]
vWFThrombosisTherapeuticwww.ablynx.com
TNFα, IL-6R, IgERheumatoid arthritisTherapeuticwww.ablynx.com
RANKLBone metastasisTherapeuticwww.ablynx.com
RSVbronchiolitis and pneumonia Therapeuticwww.ablynx.com
DR5Solid tumorsTherapeuticwww.ablynx.com
Not statedAlzheimer’s diseaseTherapeuticwww.ablynx.com
Table 3. The applications of shark VNAR against specified antigens from various diseases.
Table 3. The applications of shark VNAR against specified antigens from various diseases.
Target AntigensDiseasesApplicationsReference
AMA1 (P. falciparum)MalariaDiagnostic[313,314]
Zaire ebolavirus viral nucleoproteinEbolavirus Haemorrhagic FeverDiagnostic[262]
Cholera toxinToxinDiagnostic[258]
Tom70Human immunosilent target processesDetection[323]
BoNT/AToxinSensor and diagnostic[322]
RicinToxinSensor and diagnostic[322]
SEBToxinSensor and diagnostic[322]
HBeAgHepatitis B virusImmunolocalization and diagnostic[324]
Kgp protease (P. gingivalis)Periodontal diseaseNeutralization[318]
Nonfibrillar oligomer formationAlzheimer’s diseaseModelling[325]
rhTNFαpro-inflammatory cytokineTherapeutic[260,326]
mAb idiotopeCancerTherapeutic[327]
GPCR’s ion channels Therapeuticwww.adalta.com.au
Anti-thrombotic drug targetsCardiovacular diseaseDiagnostic and therapeuticwww.adalta.com.au
Idiopathic pulmonary fibrosisInflammationTherapeuticwww.adalta.com.au
Multiple sclerosisCentral neuron system diseaseTherapeuticwww.ossianix.com
Botulinum toxin light chain proteaseGastrointestinal tractTherapeuticwww.ossianix.com
MyostatinNeurological diseaseTherapeuticwww.ossianix.com
UveitisEye inflammatoryTherapeuticwww.elasmogen.com

Share and Cite

MDPI and ACS Style

Leow, C.H.; Fischer, K.; Leow, C.Y.; Cheng, Q.; Chuah, C.; McCarthy, J. Single Domain Antibodies as New Biomarker Detectors. Diagnostics 2017, 7, 52. https://doi.org/10.3390/diagnostics7040052

AMA Style

Leow CH, Fischer K, Leow CY, Cheng Q, Chuah C, McCarthy J. Single Domain Antibodies as New Biomarker Detectors. Diagnostics. 2017; 7(4):52. https://doi.org/10.3390/diagnostics7040052

Chicago/Turabian Style

Leow, Chiuan Herng, Katja Fischer, Chiuan Yee Leow, Qin Cheng, Candy Chuah, and James McCarthy. 2017. "Single Domain Antibodies as New Biomarker Detectors" Diagnostics 7, no. 4: 52. https://doi.org/10.3390/diagnostics7040052

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