Next Article in Journal
Dual-energy X-ray Absorptiometry of Both Hips Helps Appropriate Diagnosis of Low Bone Mineral Density and Osteoporosis
Next Article in Special Issue
New Biomarkers and Diagnostic Tools for the Management of Fever in Low- and Middle-Income Countries: An Overview of the Challenges
Previous Article in Journal
Ultrasonographic Diagnosis of Thoracic Outlet Syndrome Secondary to Brachial Plexus Piercing Variation
Previous Article in Special Issue
Analytical Tools to Improve Optimization Procedures for Lateral Flow Assays
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancing Point-of-Care (PoC) Testing Using Human Saliva as Liquid Biopsy

by
Rabia Sannam Khan
1,
Zohaib Khurshid
2,* and
Faris Yahya Ibrahim Asiri
3
1
Department of Oral Pathology, College of Dentistry, Baqai University, Super Highway, P.O.Box: 2407, Karachi 74600, Pakistan
2
Prosthodontics and Implantology, College of Dentistry, King Faisal University, Al-Ahsa 31982, Saudi Arabia
3
Department of Preventive Dentistry, College of Dentistry, King Faisal University, Al-Ahsa 31982, Saudi Arabia
*
Author to whom correspondence should be addressed.
Diagnostics 2017, 7(3), 39; https://doi.org/10.3390/diagnostics7030039
Submission received: 30 May 2017 / Revised: 24 June 2017 / Accepted: 30 June 2017 / Published: 4 July 2017
(This article belongs to the Special Issue Novel Point-of-Care Technologies in Diagnostics)

Abstract

:
Salivary diagnostics is an emerging field for the encroachment of point of care technology (PoCT). The necessity of the development of point-of-care (PoC) technology, the potential of saliva, identification and validation of biomarkers through salivary diagnostic toolboxes, and a broad overview of emerging technologies is discussed in this review. Furthermore, novel advanced techniques incorporated in devices for the early detection and diagnosis of several oral and systemic diseases in a non-invasive, easily-monitored, less time consuming, and in a personalised way is explicated. The latest technology detection systems and clinical utilities of saliva as a liquid biopsy, electric field-induced release and measurement (EFIRM), biosensors, smartphone technology, microfluidics, paper-based technology, and how their futuristic perspectives can improve salivary diagnostics and reduce hospital stays by replacing it with chairside screening is also highlighted.

Graphical Abstract

1. Introduction

Laboratory testing remains the dominant mainstay for analytical processes of a large number of samples involving the disciplines of biochemistry, haematology, microbiology, anatomical pathology, and much more [1]. Due to the limitations and pressure on healthcare budgets faced by a very large number of countries, primary care is best suited for the world to reduce expenses instead of secondary and tertiary hospitals. Poverty, chronic disease, infections lead to significant problems in developing the world, and adequate diagnostic testing turns out to be difficult to meet the needs. Hence, consequently, initiatives in making solid models using point-of-care technology (PoCT) came into existence [2].

1.1. Paradigm Shift from Central Laboratory (CL) to Point-of-Care

A self-monitoring blood glucose meter, coagulation (INR), and pregnancy testing kits using urine samples are well-known examples of PoCT and has become over-the-counter products to be sold in the market. Saliva is predicted to be a substitute for blood, collected non-invasively for the diagnosis of oral and systemic diseases. Thus, PoCT replaces the specialist testing centres by using the samples other than blood and urine [3]. For the development of PoCT devices, minimum risk of infection with no mental and physical pain is of utmost importance to consider, in addition to automation, integration, multiplexed detection ability, quick analysis, small sample size, and minimal training as the primary goals of modern medicine [4]. With the advent of the struggle in the growing potential of developing PoCT, the World Health Organization (WHO) provided guidelines which had the features for designing devices, known as the ASSURED criteria, which indicated that devices had to be affordable, sensitive, specific, user-friendly, rapid and robust, with no complex equipment, and be delivered to end users efficiently.
The precision to central lab testing is being facilitated by the use of biomarkers and saliva is a viable biofluid for diagnostic applications. The human saliva revolution in medical and dental sciences through its property as a “mirror of body health” in the last decade brought many disease detections through its compositional changes in disease conditions [5]. Human oral cavity consists of different sources, such as salivary glands (major and minor), gingival crevicular fluid (GCF), microbes, and oral epithelial sheds for the production of whole mouth fluid (WMF) [6,7,8,9,10,11,12]. This fluid is 99% water, but 1% consists of DNA, mRNA, microRNA, proteins, metabolites, and microbiota which are utilised as a diagnostic fluid for disease analysis and even for forensic analysis [13,14]. In this particular review, we present the current knowledge of saliva, diagnostic toolboxes, how PoC technology works, developed PoC devices for detection of various diseases, and the future prospects of utilising saliva as a diagnostic tool.

1.2. Saliva in the Diagnosis of Oral and Systemic Diseases

Saliva is a complex fluid containing various enzymes, electrolytes, proteins, nucleic acids, antimicrobial constituents, hormones, cytokines, and antibodies. Its composition virtually reflects the entire state of health and disease in a body and it has the potential of being a diagnostic medium for a broad range of diseases [15,16], such as in the detection of periodontal diseases, caries risk assessments, breast cancer, oral cancers, salivary gland diseases, HIV, and much more [16]. However, the effects of alteration in the salivary composition are seen in the lipid profile of cystic fibrosis patients, which is markedly changed in comparison to healthy subjects [17]. The submandibular gland saliva of cystic fibrosis patients contains 66% more lipids per 100 mL of saliva than that of a healthy subject. The salivary fatty acid profile can be a good indicator for the early detection of tumorigenesis processes and cardiovascular diseases. The increase in production of salivary arachidonic acid, relevant for their eicosanoid production related to the tumorigenesis process and cardiovascular diseases, is influenced by dietary fat intake [18]. Figure 1 portrays alterations of biomarkers in various body organs in diseased states through which saliva can detect a variety of oral and systemic diseases.
Similarly, in Sjogren’s syndrome (SS) the saliva flow is compromised in the patient due to ductal changes by lymphocytic infiltration and fibrosis of the salivary glands and the patient also suffers from the consequences of dental caries, infections dysphagia, and other oral pain [19]. The lipid profile of SS patients is twice higher than an average healthy person and elevated level of cytokines (IgA, IgG, interleukin-6, and prostaglandins-E2) and antibodies. Many salivary proteins were reported as biomarkers for SS diagnosis; for example, profilin [20], anhydrase-I [21], IL-4, IL-5 [22], MxA [23], and CXCL13 [24,25]. Human saliva has a proven diagnostic role in the detection of cardiovascular diseases, reported in studies examining whole mouth salivary biomarkers e.g., C-reactive proteins (CRP) [26], cardiac troponin (cTn) [27], creatine phosphokinase [28], and NT-ProBNP [29]. Diabetes is another common disease faced all around the world and developing rapidly due to dietary habits, genetic, and other systematic disease-related complications [30]. Due to its non-invasiveness, a cheap and easy sampling of saliva is attractive as a diagnostic fluid for diabetes analysis [31]. Previously reported studies concluded the different biomarkers for diabetes detection in early stages. A recently-reported study saw a marked alteration in levels of salivary glucose, amylase, calcium, and phosphorus in comparison to serum from diabetic and non-diabetic patients [32], as seen in Table 1 below.
The current review presents a broad overview on salivary diagnostics, the validation of biomarkers (diagnostic targets) through diagnostic toolboxes, discussion related to new biomarker-related PoC platforms, the latest emerging PoC technologies including highlights on biosensors, biological micro-electro-mechanical systems (BioMEMS), microfluidics/paper based technology, electric field-induced release and measurement (EFIRM), and smartphone-based biosensors their functions and clinical utility in medical field.

2. Point-of-Care Technology: An Overview

Point-of-care (PoC) technology in diagnostics tends to evaluate biomarkers that are suggestive of underlying biological or physical characteristics of an individual. Therefore, biomarkers determine the risk and severity of the disease, as well as the individual’s response to treatment [15]. In addition to it, diagnostic methodologies other than biomarkers also include biochip and biosensor systems. Biochip systems obtain the requisite volume of saliva for testing whereas the biosensor system is the analytical high-sensitive technology for the detection of biomarkers [50]. Accurate PoC diagnostics needs no pre-processing and screening for biomarker identification on top of it non-invasive testing, as seen in already-patented devices, such as for the detection of oral cancer known as the Oral Fluid NanoSensor Test (OFNASET), the detection of human papillomavirus (HPV) infection OraRisk HPV test, and the diagnosis of periodontal diseases PerioPath [51,52]. Moreover, measuring the molecular level biomarkers in the form of proteins, mRNA, DNA, electrolytes, and small molecules requires techniques of microfabrication, such as in the developed micro/nanoelectromechanical systems (MEMS/NEMS) [53], and current emerging technologies provide new avenues of PoC diagnostics in the variety of “lab-on-chip” techniques which integrates the complexities of lab procedures on a computer chip in the size of a device that, hence, gives an opportunity to detect and diagnose multiple diseases simultaneously with the help of biomarkers [54].

2.1. Diagnostic Targets

Salivary diagnostics needs appropriate identification and validation of biomarkers for the detection of diseases, and a biomarker is a quantifiable parameter that can interact physiologically and biochemically at a molecular or cellular level, which sequentially acts as an indicator of normal, pathological, and interventional behaviours of the body’s response [55,56]. Biomarkers include several classes, such as proteins, DNA, RNA, metabolites, and microbes, so collectively these all are used for diagnosis of several diseases and are called a molecular signature [57].

2.2. Diagnostic Toolboxes

Major salivary diagnostic tool boxes include proteomes, metabolomes, genomes (transcriptome, epigenome), microbiomes, and immunologic categories. Table 2 shows the variety of methods used to analyse molecules for the investigation and validation of biomarkers. Proteomes make up the biological system and could be employed for the detection of diabetes, periodontitis, caries, cystic fibrosis, AIDS, OSCC, breast cancer, lung cancer, pancreatic cancer, Sjogren’s syndrome, and many more through mass spectroscopy, 2D gel electrophoresis, ELISA and protein immunoblot techniques [58,59,60,61]. However, mRNA and DNA come under salivary transcriptomes and genomes, and their profiling through gene chip arrays, DNA hybridization, qPCR, and gel electrophoresis helps in the detection of OSCC, as conducted by Li et al. [62], and in Sjogren’s syndrome, hepatitis, HIV, etc. The metabolic investigation, on the other hand, requires and uses gas chromatography mass spectrometry, nuclear magnetic resonance spectroscopy, and high-performance liquid chromatography [63] for the detection of diabetes, lung, pancreatic, breast cancers, and Sjogren’s syndrome [64,65]. In the same way, the salivary microbiome and immnomics used different methods, as mentioned in the table for the detection of infectious diseases, HIV, hepatitis, malaria, dengue, Ebola virus, cytomegalovirus, herpes infection, and countless other diseases [66,67,68].

2.3. Salivary Biomarker-Based PoC Platforms

In Table 3, shows analysis system based on different technologies applied on saliva for the detection of biomarkers to reduce the time duration and early diagnosis of certain diseases. As shown in table it is revealed that single and multiplexed systems were being used such as MEMS, ORI, chromatography test strips and several salivary diagnostics devices (USA) for the detection of proteins solely, also proteins and nucleic acids altogether (for e.g., IL-8, MMP-8, a-amylase, HIV, HCV) with lowering the time limit as much little as one minute. These technologies, therefore, reduces the invasive procedures significantly to a larger level [16].

3. Emerging Novel PoC Technologies

3.1. Biosensors

The biosensor is a bioanalytical device which has the ability to mimic any biological material, i.e., antibodies/antigens, nucleic acids, cellular structures or enzymes. That organic material is integrated into a transducing microsystem. The transducer could be electrochemical, thermometric, optical, piezoelectric, or magnetic, and this is called label-free detection [74], while label-based detection includes fluorescent immunoassays, FRET, and quantum dots. Biosensors function with the biorecognition of particular elements for particular targeted analytes and the maintenance of selectivity and sensitivity in the existence of other interfering compounds [75]. In the medical field, biosensor applications are growing rapidly, such as for the diagnosis of diabetes mellitus, urinary tract infections (UTI), identification of end-stage heart failure, and acute leukaemias (Table 1) [76].

3.2. Fluorescent Biosensors

Fluorescent biosensors can be used for cancer, drug discovery, arthritis, cardiovascular and neurodegenerative diseases, viral infections, chronic myeloid leukemia, and many more, by using the principal of high throughput screening approaches, the use of fluorescent probes in gene expression, localization of protein in cell cycle, apoptosis, signal transduction, and transcription. For the detection of chronic myeloid leukaemia, a genetically-encoded FRET biosensor was developed to detect Bcr-Abl kinase activity to see the correlation, it was further used to check the response to treatment, drug resistance, and predictive values for alternative therapeutics [77]. Moreover, Lee et al. applied hafnium oxide in a novel biosensor for the detection of human interleukin (IL-10) (atomic layer deposited hafnium oxide gate dielectrics for charge-based biosensors) [78]. Applications of nanomaterial biosensors give opportunities for a new generation of biosensor technologies that can be broadly used in monitoring, diagnosis, control, and analysis.

3.3. Biological Micro-Electro-Mechanical Systems (BioMEMS)

PoC lab-on-chip systems use small and simply-constructed (BioMEMS) devices for the detection of biological and chemical agents. BioMEMS are utilized for the detection of cells, proteins, microorganisms, viruses, and DNA in biological samples. They are based on micro/nanoscale fabrication systems which help in increasing the sensitivity of results from sensors, increased reliability, increased performance, reduced detection time, and cost effectiveness. It has label-free detection techniques, including micro-cantilevers, surface plasmon resonance (SPR), quartz crystal microbalances (QCM), and organic field-effect transistors (BioFETs) [79]. BioMEMS are used for a range of applications, such as for drug delivery, cardio MEMS to monitor heart patients, hearing aids, insulin micropumps, endoscopic pills, and retinal prosthesis [80,81] (Table 3). By blending molecular biology with computational systems, the major nanotechnology achievement could be bio-nano-electro-mechanical systems (BioNEMS) in the future for further improvements in the medical sector.

3.4. Microfluidics/Paper-Based Technology

Microfluidic applications operate on integrated microfabrication and specific physiochemical properties. Initially, the use of silicon, inorganic glass, and ceramic was used in microfluidic devices which have been vastly replaced by the soft and rigid thermostatic and thermoplastic materials and, finally, into paper-based technologies, using biodegradable and hydrogel materials [82]. At Harvard University in 2007 microfluidic paper-based analytical devices (μPADs) were pioneered by Whitesides. Paper is porous and hydrophilic, therefore, it provides a platform for the fabrication of microfluidic channels by patterning the paper with 2D and 3D μPADs and having a variety of assay designs. They are used in the detection of urine metabolites, blood glucose, pH value, liver function, and infectious agents, and they are also widely used in pregnancy test kits [83].

3.5. Electric Field-Induced Release and Measurement (EFIRM)

A liquid biopsy technique called EFIRM, uses readout enzymes and immobilised probes for the capturing and detection of biomarkers from biofluids. EFIRM uses the electrochemical method to facilitate nucleic acid hybridization. This approach is advantageous for accurate detection of RNA, protein biomarker targets in exosomes. Additionally, without the extraction of DNA and nucleic acid, EFIRM can analyse the mutation status within an hour. IL-8 protein and IL-8 mRNA markers for oral cancer, non-squamous cell lung cancer (NSCLC) oncogenic mutation, and epidermal growth factor receptor (EGFR) mutation in non-small cell lung cancer can be detected through this EFIRM system [57,84].

3.6. Smartphone-Based Biosensors

Smartphones operate similarly to miniature computers, acting as cheap, portable analytical laboratory devices. They are helpful for the detection, and diagnosis, of various diseases, such as cancer, tuberculosis, and the self-monitoring of blood glucose. Figure 2 illustrates the few shapes and designs of currently-used PoC technologies while Table 4 shows all the functions, techniques, and clinical utilities of these techniques in a concise manner.
Recent advancements in smartphones electronics and new app development have made its use as a smart detector by having all the mixed optical methods, i.e., fluorescence, surface plasmon resonance (SPR), reflectance, absorbance, bio-chemiluminescence, and electrochemiluminescence [85]. Amongst them all SPR gained importance because of its high sensitivity, label-free, microfluidic technique. Conventional SPR utilizes planar thin gold film while localized SPR (LSPR) contains metal nanostructures [86]. Hence, SPR becomes the powerful tool in biomedical application such as for the study of several DNA, RNA, proteins, lipids, carbohydrates, even mutation detection [87]. Moreover, Lee et al. described the smartphone’s function as a compact microscope in which ambient illumination as a light source was being used instead of a chip-scale method. This lensless imaging scheme allows sub-micron resolution and the built-in android application brings simplicity, robustness, and employability for several field applications [88].

4. Future Direction and Conclusions

This article gives an overview of research in molecular diagnostics, microbiology, and immunology. Routine laboratory testing includes the majority of haematology testing, clinical chemistry, and immunochemistry by using high-throughput instrumentation. Therefore, salivary PoCT diagnostics is replacing the central laboratory and offers efficient, fast, quick and easy automation. Since the emphasis is switching more towards prevention and early detection of a variety of diseases, development of small wireless devices has made a dramatic impact on healthcare services. The next decade will bring breakthroughs in terms of precision, efficiency, and bedside monitoring instead of hospital setups.
Personalised medicine, with the help of biosensors, lab-on-chip systems, individual genetics, smartphones monitoring parameters, and microfluidic devices, will improve the primary healthcare system. Moreover, it allows clinicians to be accurate, to be more consistent, to capture clinical data quickly, provide patient satisfaction, and streamline workflow. Salivary diagnostics’ impact on the healthcare system is enormous, being non-invasive, convenient, and well-credentialed, while bioinformatics introduction will make standards and performance higher and improved.

Acknowledgments

We are thankful to Pakistan Human Saliva Research Group (PHSRG) for helping us with the write-up and compilation of all knowledge on one platform.

Author Contributions

Rabia Sannam Khan brought the idea of this innovative topic and helped in whole paper write-up. Zohaib Khurshid and Faris Yahya Ibrahim Asiri supported in the compilation of tables, figures and English improvement.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. St John, A.; Price, C.P. Existing and emerging technologies for point-of-care testing. Clin. Biochem. Rev. Aust. Assoc. Clin. Biochem. 2014, 35, 155–167. [Google Scholar]
  2. Jani, I.V.; Peter, T.F. How point-of-care testing could drive innovation in global health. New Engl. J. Med. 2013, 368, 2319–2324. [Google Scholar] [CrossRef] [PubMed]
  3. Shetty, V.; Yamaguchi, M. Salivary biosensors for screening trauma-related psychopathology. Oral Maxillofac. Surg. Clin. North Am. 2010, 22, 269–278. [Google Scholar] [CrossRef] [PubMed]
  4. Khanna, P.; Walt, D.R. Salivary diagnostics using a portable point-of-service platform: A review. Clin. Ther. 2015, 37, 498–504. [Google Scholar] [CrossRef] [PubMed]
  5. Khurshid, Z.; Zohaib, S.; Najeeb, S.; Zafar, M.; Rehman, R.; Rehman, I. Advances of proteomic sciences in dentistry. Int. J. Mol. Sci. 2016, 17, 728. [Google Scholar] [CrossRef] [PubMed]
  6. Sahibzada, H.A.; Khurshid, Z.; Khan, R.S.; Naseem, M.; Siddique, K.M.; Mali, M.; Zafar, M.S. Salivary IL-8, IL-6 and TNF-α as potential diagnostic biomarkers for oral cancer. Diagnostics 2017, 7, 21. [Google Scholar] [CrossRef] [PubMed]
  7. Sannam Khan, R.; Khurshid, Z.; Akhbar, S.; Faraz Moin, S. Advances of salivary proteomics in oral squamous cell carcinoma (OSCC) detection: An update. Proteomes 2016, 4, 41. [Google Scholar] [CrossRef] [PubMed]
  8. Khurshid, Z.; Najeeb, S.; Mali, M.; Moin, S.F.; Raza, S.Q.; Zohaib, S.; Sefat, F.; Zafar, M.S. Histatin peptides: Pharmacological functions and their applications in dentistry. Saudi Pharm. J. 2015, 25, 25–31. [Google Scholar] [CrossRef] [PubMed]
  9. Khurshid, Z.; Mali, M.; Naseem, M.; Najeeb, S.; Zafar, M. Human gingival crevicular fluids (GCF) proteomics: An overview. Dent. J. 2017, 5, 12. [Google Scholar] [CrossRef]
  10. Khurshid, Z.; Haq, J.A.; Khan, R.S.; Sohail Zafar, M.; Altaf, M.; Najeeb, S. Human saliva and its role in oral & systemic health. J. Pak. Dent. Assoc. 2016, 25, 170–174. [Google Scholar]
  11. Khurshid, Z.; Naseem, M.; Sheikh, Z.; Najeeb, S.; Shahab, S.; Zafar, M.S. Oral antimicrobial peptides: Types and role in the oral cavity. Saudi Pharm. J. 2015, 24, 515–524. [Google Scholar] [CrossRef] [PubMed]
  12. Khurshid, Z.; Zohaib, S.; Najeeb, S.; Zafar, M.; Slowey, P.; Almas, K. Human saliva collection devices for proteomics: An update. Int. J. Mol. Sci. 2016, 17, 846. [Google Scholar] [CrossRef] [PubMed]
  13. Tabak, L.A. In defense of the oral cavity: The protective role of the salivary secretions. Pediatr. Dent. 2006, 28, 110–198. [Google Scholar] [PubMed]
  14. Tsuchida, S.; Satoh, M.; Sogawa, K.; Kawashima, Y.; Kado, S.; Ishige, T.; Beppu, M.; Sawai, S.; Nishimura, M.; Kodera, Y.; et al. Application of proteomic technologies to discover and identify biomarkers for periodontal diseases in gingival crevicular fluid: A review. Proteom. Clin. App. 2014, 8, 232–240. [Google Scholar] [CrossRef]
  15. Tabak, L.A. Point-of-care diagnostics enter the mouth. Ann. N. Y. Acad. Sci. 2007, 1098, 7–14. [Google Scholar] [CrossRef] [PubMed]
  16. Wei, F.; Wong, D.T.W. Point-of-care platforms for salivary diagnostics. Chin. J. Dent. Res. 2012, 15, 7–15. [Google Scholar] [PubMed]
  17. Slomiany, B.L.; Murty, V.L.N.; Slomiany, A. Salivary lipids in health and disease. Prog. Lipid Res. 1985, 24, 311–324. [Google Scholar] [CrossRef]
  18. Actis, A.B.; Perovic, N.R.; Defagó, D.; Beccacece, C.; Eynard, A.R. Fatty acid profile of human saliva: A possible indicator of dietary fat intake. Arch. Oral Biol. 2005, 50, 1–6. [Google Scholar] [CrossRef] [PubMed]
  19. Javaid, M.A.; Ahmed, A.S.; Durand, R.; Tran, S.D. Saliva as a diagnostic tool for oral and systemic diseases. J. Oral Biol. Craniofac. Res. 2016, 6, 67–76. [Google Scholar] [CrossRef] [PubMed]
  20. Deutsch, O.; Krief, G.; Konttinen, Y.T.; Zaks, B.; Wong, D.T.; Aframian, D.J.; Palmon, A. Identification of Sjogren’s syndrome oral fluid biomarker candidates following high-abundance protein depletion. Rheumatology 2015, 54, 884–890. [Google Scholar] [CrossRef] [PubMed]
  21. Delaleu, N.; Mydel, P.; Kwee, I.; Brun, J.G.; Jonsson, M.V.; Jonsson, R. High fidelity between saliva proteomics and the biologic state of salivary glands defines biomarker signatures for primary Sjgren’s syndrome. Arthritis Rheumatol. 2015, 67, 1084–1095. [Google Scholar] [CrossRef] [PubMed]
  22. Langie, S.A.S.; Szarc Vel Szic, K.; Declerck, K.; Traen, S.; Koppen, G.; Van Camp, G.; Schoeters, G.; Vanden Berghe, W.; De Boever, P. Whole-genome saliva and blood DNA methylation profiling in individuals with a respiratory allergy. PLoS ONE 2016, 11, e0151109. [Google Scholar] [CrossRef] [PubMed]
  23. Maria, N.I.; Brkic, Z.; Waris, M.; van Helden-Meeuwsen, C.G.; Heezen, K.; van de Merwe, J.P.; van Daele, P.L.; Dalm, V.A.S.H.; Drexhage, H.A.; Versnel, M.A. MxA as a clinically applicable biomarker for identifying systemic interferon type I in primary Sjogren’s syndrome. Ann. Rheum. Dis. 2014, 73, 1052–1059. [Google Scholar] [CrossRef] [PubMed]
  24. Kramer, J.M.; Klimatcheva, E.; Rothstein, T.L. CXCL13 is elevated in Sjogren’s syndrome in mice and humans and is implicated in disease pathogenesis. J. Leukoc. Biol. 2013, 94, 1079–1089. [Google Scholar] [CrossRef] [PubMed]
  25. Lee, H.T.; Shiao, Y.M.; Wu, T.H.; Chen, W.S.; Hsu, Y.H.; Tsai, S.F.; Tsai, C.Y. Serum BLC/CXCL13 concentrations and renal expression of CXCL13/CXCR5 in patients with systemic lupus erythematosus and lupus nephritis. J. Rheum. 2010, 37, 45–52. [Google Scholar] [CrossRef] [PubMed]
  26. Out, D.; Hall, R.J.; Granger, D.A.; Page, G.G.; Woods, S.J. Assessing salivary C-reactive protein: Longitudinal associations with systemic inflammation and cardiovascular disease risk in women exposed to intimate partner violence. Brain Behav. Immun. 2012, 26, 543–551. [Google Scholar] [CrossRef] [PubMed]
  27. Mirzaii-Dizgah, I.; Riahi, E. Salivary high-sensitivity cardiac troponin T levels in patients with acute myocardial infarction. Oral Dis. 2013, 19, 180–184. [Google Scholar] [CrossRef] [PubMed]
  28. Mirzaii-Dizgah, I.; Jafari-Sabet, M. Unstimulated whole saliva creatine phosphokinase in acute myocardial infarction. Oral Dis. 2011, 17, 597–600. [Google Scholar] [CrossRef] [PubMed]
  29. Foo, J.Y.Y.; Wan, Y.; Kostner, K.; Arivalagan, A.; Atherton, J.; Cooper-White, J.; Dimeski, G.; Punyadeera, C. NT-ProBNP levels in saliva and its clinical relevance to heart failure. PLoS ONE 2012, 7, e48452. [Google Scholar] [CrossRef] [PubMed]
  30. Chee, C.S.; Chang, K.M.; Loke, M.F.; Angela Loo, V.P.; Subrayan, V. Association of potential salivary biomarkers with diabetic retinopathy and its severity in type-2 diabetes mellitus: A proteomic analysis by mass spectrometry. PeerJ 2016, 4, e2022. [Google Scholar] [CrossRef] [PubMed]
  31. Zhang, C.Z.; Cheng, X.Q.; Li, J.Y.; Zhang, P.; Yi, P.; Xu, X.; Zhou, X.D. Saliva in the diagnosis of diseases. Int. J. Oral Sci. 2016. [Google Scholar] [CrossRef] [PubMed]
  32. Ladgotra, A. Estimation of salivary and serum biomarkers in diabetic and non diabetic patients—A comparative study. J. Clin. Diagn. Res. 2016, 10, ZC56–ZC61. [Google Scholar] [CrossRef] [PubMed]
  33. Shetty, V.; Zigler, C.; Robles, T.F.; Elashoff, D.; Yamaguchi, M. Developmental validation of a point-of-care, salivary? Amylase biosensor. Psychoneuroendocrinology 2011, 36, 193–199. [Google Scholar] [CrossRef] [PubMed]
  34. Gaydos, C.A.; Solis, M.; Hsieh, Y.H.; Jett-Goheen, M.; Nour, S.; Rothman, R.E. Use of tablet-based kiosks in the emergency department to guide patient HIV self-testing with a point-of-care oral fluid test. Int. J. STD AIDS 2013, 24, 716–721. [Google Scholar] [CrossRef] [PubMed]
  35. Zachary, D.; Mwenge, L.; Muyoyeta, M.; Shanaube, K.; Schaap, A.; Bond, V.; Kosloff, B.; de Haas, P.; Ayles, H. Field comparison of oraquick advance rapid HIV-1/2 antibody test and two blood-based rapid HIV antibody tests in Zambia. BMC Infect. Dis. 2012, 12, 183. [Google Scholar] [CrossRef] [PubMed]
  36. Tucker, J.D.; Bien, C.H.; Peeling, R.W. Point-of-care testing for sexually transmitted infections: Recent advances and implications for disease control. Curr. Opin. Infect. Dis. 2013, 26, 73–79. [Google Scholar] [CrossRef] [PubMed]
  37. Teengam, P.; Siangproh, W.; Tuantranont, A.; Vilaivan, T.; Chailapakul, O.; Henry, C.S. Multiplex paper-based colorimetric DNA sensor using pyrrolidinyl peptide nucleic acid-induced AgNPs aggregation for detecting MERS-CoV, MTB, and HPV oligonucleotides. Anal. Chem. 2017, 89, 5428–5435. [Google Scholar] [CrossRef] [PubMed]
  38. Tlili, C.; Myung, N.V.; Shetty, V.; Mulchandani, A. Label-free, chemiresistor immunosensor for stress biomarker cortisol in saliva. Biosens. Bioelectron. 2011, 26, 4382–4386. [Google Scholar] [CrossRef] [PubMed]
  39. Ji, S.; Choi, Y. Point-of-care diagnosis of periodontitis using saliva: Technically feasible but still a challenge. Front. Cell. Infect. Microbiol. 2015, 5, 1–9. [Google Scholar] [CrossRef] [PubMed]
  40. Floriano, P.N.; Christodoulides, N.; Miller, C.S.; Ebersole, J.L.; Spertus, J.; Rose, B.G.; Kinane, D.F.; Novak, M.J.; Steinhubl, S.; Acosta, S.; et al. Use of saliva-based nano-biochip tests for acute myocardial infarction at the point of care: A feasibility study. Clin. Chem. 2009, 55, 1530–1538. [Google Scholar] [CrossRef] [PubMed]
  41. Blicharz, T.M.; Siqueira, W.L.; Helmerhorst, E.J.; Oppenheim, F.G.; Wexler, P.J.; Little, F.F.; Walt, D.R. Fiber-optic microsphere-based antibody array for the analysis of inflammatory cytokines in saliva. Anal. Chem. 2009, 81, 2106–2114. [Google Scholar] [CrossRef] [PubMed]
  42. Torrente-Rodrguez, R.M.; Campuzano, S.; Ruiz-Valdepeas Montiel, V.; Gamella, M.; Pingarrn, J.M. Electrochemical bioplatforms for the simultaneous determination of interleukin (IL)-8 mRNA and IL-8 protein oral cancer biomarkers in raw saliva. Biosens. Bioelectron. 2016, 77, 543–548. [Google Scholar] [CrossRef] [PubMed]
  43. Walt, D.R.; Blicharz, T.M.; Hayman, R.B.; Rissin, D.M.; Bowden, M.; Siqueira, W.L.; Helmerhorst, E.J.; Grand-Pierre, N.; Oppenheim, F.G.; Bhatia, J.S.; et al. Microsensor arrays for saliva diagnostics. Ann. N. Y. Acad. Sci. 2007, 1098, 389–400. [Google Scholar] [CrossRef] [PubMed]
  44. Barnfather, K.D.; Cope, G.F.; Chapple, I.L. Effect of incorporating a 10 min point of care test for salivary nicotine metabolites into a general practice based smoking cessation programme: Randomised controlled trial. BMJ Clin. Res. 2005, 331, 999. [Google Scholar] [CrossRef] [PubMed]
  45. Ching, K.H.; Burbelo, P.D.; Gonzalez-Begne, M.; Roberts, M.E.P.; Coca, A.; Sanz, I.; Iadarola, M.J. Salivary anti-Ro60 and anti-Ro52 antibody profiles to diagnose Sjogren’s Syndrome. J. Dent. Res. 2011, 90, 445–449. [Google Scholar] [CrossRef] [PubMed]
  46. Du, Y.; Zhang, W.; Wang, M.L. Sensing of salivary glucose using nano-structured biosensors. Biosensors 2016, 6, 10. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, W.; Du, Y.; Wang, M.L. Noninvasive glucose monitoring using saliva nano-biosensor. Sens. BioSens. Res. 2015, 4, 23–29. [Google Scholar] [CrossRef]
  48. Christodoulides, N.; Pierre, F.N.; Sanchez, X.; Li, L.; Hocquard, K.; Patton, A.; Muldoon, R.; Miller, C.S.; Ebersole, J.L.; Redding, S.; et al. Programmable bio-nanochip technology for the diagnosis of cardiovascular disease at the point-of-care. Methodist DeBakey Cardiovasc. J. 2012, 8, 6–12. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, L.; Xiong, Q.; Xiao, F.; Duan, H. 2D nanomaterials based electrochemical biosensors for cancer diagnosis. Biosens. Bioelectron. 2017, 89, 136–151. [Google Scholar] [CrossRef] [PubMed]
  50. Yamaguchi, M. Salivary sensors in point-of-care testing. Sens. Mater. 2010, 22, 143–153. [Google Scholar]
  51. Cone, E.J.; Clarke, J.; Tsanaclis, L. Prevalence and disposition of drugs of abuse and opioid treatment drugs in oral fluid. J. Anal. Toxicol. 2007, 31, 424–433. [Google Scholar] [CrossRef] [PubMed]
  52. Gau, V.; Wong, D. Oral fluid nanosensor test (OFNASET) with advanced electrochemical-based molecular analysis platform. Ann. N. Y. Acad. Sci. 2007, 1098, 401–410. [Google Scholar] [CrossRef] [PubMed]
  53. Kaczor-Urbanowicz, K.E.; Martin Carreras-Presas, C.; Aro, K.; Tu, M.; Garcia-Godoy, F.; Wong, D.T. Saliva diagnostics—Current views and directions. Exp. Biol. Med. 2016, 242, 459–472. [Google Scholar] [CrossRef] [PubMed]
  54. Chin, C.D.; Linder, V.; Sia, S.K.; Daar, A.S.; Thorsteinsdottir, H.; Martin, D.K. Lab-on-a-chip devices for global health: Past studies and future opportunities. Lab Chip 2007, 7, 41–57. [Google Scholar] [CrossRef] [PubMed]
  55. Silberring, J.; Ciborowski, P. Biomarker discovery and clinical proteomics. Trends Anal. Chem. 2010, 29, 128–140. [Google Scholar] [CrossRef] [PubMed]
  56. Ilyin, S.E.; Belkowski, S.M.; Plata-Salamán, C.R. Biomarker discovery and validation: Technologies and integrative approaches. Trends Biotechnol. 2004, 22, 411–416. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, A.; Wang, C.; Tu, M.; Wong, D. Oral biofluid biomarker research: Current status and emerging frontiers. Diagnostics 2016, 6, 45. [Google Scholar] [CrossRef] [PubMed]
  58. St. John, M.A.R.; Li, Y.; Zhou, X.; Denny, P.; Ho, C.M.C.M.; Montemagno, C.; Shi, W.; Qi, F.; Wu, B.; Sinha, U.; et al. Interleukin 6 and interleukin 8 as potential biomarkers for oral cavity and oropharyngeal squamous cell carcinoma. Arch. Otolaryngol. Head Neck Surg. 2004, 130, 929–935. [Google Scholar] [CrossRef] [PubMed]
  59. Landrum, M.L.; Wilson, C.H.; Perri, L.P.; Hannibal, S.L.; O’Connell, R.J. Usefulness of a rapid human immunodeficiency virus-1 antibody test for the management of occupational exposure to blood and body fluid. Infect. Control Hosp. Epidemiol. 2005, 26, 768–774. [Google Scholar] [CrossRef] [PubMed]
  60. Genco, R.J.; Grossi, S.G.; Ho, A.; Nishimura, F.; Murayama, Y. A proposed model linking inflammation to obesity, diabetes, and periodontal infections. J. Periodontol. 2005, 76, 2075–2084. [Google Scholar] [CrossRef] [PubMed]
  61. Huang, C.M. Comparative proteomic analysis of human whole saliva. Arch. Oral Biol. 2004, 49, 951–962. [Google Scholar] [CrossRef] [PubMed]
  62. Li, Y.; St. John, M.A.R.; Zhou, X.; Kim, Y.; Sinha, U.; Jordan, R.C.K.; Eisele, D.; Abemayor, E.; Elashoff, D.; Park, N.H.; et al. Salivary transcriptome diagnostics for oral cancer detection. Clin. Cancer Res. 2004, 10, 8442–8450. [Google Scholar] [CrossRef] [PubMed]
  63. Roberts, L.D.; Souza, A.L.; Gerszten, R.E.; Clish, C.B. Targeted metabolomics. Curr. Protoc. Mol. Biol. 2012. [Google Scholar] [CrossRef]
  64. Kageyama, G.; Saegusa, J.; Irino, Y.; Tanaka, S.; Tsuda, K.; Takahashi, S.; Sendo, S.; Morinobu, A. Metabolomics analysis of saliva from patients with primary Sjgren’s syndrome. Clin. Exp. Immunol. 2015, 182, 149–153. [Google Scholar] [CrossRef] [PubMed]
  65. Sugimoto, M.; Wong, D.T.; Hirayama, A.; Soga, T.; Tomita, M. Capillary electrophoresis mass spectrometry-based saliva metabolomics identified oral, breast and pancreatic cancer-specific profiles. Metabolomics 2010, 6, 78–95. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, C.; Li, J. Pathogenic microorganisms and pancreatic cancer. Gastrointest. Tumors 2015, 2, 41–47. [Google Scholar] [CrossRef] [PubMed]
  67. Hu, S.; Vissink, A.; Arellano, M.; Roozendaal, C.; Zhou, H.; Kallenberg, C.G.; Wong, D.T. Identification of autoantibody biomarkers for primary Sjogren’s syndrome using protein microarrays. Proteomics 2011, 11, 1499–1507. [Google Scholar] [CrossRef] [PubMed]
  68. Sakamoto, M.; Umeda, M.; Benno, Y. Molecular analysis of human oral microbiota. J. Periodontal Res. 2005, 40, 277–285. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, L.; Xiao, H.; Karlan, S.; Zhou, H.; Gross, J.; Elashoff, D.; Akin, D.; Yan, X.; Chia, D.; Karlan, B.; et al. Discovery and preclinical validation of salivary transcriptomic and proteomic biomarkers for the non-invasive detection of breast cancer. PLoS ONE 2010, 5, e15573. [Google Scholar] [CrossRef] [PubMed]
  70. Hu, Z.; Zimmermann, B.G.; Zhou, H.; Wang, J.; Henson, B.S.; Yu, W.; Elashoff, D.; Krupp, G.; Wong, D.T. Exon-level expression profiling: A comprehensive transcriptome analysis of oral fluids. Clin. Chem. 2008, 54, 824–832. [Google Scholar] [CrossRef] [PubMed]
  71. Idle, J.R.; Gonzalez, F.J. Metabolomics. Cell Metab. 2007, 6, 348–351. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, L.; Liu, F.; Fan, X.; Gao, J.; Chen, N.; Wong, T.; Wu, J.; Wen, S.W. Detection of hepatitis B surface antigen, hepatitis B core antigen, and hepatitis B virus DNA in parotid tissues. Int. J. Infect. Dis. 2009, 13, 20–23. [Google Scholar] [CrossRef] [PubMed]
  73. Nasidze, I.; Quinque, D.; Li, J.; Li, M.; Tang, K.; Stoneking, M. Comparative analysis of human saliva microbiome diversity by barcoded pyrosequencing and cloning approaches. Anal. Biochem. 2009, 391, 64–68. [Google Scholar] [CrossRef] [PubMed]
  74. Soper, S.A.; Brown, K.; Ellington, A.; Frazier, B.; Garcia-Manero, G.; Gau, V.; Gutman, S.I.; Hayes, D.F.; Korte, B.; Landers, J.L.; et al. Point-of-care biosensor systems for cancer diagnostics/prognostics. Biosens. Bioelectron. 2006, 21, 1932–1942. [Google Scholar] [CrossRef] [PubMed]
  75. Collings, A.F.; Caruso, F. Biosensors: Recent advances. Rep. Prog. Phys. 1997, 60, 1397–1445. [Google Scholar] [CrossRef]
  76. McRae, M.P.; Simmons, G.W.; Wong, J.; Shadfan, B.; Gopalkrishnan, S.; Christodoulides, N.; McDevitt, J.T. Programmable bio-nano-chip system: A flexible point-of-care platform for bioscience and clinical measurements. Lab Chip 2015, 15, 4020–4031. [Google Scholar] [CrossRef] [PubMed]
  77. Mehrotra, P. Biosensors and their applications—A review. J. Oral Biol. Craniofac. Res. 2016, 6, 153–159. [Google Scholar] [CrossRef] [PubMed]
  78. Lee, M.; Zine, N.; Baraket, A.; Zabala, M.; Campabadal, F.; Caruso, R.; Trivella, M.G.; Jaffrezic-Renault, N.; Errachid, A. A novel biosensor based on hafnium oxide: Application for early stage detection of human interleukin-10. Sens. Actuators B Chem. 2012, 175, 201–207. [Google Scholar] [CrossRef]
  79. Vasan, A.S.S. Point-of-care biosensor system. Front. Biosci. 2013, S5, S357. [Google Scholar] [CrossRef]
  80. Mastrangelo, C.H.; Burns, M.A.; Burke, D.T. Microfabricated devices for genetic diagnostics. Proc. IEEE 1998, 86, 1769–1786. [Google Scholar] [CrossRef]
  81. Gilleo, K. MEMS in medicine. Circuits Assem. 2005, 16, 32–33. [Google Scholar]
  82. Naher, S.; Orpen, D.; Brabazon, D.; Morshed, M.M. An overview of microfluidic mixing application. Adv. Mater. Res. 2009, 83–86, 931–939. [Google Scholar] [CrossRef]
  83. Hu, J.; Wang, S.Q.; Wang, L.; Li, F.; Pingguan-Murphy, B.; Lu, T.J.; Xu, F. Advances in paper-based point-of-care diagnostics. Biosens. Bioelectron. 2014, 54, 585–597. [Google Scholar] [CrossRef] [PubMed]
  84. Wei, F.; Yang, J.; Wong, D.T.W. Detection of exosomal biomarker by electric field-induced release and measurement (EFIRM). Biosens. Bioelectron. 2013, 44, 115–121. [Google Scholar] [CrossRef] [PubMed]
  85. Roda, A.; Michelini, E.; Zangheri, M.; Di Fusco, M.; Calabria, D.; Simoni, P. Smartphone-based biosensors: A critical review and perspectives. Trends Anal. Chem. 2016, 79, 317–325. [Google Scholar] [CrossRef]
  86. Wang, D.S.; Fan, S.K. Microfluidic surface plasmon resonance sensors: From principles to point-of-care applications. Sensors 2016, 16, 1175. [Google Scholar] [CrossRef] [PubMed]
  87. Nguyen, H.H.; Park, J.; Kang, S.; Kim, M. Surface plasmon resonance: A versatile technique for biosensor applications. Sensors 2015, 15, 10481–10510. [Google Scholar] [CrossRef] [PubMed]
  88. Lee, S.A.; Yang, C.A. Smartphone-based chip-scale microscope using ambient illumination. Lab Chip 2014, 14, 3056. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Depiction of the detection of various oral and systemic diseases through salivary biomarkers. Acquired Immuno Defeciency Syndrome (AIDS), Human Papilloma Virus (HPV), Myocardial infarction (MI), Chronic Obstructive Pulmonary Disease (COPD), Sexually Transmitted Infection (STI), End Stage Renal Failure (ESRD) and Oral Squamous Muccous Fibrosis (OSMF).
Figure 1. Depiction of the detection of various oral and systemic diseases through salivary biomarkers. Acquired Immuno Defeciency Syndrome (AIDS), Human Papilloma Virus (HPV), Myocardial infarction (MI), Chronic Obstructive Pulmonary Disease (COPD), Sexually Transmitted Infection (STI), End Stage Renal Failure (ESRD) and Oral Squamous Muccous Fibrosis (OSMF).
Diagnostics 07 00039 g001
Figure 2. Illustration of a range of promising emerging methods of PoC technologies.
Figure 2. Illustration of a range of promising emerging methods of PoC technologies.
Diagnostics 07 00039 g002
Table 1. Description of Point-of-care (PoC) devices for detection of diseases through specific salivary biomarkers.
Table 1. Description of Point-of-care (PoC) devices for detection of diseases through specific salivary biomarkers.
Salivary BiomarkersDiseases/ConditionsDeveloped PoCReferences
A-amylaseClinical judgment for stress-induced diseaseSalivary α-Amylase (sAA) biosensor system[33]
HIVAIDSOraquick, tablet-based kiosks[34,35]
Hep CHepatitisOraQuick[36]
HPVHPV-associated cancers, sexually transmitted diseasessimple fluorescent and colorimetric assay that enables DNA and RNA detection[37]
CortisolStress levelsLabel-free chemiresistor immuno-sensor[38]
Proteins (Dipeptidyl peptidase etc.), metabolites, DNAPeriodontitisIntegrated Microfluidic Platform for Oral Diagnostics (IMPOD), lab-on-a-chip (LOC)[39]
C-reactive protein, myoglobin, and myeloperoxidaseAcute Myocardial InfarctionLuminex, lab-on-a-chip methods[40]
CytokinesAsthma and chronic obstructive pulmonary disease (COPD)Fiber-optic microsphere-based antibody array[41]
IL-8, IL-8mRNAOral CancerElectrochemical magneto biosensors[42]
(NO2− and uric acid), and pulmonary inflammation biomarkersEnd-stage renal disease (ESRD), asthma and chronic obstructive pulmonary disease (COPD) patientsOptical fibre microarrays[43]
Salivary nicotine metabolitesSmoking/tobacco usePoint of care test for salivary nicotine metabolites[44]
Porphyromonas gingivalischronic periodontitisP. gingivalis saliva kit
Gonorrhoea and chlamydiaSexually transmitted infections (STIs)Oral STI point-of-care (PoC)[36]
Salivary anti-Ro60 and anti-Ro52 Antibody ProfilesSjögren’s SyndromeLuciferase Immunoprecipitation Systems (LIPS)[45]
Salivary glucoseDiabetesGlucose monitoring using saliva nanostructured biosensor[46,47]
cRP, MPo, ctnl, Myo, cK-MB, d-dimer, apoa1, apoB, BnP, nt-proBnP, scd40l, McP-1, adiponectinCardiovascular disease (CVD)Programmable bio-nanochip (P-BNC) system[48]
cea, ca125, Her2-neu, Psa (free and complexed)CancerProgrammable bio-nanochip (P-BNC) system, 2D nanomaterials[48,49]
Table 2. Methods used for the evaluation of diagnostic toolboxes.
Table 2. Methods used for the evaluation of diagnostic toolboxes.
Diagnostic ToolboxMethods of EvaluationMolecules to Be AnalysedReferences
ProteomicsMass spectroscopy and 2D gel electrophoresis, ELISA, protein immunoblot techniquesPost-translational modifications and protein-enzyme complexes[69]
Genomics (Transcriptomics and Epigenomics)Gene chip arrays, DNA hybridization, qPCR, and gel electrophoresisDNA, RNA and mRNA[70]
MetabolomicsNuclear magnetic resonance spectroscopy (NMR), gas chromatography-mass spectrometry, direct flow injection/liquid chromatography-mass spectrometry, inductively coupled plasma mass spectrometry, and high-performance liquid chromatography (HPLC), capillary electrophoresis time of flight mass spectroscopySmall molecules end products of metabolic processes in the body such as organic species, together with non-protein hormones (epinephrine, peptide hormones and cortisol).[71]
MicrobiomeBacterial microarrays, DNA hybridization, PCR, next-generation sequencing, and quantitative 16S rRNA gene sequencing, oligonucleotide microarray based on 16S rRNA, aptly named human-microbe identification microarrays (HOMIM)Bacterial species (Streptococcus, Staphylococcus)[68,72,73]
ImmunomicsImmunologic analysisImmunological markers (IgM, IgA, and IgG tests, and hepatitis B virus and hepatitis C virus, IgG)[67]
Table 3. Examples of single and multiplexed salivary biomarker-based PoC diagnostics [9].
Table 3. Examples of single and multiplexed salivary biomarker-based PoC diagnostics [9].
PoC PlatformsSystem UsedBiomarkerTest DurationRegion of Origin
SingleMicroelectromechanical technology (MEMS), optical fluorescent system followed by electrophoresisMatrix metalloproteinase-8 MMP10 minSandia National Lab (USA)
Oral risk indicator ORIMMP-8Less than 10 minDentognostics (Germany)
Chromatography test stripsHIV1&2, HCV, influenza20 minOrasure Technologies (USA)
Handheld deviceCortisol, a-amylase1 minNipro (Japan)
MultiplexedSalivary diagnosticsSalivary proteins and nucleic acidsLess than 15 minSDx (USA)
Salivary diagnosticsIL-8 mRNA, IL-8 proteinLess than 15 minSDx (USA)
Table 4. Variety of emerging PoC technologies and their clinical utility and functions.
Table 4. Variety of emerging PoC technologies and their clinical utility and functions.
Types of Emerging TechnologiesBiomarkers/Clinical UtilityTechnique UsedFunctionReferences
BiosensorsDiabetes mellitus, (glucose biosensors), UTI, cardiac markers, acute leukaemiasElectrochemicalDrug delivery, cardio MEMS to monitor heart patients, hearing aids, insulin micropumps, endoscopic pills, retinal prosthesis[79]
Fluorescent biosensors/FRET biosensorDrug discovery, arthritis, cancers, cardiovascular and neurodegenerative diseases, viral infections, chronic myeloid leukaemiaFluorescent probes are mounted through a receptorThey are able to probe gene expression, localisation of protein, signal transduction, transcription and cell cycle apoptosis[77]
Biological Microelectromechanical Systems (BioMEMS)Drug delivery, cardio MEMS, insulin micro pumps, endoscopic pills, retinal prosthesislab-on-a-chip systems/micro/nano-scale fabricationDetection of, proteins, viruses, DNA and microorganisms[80,81]
Microfluidics/paper based technologyStomach cancer biomarkers (H. pylori), detection of urine metabolites, blood glucose, pH value, liver function, infectious agentsOptoelectronic and microfluidic systemDNA extraction, polymerase chain reaction (PCR) amplification[83]
Electric field induced release and measurement EFIRMIL-8 protein and IL-8 mRNA markers for oral cancer, non-squamous cell lung cancer (NSCLC) oncogenic mutation, EGFR mutation in no small cell lung cancerElectrochemicalLiquid biopsy technique, selective hybridization[57,84]
Smartphone based biosensorsBlood samples of falciparum malaria infected and fluorescent images M. tuberculosis-positive sputum smears, self-monitoring of blood glucose, cancerMetal-oxide semiconductor (CMOS)-based photo cameras, optical-based methods including absorbance, chemiluminescence, fluorescence, reflectance, surface plasmon resonance (SPR), bio- and electrochemilumines-cenceDetector system for reflectance, colorimetry and luminescence[85]

Share and Cite

MDPI and ACS Style

Khan, R.S.; Khurshid, Z.; Yahya Ibrahim Asiri, F. Advancing Point-of-Care (PoC) Testing Using Human Saliva as Liquid Biopsy. Diagnostics 2017, 7, 39. https://doi.org/10.3390/diagnostics7030039

AMA Style

Khan RS, Khurshid Z, Yahya Ibrahim Asiri F. Advancing Point-of-Care (PoC) Testing Using Human Saliva as Liquid Biopsy. Diagnostics. 2017; 7(3):39. https://doi.org/10.3390/diagnostics7030039

Chicago/Turabian Style

Khan, Rabia Sannam, Zohaib Khurshid, and Faris Yahya Ibrahim Asiri. 2017. "Advancing Point-of-Care (PoC) Testing Using Human Saliva as Liquid Biopsy" Diagnostics 7, no. 3: 39. https://doi.org/10.3390/diagnostics7030039

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