C278 Luong Figure 1

Point-of-care testing: state-of-the-art and emerging trends

Point-of-care testing (POCT) enables quick test results with minimal manual interference nearer to the site of patient care, which leads to better health outcomes via rapid diagnosis, quick clinical decisions and the early start of treatment. The emerging technologies would further improve POCT by low-cost analysis with increased performance characteristics.

by Dr Sandeep Kumar Vashist and Prof. John H.T. Luong

Existing point-of-care testing (POCT) technologies
A variety of POCT technologies are being used such as POCT analysers, biosensor devices, lab-on-chips (LOC), test strips, and lateral flow assay (LFA) cartridges. Such cost-effective technologies offer rapid analysis in just a few minutes using minimal sample volumes. As well as at a patient’s bedside, POCT have been employed in the operating theatre, emergency department and critical care/maternity unit. Other deployments include nursing homes, physician’s office, prison, emergency vehicles, etc. Local pharmacies have adopted the POCT technology to provide a one-stop service for glucose, cholesterol, pregnancy, etc.

The POCT analysers are standard bench-top devices that can determine a broad range of analytes, based on spectrophotometry, reflectometry, immunoassay, turbidimetry, potentiometry/amperometry, oximetry and hematological particle counting. The target analytes are small metabolites, enzymes, drugs-of-abuse, inflammation biomarkers, heart and kidney injury biomarkers, infectious agents, humoral and cellular coagulation markers, hematological parameters, etc.

The biosensor-based devices, notably the blood glucose meters, are the most common POCT technology, which have been widely used for the detection of glucose. Different LOC platforms are being used in various POCT technologies. They are fully automated platforms that integrate all microfluidics-based bioanalytical steps such as sample treatment, separation, biomolecular detection, washing, signal detection, and data processing, storage, and transmission. The signal detection in most LOC platforms employs optical readout. The technologies and materials used for the production of LOC platforms have been fully characterized and standardized.

Test strips are another prominent POCT technology for detecting different analytes in a patient’s blood or urine sample. They are easy to use and easy to read, leading  to immediate on-the-spot analysis. The test strip comprises a solid support onto which porous matrices with dried assay reagents are integrated. The reaction starts as the biorecognition element present on the test strip detects the analyte, which leads to a visual change in colour on the test strip. The signal is read by inserting the test strip into a reader device.

LFA, one of the most widely used POCT technologies, is based on an immunochromatography format where the assay reagents are stored in dried form on various porous materials. Once the sample (urine or diluted blood) is dispensed at the designated area of the LFA cartridge, the sample flows in the lateral direction by capillary forces. It first interacts with the capture antibodies spotted at the reaction area leading to the formation of the immune complex, which is followed by binding to the detection antibodies immobilized at another area of the cartridge, thereby resulting in the formation of the sandwich immune complex. This leads to a visible colour change in the test and control lines, which facilitates rapid qualitative or semi-quantitative analysis. The POC pregnancy testing is done exclusively by LFA.

The multiplex analysis using DNA and protein microarrays is also being intensively investigated although the technology has not yet been commercialized for clinical POCT. It is envisaged that this upcoming technology would be fully automated, which would involve advanced microfluidics and the signal readout by electrochemical, chemiluminescence, fluorescence or evanescent wave techniques.

Emerging POCT technologies
Various POCT technologies have emerged during the last decade, which have tremendous potential for next-generation healthcare monitoring and management [1]. In 2011, the estimated total POCT market was US$15 billion and projected to reach US$18 billion by 2016. Of the total POCT market in 2011, 55% of it was in the US market, followed by 30% in Europe and 12% in Asia [2].

Cellphone (CP)-based devices
The most prospective emerging technology is CP-based devices. CPs have become ubiquitous with more than 7 billion global users that account for more than 95% of the world’s population. Moreover, about 70% of the CP users reside in the developing countries, where there is an imminent need for mobile healthcare (mH). The current generation of CPs are cost-effective and equipped with all the desired advanced features that facilitate personalized mH monitoring and management. The spatiotemporal tagging of the data by CP enables the real-time active response to epidemics and emergency situations. Various FDA approved and CE certified CP-based personalized healthcare devices have already been commercialized for the monitoring of basic physiological parameters such as blood glucose, blood pressure, pulse rate, blood oxygen saturation, body weight, body analysis parameters, electrocardiogram, physical activity, sleep and cardiac parameters (Fig. 1) [3]. Most of such commercial CP-based devices are developed by iHealth Labs, France. Similarly, CP-based technologies have been prepared for many POCT applications [4]. Cellmic, USA has developed a compact CP-based rapid-diagnostic-test reader for the readout of colorimetric and fluorometric LFA (Fig. 2). Of notice is the conversion of the CP into a compact and lightweight computational microscope for bright field, fluorescence, darkfield, transmission and polarized microscopy modes. Moreover, a CP-based flow cytometer based on optofluidic fluorescent imaging enabled the screening of pathogens in whole blood or water samples [5]. Another similar endeavour is the development of smartphone-based spectrophotometers for the detection of absorbance, fluorescent or chemiluminescent signals [6]. Various CP-based colorimetric readers [7], electrochemical sensing platform [8], angle-resolved surface plasmon resonance (SPR) system [9], and multiplex assays have also been developed [10].

Paper-based diagnostics
Considering simplicity and cost-effectiveness, paper-based diagnostics (PBD) are available in various formats: LFA, dipstick and microfluidic paper-based analytical devices (µPADs) [11, 12]. LFA are widely used in home pregnancy test strips to detect human chorionic gonadotropin in urine. It employs the dispensing of the sample onto the sample pad of the LFA test strip (fabricated from a nitrocellulose membrane). The sample flows laterally over a conjugate pad due to the capillary action provided by the absorbent pad, which leads to the binding of the analyte to conjugate particles (gold nanoparticles and upconversion nanoparticles). The signal detection can be visual, colorimetric, electrochemical, photoelectrochemical, chemiluminescent and electrochemiluminescent. The colorimetric PBD provide qualitative analysis by comparing the colour against a predetermined score chart. But the colour intensity can also be quantified using cameras, scanners, commercial test strip readers and hand-held colorimeters. A CP-based rapid-diagnostic-test reader is the most recent development that enables precise determination of colour intensity [13].

Paper can be patterned to fabricate two-dimensional (2D) or three-dimensional (3D) µPADs. The 3D µPADs, formed by stacking layers of the 2D paper, are ideal for multiplexing. The microfluidic paper-based electrochemical devices (µPEDs) can be fabricated by printing electrodes on paper.

The sensitivity of PBDs can be increased by employing enzymes and nanomaterials based signal enhancement strategies, which increases the costs and assay duration, and decreases the shelf-life. However, PBDs suffer from poor reproducibility, non-uniformity and variable accuracy on µPADs due to the passive capillary transport in paper substrates.

Lab-on-a-chip platforms
Various LOC platforms, such as the most widely used blood glucose testing strips, have been made for POCT. These platforms enable fully automated analysis by integrating all process steps in the operational procedure. The Piccolo Xpress™ whole blood chemistry analyser, developed by Abaxis Inc, is a prospective LOC-based POCT device that performs 14 tests on a single reagent LabDisk (8 cm diameter, barcoded).

Rapid assay formats

A prospective assay format is Optimiser™ ELISA [14] by Siloam Biosciences Inc,  which employs a novel microfluidic microtiter plate. It detects an analyte in just a few minutes using minimal reagent volumes and least number of steps. Other prospective formats are the wash-free AlphaLISA® by Perkin Elmer, wash-free electrochemiluminescent ELISA by Meso Scale Diagnostics LLC, rapid one-step kinetics-based formats [15], CP-based easy immunoassay platforms [16], and COBAS® Lab-in-a-Tube (LIAT) system by Roche Diagnostics.

Prolonged reagent storage strategies
The most prospective prolonged reagent storage strategies are the use of polysaccharides and saccharides (such as pullulan and trehalose), sugar alcohols, stabilizers, freeze drying, lyophilization, and reagent pouches. Use of POCT in remote areas, particularly in developing countries might be problematic due to inconsistent electrical power, lighting, and refrigeration.

Conclusions
The next decade will witness the revolutionary breakthroughs in POCT that will drastically cut down the costs and lead to considerably improved analysis with increased bioanalytical performance and capabilities. Multi-channel, high-throughput instruments are expected to expand the new concept of POCT, and there is an obvious need for the combination of POCT technology and communication technology. Non-invasive blood glucose monitoring remains the most important market for POCT, followed by coagulation, blood gas, chemistry, hematology, urinalysis, and cardiac. Also, molecular POCT technology has matured and began to move toward commercialization.

Future trends for POCT will rise due to new emerging technologies to make them well suited for low-resource or remote areas. POCT vendors increasingly offer more available types of tests with improved accuracy and minimal turnaround times. Thus, POCT will be more widely accepted as an addition to the current method of managing patients. Considering its limited test menus, clinicians still have to wait for other sophisticated laboratory tests before the action can be taken for treatment or further testing [17]. POCT evolves continuously and becomes an ‘extension’ of laboratory services, not a replacement of routine core laboratory testing.

References
1. Vashist SK, Luppa PB, Yeo LY, Ozcan A, Luong JHT. Emerging technologies for next-generation point-of-care testing. Trends Biotechnol. 2015; 33(11): 692–705.
2. Scientia Advisors. The point-of-care diagnostics market. 2013, Cambridge, MA, USA. http://www.businesswire.com/news/home/20100915005199/en/Scientia-Advisors-Projects-Sustainable-Growth-Point-of-Care
3. Vashist S, Schneider E, Luong JHT. Commercial smartphone-based devices and smart applications for personalized healthcare monitoring and management. Diagnostics 2014; 4(3): 104–128.
4. Vashist SK, Mudanyali O, Schneider EM, Zengerle R, Ozcan A. Cellphone-based devices for bioanalytical sciences. Anal Bioanal Chem. 2014; 406(14): 3263–3277.
5. Zhu H, Mavandadi S, Coskun AF, Yaglidere O, Ozcan A. Optofluidic fluorescent imaging cytometry on a cell phone. Anal Chem. 2011; 83(17): 6641–6647.
6. Yu H, Tan Y, Cunningham BT. Smartphone fluorescence spectroscopy. Anal Chem. 2014; 86(17): 8805–8813.
7. Vashist SK, van Oordt T, Schneider EM, Zengerle R, von Stetten F, Luong JHT. A smartphone-based colorimetric reader for bioanalytical applications using the screen-based bottom illumination provided by gadgets. Biosens Bioelectron. 2015; 67: 248–255.
8. Lillehoj PB, Huang MC, Truong N, Ho CM. Rapid electrochemical detection on a mobile phone. Lab Chip. 2013; 13(15): 2950–2955.
9. Preechaburana P, Gonzalez MC, Suska A, Filippini D. Surface plasmon resonance chemical sensing on cell phones. Angew Chem Int Ed Engl. 2012; 51(46): 11585–11588.
10. Laksanasopin T, Guo TW, Nayak S, Sridhara AA, Xie S, Olowookere OO, Cadinu P, Meng F, Chee NH, et al. A smartphone dongle for diagnosis of infectious diseases at the point of care. Sci Transl Med. 2015; 7(273): 273re1.
11. Mao X, Huang TJ. Microfluidic diagnostics for the developing world. Lab Chip 2012; 12(8): 1412–1416.
12. Li X, Ballerini DR, Shen W. A perspective on paper-based microfluidics: current status and future trends. Biomicrofluidics 2012; 6(1): 11301–11301-13.
13. Mudanyali O, Dimitrov S, Sikora U, Padmanabhan S, Navruz I, Ozcan A. Integrated rapid-diagnostic-test reader platform on a cellphone. Lab Chip 2012; 12(15): 2678–2686.
14. Kai J, Puntambekar A, Santiago N, Lee SH, Sehy DW, Moore V, Han J, Ahn CH. A novel microfluidic microplate as the next generation assay platform for enzyme linked immunoassays (ELISA). Lab Chip 2012; 12(21): 4257–4262.
15. Vashist SK, Czilwik G, van Oordt T, von Stetten F, Zengerle R, Marion Schneider E, Luong JHT. One-step kinetics-based immunoassay for the highly sensitive detection of C-reactive protein in less than 30min. Anal Biochem. 2014; 456: 32–37.
16. Vashist SK, Czilwik G, Venkatesh AG. Elisa system and related methods. WIPO Patent Pub No WO/2014/198836; 2014.
17. Boonlert W, Lolekha PH, Kost GJ, Lolekha S. Comparison of the performance of point-of-care and device analyzers to hospital laboratory instruments. Point of Care 2003; 2(3): 172–178.

The authors
Sandeep Kumar Vashist*1 PhD, John H.T. Luong2 PhD
1Vallo Med Health Care GmbH, Castrop-Rauxel, Germany
2Innovative Chromatography Group, Irish Separation Science Cluster (ISSC), Department of Chemistry and Analytical, Biological Chemistry Research Facility (ABCRF), University College Cork, Cork, Ireland

*Corresponding author
E-mail: sandeep.vashist@vallomed.com

C276 Grewal Jenum Figure 050916

Recent progress in host transcriptome profiling paves the way for a future point-of-care test for pediatric TB

A diagnostic point-of care (POC) test would greatly improve case detection and management of pediatric tuberculosis (TB). Herein, we provide a brief overview of the challenges associated with diagnosing TB in children and recent evidence that points towards a future POC test based on transcriptomes in peripheral blood.

by Dr S. Jenum, Dr J.E. Gjøen, R. Bakken, Dr D. Sivakumaran and Prof. H.M.S. Grewal

Introduction
Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb), is a leading cause of childhood death and morbidity worldwide estimated to cause 1 million new cases in children <15 years of age in 2014 [1]. The gold standard for a diagnosis of pulmonary TB is growth of Mtb from respiratory specimens. However, culture-confirmed TB only occurs in about 30% of pediatric TB cases because of the paucibacillary nature of the disease and the difficulties in obtaining representative specimens [2]. For these reasons, children have been perceived less contagious and, therefore, not considered a priority for case finding within national TB programmes. This has resulted in detection rates being as low as 35% [3], with evident consequences for childhood death and morbidity. An accurate diagnostic test that provides a rapid, sensitive and specific result, enabling early initiation of treatment at the point-of-care (POC), ‘a diagnostic POC test’, could prevent this. The declaration by the World Health Organization (WHO) of pediatric TB as a neglected area of science and the stated urgent need for improved diagnostics [4], has spurred the search for TB diagnostic biomarkers in children. We provide a brief overview of the challenges associated with diagnosing TB in children and recent evidence that points towards a future POC test based on transcriptomes in peripheral blood.

Diagnosing TB in children
Diagnostic approaches and criteria for pediatric TB have recently been extensively reviewed [5]. In summary, sampling of respiratory specimens for confirmation of diagnosis by culture is strongly recommended, but requires repeated sampling of induced sputum and gastric aspirates, which are cost and labour intensive [5]. Notably, as many as 70% of children are culture negative even under optimized study conditions [2]. Therefore, in most cases, a diagnosis is based on a diagnostic algorithm including clinical signs and symptoms, radiological findings suggestive of TB and immunological evidence of previous Mtb sensitization [5]. Clinical diagnostic algorithms have not being adequately validated [6], and radiological findings are often unspecific [5]. The tuberculin skin test (TST) and the newer interferon-γ release assays used as a surrogate for Mtb infection cannot differentiate between infection and disease [7], and have reduced sensitivity in young and malnourished children [8, 9], but may be complementary by improving sensitivity in such clinical contexts [5].

Requirements for a POC test
Direct microscopy of sputum smears looking for acid-fast bacilli, is widely implemented at primary diagnostic centres for the diagnosis of pulmonary TB in adults, but the sensitivity is highly variable (20–80%) [10]. Since 2010, the use of the Xpert MTB/RIF assay on sputum samples has expanded in low- and middle-income countries [11], serving as a POC test in adults. The assay is based on direct identification of Mtb in specimens by a nucleic acid amplification test and detects in children three times the cases detected by direct microscopy and ~70% of the cases detected by liquid culture [12]. The WHO recommends that a POC triage test, meant to narrow down the population that needs further diagnostic work-up, should achieve a sensitivity of 90% and a specificity of 70%. For a diagnostic POC test, the sensitivity should be >95% and the specificity >98% [13]. A POC test based on Mtb detection will never meet these requirements in children because of the high proportion of culture-negative pulmonary TB [2], as well as the significant proportion of extrapulmonary TB [5]. Host biomarkers reflecting the ongoing pathological processes resulting from Mtb infection may hold greater promise for this purpose. Moreover, peripheral whole blood (WB) is more easily available for diagnosis than respiratory or tissue samples, particularly in children. Therefore, biomarkers based on transcriptomes in WB have gained increasing interest.

Genome-wide analysis of RNA expression
A landmark study by Berry et al., based on genome-wide analysis of RNA expression in unstimulated WB from adults, identified an 86-transcript signature able to discriminate active TB from other inflammatory and infectious diseases [14]. This transcriptomic signature consisted mainly of neutrophil-driven interferon (IFN)-inducible genes, which until then, had not been considered essential players in TB immunity. Further, a decreased abundance of B-cell transcripts in TB patients [14], challenged the paradigm of B-cells and humoral immunity being of little importance in protection against Mtb. Following this study, the upregulation of type I IFN-inducible genes and altered expression of B-cell related genes has been confirmed in adults [15–19] and adolescents [20].

To our knowledge, two previous studies in children have been published based on genome-wide transcriptional profiling of WB in analysis of RNA expression in Warao Amerindians [21], and in three African cohorts [22]. Verhagen et al. addressed the diagnostic gap in discriminating TB disease from latent TB, and identified a 5-transcript signature with a prediction error of 11% in discriminating active from latent TB. The signature classified correctly 78% of TB cases and 100% of latent TB cases in a validation cohort comprising children with non-TB pneumonia [21]. Interestingly, the 5-transcript signature, when applied on transcriptional data-sets generated by microarray analyses of WB from adults performed similarly, whereas the gene signatures identified in these adults were useless in the cohort of Warao Amerindian children [21]. This questions the validity in extrapolating results from transcriptional TB biomarker research in adults to children, highlighting the importance of studies in pediatric populations.

Applying a more real-life diagnostic setting with inclusion of children evaluated for suspected TB applying a graded diagnosis of TB in line with the consensus statement [6], Anderson et al. identified a 51-transcript signature that distinguished TB from other diseases in South African and Malawian children, subsequently validated in Kenyan children. A risk score based on the signature identified confirmed TB with a sensitivity of 82.9% and a specificity of 83.6%, and discriminated between other disease and culture-negative TB with a sensitivity ranging from 35–82% [22].

A more cost-effective method for assessing transcriptomes
Genome-wide analyses of transcriptomes are costly and resource-intensive but can be seen as a necessary first step in identifying markers with potential for subsequent refinement as POC tests. A test more suitable for hypothesis-testing or more high-throughput screening of transcriptional signatures with relevance in TB, has recently been developed [23]. The dual-colour Reverse Transcriptase-Multiplex Ligation-dependent Probe Amplification (dcRT-MLPA) can rapidly profile multiple host genes with a dynamic range and accuracy comparable to real-time qPCR and RNA sequencing at a cost of €5–7 per sample [24], most likely simplifying the translation of findings into diagnostic tools in resource-poor settings. The dcRT-MLPA method has been applied in different studies assessing TB biomarkers [19, 23–25]. Recently, incorporating the results from unbiased gene-expression profiling, the gene panels adapted for the dcRT-MLPA platform have been expanded to include type I IFN-inducible genes as well as genes covering adaptive immunity, including B-cells [25].

The dcRT-MLPA method in the context of pediatric TB
We have applied the dcRT-MLPA method to identify TB diagnostic transcriptome signatures in pediatric TB settings in India [26, 27]. We have also examined the immunological basis for discordant TST and Quantiferon® responses [28], as well as the potential interference of non-tuberculous mycobacteria on transcriptional biomarkers [29]. The younger the child, the more challenging the diagnosis of TB. We therefore explored transcriptional biomarkers in Indian children aged <3 years referred for a TB diagnostic work-up in a prospective cohort study of BCG-vaccinated neonates. We found that RAB33A alone discriminated between clinical TB and Mtb infection with an area under the curve (AUC) of 77.5%, whereas a 5-transcript signature effectively discriminated between clinical TB and controls (AUC 91.7%) [26]. Then, in a cohort of older Indian children (mean age ~9 years) with intrathoracic TB and their siblings, we assessed transcriptional biomarkers across the spectrum of TB disease. We identified 12 biomarkers consistently associated with clinical groups ‘upstream’ towards confirmed TB or ‘downstream’ towards a decreased likelihood of TB disease (Fig. 1), suggesting a correlation with Mtb-related pathology and high relevance to a future POC test. Furthermore, an 8-transcript signature separated children with TB from asymptomatic siblings (AUC 88%) [27]. Notably, these transcriptome signatures provided a superior sensitivity for confirmed TB compared with the Xpert MTB/RIF performed on gastric lavage [2]. Many of the biomarkers within these signatures have been confirmed in adults with Mtb infection or disease [19, 23, 24].

Future perspectives
We are currently exploring the performance of the extended dcRT-MLPA panels of genes which include type I IFN-signalling, myeloid cell activation, general inflammation and B-cell related genes in Indian children with intrathoracic TB. Transcriptome signatures identified in a discovery set consisting of randomly assigned TB cases and their healthy siblings will be validated in a diagnostic setting of symptomatic children aged <3 years. We are also exploring the association between transcriptional biomarkers and the extent of TB disease and the outcome of TB treatment in children. The monitoring and evaluation of TB treatment is hampered by the limited tools available to guide decision-making during the long treatment period. Based on our unpublished results, as well as from findings in adults [16, 17, 23] we believe that treatment monitoring by means other than a negative culture result at 2 months post-treatment [30] will be possible. Conclusions
Transcriptome-based tests meeting the requirements for both a screening and a diagnostic POC test would greatly improve case detection in children and subsequently reduce morbidity and mortality attributable to TB [13]. Further, a POC test capable of guiding treatment would prevent treatment failure and the subsequent emergence of resistant Mtb strains.

References
1. WHO. Global tuberculosis report 2015. ISBN 978-9241565059. http://apps.who.int/iris/bitstream/10665/191102/1/9789241565059_eng.pdf.
2. Dodd LE, Wilkinson RJ. Diagnosis of paediatric tuberculosis: the culture conundrum. Lancet Infect Dis. 2013; 13(1): 3–4.
3. Dodd PJ, Gardiner E, Coghlan R, Seddon JA. Burden of childhood tuberculosis in 22 high-burden countries: a mathematical modelling study. Lancet Glob Health 2014; 2(8): e453–459.
4. WHO. Roadmap for childhood tube culosis: towards zero deaths. WHO 2013;http://apps.who.int/iris bitst eam/10665/89506/1/9789241506137_eng pdf.
5. Perez-Velez CM, Marais BJ. Tuberculosis in children. N Engl J Med. 2012; 367(4): 348–361.
6. Graham SM, Ahmed T, Amanullah F, Browning R, Cardenas V, Casenghi M, Cuevas LE, Gale M, Gie RP, et al. Evaluation of tuberculosis diagnostics in children: 1. Proposed clinical case definitions for classification of intrathoracic tuberculosis disease. Consensus from an expert panel. J Infect Dis. 2012; 205 Suppl 2: S199–S208.
7. Pai M, Denkinger CM, Kik SV, Rangaka MX, Zwerling A, Oxlade O, Metcalfe JZ, Cattamanchi A, Dowdy DW, et al. Gamma interferon release assays for detection of Mycobacterium tuberculosis infection. Clin Microbiol Rev. 2014; 27(1): 3–20.
8. Machingaidze S, Wiysonge CS, Gonzalez-Angulo Y, Hatherill M, Moyo S, Hanekom W, Mahomed H. The utility of an interferon gamma release assay for diagnosis of latent tuberculosis infection and disease in children: a systematic review and meta-analysis. Pediatr Infect Dis J. 2011; 30(8): 694–700.
9. Jenum S, Selvam S, Mahelai D, Jesuraj N, Cardenas V, Kenneth J, Hesseling AC, Doherty TM, Vaz M, Grewal HM. Influence of age and nutritional status on the performance of the tuberculin skin test and QuantiFERON-TB gold in-tube in young children evaluated for tuberculosis in Southern India. Pediatr Infect Dis J. 2014; 33(10): e260–269.
10. WHO. Report of the Tenth Meeting of the Strategic and Technical Advisory Group for Tuberculosis (STAG-TB) 2010. WHO 2010;  http://www.who.int/tb/advisory_bodies/stag_tb_report_2010.pdf.
11. Qin ZZ, Pai M, Van GW, Sahu S, Ghiasi M, Creswell J. How is Xpert MTB/RIF being implemented in 22 high tuberculosis burden countries? Eur Respir J. 2015; 45(2): 549–554.
12. Nicol MP, Workman L, Isaacs W, Munro J, Black F, Eley B, Boehme CC, Zemanay W, Zar HJ. Accuracy of the Xpert MTB/RIF test for the diagnosis of pulmonary tuberculosis in children admitted to hospital in Cape Town, South Africa: a descriptive study. Lancet Infect Dis. 2011; 11(11): 819–824.
13. WHO. Global Tuberculosis Report. WHO 2014;  http://apps.who.int/iris/bitst eam/10665/137094/1/9789241564809_eng.pdf?ua=1.
14. Berry MP, Graham CM, McNab FW, Xu Z, Bloch SA, Oni T, Wilkinson KA, Banchereau R, Skinner J, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature 2010; 466(7309): 973–977.
15. Ottenhoff TH, Dass RH, Yang N, Zhang MM, Wong HE, Sahiratmadja E, Khor CC, Alisjahbana B, van Crevel R, et al. Genome-wide expression profiling identifies type 1 interferon response pathways in active tuberculosis. PLoS One 2012; 7(9): e45839.
16. Bloom CI, Graham CM, Berry MP, Wilkinson KA, Oni T, Rozakeas F, Xu Z, Rossello-Urgell J, Chaussabel D, et al. Detectable changes in the blood transcriptome are present after two weeks of antituberculosis therapy. PLoS One 2012; 7(10): e46191.
17. Cliff JM, Lee JS, Constantinou N, Cho JE, Clark TG, Ronacher K, King EC, Lukey PT, Duncan K, et al. Distinct phases of blood gene expression pattern through tuberculosis treatment reflect modulation of the humoral immune response. J Infect Dis. 2013; 207(1): 18–29.
18. Kaforou M, Wright VJ, Oni T, French N, Anderson ST, Bangani N, Banwell CM, Brent AJ, Crampin AC, et al. Detection of tuberculosis in HIV-infected and -uninfected African adults using whole blood RNA expression signatures: a case-control study. PLoS Med. 2013; 10(10): e1001538.
19. Sutherland JS, Loxton AG, Haks MC, Kassa D, Ambrose L, Lee JS, Ran L, van Baarle D, Maertzdorf J, et al. Differential gene expression of activating Fcγ receptor classifies active tuberculosis regardless of human immunodeficiency virus status or ethnicity. Clin Microbiol Infect. 2014; 20(4): O230–238.
20. Zak DE, Penn-Nicholson A, Scriba TJ, Thompson E, Suliman S, Amon LM, Mahomed H, Erasmus M, Whatney W, et al. A blood RNA signature for tuberculosis disease risk: a prospective cohort study. Lancet 2016; 387(10035): 2312–2322.
21. Verhagen LM, Zomer A, Maes M, Villalba JA, Del Nogal B, Eleveld M, van Hijum SA, de Waard JH, Hermans PW. A predictive signature gene set for discriminating active from latent tuberculosis in Warao Amerindian children. BMC Genomics 2013; 14: 74.
22. Anderson ST, Kaforou M, Brent AJ, Wright VJ, Banwell CM, Chagaluka G, Crampin AC, Dockrell HM, French N, et al. Diagnosis of childhood tuberculosis and host RNA expression in Africa. New Eng J Med. 2014; 370(18): 1712–1723.
23. Joosten SA, Goeman JJ, Sutherland JS, Opmeer L, de Boer KG, Jacobsen M, Kaufmann SH, Finos L, Magis-Escurra C, et al. Identification of biomarkers for tuberculosis disease using a novel dual-color RT-MLPA assay. Genes Immun. 2012; 13(1): 71–82.
24. Haks MC, Goeman JJ, Magis-Escurra C, Ottenhoff TH. Focused human gene expression profiling using dual-color reverse transcriptase multiplex ligation-dependent probe amplification. Vaccine 2015; 33(40): 5282–5288.
25. Sloot R, Schim van der Loeff MF, van Zwet EW, Haks MC, Keizer ST, Scholing M, Ottenhoff TH, Borgdorff MW, Joosten SA. Biomarkers can identify pulmonary tuberculosis in HIV-infected drug users months prior to clinical diagnosis. EBioMedicine 2015; 2(2): 172–179.
26. Dhanasekaran S, Jenum S, Stavrum R, Ritz C, Faurholt-Jepsen D, Kenneth J, Vaz M, Grewal HM, Doherty TM; TB Trials Study Group. Identification of biomarkers for Mycobacterium tuberculosis infection and disease in BCG-vaccinated young children in Southern India. Genes Immun. 2013; 14(6): 356–364.
27. Jenum S, Dhanasekaran S, Lodha R, Mukherjee A, Kumar Saini D, Singh S, Singh V, Medigeshi G, Haks MC, et al. Approaching a diagnostic point-of-care test for pediatric tuberculosis through evaluation of immune biomarkers across the clinical disease spectrum. Sci Rep. 2016; 6: 18520.
28. Dhanasekaran S, Jenum S, Stavrum R, Ritz C, Kenneth J, Vaz M, Doherty TM, Grewal HM; TB Trials Study Group. Concordant or discordant results by the tuberculin skin test and the quantiFERON-TB test in children reflect immune biomarker profiles. Genes Immun. 2014; 15(5): 265–274.
29. Dhanasekaran S, Jenum S, Stavrum R, Wiker HG, Kenneth J, Vaz M, Doherty TM, Grewal HM; TB Trials Study Group. Effect of non-tuberculous Mycobacteria on host biomarkers potentially relevant for tuberculosis management. PLoS Negl Trop Dis. 2014; 8(10): e3243.
30. Wallis RS, Kim P, Cole S, Hanna D, Andrade BB, Maeurer M, et al. Tuberculosis biomarkers discovery: developments, needs, and challenges. Lancet Infect Dis. 2013; 13(4): 362–372.

The authors
Synne Jenum*1 MD, PhD; John Espen Gjøen2 MD, Rasmus Bakken2; Dhanasekaran Sivakumaran2 PhD; Harleen M.S. Grewal2 MD, PhD, DTMH
1Department of Infectious Diseases, Oslo University Hospital, Pb 4950, N-0424 Oslo, Norway
2Department of Clinical Science, University of Bergen, Pb 7804, N-5021 Bergen, Norway

*Corresponding author
E-mail: synnejenum@gmail.com

C264 Goswami Figure 1 BW

Molecular diagnostics from stained cytology smears

Molecular testing is increasingly recognized for guiding patient management and development of new targeted therapies. Meanwhile, there is an increased demand to perform testing on smaller volumes of tissue. Recent literature reports the use of ultrasensitive techniques to detect DNA mutations and translocations from stained cytology smears [1].

by Dr Laleh Hakima, Dr Maja H. Oktay and Prof. Sumanta Goswami

Introduction
Mutational analyses are crucial for guiding treatment decisions. This is particularly important for targeted therapies with tyrosine kinase inhibitors (TKI) for lung non-small cell carcinoma (NSCC) and for surgical management of thyroid nodules with indeterminate cytological diagnoses. Both lung and thyroid lesions are frequently diagnosed using fine need aspiration (FNA). FNA is a preferred method of obtaining diagnostic samples compared to surgical excisions or core biopsies. The FNA procedure is minimally invasive, rapid, cost-effective and has reduced procedure-related complications.

Molecular diagnostic tests of cytological samples are often performed using cytology cell block preparations. However, insufficient cellularity of cell blocks is a frequent limitation to these tests. In addition, errors introduced by formalin fixation may also interfere with accurate detection of mutations. Recent studies have reported successful testing using cytology smears stained with Diff-Quik®, which are air dried and fixed in alcohol. This approach has several advantages compared to testing on paraffin-embedded tissue (PET). Alcohol is a great preservative for DNA and RNA. The sample quality is superior to PET because FNA samples are typically enriched for cancer cells and cytology smears yield intact whole nuclei rather than the nuclear fragments that are obtained by cutting PET [2]. Lastly, testing performed directly on microdissected tumour cells ensures that material is sufficient and representative of the tumour and results in a faster turnaround time. We used cytological direct smears stained with Diff-Quik® and Papanicolaou (Pap) to detect commonly encountered mutations in non-small cell lung carcinoma and thyroid carcinoma [1].

Lung- and thyroid-carcinoma mutations and fusion gene testing targets
Approximately 64% of all lung adenocarcinomas harbour somatic driver mutations. According to The Lung Cancer Mutation Consortium, the frequency of EGFR and KRAS mutations are 23% and 25%, respectively. The incidence of EML4–ALK translocation, mainly detected in non-smoker patients with wild-type EGFR and KRAS genes, is approximately 6% [2, 3]. Other less frequent mutations include: BRAF ~3%, PIK3CA ~3%, MET amplifications ~2%, ERBB2 (HER2/NEU) ~1%, MAP2K1 ~0.4%, and NRAS ~0.2% [2]. Additionally, new driver mutations have recently been identified in lung cancer patients [3]. The most common mutations in thyroid carcinoma involve BRAF, KRAS, and RET/PTC genes. The mutations in BRAF and KRAS are point mutations, whereas RET (PTC) mutations are gene rearrangements that result in fusion of the tyrosine kinase domain of the RET gene to various unrelated genes [4–7].

The EGFR mutation status of the cancer is associated with its responsiveness or resistance to EGFR TKI therapy. The EGFR gene is located on chromosome 7p11.2, spans about 200kb, and contains 28 exons. The gene encodes a protein of 464 amino acids. The EGFR protein is composed of an N-terminal extracellular ligand-binding domain, a transmembrane lipophilic segment, and a C-terminal intracellular region containing a tyrosine kinase domain. The EGFR tyrosine kinase modulates cell proliferation and survival. Activation of EGFR initiates signalling cascades involving several downstream pathways, including Ras GTPases, which induce crucial cellular responses, such as proliferation, differentiation, motility, and survival. EGFR mutations associated with objective responses to single-agent TKI therapy in lung adenocarcinomas are preferentially observed in females of East Asian ethnicity who are never smokers and have adenocarcinoma with lepidic growth pattern (formerly bronchioloalveolar carcinoma). In adenocarcinomas, the majority of mutations have been identified in exons 18–21 of the EGFR gene. These mutations can be roughly classified into three major categories: in-frame deletions in exon 19, insertion mutations in exon 20, and missense mutations in exons 18–21 [8].

The fusion of the echinoderm microtubule-associated protein-like 4 (EML4) gene to the anaplastic lymphoma kinase (ALK) gene, EML4–ALK, is the most common fusion and results from the joining of exons 1–13 of EML4 to exons 20–29 of ALK. At least seven EML4–ALK variants (V1–V7) have been identified in lung adenocarcinomas. All seven variants are formed through the fusion of the intracellular tyrosine kinase domain of ALK with a variably truncated EML4 gene promoter. Activated ALK is involved in the inhibition of apoptosis and the promotion of cellular proliferation through activation of downstream PI3K/AKT1- and MAPK1-signalling pathways. The key downstream effectors on the ALK pathway include the Ras-activated protein, mitogen-activated protein kinase 1 [MAPK1; also known as extracellular signal regulated kinase (ERK)], phosphatidylinositol 3-kinase (PI3K), and signal transducer and activator of transcription 3 (STAT3) signalling pathways. Ras/MAPKK1/MAPK1 pathways are critical for cell proliferation, whereas the PI3K/AKT1 and STAT3 pathways are important for cell survival. The histology of these tumours is typically characterized by mucin production and either a solid growth pattern containing signet ring cells in Western patients or an acinar growth pattern in Asian patients. Compared with patients with wild-type ALK and EGFR, patients with the EML4–ALK fusion gene tend to be younger, of Asian ethnicity, diagnosed at an advanced clinical stage at presentation, male dominant, and more likely to be never smokers, but with a comparable response rate to chemotherapy and overall survival. The EML4–ALK fusion gene is typically detected by fluorescence in-situ hybridization (FISH). It has been reported that although ALK-fusion-positive lung cancers are resistant to the EGFR TKIs, gefitinib, and erlotinib, they are sensitive to small molecule TKIs against ALK [8].

The BRAFV600E point mutation involves nucleotide 1799 and results in a valine-to-glutamate substitution at residue 600 (V600E). B-Raf is a serine/theronine protein kinase involved in MAPK/ ERK signalling pathway and in regulation of cell proliferation and differentiation. It is found in approximately 40–45% of papillary thyroid carcinomas. Not all variants are equally affected; 60% of classic papillary, 80% of tall cell variant, and 10% of follicular variant harbour this mutation. Its detection is clinically significant because if represents a prognostic marker for thyroid papillary carcinoma, it is associated with extrathyroidal extension, advanced tumour stage at presentation, and lymph node or distant metastases. BRAFV600E point mutation is also an independent predicator of treatment failure and tumour recurrence, even with patients with low-stage disease [4].

Case selection and molecular analysis techniques
Thirty-one cases of lung adenocarcinomas and 26 thyroid carcinomas (17 classic papillary, 7 follicular variant, and 2 follicular carcinomas) were selected from the archives. Molecular analysis was performed on PET and from either Pap or Diff-Quik® stained smears for each case. The following mutations in lung adenocarcinomas were tested: EGFR point mutations in exons 20 and 21, in-frame deletions in exon 19; KRAS point mutations in codons 12, 13, and 61; and EML4–ALK translocation. Thyroid carcinomas were tested for the BRAFV600E point mutation.

Smears were reviewed by two cytopathologists and areas containing at least 50 cancer cells without necrosis or inflammation were marked for analysis [9, 10]. Tumour cells from marked areas were microdissected using RNA/DNA co-purification solution (Zymo Technologies) [5, 6]. QClamp xenonucleic acid (XNA) technology was used to detect mutations in EGFR, KRAS and BRAF genes. The qClamp, a wild-type sequence-specific XNA probe, has a melting point higher than 72 °C and remains attached to the wild-type DNA during the PCR extension stage. If there is a mutation, the probe dissociates from the template, which allows amplification. The technology was optimized using wild-type DNA (Promega, Madison, WI) resulting in a DNA shift of more than seven cycle thresholds (CTs) with the XNA clamp. In samples with mutations, the shift was less than 5 CTs (Fig. 1).

Quantitative RT-PCR (RT-qPCR) was used to detect EML–ALK translocations. PCR primers were used to amplify both the 3ʹ and 5ʹ ends of the ALK transcript (Aanera Biotech). In the presence of the translocation, the transcription of the EML4–ALK fusion gene (under the control of a stronger EML promoter) resulted in higher than expected 3ʹ ends than 5ʹ ends which lead to lower CT values for the 3ʹ transcript. A difference of more than 10 CTs between 3ʹ and 5ʹ ends was considered positive (Fig. 1).

The sensitivity of our assay was determined using two lung cancer cell lines, H1975 and H2228 (ATCC), harbouring the L858R mutation in the EGFR gene and the EML4–ALK translocation, respectively. The mutation detection rate was 1%.

Results
Approximately 80.6% of lung cases had some form of molecular alteration detected. There was 100% concordance between PET and cytology smears. All cases tested positive by our laboratory were also positive by the reference laboratory. However, seven cases that tested negative for EGFR mutation by the reference laboratory were found to be positive in our lab. In other words, clamp qPCR methodology detected approximately 20% more EGFR mutations than the reference laboratory. Likewise, three cases that were negative for KRAS by the reference laboratory, tested positive in our laboratory. In addition, there were six cases (19%) that had more than one molecular alteration in our cohort. Five cases had two mutations (three had EGFR exon 19 and KRAS codon 12 mutations, two had KRAS codon 12 and EML4–ALK mutations) and one case had three mutations (EGFR exon 19 and 21 and KRAS codon 12 mutation). Although EGFR and KRAS mutations were previously thought to be mutually exclusive, our results confirm recent reports of simultaneous mutations detected in study samples [7]. Given the known heterogeneity of cancers this finding may be expected, but might not have been detected previously because most analyses are performed using standard-sensitivity techniques.

Of 26 thyroid cases, 18 (81%) were positive for BRAFV600E mutation on both PET and cytology smears. Concordant results were obtained from both cytology smears and PET from cases tested by us and the reference laboratory. Both follicular carcinoma cases tested negative on cytology smears and PET for BRAFV600E. The percentage of patients with a BRAF mutation detected by us was higher than expected from the literature (81% versus 50%) reflecting our ultrasensitive approach to mutation detection [10].

Conclusion
Our results indicate that qClamp technology and the RT-qPCR approach can be used successfully to detect most common targetable molecular alterations from cytology smears of lung and thyroid carcinomas. We report successful DNA and RNA isolation from both Diff-Quik® and Pap stained smears. This methodology represents an ultrasensitive and accurate approach with a 1% mutation detection rate and a decreased turnaround time of 1 to 2 days. Such an ultrasensitive method for molecular testing is essential as smaller amounts of diagnostic material become available and targeted approaches will be aimed at both molecular alterations present in the majority of cells, and at those present in a minority of cells that potentially may represent a subpopulation susceptible to recurrence [11].

Abbreviations
Table 1 shows the gene symbols and the names and symbols of the proteins encoded by those genes.

References
1. Oktay MH, Adler E, Hakima L, Grunblatt E, Pieri E, Seymour A, Khader S, Cajigas A, Suhrland M, Goswami S. The application of molecular diagnostics to stained cytology smears. J Mol Diagn. 2016; 18(3): 407–415.
2. Tsao MS, Sakurada A, Cutz JC, Zhu CQ, Kamel-Reid S, Squire J, Lorimer I, Zhang T, Liu N, Daneshmand M, Marrano P, da Cunha Santos G, Lagarde A, Richardson F, Seymour L, Whitehead M, Ding K, Pater J, Shepherd FA. Erlotinib in lung cancer – molecular and clinical predictors of outcome. N Engl J Med. 2005; 353: 133–144.
3. Campbell JD, Alexandrov A, Kim J, Wala J, Berger AH, Pedamallu CS, Shukla SA, Guo G, Brooks AN, Murray BA, Imielinski M, Hu X, Ling S, Akbani R, Rosenberg M, Cibulskis C, Ramachandran A, Collisson EA, Kwiatkowski. DJ, Lawrence MS, Weinstein JN, Verhaak RG, Wu CJ, Hammerman PS, Cherniack AD, Getz G, Cancer Genome Atlas Research Network, Artyomov MN, Schreiber R, Govindan R, Meyerson M. Distinct patterns of somatic genome alterations in lung adenocarcinomas and squamous cell carcinomas. Nat Genet. 2016; 48(6): 607–616.
4. Nikiforov YE. Molecular diagnostics of thyroid tumors. Arch Pathol Lab Med. 2011; 135(5): 569–577.
5. Gupta N, Dasyam AK, Carty SE, Nikiforova MN, Ohori NP, Armstrong M, Yip L, LeBeau SO, McCoy KL, Coyne C, Stang MT, Johnson J, Ferris RL, Seethala R, Nikiforov YE, Hodak SP. RAS mutations in thyroid FNA specimens are highly predictive of predominantly low-risk follicular-pattern cancers. J Clin Endocrinol Metab. 2013; 98(5): 914–922.
6. Nikiforov YE, Ohori NP, Hodak SP, Carty SE, LeBeau SO, Ferris RL, Yip L, Seethala RR, Tublin ME, Stang MT, Coyne C, Johnson JT, Stewart AF, Nikiforova MN. Impact of mutational testing on the diagnosis and management of patients with cytologically indeterminate thyroid nodules: a prospective analysis of 1056 FNA samples. J Clin Endocrinol Metab. 2011; 96(11): 3390–3397.
7. Nikiforov YE, Yip L, Nikiforova MN. New strategies in diagnosing cancer in thyroid nodules: impact of molecular markers. Clin Cancer Res. 2013; 19(9):2283–2288.
8. Cheng L, Alexander RE, Maclennan GT, Cummings OW, Montironi R, Lopez-Beltran A, Cramer HM, Davidson DD, Zhang S. Molecular pathology of lung cancer: key to personalized medicine. Mod Pathol. 2012; 25: 347–369.
9. Marchetti A, Milella M, Felicioni L, Cappuzzo F, Irtelli L, Del Grammastro M, Sciarrotta M, Malatesta S, Nuzzo C, Finocchiaro G, Perrucci B, Carlone D, Gelibter AJ, Ceribelli A, Mezzetti A, Iacobelli S, Cognetti F, Buttitta F. Clinical implications of KRAS mutations in lung cancer patients treated with tyrosine kinase inhibitors: an important role for mutations in minor clones. Neoplasia. 2009; 11: 1084–1092.
10. Russo M, Malandrino P, Nicolosi ML, Manusia M, Marturano I, Trovato MA, Pellegriti G, Frasca F, Vigneri R. The BRAF (V600E) mutation influences the short and medium-term outcomes of classic papillary thyroid cancer, but is not an independent predictor of unfavorable outcome. Thyroid 2014; 24: 1267–1274.
11. Pon JR, Marra MA. Driver and passenger mutations in cancer. Annu Rev Pathol. 2015; 10: 25–50.

The authors
Laleh Hakima1 DO; Maja H. Oktay1 MD, PhD; Sumanta Goswami*2 PhD
1Department of Cytopathology, Montefiore Medical Center Bronx, NY, USA
2Department of Biology, Yeshiva University, New York, NY, USA

*Corresponding author
E-mail: Goswami@yu.edu

p22 04

Using genetic risk factors for predicting type 1 diabetes progression and prognosis

Type 1 diabetes is a multigenic disease in which the pancreatic β-cells are destroyed by an autoimmune process. At time of diagnosis, only poorly functional β-cell mass exists. Prediction of type 1 diabetes progression and prognosis using genetic markers may improve treatment strategies and increase the patient´s life quality.

by Dr Caroline A. Brorsson and Dr Joachim Størling

Type 1 diabetes
Type 1 diabetes (T1D) is a chronic disease that results from an autoimmune destruction of the insulin-producing pancreatic β-cells in the islets of Langerhans. Worldwide, T1D is affecting an increasing number of people and the strongest increase in incidence is observed among young children. The disease is complex and caused by an interplay between genetic and environmental risk factors. Genes of the human leukocyte antigen (HLA) locus are the most prominent risk-conferring genes, but dozens (>50) of other risk loci have now been established to influence the risk of T1D [1, 2]. The exact mechanisms, however, by which HLA and other associated loci affect T1D risk, islet autoimmunity and the time course of β-cell destruction, remain elusive. T1D is preceded by a pre-clinical phase characterized by the appearance of autoantibodies directed against islet antigens. Several studies have confirmed the strong predictive effect of islet autoantibodies for the risk of developing T1D in genetically susceptible individuals. Still the rate of progression from autoantibody positivity to clinical onset is highly variable between individuals, and likely influenced by a combination of genetic and environmental risk factors [3].

As any immune-modulating interventions are possible only after the first signs of autoimmunity have occurred, there is a high risk that most of the β-cells have already been destroyed. Therefore, there is a need for more precise, and preferably earlier, methods for predicting disease risk and progression in order to preserve the residual β-cell mass and to choose optimal treatment regimens. Genetic markers can be measured before the onset of autoimmunity and offers an opportunity for early screening. Also, after the onset of T1D, preservation of β-cell function, as assessed by higher C-peptide levels, has been associated with decreased risk of diabetes complications including acute hypoglycemic events and long-term microvascular complications [4–6].  
 
Prediction of T1D progression in high-risk individuals
Children with a high risk of diabetes, as characterized by either by carrying high-risk HLA genotypes or by having first-degree relatives with T1D, have been followed from birth until autoantibody development and diabetes onset in several cohort studies. A few of these studies have investigated the predictive effect of non-HLA genetic variants for islet autoimmunity and progression to T1D. Steck et al. studied the largest such prospective cohort from the U.S. population (the Diabetes Autoimmunity Study of the Young; DAISY) consisting of 861 first-degree relatives and 882 high-risk children from the general population [7]. They found that the risk alleles for PTPN22 and UBASH3A predicted both islet autoimmunity and diabetes, whereas PTPN2 predicted islet autoimmunity alone and INS predicted diabetes alone.

Studying a similar population of 1650 children of type 1 diabetic parents in the German BABYDIAB cohort, Winkler and co-workers showed that the cumulative sum of risk alleles of 12 T1D risk variants (a so-called genetic risk score; GRS) could stratify the risk of developing islet autoantibodies and diabetes, and progression from islet autoimmunity to diabetes [8]. In a subsequent study, Bonifacio et al. studied the rate of progression from the development of islet autoantibodies to diabetes in the same cohort of high-risk children [9]. They found that the genetic risk score of 12 genes could only marginally predict the risk of islet autoimmunity, but could significantly modify the risk of progressing from autoantibody positivity to diabetes. The most predictive power had a genetic risk score constructed from the five risk variants in INS, IFIH1, IL18RAP, CD25 and IL2, which could identify 80% of islet autoantibody-positive children who progressed to diabetes within 6 years and discriminate high risk (63% within 6 years) and low risk (11% within 6 years) antibody-positive children.

Achenbach and colleagues used the same cohort to investigate whether the 12 genetic variants could discriminate between slow and rapid progression to T1D in multiple autoantibody-positive children (3). Among the 1650 children, 23 developed multiple autoantibodies and progressed to diabetes within 3 years, while 24 developed multiple autoantibodies but did not progress to diabetes during more than 10 years of follow-up. The slow and rapid progressors were similar in regards to HLA risk genotypes, development of autoantibodies to insulin (IAA), glutamic acid decarboxylase (GADA) and zinc transporter 8 (ZnT8A), and progression to multiple autoantibodies. However, autoantibodies to insulinoma-associated antigen-2 (IA-2A) developed significantly later in children who progressed slowly. The GRS could clearly discriminate between the two groups of progressors. Best discriminatory power had a GRS including seven of the 12 risk variants (for the genes IL2, CD25, INS, IL18RA, IL10, IFIH1, and PTPN22). Interestingly, the risk score did particularly well in discriminating between children that carried high-risk HLA genotypes.

Prediction of T1D prognosis in new-onset patients
The Hvidoere Study Group for Childhood Diabetes (HSG) has collected a cohort of 275 newly diagnosed children with the purpose of identifying factors that control changes in β-cell function and glycemic control over time. All children underwent a standardized mixed-meal test at 1, 6 and 12 months after the diagnosis of T1D, to assess the stimulated C-peptide response at these time-points. Mortensen et al. (10) were able to demonstrate several factors that predict lower β-cell function at 12 months after diagnosis, including younger age and ketoacidosis at diagnosis, and stimulated C-peptide levels, post-meal blood glucose levels, and IAA and GADA autoantibodies at 1 month.   

Only a few studies have investigated the genetic effect on prognosis after disease onset in T1D, including the cohort collected by the HSG. Candidate gene studies of single genetic variants have shown that the INS and PTPN22 risk variants are associated with residual β-cell function, glycemic control, autoantibody titres and proinsulin in new-onset T1D [11–13]. Furthermore, in one of the first studies that used a combination of cell biology experiments and clinical observations to study the impact of a T1D risk gene, we investigated the function of CTSH. That study showed that the risk variant of CTSH was associated with β-cell function and insulin dose in the children one year after diagnosis [14]. Interestingly, it was observed that within the β-cells, CTSH is a protective gene that inhibitsβ-cell death induced by pro-inflammatory cytokines – believed to contribute to β-cell killing in T1D – thus providing a mechanistic explanation for how genetic variation in CTSH affects T1D risk. In a separate cohort studying children diagnosed with T1D before the age of 11 years, we demonstrated that the risk variant in ERBB3 was associated with better β-cell function and lower HbA1c levels, and thereby a better glycemic control, after controlling for the effects of sex, age at diagnosis and duration of diabetes [15]. In that study, we also found that ERBB3 is regulating β-cell death in response to pro-inflammatory cytokines providing a possible mechanistic link.

In our most recent study on the HSG cohort, we investigated the impact of an increasing GRS on β-cell function and glycemic control during the first year after diabetes onset (16). The GRS was constructed from 11 T1D risk genes that we found to be expressed in human pancreatic islets, and whose expression changed upon stimulation with cytokines. We chose to focus strictly on the islet-expressed risk genes because we hypothesized that these would be the best predictors of islet (β-cell) function. We found that for each additional risk variant, i.e. for each unit increase in the GRS, a decreased β-cell function and a worsened glycemic control from 6 to 12 months after onset were observed, after controlling for the effect of age at diagnosis, sex and HLA risk groups. Further, we found that several of the genes used in the GRS interacted in a network suggesting that they may cooperate to regulate important processes within the β-cells. The results from these reviewed studies are summarized in Table 1.

Benefits for patients
Use of genetics in prediction models could lead to earlier prediction useful for immune-modulatory interventions to preserve residual β-cell mass and will be beneficial both in the pre-clinical phase and after diagnosis. Better stratification for fast and slow progressors both from autoantibody positivity to diabetes and disease progression after diagnosis would be a major achievement in diabetes care. Being able to foresee which genetically-predisposed individuals progress to T1D and these patients’ remaining β-cell function at time of diagnosis and first year(s) to come would have a tremendous impact on the individual patient’s health burden and quality of life, due to lowering the risk for hypo- and hyperglycemia and long-term complications.

Conclusion
In summary, profiling of selected genetic variants may hold promise to better predict T1D progression in risk individuals and residual β-cell function in new-onset type 1 diabetics. Such knowledge may in the future be exploited to offer personalized medicine to optimize treatment regimens to increase patient care and reduce severe long-term complications.

References
1. Barrett JC, Clayton DG, Concannon P, et al. Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat Genet. 2009; 41: 703–707.
2. Onengut-Gumuscu S, Chen WM, Burren O, et al. Fine mapping of type 1 diabetes susceptibility loci and evidence for colocalization of causal variants with lymphoid gene enhancers. Nat Genet. 2015; 47: 381–386.
3. Achenbach P, Hummel M, Thumer L, et al. Characteristics of rapid vs slow progression to type 1 diabetes in multiple islet autoantibody-positive children. Diabetologia 2013; 56: 1615–1622.
4. Steffes MW, Sibley S, Jackson M, Thomas W. Beta-cell function and the development of diabetes-related complications in the diabetes control and complications trial. Diabetes Care 2003; 26: 832–836.
5. Panero F, Novelli G, Zucco C, et al. Fasting plasma C-peptide and micro- and macrovascular complications in a large clinic-based cohort of type 1 diabetic patients. Diabetes Care 2009; 32: 301–305.
6. Pinckney A, Rigby MR, Keyes-Elstein L, et al. Correlation among hypoglycemia, glycemic variability, and C-Peptide preservation after alefacept therapy in patients with type 1 diabetes mellitus: analysis of data from the immune tolerance network T1DAL trial. Clin Ther. 2016; doi: 10.1016/j.clinthera.2016.04.032. [Epub ahead of print].
7. Steck AK, Wong R, Wagner B, et al. Effects of non-HLA gene polymorphisms on development of islet autoimmunity and type 1 diabetes in a population with high-risk HLA-DR,DQ genotypes. Diabetes 2012; 61: 753–758.
8. Winkler C, Krumsiek J, Lempainen J, et al. Ziegler AG. A strategy for combining minor genetic susceptibility genes to improve prediction of disease in type 1 diabetes. Genes Immun. 2012; 13: 549–555.
9. Bonifacio E, Krumsiek J, Winkler C, et al. Ziegler AG. A strategy to find gene combinations that identify children who progress rapidly to type 1 diabetes after islet autoantibody seroconversion. Acta Diabetol. 2014; 51: 403–411.
10. Mortensen HB, Swift PG, Holl RW, et al. Multinational study in children and adolescents with newly diagnosed type 1 diabetes: association of age, ketoacidosis, HLA status, and autoantibodies on residual beta-cell function and glycemic control 12 months after diagnosis. Pediatr Diabetes 2010; 11: 218–226.
11. Nielsen LB, Mortensen HB, Chiarelli F, et al. Impact of IDDM2 on disease pathogenesis and progression in children with newly diagnosed type 1 diabetes: reduced insulin antibody titres and preserved beta cell function. Diabetologia 2006; 49: 71–74.
12. Nielsen LB, Porksen S, Andersen ML, et al. The PTPN22 C1858T gene variant is associated with proinsulin in new-onset type 1 diabetes. BMC Med Genet. 2011; 12: 41.
13. Petrone A, Spoletini M, Zampetti S, et al. The PTPN22 1858T gene variant in type 1 diabetes is associated with reduced residual beta-cell function and worse metabolic control. Diabetes Care 2008; 31: 1214–1218.
14. Floyel T, Brorsson C, Nielsen LB, et al. CTSH regulates beta-cell function and disease progression in newly diagnosed type 1 diabetes patients. Proc Natl Acad Sci. U S A 2014; 111: 10305–10310.
15. Kaur S, Mirza AH, Brorsson CA, et al. The genetic and regulatory architecture of ERBB3-type 1 diabetes susceptibility locus. Mol Cell Endocrinol. 2016; 419: 83–91.
16. Brorsson CA, Nielsen LB, Andersen ML, et al. Hvidoere Study Group On Childhood Diabetes. Genetic risk score modelling for disease progression in new-onset type 1 diabetes patients: increased genetic load of islet-expressed and cytokine-regulated candidate genes predicts poorer glycemic control. J Diabetes Res. 2016; 2016: 9570424.

The authors
Caroline A Brorsson*1 PhD and Joachim Størling2 PhD
1Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
2Copenhagen Diabetes Research Center, Pediatric Department E, University Hospital Herlev, Herlev, Denmark

*Corresponding author
E-mail: caroline@cbs.dtu.dk

C260 Kawano Fig p23

Diagnosis and other aspects of uromodulin kidney disease

Uromodulin kidney disease is a rare autosomal dominant kidney disease, characterized by hyperuricemia, gout and progressive kidney failure. Affected patients typically need renal replacement therapy in middle age. A considerable number of patients may reach end-stage kidney disease without a correct diagnosis, making improvements in diagnostic methods of vital importance.

by Dr Tamehito Onoe and Dr Mitsuhiro Kawano

Introduction
Uromodulin (UMOD), also known as Tamm–Horsfall protein, is the most abundant protein in healthy human urine. UMOD protein is a kidney-specific protein which is exclusively produced at the epithelial cells lining the thick ascending limb (TAL) of Henle’s loop.

The roles of urinary UMOD protein are assumed to be to protect against urinary tract infection, prevent urolithiasis formation and ensure water impermeability to create the countercurrent gradient. However, the accurate function and significance of UMOD protein are not yet fully elucidated [1].

Uromodulin kidney disease
Uromodulin kidney disease (UKD) is an inherited disease caused by UMOD gene mutations. So far, more than 100 mutations of the UMOD gene have been reported from all over the world [2]. Familial juvenile hyperuricemic nephropathy (FJHN), medullary cystic kidney disease type2 (MCKD2) and glomerulocystic kidney disease (GCKD), which are considered to be different diseases, have been proved to be caused by UMOD gene mutations [3]. Subsequently, because multiple names for one condition would be confusing and misleading, and also cysts are not pathognomonic for this disease, a new term, ‘Autosomal dominant tubulointerstitial kidney disease’ (ADTKD) was proposed in 2015 [4]. Mutations of renin (REN), hepatocyte nuclear factor 1β (HNF1β), and mucin-1 (MUC1) are also responsible for ADTKD besides UMOD. They all share common clinical characteristics, which are progressive kidney failure, tubulointerstitial nephritis and inheritance compatible with autosomal dominant trait with only trivial clinical differences. When UMOD mutation is identified in an ADTKD patient, the official diagnostic term is ADTKD-UMOD. However, UKD is also used to facilitate communication with patients, and so this term is used in the present article.

Patients with UKD have urinary concentration defect, hyperuricemia and gout from a young age. Their kidney function gradually deteriorates, and reaches end-stage kidney disease (ESKD) from 25 to 75 years of age. Their kidneys are usually of normal size or small and there are sometimes cysts, although the frequency of cysts does not differ from that of ‘non-cystic’ kidney diseases. Their urine tests usually show no or only very mild proteinuria or hematuria. The most prominent characteristic of UKD is a marked abundance of chronic kidney disease (CKD) patients in their pedigree, compatible with an autosomal dominant trait. Our group detected a novel A247P UMOD mutation in a UKD family (Fig. 1), many of whose members have hyperuricemia, CKD and ESKD, and are on hemodialysis (HD) therapy [5].

UKD is reported to be a rare disease, with a frequency of about 1.5 cases per million population. However, because hyperuricemia is a frequent complication in all CKD patients, when their family history is absent or unknown, it is difficult to suspect UKD, and so the frequency of UKD may be underestimated. This means that a certain proportion of UKD patients may reach ESKD without a correct diagnosis.

Diagnosis of uromodulin kidney disease
Clinically UKD should be suspected when a CKD patient has an abundant family history compatible with autosomal dominant trait, hyperuricemia, gout and bland urine findings. The final diagnosis of UKD is made by genetic test, which is, however, not commercially available, and only a limited number of laboratories are capable of performing it. So easier laboratory tests supportive of genetic tests would be helpful for the diagnosis of UKD and are awaited.

The renal histology of UKD patients shows nonspecific interstitial fibrosis, tubular atrophy and normal glomeruli. So it is difficult to make a diagnosis of UKD by ordinary histological methods. Moreover, not many UKD patients seem to undergo renal biopsy because their urine sediment shows no or only slight abnormalities and so clinicians may hesitate to undertake this invasive test. However, we believe that renal pathological examination is very informative not only for ruling out other kidney diseases but also for the diagnosis of UKD. UMOD proteins synthesized from mutated UMOD gene have protein folding disability and cannot escape from the endoplasmic reticulum (ER) of the epithelial cells. Immunostaining using anti-UMOD antibody in kidney sections of UKD patients shows massive UMOD accumulation in their epithelial cells (Fig. 2). Because of the question of whether there are any UKD patients among those who received kidney biopsy and were diagnosed as having nephrosclerosis or interstitial nephritis, we performed the following investigation.

In a 3787-sample kidney biopsy database of Kanazawa University, patients meeting all of the following criteria were selected for UMOD immunostaining. (1) Renal insufficiency (serum creatinine >1.0 mg/dL) below 50 years of age; (2) hyperuricemia: serum uric acid higher than 7mg/dl or under treatment for hyperuricemia; (3) no or only very mild abnormalities in urinalysis; and (4) no other apparent renal disease present clinically or histopathologically. Finally, 15 patients were selected and abnormal UMOD accumulations were detected in three independent patients by UMOD immunostaining. A247P UMOD gene mutations were detected in the proband of the family in Figure 1 and the other independent patient, indicating that they may share the same ancestor. The other patient had no family history of CKD. These results show that there may be more UKD patients than expected before, and also indicate that when kidney biopsy shows only nonspecific interstitial fibrosis in patients with renal insufficiency, UMOD immunostaining may be considered to detect UKD with or without a family history of CKD, especially with hyperuricemia and bland urinary findings.

Most of the synthesized UMOD protein is carried to the apical membrane of epithelial cells and excreted in the urine. However, a low but considerable amount of UMOD protein goes to the basolateral membrane and is secreted into the serum [6]. Serum UMOD protein concentrations are reported to be 45–490 ng/mL, while urine UMOD protein concentrations are 1000–80 000 ng/mL. The functions and significance of serum UMOD proteins are unknown.

Some results of animal experiments indicate that UMOD protein has a renoprotective effect against various types of injury. A renal ischemia-reperfusion experiment in UMOD knockout mice showed significantly worse results than in wild-type animals [7]. It is well known that urinary UMOD concentrations in UKD patients are decreased. The authors recently reported that serum UMOD protein concentrations are also significantly decreased in UKD patients besides urinary UMOD (Fig. 3). Serum and urinary UMOD concentrations decline in parallel with the decrease of estimated glomerular filtration rate (eGFR) due to the diminishment of UMOD producing epithelial cells in CKD patients. In UKD patients, the serum and urine UMOD concentrations were significantly lower compared with CKD patients beyond their eGFRs. Decreased serum and urinary UMOD concentrations may be good clues to suspect and diagnose UKD; however, verification in more UKD patients with various mutations will be indispensable.

Conclusions
So far no treatment has been devised that slows the rate of renal functional deterioration of UKD. At present, management for UKD patients is not different from that for other CKD patients. Anti-hyperuricemia drugs or anti-hypertensive therapy is used when necessary and the appropriate renal replacement therapy or renal transplantation should be considered when ESKD is reached. To clarify the pathogenesis and achieve effective treatment for UKD, establishment of more efficient diagnostic methods for UKD is expected. UMOD immunostaining for renal sections and measurement of serum and urinary UMOD concentrations are considered to be good modalities for the diagnosis of UKD. It is expected that through these tests, more UKD patients will be diagnosed at earlier stages and will be able to benefit from starting appropriate therapy before ESKD.

Recently some particular SNPs of UMOD promoter areas have been proved to be associated with hypertension or renal insufficiency from the genome-wide association study [8]. UMOD will likely attract greater attention as a renal-prognostic marker for not only UKD patients but also the general population.

References
1. Lhotta K, Piret SE, Kramar R, Thakker RV, Sunder-Plassmann G, Kotanko P. Epidemiology of uromodulin-associated kidney disease – results from a nation-wide survey. Nephron Extra 2012; 2: 147–158.
2. Scolari F, Izzi C, Ghiggeri GM. Uromodulin: from monogenic to multifactorial diseases. Nephrol Dial Transplant. 2015; 30: 1250–1256.
3. Hart TC, Gorry MC, Hart PS, Woodard AS, Shihabi Z, Sandhu J, Shirts B, Xu L, Zhu H, Barmada MM, Bleyer AJ. Mutations of the UMOD gene are responsible for medullary cystic kidney disease 2 and familial juvenile hyperuricaemic nephropathy. J Med Genet. 2002; 39: 882–892.
4. Eckardt KU, Alper SL, Antignac C, Bleyer AJ, Chauveau D, Dahan K, Deltas C, Hosking A, Kmoch S, Rampoldi L, Wiesener M, Wolf MT, Devuyst O. Autosomal dominant tubulointerstitial kidney disease: diagnosis, classification, and management-A KDIGO consensus report. Kidney Int. 2015; 88(4): 676–683.
5. Onoe T, Yamada K, Mizushima I, Ito K, Kawakami T, Daimon S, Muramoto H, Konoshita T, Yamagishi M, Kawano M. Hints to the diagnosis of uromodulin kidney disease. Clin Kidney J. 2016; 9: 69–75.
6. Bachmann S, Koeppen-Hagemann I, Kriz W. Ultrastructural localization of Tamm-Horsfall glycoprotein (THP) in rat kidney as revealed by protein A-gold immunocytochemistry. Histochemistry 1985; 83: 531–538.
7. El-Achkar TM, Wu XR, Rauchman M, McCracken R, Kiefer S, Dagher PC. Tamm-Horsfall protein protects the kidney from ischemic injury by decreasing inflammation and altering TLR4 expression. Am J Physiol Renal Physiol. 2008; 295: F534–544.
8. Trudu M, Janas S, Lanzani C, Debaix H, Schaeffer C, Ikehata M, Citterio L, Demaretz S, Trevisani F, Ristagno G, Glaudemans B, Laghmani K, Dell’Antonio G, Loffing J, Rastaldi MP, Manunta P, Devuyst O, Rampoldi L. Common noncoding UMOD gene variants induce salt-sensitive hypertension and kidney damage by increasing uromodulin expression. Nat Med. 2013; 19: 1655–1660.

The authors
Tamehito Onoe MD, PhD and Mitsuhiro Kawano* MD, PhD
Division of Rheumatology,
Department of Internal Medicine,
Kanazawa University Hospital,
Kanazawa, 920-8641,
Japan

*Corresponding author
E-mail: sk33166@gmail.com

fig1

Improved productivity and workflow efficiencies enhance viral load service provision

A series of independent evaluations of the Beckman Coulter DxN VERIS Molecular Diagnostics System demonstrated how workflow efficiencies that can be achieved with the DxN VERIS have the potential to improve productivity, while making the best use of existing space and staffing levels.

Background
Virology laboratories throughout the world face a number of challenges that need to be addressed in order to meet service user requirements. These include:

  • Expanding molecular diagnostic workloads
    Include an increase in requests for viral load assays for targets, such as human immunodeficiency virus type 1 (HIV-1), hepatitis C virus (HCV), hepatitis B virus (HBV) and cytomegalovirus (CMV). This growth may be attributed, in part, to the development of new therapeutic strategies or the move away from other traditional “non-molecular” methods.
  • Limited space and workflowinefficiencies
    Many laboratories have to cope with these expanding workloads in already crowded working environments.
    Many existing methods used for obtaining HIV-1, HCV, HBV and CMV viral loads involve multiple platforms and are thus using valuable laboratory space.

Evaluation of a new automated molecular diagnostics method
In 2014/2015, a number of hospital laboratories across Europe became beta trial sites for a new, fully automated molecular diagnostics system, the DxN VERIS Molecular Diagnostics System (Beckman Coulter Inc.), including the virology section at the Hospital Clinic of Barcelona, Spain, the department of laboratory medicine at Niguarda Hospital, Milan, Italy, the department of clinical microbiology at the Hospital Universitario 12 de Octubre, Madrid, Spain and the virology department at Sheffield Teaching Hospitals NHS Foundation Trust, UK.
The DxN VERIS System, launched at ECCMID 2015, consolidates nucleic acid extraction, amplification, quantification and detection onto a single automated instrument for a number of molecular targets, including HIV-1, HCV, HBV and CMV. The system offers single sample random access and the potential to improve clinical laboratory workflow efficiency.
The performances of the VERIS assays for CMV, HBV, HCV and HIV-1 were evaluated using standard and control samples, as well as clinical samples, and were compared to various existing viral load methods in each laboratory. In addition, a series of independent time/workflow analysis studies were performed by Nexus Global Solutions (Plano, Texas, USA).

Productivity and workflow improvements
The results of the comparative workflow studies performed in these participating laboratories are summarized below:

  • The DxN VERIS System workflow involved far fewer steps and consumables than existing methods, particularly in the pre-analytical phase, and required reduced hands-on time, which resulted in significant savings in the time that staff are tied to the process (examples shown in Figure 1).
  • Significant improvement in time to first result. This is in contrast to the current methods, where results are not available until the end of the assay run.
  • The DxN VERIS System allows true, single sample random access which, combined with short assay runtimes, ensures the rapid turnaround of results.
  • The DxN VERIS system allowed much faster turnaround of results in a normal working week, with all results being reported within 8-24 hours of receipt (depending on the laboratory), unlike existing methods which often required several days (example in Figure 2).

Duncan Whittaker, Laboratory Manager Virology at Sheffield Teaching Hospitals NHS Foundation Trust, shared his experience:
“Hands on time requirements were measured specifically for the HIV-1 and CMV assays. If these two assays alone were consolidated onto the DxN VERIS it would save around 2 hours manual time per day. If all four parameters were consolidated onto the DxN VERIS system, it is estimated that this would ultimately save at least 0.6 whole time equivalent (WTE) biomedical scientists.”
Diana Fanti, Molecular Biology Laboratory Manager at Niguarda Hospital, Milan, commented:
“By reducing manual intervention and automating processes from sample loading to reporting of results, the DxN VERIS offers the potential to transform clinical laboratory workflows. Each assay is supplied in a unique, single cartridge system, and all consumables and reagents are stored on-board, which cuts preparation time compared to alternative methods”.
Rafael Delgado, Head of Clinical Microbiology at the Hospital Universitario 12 de Octubre, Madrid, agreed:
“One of the most important aspects of the system for our laboratory is the ability to process samples as they are received in the laboratory. With our pervious method, we had to work in batches of 24 or 48, collecting and storing samples throughout the day (or overnight) until we had sufficient for a single run. Then results were not available until the entire run was completed. Now, with the random access capabilities of the DxN VERIS system, this has changed. We receive samples around the clock and we are able to run them straight away. This has improved our response times significantly, from 24-28 hours to just 4-5 hours from sample receipt, and with comparable quality of results compared to our previous method.”

Driving efficiency
DxN VERIS assays are supplied in a unique single cartridge system, which saves further preparation time and effort compared to alternative methods.
The DxN VERIS System also allows more efficient use of staff. Dr Fanti commented:
“By transforming laboratory organization and workflows and reducing manual intervention, viral loads (which account for about 50% of the molecular workload) could be completed in a single day using the DxN VERIS. Requiring fewer people to be dedicated to this purpose, this makes it possible to accomplish more work with the same number of staff.”
Duncan Whittaker agreed, stating:
“The ease of use of the DxN VERIS would help to address staffing issues, as routine operation could be performed by medical laboratory assistants, allowing biomedical scientists to be redeployed more effectively in more skilled areas. Training staff to use the DxN VERIS is very quick and straightforward, taking just 20 minutes. Furthermore, as there is less hands on intervention required, the laboratory could achieve more without any increase in staff. With an annual cost improvement package to meet, anything that helps to increase productivity is a bonus.”
Rafael Delgado also appreciated the benefits for laboratory staff, commenting:
“The DxN VERIS system has been well received by laboratory staff and has expanded our service capabilities. Fully automated from the loading of samples to obtaining results, it is easy to operate by laboratory technicians of all abilities. In addition, since it involves minimal manual intervention and fewer steps than our previous method, there is less opportunity for error and staff have more time to perform other important tasks in the laboratory.”

Conclusions
In conclusion, Duncan Whittaker continued:
“Following the workflow analysis study, it was apparent that the improved workflow and time savings that can be achieved using the DxN VERIS Molecular Diagnostics System could have an enormous impact on the challenges faced by our laboratory. In terms of addressing increasing workloads, the reduced manual intervention required for DxN VERIS would allow more work to be performed per member of staff. The more efficient workflow would free staff to perform other tasks, which would allow the laboratory to develop new services and further increase the department’s test repertoire. This, and improved turnaround times, would help the laboratory to remain competitive in an increasingly competitive environment.”
Since early 2016, the Clinical Microbiology department at the Hospital Universitario 12 de Octubre, Madrid, has also been using the DxN VERIS System routinely for HBV and HCV viral load quantifications. Rafael Delgado commented:
“Our experiences in evaluating the DxN VERIS system enabled us to appreciate its potential as an enabler for an improved molecular biology clinical service. The increased automation and random access offer workflow improvements that simplify laboratory tasks and reduce the potential for human error. Furthermore, its overall performance and ease of use facilitated the smooth introduction of the technology in our laboratory.”
“Our annual volume of HBV and HCV samples is around 7,000 and, as a clinical laboratory working closely alongside medical staff, our viral load results support timely clinical decision making and subsequent patient management. In this respect the DxN VERIS system is ideal for our needs, providing same day results to our outpatient clinics.”
Beckman Couter is commited to providing an increasing menu of assays for the DxN VERIS Molecular Diagnostics System.

Email: info@beckmanmolecular.com or visit www.beckmancoulter.com/moleculardiagnostics

Contributors
Professor Jordi Vila, Head of Department of Clinical Microbiology and Dr Angeles Marcos, Head of the Virology Section
Hospital Clinic, School of Medicine
University of Barcelona, Spain
Providing a full range of medical and surgical specialties for a local population of over half a million, the Hospital Clinic of Barcelona is also a National and International Centre of reference. The Hospital’s Department of Clinical Microbiology, also a reference laboratory for organ transplantation, operates 24 hours a day, seven days a week and, like many laboratories throughout Europe, has experienced increasing workloads in recent years. HBV HCV CMV and HIV-1 viral loads constitute an annual workload volume of nearly 19,000 tests.


Diana Fanti, Molecular Biology Laboratory Manager
Department of Laboratory Medicine, Niguarda Hospital, Milan, Italy
Niguarda Hospital in Milan is one of Italy’s leading General Hospitals, and provides an extensive range of medical disciplines for adults and children throughout the Lombardy region and beyond. The hospital’s Department of Laboratory Medicine aims to offer a complete, continuous and prompt diagnostic laboratory testing service and is committed to research into automation and analysis to ensure this is maintained. Its busy Molecular Biology Laboratory performed an estimated 40,000 tests in 2015, which is approximately 10% increase on the previous year.

Rafael Delgado, Head of Clinical Microbiology
Hospital Universitario 12 de Octubre, Madrid, Spain
With 1,300 beds and over 6,000 employees, the Hospital Universitario 12 de Octubre in Madrid is one of the largest hospitals in Spain, serving a population of more than 500,000 people in and around the capital. It is an important teaching and research center with a number of areas of expertise, including organ transplantation and the diagnosis and treatment of cancer. The hospital’s Clinical Microbiology Department has a significant serology workload, processing more than 250 serology samples every day, which includes viral load testing for targets such as cytomegalovirus (CMV), hepatitis B virus (HBV), hepatitis C virus (HCV) and human immunodeficiency virus type 1 (HIV-1).

Duncan Whittaker, Laboratory Manager Virology
Sheffield Teaching Hospitals NHS Foundation Trust
The Department of Virology at Sheffield Teaching Hospitals NHS Foundation Trust provides a valuable diagnostic testing service to the people of Sheffield, serving the local community and five teaching hospitals within the trust as well as the Sheffield Children’s Hospital. It is also a referral laboratory receiving samples from further afield for a variety of tests, including routine viral loads and molecular diagnostics. The department’s annual automated workload includes around 105,000 serology samples (more than 300,000 tests) and 65,000 samples for molecular testing (around 129,000 tests), as well as 60,000 samples for Chlamydia and Gonorrhoea testing.

C279 77E UriSed mini whole viewfield image

Evaluation of UriSed mini semi-automated urine microscopy analyser

Urinalysis may provide evidence of significant renal disease in asymptomatic patients. The microscopic urinalysis is vital to making diagnoses in many asymptomatic cases, including urinary tract infection, urinary tract tumours, occult glomerulonephritis, and interstitial nephritis.
Presence or absence of different particles in urine sediment is crucial for clinical decision making. Urine sediment cells or particles provide important information for the diagnosis of renal or urinary diseases [1]. UriSed Technology is a unique solution in the market for the automation of sediment analysis, making traditional manual microscopy automatic [2]. The UriSed analysers provide a reliable and reproducible solution since 2007 [3]. As a new category instrument based on the improved UriSed Technology, the semi-automated UriSed mini was introduced in the market in 2015. In the present study, we evaluated the analytical performance of UriSed mini Semi-Automated Urine Microscopy Analyser (Manufactured by 77 Elektronika Kft., Budapest) and compared the results to those from manual microscopy using standardized KOVA counting chambers.
UriSed mini provides quantitative Red Blood Cell (RBC) and White Blood Cell (WBC) results, and semi-quantitative results for all other particle types: Squamous Epithelial Cells (EPI), Non-squamous Epithelial Cells (renal tubular and urothelium cells) (NEC), Crystals (CRY): Calcium oxalate dihydrate (CaOxd), Calcium oxalate monohydrate (CaOxm), Uric acid (URI), and Triple-phosphate crystals (TRI), Hyaline casts (HYA), Pathological casts (PAT), Bacteria (cocci-like and rod-like) (BACc, BACr), Yeasts (YEA), Spermatozoa (SPRM) and Mucus (MUC) [4].
The instrument throughput is up to 60 samples per hour. Preparation of the UriSed mini analyser for measurement takes only some minutes, the only consumables are the patented disposable cuvettes for sample investigation and a manual pipette with appropriate pipette tip. 175 µl of urine sample is dispensed manually into the cuvette, further steps of the measurement sequence are completely automatic: spinning the cuvette for a few seconds gently deposits formed elements into a monolayer at the bottom of the cuvette. The built-in digital camera takes 15 independent images at different positions of the sediment layer. These whole viewfield images are evaluated by a neural-network based image processing software.

Material and methods
Analysis of 311 samples was performed to evaluate UriSed mini analytical performance compared to the manual microscopy urine examination method. Both measurements were carried out with the same anonymous urine samples. Fresh, native urine samples were used, that were typically held for no more than 4 hours before being analysed, as recommended in the relevant guidelines [5,6] to prevent change in the morphology of the particles. Samples were mixed until homogeneous and then split and run on each measuring procedure as close to the same time as possible. The standardized microscopic urinalysis of native samples (Level 3) was followed by using a KOVA counting chamber. The particle concentration for all particle types was evenly distributed in the evaluated urine samples. Carry-over, precision, diagnostic tests such as sensitivity, specificity, diagnostic accuracy, concordance and one category concordance were investigated according to well-established protocols [7].

Results
No carry-over was detected in any of the samples due to the single-use disposables. UriSed mini has better precision than microscopy at all of the tested RBC and WBC concentrations. The majority of all coefficients of variation obtained for within-series imprecision (CV) using UriSed mini was 4-24% while 5,5-67% in the case of manual microscopy [8]. Good correlation can be found between UriSed mini and manual counting chamber for formed elements. The Pearson correlation of quantitative parameters are 0.97 (RBC), 0.95 (WBC). The clinical evaluation of UriSed mini was based on McNemar test and concordance study. The results are shown in the following table.

Conclusion
The new UriSed mini utilizes the traditional gold standard method while eliminating the most time-consuming and operator-dependent procedures in laboratories performing manual microscopy. The semi-automated measurement process is reproducible and operator-independent. The UriSed mini semi-automated microscopy analyser requires manual sample pipetting, which makes the instrument small and simple to use. UriSed mini is a highly effective tool in a wide range of medical and clinical settings such as hospitals, clinics, accident and emergency departments and outpatient laboratories. In addition it can also serve as a backup instrument of automated sediment analysers.

References
1. Fogazzi GB. The Urinary Sediment an Integrated View Third Edition. Milano: Elsevier, 2010.
2. Barta Z., Kránicz T., Bayer G. UriSed Technology – A Standardised Automatic Method of Urine Sediment Analysis. European Infectious Disease 2011;5:139–42.
3. Zaman Z, Fogazzi GB, Garigali G, Croci MD, Bayer G, Kránicz T. Urine sediment analysis: analytical and diagnostic performances of sediMAX – a new automated microscopy image-based urine sediment analyser. Clin Chim Acta 2010; 411: 147-154.
4. Fogazzi GB, Garigali G. The Urinary Sediment by UriSed Technology. A New Approach to Urinary Sediment Examination. Milano: Elsevier, 2013.
5. Kouri T, Fogazzi G, Hallander H, Hofmann W, Guder WG, editors. European Urinalysis Guidelines. Scand J Clin Lab Invest 2000; 60 (Suppl 231): 1-96.
6. Clinical and Laboratory Standard Institute (ex NCCLS). Document GP16-A3 – Urinalysis; Approved guideline, 3rd ed. Wayne, PA: CLSI, 2009.
7. T. Kouri, A. Gyory, R.M. Rowan. ISLH Recommended Reference Procedure for the enumeration of Particles in Urine. Laboratory Hematology 9:58-63, 2003.
8. Haber MH, Galagan K, Blomberg D, Glassy EF, Ward PCJ, editors. Color Atlas of Urinary Sediment; An Illustrated Field Guide Based on Proficiency Testing. Chicago: CAP Press, 2010.

More information on UriSed mini is available from the manufacturer:
77 Elektronika Kft.,  Budapest,  HUNGARY.
Email: sales@e77.hu, web: www.e77.hu

The author
Erzsébet Nagy MD,
Honorary Associate Professor
Head Physician of Central Laboratory;
Hospitaller Brothers of St. John of God
Hospital, Budapest