Shimadzu Europe
  • News
    • Featured Articles
    • Product News
    • E-News
  • Magazine
    • About us
    • Digital edition
    • Archived issues
    • Free subscriptions
    • Media kit
    • Submit Press Release
  • White Papers
  • Events
  • Suppliers
  • E-Alert
  • Contact us
  • FREE newsletter subscription
  • Search
  • Menu Menu
Clinical Laboratory int.
  • Allergies
  • Cardiac
  • Gastrointestinal
  • Hematology
  • Microbiology
  • Microscopy & Imaging
  • Molecular Diagnostics
  • Pathology & Histology
  • Protein Analysis
  • Rapid Tests
  • Therapeutic Drug Monitoring
  • Tumour Markers
  • Urine Analysis

Archive for category: Featured Articles

Featured Articles

C328 Saeed fig1

MTHFR, hyperhomocysteinemia, CAD and T2DM

, 26 August 2020/in Featured Articles /by 3wmedia

Individuals with type 2 diabetes mellitus (T2DM) are at increased risk of coronary artery disease (CAD). The C677T mutation of the methylenetetrahydrofolate reductase (MTHFR) gene is associated with elevated plasma levels of homocysteine. The association of the MTHFR gene and the level of homocysteine with development of CAD has been studied in various population groups, including patients with T2DM, but the results have been variable. In practice, plasma homocysteine may be ordered as part of a screen for people with CAD or stroke, or who are at high risk for CAD or stroke but no other known risk factors. Testing of C677T polymorphism with or without elevated homocysteine is not recommended and has no clinical utility.

by Prof. Bakri Saeed and Dr Nisreen Mohammed

Type 2 diabetes mellitus and coronary artery disease
Type 2 diabetes mellitus (T2DM) is a major health problem throughout the world. It is a polygenic and multifactorial disease that is a major risk factor for cardiovascular disease. Cardiovascular disease (CVD) comprises coronary artery disease (CAD), also referred to as coronary heart disease (CHD), or ischemic heart disease (IHD), and cerebrovascular disease.

CAD due to atherosclerosis is a cause of significant morbidity and mortality, and is the leading cause of death worldwide. There are several risk factors for CAD. The well-stablished risk factors for CAD include diabetes mellitus, hypertension, smoking and dyslipidemia. There is growing interest in emerging risk factors for improved understanding of the mechanisms that underline cardiovascular disorders and CAD.

T2DM increases the risk for CAD by 2–4-fold compared to people without diabetes. CVD accounts for about 70% of deaths in people with diabetes. Identification and management of risk factors for CAD is an important aspect of management of diabetes mellitus.
Hyperhomocysteinemia and MTHFR polymorphism
Homocysteine is a sulfur-containing amino acid formed from demethylation of methionine. Methionine is the precursor to S-adenosyl methionine (SAMe) and is one of the essential amino acids. SAMe is a major methyl donor and is involved in numerous biological reactions. Homocysteine is metabolized by either remethylation to methionine or transsulfuration to cystathionine. The former reaction is catalysed by the vitamin B12-dependent methionine synthase. The latter reaction is catalysed by the enzyme cystathionine beta-synthase, which requires vitamin B6.

The methyl donor in the remethylation of homocysteine to methionine is 5-methyltetrahydrofolate. The 5,10-methylene-tetrahydrofolate reductase (MTHFR) enzyme catalyses the reduction of 5,10-methylene-tetrahydrofolate to 5-methyltetrahydrofolate. The enzyme requires B2 (riboflavin) as a cofactor (Fig. 1).

Therefore, hyperhomocysteinemia can result from reduced activity of the enzymes involved in homocysteine metabolism or from deficiency of the vitamins which are needed as cofactors in homocysteine metabolic reactions: folate, vitamin B6 and vitamin B12.

Several mutations in the MTHFR gene have been identified and some of them affect the activity of the enzyme. The commonest MTHFR gene mutation is a cytosine-to-thymidine substitution at nucleotide 677 (C677T), which changes alanine into valine, resulting in a thermolabile enzyme with impaired enzymatic activity and leading to hyperhomocysteinemia.

There are two copies of each gene. Therefore, an individual can be homozygous for the mutated gene or can be heterozygous, having one copy of the C677T variant and one normal copy. The C677T homozygous variant enzyme is thermolabile and demonstrates 70% reduced enzyme activity in vitro. The heterozygous C677T MTHFR enzyme has 35% reduced activity in vitro.

Worldwide, the frequency of MTHFR gene mutations varies among racial and ethnic groups, in Africa MTHFR gene polymorphism is markedly low (below 10%) for the C677T allele. In the European and Asian population, estimates of 18.6% and 20.8% were reported [1].

Association with CAD
In recent years hyperhomocysteinemia has been implicated as a risk factor for CAD, independent of other known risk factors. The primary mechanism by which homocysteine promotes atherosclerosis is by impairing endothelial function, which initiates the chain of events resulting in atherosclerotic plaque formation.

Numerous studies looked into the possible association between MTHFR genotypes and plasma homocysteine levels and the incidence of different MTHFR genotypes and hyperhomocysteinemia in CAD patients [2–5]. The results of these studies have been controversial. Several studies have shown the link between the MTHFR C677T gene polymorphism and the risk for CAD but many other studies failed to show association between MTHFR genotypes and plasma homocysteine levels and their role in CAD.

Previous studies in T2DM patients were also controversial. MTHFR polymorphism and hyperhomocysteinemia were shown to be predictors of cardiovascular events among diabetic patients [6, 7], whereas other studies failed to show a role for MTHFR polymorphic variants and homocysteine in increasing susceptibility to cardiovascular disease [8, 9].

Our study
We recently screened 226 consecutive patients with T2DM, <60 years of age, diagnosed according to WHO criteria. Of these, 113 had CAD confirmed by angiography and electrocardiography (ECG) and 113 had no evidence of CAD [10]. PCR and restriction fragment length polymorphism (RFLP) using Hinf1 restriction enzyme were used to determine MTHFR genotypes.

In our study, the T allele had a significant effect on homocysteine level (P value <0.05) and showed strong association with CAD among T2DM patients (odds ratio 6.2, P <0.0001).

Our study indicates that the C677T polymorphism of the MTHFR gene is associated with hyperhomocysteinemia, and the two are independently associated with the presence of CAD in patients with T2DM.

Reasons for controversy
The outcome of these numerous studies and meta-analysis remained contradictory. There was no agreement on the association between MTHFR genotypes and plasma homocysteine levels or the incidence of different MTHFR genotypes and hyperhomocysteinemia in CAD patients.

Plasma homocysteine levels are dependent on interacting nutritional and genetic factors. Some studies suggested that people homozygous for MTHFR C667T polymorphism tend to have hyperhomocysteinemia in the context of low folic acid levels. Supplementation with the vitamins involved in homocysteine metabolism was found to lower plasma homocysteine levels.

Therefore, geographic heterogeneity, nutritional and environmental factors could affect the relationship between MTHFR genotypes and CVD risk in different populations.

Practical points
Homocysteine may be ordered as part of a screen for people with or at high risk of CAD or stroke, especially if there is family history of CAD or stroke but no other known risk factors, such as diabetes, smoking, hypertension, or dyslipidemia. Routine screening of homocysteine, like that of cholesterol, has not been recommended.

Plasma homocysteine concentration may be elevated in B12 and folate deficiency and its measurement has been suggested to give an early indicator of deficiency.

In new-born testing, greatly increased concentrations of homocysteine in the urine and blood suggests a diagnosis of homocystinuria and indicates the need for confirmation of the cause of raised levels.

Most laboratories report normal homocysteine levels in the blood between 5 and 15 µmol/L. Any measurement above 15 µmol/L is considered high.

However, it should be noted that normal levels will vary between ethnic groups and populations. Homocysteine levels increase with age, are lower in pregnancy and are influenced by drugs. These factors should be taken into consideration when interpreting results.

Testing of C677T polymorphism with or without elevated homocysteine is not recommended in patients with CAD or other diseases where MTHFR variants have been implicated, such as thrombophilia or recurrent pregnancy loss.
References
1. Schneider JA, Rees DC, Liu YT, Clegg JB. Worldwide distribution of a common methylenetetrahydrofolate reductase mutation. Am J Hum Genet 1998; 62: 1258–1260.
2. Chehadeh SWEH, Jelinek HF, Al Mahmeed WA, Tay GK, Odama UO, Elghazali GE, et al. Relationship between MTHFR C677T and A1298C gene polymorphisms and complications of type 2 diabetes mellitus in an Emirati population. Meta gene 2016; 9: 70–75.
3. Bickel C, Schnabel R, Zengin E, Lubos E, Rupprecht H, Lackner K, et al. Homocysteine concentration in coronary artery disease: Influence of three common single nucleotide polymorphisms. Nutr Metab Cardiovascular Dis 2017; 27(2): 168–175.
4. Yilmaz H, Isbir S, Agachan B, Ergen A, Farsak B, Isbir T. C677T mutation of methylenetetrahydrofolate reductase gene and serum homocysteine levels in Turkish patients with coronary artery disease. Cell Biochem Funct 2006; 24(1): 87–90.
5. Meisel C, Cascorbi I, Gerloff T, Stangl V, Laule M, Müller JM, et al. Identification of six methylenetetrahydrofolate reductase (MTHFR) genotypes resulting from common polymorphisms: impact on plasma homocysteine levels and development of coronary artery disease. Atherosclerosis 2001; 154(3): 651–658.
6. Lewis SJ, Ebrahim S, Smith GD. Meta-analysis of MTHFR 677C→T polymorphism and coronary heart disease: does totality of evidence support causal role for homocysteine and preventive potential of folate? BMJ 2005; 331(7524): 1053–1058.
7. Bennouar N, Allami A, Azeddoug H, Bendris A, Laraqui A, El Jaffali A, et al. Thermolabile methylenetetrahydrofolate reductase C677T polymorphism and homocysteine are risk factors for coronary artery disease in Moroccan population. J Biomed Biotechnol 2007(1); 80687.
8. Bahadır A, Eroz R, Türker Y. Does the MTHFR C677T gene polymorphism indicate cardiovascular disease risk in type 2 diabetes mellitus patients? Anatolian J Cardiol 2015; 15(7): 524–530.
9. Rahimi Z, Nomani H, Mozafari H, Vaisi-Raygani A, Madani H, Malek-Khosravi S, et al. Factor V G1691A, prothrombin G20210A and methylenetetrahydrofolate reductase polymorphism C677T are not associated with coronary artery disease and type 2 diabetes mellitus in western Iran. Blood Coagul Fibrinolysis 2009; 20(4): 252–256.
10. Mohammed NO, Ali IA, Elamin BK and Saeed BO. The association of methylenetetrahydrofolate reductase gene polymorphism and hyperhomocysteinaemia with coronary artery disease in Sudanese patients with type 2 diabetes. Poster at Focus 2017, Association of Clinical Biochemistry annual meeting.

The authors

Bakri Osman Saeed*1 PhD, MD, FRCPath, FRCP; Nisreen Osman Mohamed2 PhD
1Faculty of Medicine, Sudan International University, Khartoum, Sudan
2Ahfad Centre for Science and Technology, Ahfad University for Women, Khartoum, Sudan

*Corresponding author
E-mail: saeedbakri@hotmail.com

https://clinlabint.com/wp-content/uploads/sites/2/2020/08/C328_Saeed_fig1.jpg 706 1000 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:30MTHFR, hyperhomocysteinemia, CAD and T2DM

Literarure Review: Brain Biomarkers

, 26 August 2020/in Featured Articles /by 3wmedia

Prognostic value of molecular and imaging biomarkers in patients with supratentorial glioma

Lopci E, Riva M, Olivari L, Raneri F, Soffietti R, et al. Eur J Nucl Med Mol Imaging 2017; 44(7): 1155–1164

PURPOSE: We evaluated the relationship between 11C-methionine PET (11C-METH PET) findings and molecular biomarkers in patients with supratentorial glioma who underwent surgery.
METHODS: A consecutive series of 109 patients with pathologically proven glioma (64 men, 45 women; median age 43 years) referred to our Institution from March 2012 to January 2015 for tumour resection and who underwent preoperative 11C-METH PET were analysed. Semi-quantitative evaluation of the 11C-METH PET images included SUVmax, region of interest-to-normal brain SUV ratio (SUVratio) and metabolic tumour volume (MTV). Imaging findings were correlated with disease outcome in terms of progression-free survival (PFS), and compared with other clinical biological data, including IDH1 mutation status, 1p/19q codeletion and MGMT promoter methylation. The patients were monitored for a mean period of 16.7 months (median 13 months).
RESULTS: In all patients, the tumour was identified on 11C-METH PET. Significant differences in SUVmax, SUVratio and MTV were observed in relation to tumour grade (P<0.001). IDH1 mutation was found in 49 patients, 1p/19q codeletion in 58 patients and MGMT promoter methylation in 74 patients. SUVmax and SUVratio were significantly inversely correlated with the presence of IDH1 mutation (P<0.001). Using the 2016 WHO classification, SUVmax and SUVratio were significantly higher in patients with primary glioblastoma (IDH1-negative) than in those with other diffuse gliomas (P<0.001). Relapse or progression was documented in 48 patients (median PFS 8.7 months). Cox regression analysis showed that SUVmax and SUVratio, tumour grade, tumour type on 2016 WHO classification, IDH1 mutation status, 1p/19q codeletion and MGMT promoter methylation were significantly associated with PFS. None of these factors was found to be an independent prognostic factor in multivariate analysis.
CONCLUSION: 11C-METH PET parameters are significantly correlated with histological grade and IDH1 mutation status in patients with glioma. Grade, pathological classification, molecular biomarkers, SUVmax and SUVratio were prognostic factors for PFS in this cohort of patients. The trial was registered with ClinicalTrials.gov (registration: NCT02518061).

Expression of cell cycle regulators and biomarkers of proliferation and regrowth in human pituitary adenomas

Gruppetta M Formosa R, Falzon S, Ariff Scicluna S, Falzon E, et al. Pituitary 2017; 20(3): 358–371

PURPOSE: The pathogenesis of pituitary adenomas (PA) is complex. Ki-67, pituitary tumour transforming gene (PTTG), vascular endothelial growth factor (VEGF), cyclin D1, c-MYC and pituitary adenylate cyclase-activating peptide (PACAP) protein expression was analysed and correlated with tumour and patient characteristics.
METHODS: 74 pituitary tumour samples (48 non-functional PA, 26 functional PAs); immunohistochemical analysis of protein expression, retrospective analysis of MR images and in vitro analysis of octreotide treatment was carried out on GH3 cells.
RESULTS: PTTG expression was negatively associated with age and positively with PA size, regrowth and Ki-67 index. Cyclin D1 correlated with Ki-67 and tumour size. c-MYC negatively correlated with size of tumour and age, and correlated with PTTG expression. Somatostatin analogue treatment was associated with lower Ki-67, PTTG and cyclin D1 expression while T2 hypointense PAs were associated with lower PTTG, cyclin D1, c-MYC and Ki-67. In vitro analyses confirmed the effect of somatostatin analogue treatment on PTTG and cyclin D1 expression.
CONCLUSIONS: Interesting and novel observations on the differences in expression of tumour markers studied are reported. Correlation between Ki-67 expression, PTTG nuclear expression and recurrence/regrowth of PAs, emphasizes the role that Ki-67 and PTTG expression have as markers of increased proliferation. c-MYC and PTTG nuclear expression levels were correlated providing evidence that PTTG induces c-MYC expression in PAs and we propose that c-MYC might principally have a role in early pituitary tumorigenesis. Evidence is shown that the anti-proliferative effect of somatostatin analogue treatment in vivo occurs through regulation of the cell cycle.

Comparison of multiple tau PET measures as biomarkers in aging and Alzheimer’s Disease

Maass A, Landau S, Baker SL, Horng A, Lockhart SN, et al. Neuroimage 2017; 157: 448–463

The recent development of tau-specific positron emission tomography (PET) tracers enables in vivo quantification of regional tau pathology, one of the key lesions in Alzheimer’s disease (AD). Tau PET imaging may become a useful biomarker for clinical diagnosis and tracking of disease progression but there is no consensus yet on how tau PET signal is best quantified. The goal of the current study was to evaluate multiple whole-brain and region-specific approaches to detect clinically relevant tau PET signal. Two independent cohorts of cognitively normal adults and amyloid-positive (Aβ+) patients with mild cognitive impairment (MCI) or AD-dementia underwent [18F]AV-1451 PET. Methods for tau tracer quantification included: (i) in vivo Braak staging, (ii) regional uptake in Braak composite regions, (iii) several whole-brain measures of tracer uptake, (iv) regional uptake in AD-vulnerable voxels, and (v) uptake in a priori defined regions. Receiver operating curves characterized accuracy in distinguishing Aβ− controls from AD/MCI patients and yielded tau positivity cut-offs. Clinical relevance of tau PET measures was assessed by regressions against cognition and MR imaging measures. Key tracer uptake patterns were identified by a factor analysis and voxel-wise contrasts. Braak staging, global and region-specific tau measures yielded similar diagnostic accuracies, which differed between cohorts. While all tau measures were related to amyloid and global cognition, memory and hippocampal/entorhinal volume/thickness were associated with regional tracer retention in the medial temporal lobe. Key regions of tau accumulation included medial temporal and inferior/middle temporal regions, retrosplenial cortex, and banks of the superior temporal sulcus. Our data indicate that whole-brain tau PET measures might be adequate biomarkers to detect AD-related tau pathology. However, regional measures covering AD-vulnerable regions may increase sensitivity to early tau PET signal, atrophy and memory decline.

C-terminal fragments of the amyloid precursor protein in cerebrospinal fluid as potential biomarkers for Alzheimer disease

García-Ayllón MS, Lopez-Font I, Boix CP, Fortea J, Sánchez-Valle R, et al. Sci Rep. 2017; 7(1): 2477

This study assesses whether C-terminal fragments (CTF) of the amyloid precursor protein (APP) are present in cerebrospinal fluid (CSF) and their potential as biomarkers for Alzheimer’s disease (AD). Immunoprecipitation and simultaneous assay by Western blotting using multiplex fluorescence imaging with specific antibodies against particular domains served to characterize CTFs of APP in human CSF. We demonstrate that APP-CTFs are detectable in human CSF, being the most abundant a 25-kDa fragment, probably resulting from proteolytic processing by η-secretase. The level of the 25-kDa APP-CTF was evaluated in three independent CSF sample sets of patients and controls. The CSF level of this 25-kDa CTF is higher in subjects with autosomal dominant AD linked to PSEN1 mutations, in demented Down syndrome individuals and in sporadic AD subjects compared to age-matched controls. Our data suggest that APP-CTF could be a potential diagnostic biomarker for AD.

Blood-based biomarkers for the identification of sports-related concussion

Anto-Ocrah M, Jones CMC, Diacovo D, Bazarian JJ. Neurol Clin 2017; 35(3): 473–485

Sports-related concussions (SRCs) are common among athletes in the United States. Most athletes who sustain an SRC recover within 7 to 10 days; however, many athletes who sustain the injury do not recover as expected and experience prolonged, persistent symptoms. In this document, the authors provide an overview of the empirical evidence related to the use of blood-based brain biomarkers in the athlete population for diagnosis of SRCs, prognosis of recovery and return to play guidelines, and indications of neurodegeneration. The authors also provide a summary of research challenges, gaps in the literature, and future directions for research.

Brain biomarkers and pre-injury cognition are associated with long-term cognitive outcome in children with traumatic brain injury

Wilkinson AA, Dennis M, Simic N, Taylor MJ, Morgan BR, et al. BMC Pediatr 2017; 17(1): 173

BACKGROUND: Children with traumatic brain injury (TBI) are frequently at risk of long-term impairments of attention and executive functioning but these problems are difficult to predict. Although deficits have been reported to vary with injury severity, age at injury and sex, prognostication of outcome remains imperfect at a patient-specific level. The objective of this proof of principle study was to evaluate a variety of patient variables, along with six brain-specific and inflammatory serum protein biomarkers, as predictors of long-term cognitive outcome following pediatric TBI.
METHOD: Outcome was assessed in 23 patients via parent-rated questionnaires related to attention deficit hyperactivity disorder (ADHD) and executive functioning, using the Conners 3rd Edition Rating Scales (Conners-3) and Behaviour Rating Inventory of Executive Function (BRIEF) at a mean time since injury of 3.1 years. Partial least squares (PLS) analyses were performed to identify factors measured at the time of injury that were most closely associated with outcome on (1) the Conners-3 and (2) the Behavioural Regulation Index (BRI) and (3) Metacognition Index (MI) of the BRIEF.
RESULTS: Higher levels of neuron specific enolase (NSE) and lower levels of soluble neuron cell adhesion molecule (sNCAM) were associated with higher scores on the inattention, hyperactivity/impulsivity and executive functioning scales of the Conners-3, as well as working memory and initiate scales of the MI from the BRIEF. Higher levels of NSE only were associated with higher scores on the inhibit scale of the BRI.
CONCLUSIONS: NSE and sNCAM show promise as reliable, early predictors of long-term attention-related and executive functioning problems following pediatric TBI.

Biomarkers of stroke recovery: Consensus-based core recommendations from the Stroke Recovery and Rehabilitation Roundtable

Boyd LA, Hayward KS, Ward NS, Stinear CM, Rosso C, et al. Int J Stroke 2017; 12(5): 480-493

The most difficult clinical questions in stroke rehabilitation are “What is this patient’s potential for recovery?” and “What is the best rehabilitation strategy for this person, given her/his clinical profile?” Without answers to these questions, clinicians struggle to make decisions regarding the content and focus of therapy, and researchers design studies that inadvertently mix participants who have a high likelihood of responding with those who do not. Developing and implementing biomarkers that distinguish patient subgroups will help address these issues and unravel the factors important to the recovery process. The goal of the present paper is to provide a consensus statement regarding the current state of the evidence for stroke recovery biomarkers. Biomarkers of motor, somatosensory, cognitive and language domains across the recovery timeline post-stroke are considered; with focus on brain structure and function, and exclusion of blood markers and genetics. We provide evidence for biomarkers that are considered ready to be included in clinical trials, as well as others that are promising but not ready and so represent a developmental priority. We conclude with an example that illustrates the utility of biomarkers in recovery and rehabilitation research, demonstrating how the inclusion of a biomarker may enhance future clinical trials. In this way, we propose a way forward for when and where we can include biomarkers to advance the efficacy of the practice of, and research into, rehabilitation and recovery after stroke.

Brain biomarkers of vulnerability and progression to psychosis

Cannon TD. Schizophr Bull 2016; 42(Suppl 1): S127–132

Identifying predictors and elucidating the fundamental mechanisms underlying onset of psychosis are critical for the development of targeted pre-emptive interventions. This article presents a selective review of findings on risk prediction algorithms and potential mechanisms of onset in youth at clinical high-risk for psychosis, focusing principally on recent findings of the North American Prodrome Longitudinal Study (NAPLS). Multivariate models incorporating risk factors from clinical, demographic, neurocognitive, and psychosocial assessments achieve high levels of predictive accuracy when applied to individuals who meet criteria for a prodromal risk syndrome. An individualized risk calculator is available to scale the risk for newly ascertained cases, which could aid in clinical decision making. At risk individuals who convert to psychosis show elevated levels of proinflammatory cytokines, as well as disrupted resting state thalamo-cortical functional connectivity at baseline, compared with those who do not. Further, converters show a steeper rate of grey matter reduction, most prominent in prefrontal cortex, that in turn is predicted by higher levels of inflammatory markers at baseline. Microglia, resident immune cells in the brain, have recently been discovered to influence synaptic plasticity in health and impair plasticity in disease. Processes that modulate microglial activation may represent convergent mechanisms that influence brain dysconnectivity and risk for onset of psychosis and thus may be targetable in developing and testing preventive interventions.

https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 0 0 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:42Literarure Review: Brain Biomarkers
27559 Biohit CLI ad 92x132 271017

GastroPanel Quick Test

, 26 August 2020/in Featured Articles /by 3wmedia
https://clinlabint.com/wp-content/uploads/sites/2/2020/08/27559-Biohit-CLI_ad_92x132_271017.jpg 1500 1051 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:33GastroPanel Quick Test
27534 Hologic Panther Fusion CLI Ad CROPS

Join the evolution – Panther Fusion

, 26 August 2020/in Featured Articles /by 3wmedia
https://clinlabint.com/wp-content/uploads/sites/2/2020/08/27534-Hologic-Panther-Fusion-CLI-Ad-CROPS.jpg 1500 1017 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:45Join the evolution – Panther Fusion
27439 Greiner 140x204 MiniCollect System 2017 2 en print

MiniCollect® Capillary Blood Collection System

, 26 August 2020/in Featured Articles /by 3wmedia
https://clinlabint.com/wp-content/uploads/sites/2/2020/08/27439-Greiner-140x204_MiniCollect-System-2017-2_en_print.jpg 1500 1028 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:27MiniCollect® Capillary Blood Collection System
27530 Messe Duesseldorf 2017 08 01 MEDICA 2017 International Labor 92 x 270mm Clinical Laborator.

Medica 2017, 13-16 November, Düsseldorf

, 26 August 2020/in Featured Articles /by 3wmedia
https://clinlabint.com/wp-content/uploads/sites/2/2020/08/27530-Messe-Duesseldorf-2017_08_01_MEDICA_2017_International_Labor_92_x_270mm_Clinical_Laborator..jpg 1500 508 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:46Medica 2017, 13-16 November, Düsseldorf
27450 Randox QC CLI Full Page Advert June Issue 2017

Complete QC solutions for results you can trust

, 26 August 2020/in Featured Articles /by 3wmedia
https://clinlabint.com/wp-content/uploads/sites/2/2020/08/27450-Randox-QC-CLI-Full-Page-Advert-June-Issue-2017.jpg 1500 1061 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:49Complete QC solutions for results you can trust
C327 Parfitt Fig

Chest pain and elevated cardiac troponin: a typical case of myocardial infarction? Perhaps not…

, 26 August 2020/in Featured Articles /by 3wmedia

Chest pain is a common presentation in Emergency Medicine and can cause a diagnostic dilemma. Does the patient need urgent admission for percutaneous coronary intervention following a myocardial infarction? Or will discharge with reassurance and a bottle of antacids sort out their indigestion? Measurement of cardiac troponin has become an essential part of emergency assessment of chest pain. Despite this, clinicians and laboratory professionals should be conscious that elevated cardiac troponin is not inevitably indicative of cardiac pathology – and in some cases, may be deceptive…

by Ceri Parfitt and Dr Christopher Duff

Troponin and assessment of acute chest pain
The Universal Definition of Myocardial Infarction and Injury has incorporated measurement of cardiac troponin (cTn) since 2000, with the most recent revision in 2012 [1]. In the UK, the National Institute of Health and Care Excellence (NICE) the ‘Chest pain of recent onset: assessment and diagnosis’ pathway [2] is well established in clinical practice, and Emergency Department clinicians are confident in the use of cTn assays to identify patients with acute coronary syndrome (ACS).

The troponin complex comprises three regulatory proteins – troponin C, troponin T and troponin I – essential for skeletal and cardiac muscle contraction, through regulation of actin/myosin filaments. Cardiac-specific isoforms of both troponin T (cTnT) and troponin I (cTnI) have been identified. cTn complexes can be detected in the blood within 2–3 hours of myocardial damage, peak within 24 hours and persist for 1–2 weeks [3, 4]. This characteristic pattern of release and specificity for cardiac injury has cemented cTn as the biomarker of choice for diagnosis of ACS, replacing creatinine kinase, aspartate transaminase and lactate dehydrogenase. Measurement of cTn is a standard assay available on an urgent basis in the majority of modern clinical laboratories. Both cTnT and cTnI can be measured by two-site ‘sandwich’ immunoassay, based on formation of complexes of cTn, a ‘capture’ anti-cTn antibody and a ‘label’ anti-cTn antibody (Fig. 1a). Owing to patent regulations, Roche Diagnostics is the sole distributor of cTnT immunoassays, whereas various companies offer cTnI immunoassays on several different platforms. Although results cannot be directly compared, the two assays largely provide the same diagnostic information.

A patient presents with chest pain and raised troponin
Despite the widespread use of cTn in Emergency Departments worldwide, clinicians may often be unaware of spurious causes of raised cTn. Application of the pathway without considering patient history and previous presentations can potentially lead to invasive, unnecessary (and expensive) investigations. We recently encountered an example of this in our local Emergency Department. A 34-year-old male with no significant cardiac risk factors repeatedly presented to the Emergency Department complaining of chest pain over a period of 3 years, resulting in more than 40 individual analyses of cTnI (using the Siemens TnI-Ultra assay on ADVIA Centaur XP). The results were variable, but always elevated (48–4030 ng/L, decision limit 40 ng/L). The patient underwent thorough investigation, but no cardiac pathology was ever identified. The Cardiology Department contacted the laboratory to discuss the apparent mismatch between patient presentation and biochemical findings.

Has there been a laboratory error?
Spurious results can arise for a number of reasons, including mislabelled samples, sample contamination, poor sample quality, analyser malfunction, calibration/quality control errors and transcription errors [5]. Where possible, these factors are minimized by stringent sampling techniques and robust laboratory procedures, or can be identified through the technical and clinical validation processes, and highlighted to the clinical team by laboratory staff. In many cases, a simple repeat sample is sufficient to confirm/rule out error. However, clinicians should always be vigilant for results that are not consistent with patient presentation. The persistent nature of the elevated cTnI in this case – over a period of 3 years – rules out sporadic errors, such as mislabelled samples or analyser malfunction. Furthermore, no other patients with unexpectedly high cTnI concentrations were discussed with the laboratory. These factors strongly suggested that the solution to the mystery lay within the patient, rather than in management of their samples.
Alternative causes of elevated troponin: cardiac and non-cardiac
Although elevated cTn is typically associated with myocardial infarction, other cardiac pathologies also cause cTn release into the circulation through direct or hypoxic myocyte damage. These include aortic dissection, pericarditis, myocarditis, acute heart failure and cardiac contusion following trauma [4, 6]. Unsurprisingly, cardiac interventions such as defibrillator shocks, coronary angioplasty, percutaneous coronary intervention and open heart surgery are also associated with variable increase in cTn [6]. Chemotherapy agents, including anthracyclines, alkylating agents, anti-metabolites and anti-microtubules are all associated with toxic damage to myocytes [4].

In addition to cardiac sources of cTn, non-cardiac sources must also be considered. A significant proportion (estimated at 36–85 %) of patients admitted to critical care units for sepsis/systemic inflammatory response syndromes (SIRS) have elevated cTn concentrations. In these patients, cTn release is thought to be a consequence of oxygen supply-demand mismatch in the myocardium: although oxygen demand is increased, due to fever and tachycardia, systemic hypoxemia reduces oxygen supply due to respiratory failure, circulatory dysfunction and hypotension [7, 8]. Patients with end-stage renal disease (ESRD) are often noted to have chronically elevated cTn. The underlying cause for this is unclear, but various hypotheses have been presented, including reduced renal clearance of cTn microfragments, sub-clinical cardiac disease, and metabolic abnormalities [9]. cTn concentrations are also found to be elevated in patients with acute pulmonary embolism. Again, the cause has not been conclusively established, but it is believed that abrupt increases in pulmonary artery pressure leads to dilatation of the right ventricle, with associated myocyte injury [4].

For most patients, any of these situations can be identified by examining the patient records, or through further cardiac investigations. In the case presented here, after thorough analysis of the patient’s history, and given the resolutely normal cardiac investigations, we concluded that it was unlikely that the elevation in cTn was due to pathological factors, whether cardiac or non-cardiac. This led us to examine the local cTnI assay – and the patient’s samples – in more detail, to ascertain the cause of the spurious results.

Could immunoassay interference be involved?
Non-pathological causes of false positives in cTn analysis include human anti-mouse antibodies (HAMA), rheumatoid factor (RF) and heterophilic antibodies [10–12]. HAMA are characterized by strong avidity to well-defined antigens. They are thought to develop following exposure to mouse immunoglobulins, which may occur through monoclonal antibody therapy, vaccination, infection, blood transfusion, or simply through animal handling [10]. In contrast, heterophilic antibodies are endogenous antibodies present in serum/plasma produced against poorly defined antigens. They often have weak affinity, but multiple specificities. Heterophilic antibodies may develop following infection and are thought to affect up to 30% of the population [10, 12, 13]. RFs are endogenous human antibodies with heterophilic activity, often arising following autoimmune disease [12].

Genuine assay interference is less commonly encountered these days, with the development of highly specialized commercial assays. Most modern immunoassays contain agents able to block low concentrations of interfering proteins. However, the fact that interference is less frequently observed may lead laboratory staff members to believe that interference is less prevalent in the general population. This is not the case. In fact, the prevalence of assay interference may be increasing, as immunotherapy and the use of radiolabelled antibodies are established in routine practice [13]. Although heterophilic antibodies usually have little clinical significance in vivo, their ability to interfere with two-site immunoassays can have major effects on patient management. The magnitude of interference varies between samples and may even vary within a patient over time [13]. The mechanism for antibody interference is complex and has not been fully elucidated, but is likely to involve ‘bridge’ formation between the capture antibody and label antibody (Fig. 1b) [13, 14]. Several standard techniques are available to investigate assay interference [12, 13]. In this case, we used dilution studies, polyethylene glycol precipitation, heterophilic antibody blocking tubes and an alternative assay method. In all cases, samples from the patient were compared with a control sample with a similar cTnI concentration, from a patient with confirmed ACS.

Investigations into immunoassay interference
Serial dilution of samples should produce a linear decrease in concentration if interfering species are absent. However, if interfering substances are present, then non-linear recovery following dilution is commonly observed, as the interferent is ‘diluted out’. Both the patient’s serum and control serum were diluted with saline, and cTnI was measured in each dilution. As expected, the cTnI concentration in the control sample decreased proportionally to the dilution factor. In contrast, any dilution of the patient sample resulted in undetectable cTnI, consistent with presence of an interfering species (Fig. 2a). Immunoglobulins can be removed by treatment with polyethylene glycol (PEG). PEG forms poorly soluble complexes with immunoglobulins, which can be precipitated then removed by centrifugation, before the sample is re-analysed. In the patient sample, PEG precipitation resulted in undetectable levels of cTnI. Levels were also reduced in a control sample (62% recovery), though it is not uncommon to see at least some reduction in recovery using this approach, regardless of whether interfering immunoglobulins are present or not (Fig. 2b). Heterophilic blocking reagents prevent non-analyte mediated bridging of antibodies by heterophilic interference. Incubation of a patient sample in a heterophilic blocking tube (Scantibodies Laboratory Inc.) before cTnI analysis resulted in 4% recovery of cTnI, suggesting analytical interference. In the control sample, 94% of cTnI was recovered (Fig. 2c). Finally, a patient sample was sent to a referral laboratory for analysis of cTnT, as opposed to cTnI. A cTnT concentration below the decision limit (99th percentile) was obtained (cTnI result: 398 ng/L; cTnT result: 6 ng/L) (Fig. 3).

As initial investigations strongly suggested the presence of an assay interferent, further work was conducted to identify the precise nature of the interfering species. Two patient samples with elevated cTnI were analysed for presence of HAMAs, which was negative in both cases. This was not unexpected, given that the patient had no history of exposure to animal proteins in either a therapeutic or a social setting. The same sample was tested for RFs, using the local assay (Orgentec RF IgM), which returned a slightly elevated result. This slightly elevated concentration is not considered clinically significant, but may contribute to interference in the assay. Thus, heterophilic antibody interference remains the most likely cause of cTnI elevation in this patient. Further investigations have been limited, however, by recently decreasing concentrations of cTnI in this patient. Unpredictable variation in heterophilic antibody activity is a known phenomenon however, and the laboratory continues to oversee samples received from the patient.

Outcome and conclusions
The patient continues to regularly attend the Emergency Department with chest pain. No cardiac pathology has ever been identified in this patient and  his chest pain has been attributed to a combination of gastro-oesophageal reflux disease and health anxiety. Senior review is now required in the Emergency Department before the patient is investigated or admitted. If ACS is ever suspected in this patient, the laboratory is able to pre-treat samples with heterophilic blocking tubes to provide an estimate of cTnI concentration, with referral for cTnT analysis required for a definitive result. This alternate pathway ensures that the patient is protected in case of a genuine cardiac event.

This case demonstrates that clinicians must be aware of assay interference, and highlights the benefit of discussing patients with the laboratory when test results do not correlate with clinical presentation. Laboratory staff members are rarely able to visit patients and, thus, are not in a position to suspect interference without input from the clinical team. In this case, failure to identify the underlying cause of elevated cTnI at an early stage resulted in a number of unnecessary and invasive investigations, significant costs to the NHS, and continued anxiety to the patient.

References
1. Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, et al. Third universal definition of myocardial infarction. Eur Heart J 2012; 33: 2551–2567.
2. National Institute of Health and Care Excellence (NICE). Chest pain of recent onset: assessment and diagnosis. NICE clinical guideline 95, 2010 (https://www.nice.org.uk/guidance/cg95).
3. Wu AHB. Analytical issues for clinical use of cardiac troponin. In: Morrow DA (ed.) Contemporary cardiology: cardiovascular biomarkers: pathophysiology and disease management. Humana 2006, pp 27–40.
4. Korff S, Katus HA, Giannitsis E. Differential diagnosis of elevated troponins. Heart 2006; 92: 987–993.
5. Plebani M. The detection and prevention of errors in laboratory medicine. Ann Clin Biochem 2010; 47: 101–110.
6. Sara JDS, Holmes DR, Jaffe AS. Fundamental concepts of effective troponin use: important principles for internists. Am J Med 2015; 128: 111–119.
7. Ver Elst KM, Spapen HD, Nguyen DN, Garbar C, Huyghens LP, Gorus FK. Cardiac troponins I and T are biological markers of left ventricular dysfunction in septic shock 2000; 46: 650–657.
8. Spies C, Haude V, Fitzner R, Schroder K, Overbeck M, Runkel N, Schaffartzik W. Serum cardiac troponin T as a prognostic marker in early sepsis. Chest 1998; 113: 1055–1063.
9. Jaffe AS. The 10 commandments of troponin with special reference to high sensitivity assays. Heart 2011; 97: 940–946.
10. Lippi G, Aloe R, Meschi T, Borghi L, Cervellin G. Interference from heterophilic antibodies in troponin testing. Case report and systematic review of the literature. Clin Chim Acta 2013; 426: 79–84.
11. Makaryus AN, Markaryus MN, Hassid B. Falsely elevated cardiac troponin I levels. Clin Cardiol 2007; 30: 92–94.
12. Zaidi A, Cowell R. False positive cardiac troponin elevation due to heterophile antibodies: more common than we recognise? BMJ Case Rep 2010; 2010: bcr1120092477.
13. Ismail AAA, Walker PL, Cawood ML, Barth JH. Interference in immunoassay is an underestimated problem. Ann Clin Biochem 2002; 39: 266–373.
14. Kaplan IV, Levinson SS. When is a heterophile antibody not a heterophile antibody? When it is an antibody against a specific immunogen. Clin Chem 1999; 45: 616–618.

The authors
Ceri Parfitt MSc, Christopher Duff* PhD
Department of Clinical Biochemistry, Pathology Directorate, Royal Stoke University Hospital, University Hospitals of North Midlands NHS Trust, Staffordshire, UK


*Corresponding author
E-mail: Chris.duff@uhnm.nhs.uk

https://clinlabint.com/wp-content/uploads/sites/2/2020/08/C327_Parfitt_Fig__.jpg 523 1000 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:30Chest pain and elevated cardiac troponin: a typical case of myocardial infarction? Perhaps not…
C320 C

Clostridium difficile diagnosis: not always a bed of roses

, 26 August 2020/in Featured Articles /by 3wmedia

Clostridium difficile is a leading cause of nosocomial diarrhoea and one of the most common healthcare-associated infections. A dramatic worldwide increase in the incidence of C. difficile infection has occurred over the past two decades with the emergence of hypervirulent strains. Accurate and timely laboratory diagnosis of C. difficile infection is fundamental to ensure patients receive appropriate treatment and proper counter-infection measures are put in place. However, the availability of many commercial tests with different C. difficile targets contributes to uncertainty and controversy around the optimal diagnostic algorithm.

by Dr Guilherme Grossi Lopes Cançado, Prof. Rodrigo Otávio Silveira Silva and Prof. Eduardo Garcia Vilela

Introduction
Clostridium difficile is a major cause of healthcare-associated diarrhoea, and is linked to significant morbidity, economic burden and even mortality. Disagreement between diagnostic tests is an ongoing barrier to clinical decision-making and epidemiological surveillance. With the increasing number and severity of Clostridium difficile infections (CDI) after the emergence of the epidemic BI/NAP1/027 strain in the 2000s, there has been a renewed interest in optimizing the laboratory diagnosis of this infection.

Testing for C. difficile
Cytotoxin neutralization, toxigenic culture and toxin enzyme immunoassay

The two most accurate methods for CDI diagnosis are cytotoxin neutralization (CTN) and toxigenic culture (TC). CTN has long been used as a reference method for C. difficile detection, although different protocols have been proposed. Basically, this technique consists of inoculating a filtrate of a stool suspension into a cell culture (Vero, Hep2, fibroblasts, CHO or HeLa cells) and observing a cytopathic effect (such as cell rounding) as a consequence of disruption of the cell cytoskeleton 24–48 h later. CDI confirmation is obtained by the addition of a specific antiserum directed against C. difficile or against C. sordellii, neutralizing the toxin effects. Studies performed in the last 10 years, however, demonstrated that the sensitivities of CTN protocols range between 60 and 86%, when compared with toxigenic culture. Although culture was found to be the most sensitive method for detecting C. difficile in feces, it is not very specific owing to the possibility of isolating non-toxigenic strains. In order to overcome this issue, it should always be combined with a toxin detection method, such as enzyme immunoassay (EIA), direct cytopathic effect on cell lines or the identification of toxin-related genes by PCR. This notwithstanding, both techniques are time consuming, laborious and require trained personnel, reasons why they are not frequently used in daily practice [1].

In this context, commercial rapid EIA and DNA-based tests are currently the most widely used tools for CDI diagnosis. EIA for toxin detection is fast, cheap, easy to perform and does not require technical training or special equipment: characteristics which have favored its use in low income countries. However, several studies have reported low detection sensitivities of different commercial kits (approximately 50–70%), making toxin EIAs inadequate as standalone tests [2]. Some authors have suggested the analysis of sequential samples from the same patient, but this practice did not significantly increase the positive predictive value of the test and may even amplify the rate of false positives. Furthermore, toxin tests can be falsely negative, which can be due to previous antibiotic treatment, pre-analytic toxin degradation or sampling error (e.g., ileus, fecal dilution). Consequently, toxin EIA may be sufficient and cost-effective as a screening test in clinical settings where there is a low prevalence of CDI, but not for epidemic scenarios.

Detection of glutamate dehydrogenase
Another method that has been used to diagnose CDI is the detection of glutamate dehydrogenase (GDH), a metabolic enzyme constitutionally produced almost exclusively by C. difficile, which is significantly more sensitive than toxin A and B EIAs. We have recently shown that GDH rapid immunoassay presents 100% sensitivity and negative predictive value, compared with the culture-based method, in accordance with order authors [3]. Cheng et al. have advocated the use of GDH in order to improve the diagnostic capacity and control of potential outbreaks of CDI in developing countries, as this test is five to ten times cheaper than molecular assays [4]. In fact, we have recently reported a significant increase in CDI treatment rates in a university hospital of Brazil (from 53.8% to 100%) simply after replacing the toxin EIA by the rapid GDH immunoassay as a screening test [3]. This finding supports the use of GDH in countries with limited economic resources, as it is simple to perform and, unlike PCR, requires no special facilities or personnel qualifications that might restrict its use. Although sensitive, GDH lacks specificity, because it only indicates the presence of the microorganism, instead of toxin production. In this context, a concern with the overdiagnosis and overtreatment of patients with diarrhoea and C. difficile colonization may arise. A negative test can almost rule out CDI and avoid the need for more expensive toxin testing, but a positive GDH assay should preferably be followed by toxin-based diagnostic methods before one can come to a safe conclusion on infection. Asymptomatic carriage of C. difficile occurs in 5–15% of healthy adults, but may be as high as 90% in newborns and healthy infants, and up to 51% in residents in long-term care facilities [5]. In this way, the incorporation of a two-step strategy, including a sensitive organism-based test for screening, such as GDH, followed by a toxin test for confirmation of clinically significant disease, is a reasonable approach. Combined tests including GDH and toxin detection in one easy-to-use cartridge have been recently developed, but several authors have demonstrated limited sensitivity of the toxin component. In our study, the toxin component presented an even lower sensitivity than conventional toxin EIA (50% vs 58%), compared with toxigenic culture [3]. In this way, GDH+/toxin− samples would still have to be submitted to a third test (in a multistep algorithm) to rule out infection, increasing time to diagnosis and health costs. However, some studies have shown that, even without treatment, patients with toxin-negative stool specimens have shorter diarrhoea duration than those with toxin-positive stool specimens. These findings may suggest a limited need for CDI treatment for GDH-positive patients and toxin-negative stool specimens [6].

Molecular testing and rapid diagnosis
In 2009, facing an epidemic of CDI due to hypervirulent C. difficile strains, the US FDA cleared the first commercial molecular test for rapid CDI diagnosis [7]. Shortly afterwards, hospitals in North America and Europe began switching to DNA-based testing strategies as a method of choice for the diagnosis of C. difficile. Nucleic acid amplification testing (NAAT), including rapid testing PCR and loop-mediated isothermal amplification (LAMP), can detect the tcdA/tcdB genes (regulate toxin A/B production) or the tcdC gene (a negative regulator of toxin A and B production) and identify the presence of toxigenic C. difficile in a single step. It has a higher sensitivity (90–95%) and specificity (95–96%) than toxin EIAs and has a rapid turnaround time, but requires specialized equipment and personnel. It is worth noting that the Gene-Xpert® system can also simultaneously indicate the presence of the potentially ‘hypervirulent’ ribotype 027 strain, giving important epidemiological information. Nonetheless, although a PCR assay can identify toxin genes, it cannot detect the presence of toxin. Several studies have shown that toxin−/C. difficile+ patients present shorter duration of symptoms and better outcomes than toxin+/C. difficile+ individuals, demonstrating that this subpopulation may have either mild CDI or colonization, and even not need to be treated. In this way, some authors have questioned the use molecular tests as standalone tests because of the high likelihood of overdiagnosis and overtreatment [8]. However, there may be a role for identifying carriers to prevent transmission and this issue should be better addressed in future studies.

C. difficile testing protocol
Using reliable and rapid diagnostic tests, such as NAAT, practitioners could offer appropriate treatment earlier, thereby sparing patients a time-consuming evaluation and unnecessary antibiotic therapy and its complications. Despite worldwide advances in analytical technology, transport systems and computerization, many laboratories in developing countries have difficulties in improving turnaround times and diagnostic capability. The optimal diagnostic algorithm for CDI is yet to be adequately defined and may vary according to the underlying clinical and laboratory circumstances. We believe that the decision about which test(s) to use is determined by a combination of what is practical and feasible in a specific setting. In Brazil, for example, where only toxin EIA was available for CDI diagnosis until 2015, the introduction of the GDH test resulted in a dramatic increase in C. difficile treatment rates [3]. All of the reference methods, CTN, toxigenic culture, or PCR, require advanced infrastructure and expensive testing. Smaller, community-based hospitals, where much of C. difficile testing is currently performed, may not have the financial means to establish these methods nor have the staff to perform the time-consuming, highly complex assays. In this way, using GDH–toxin A/B assays may be an adequate option for diagnostic algorithms in developing countries, whereas molecular techniques, toxigenic culture or CTN may be reserved for discordant samples (Figure 1). Future studies should focus on developing simple diagnostic approaches to accurately distinguish active infection from mere colonization.

References
1. Delmée M. Laboratory diagnosis of Clostridium difficile disease. Clin Microbiol Infect 2001; 7(8): 411–416.
2. Silva RO, Vilela EG, Neves MS, Lobato FC. Evaluation of three enzyme immunoassays and a nucleic acid amplification test for the diagnosis of Clostridium difficile-associated diarrhea at a university hospital in Brazil. Rev Soc Bras Med Trop 2014; 47(4): 447–450.
3. Cançado GGL, Silva ROS, Nader AP, Lobato FCF, Vilela EG. Impact of simultaneous glutamate dehydrogenase (GDH) and toxin A/B rapid immunoassay on Clostridium difficile diagnosis and treatment in hospitalized patients with antibiotic-associated diarrhea in a university hospital of Brazil. J Gastroenterol Hepatol 2017; doi: 10.1111/jgh.13901 [Epub ahead of print].
4. Cheng JW, Xiao M, Kudinha T, Xu ZP, Sun LY, Hou X, Zhang L, Fan X, Kong F, Xu YC. The role of glutamate gehydrogenase (GDH) testing assay in the diagnosis of Clostridium difficile infections: a high sensitive screening test and an essential step in the proposed laboratory diagnosis workflow for developing countries like China. PLoS One 2015; 10(12): e0144604.
5. Furuya-Kanamori L, Marquess J, Yakob L, Riley TV, Paterson DL, Foster NF, Huber CA, Clements AC. Asymptomatic Clostridium difficile colonization: epidemiology and clinical implications. BMC Infect Dis 2015; 15: 516.
6. Yuhashi K, Yagihara Y, Misawa Y, Sato T, Saito R, Okugawa S, Moriya K. Diagnosing Clostridium difficile-associated diarrhea using enzyme immunoassay: the clinical significance of toxin negativity in glutamate dehydrogenase-positive patients. Infect Drug Resist 2016; 9: 93–99.
7. Polage CR, Turkiewicz JV, Cohen SH. The never-ending struggle with laboratory testing for Clostridium difficile infection. J Comp Eff Res 2016; 5(2): 113–116.
8. Polage CR, Gyorke CE, Kennedy MA, Leslie JL, Chin DL, Wang S, Nguyen HH, Huang B, Tang YW, Lee LW, Kim K, Taylor S, Romano PS, Panacek EA, Goodell PB, Solnick JV, Cohen SH. Overdiagnosis of Clostridium difficile infection in the molecular test era. JAMA Intern Med 2015; 175(11): 1792–1801.

The authors
Guilherme Grossi Lopes Cançado*1,2 MD, Rodrigo Otávio Silveira Silva3 PhD, and Eduardo Garcia Vilela1 MD, PhD
1Instituto Alfa de Gastroenterologia,
Federal University of Minas Gerais, Brazil.
2Department of Gastroenterology,
Hospital da Polícia Militar de Minas Gerais, Brazil.
3Veterinary School, Federal University of Minas Gerais, Brazil.

*Corresponding author
E-mail: guilhermegrossi@terra.com.br

https://clinlabint.com/wp-content/uploads/sites/2/2020/08/C320_C.jpg 1068 1000 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:43Clostridium difficile diagnosis: not always a bed of roses
27475 Coris Insertion CLI 2017 10 18

RESIST – get precise identifictaion of your CPO

, 26 August 2020/in Featured Articles /by 3wmedia
https://clinlabint.com/wp-content/uploads/sites/2/2020/08/27475-Coris-Insertion-CLI-2017_10_18.jpg 686 1500 3wmedia https://clinlabint.com/wp-content/uploads/sites/2/2020/06/clinlab-logo.png 3wmedia2020-08-26 09:41:022021-01-08 11:34:33RESIST – get precise identifictaion of your CPO
Page 117 of 144«‹115116117118119›»
Bio-Rad - Preparing for a Stress-free QC Audit

Latest issue of Clinical laboratory

November 2025

CLi Cover nov 2025
13 November 2025

New Chromsystems Product for Antiepileptic Drugs Testing

11 November 2025

Trusted analytical solutions for reliable results

10 November 2025

Chromsystems | Therapeutic Drug Monitoring by LC-MS/MS

Digital edition
All articles Archived issues

Free subscription

View more product news

Get our e-alert

The leading international magazine for Clinical laboratory Equipment for everyone in the Vitro diagnostics

Sign up today
  • News
    • Featured Articles
    • Product News
    • E-News
  • Magazine
    • About us
    • Archived issues
    • Free subscriptions
    • Media kit
    • Submit Press Release
clinlab logo blackbg 1

Prins Hendrikstraat 1
5611HH Eindhoven
The Netherlands
info@clinlabint.com

PanGlobal Media is not responsible for any error or omission that might occur in the electronic display of product or company data.

Scroll to top

This site uses cookies. By continuing to browse the site, you are agreeing to our use of cookies.

Accept settingsHide notification onlyCookie settings

Cookie and Privacy Settings



How we use cookies

We may ask you to place cookies on your device. We use cookies to let us know when you visit our websites, how you interact with us, to enrich your user experience and to customise your relationship with our website.

Click on the different sections for more information. You can also change some of your preferences. Please note that blocking some types of cookies may affect your experience on our websites and the services we can provide.

Essential Website Cookies

These cookies are strictly necessary to provide you with services available through our website and to use some of its features.

Because these cookies are strictly necessary to provide the website, refusing them will affect the functioning of our site. You can always block or delete cookies by changing your browser settings and block all cookies on this website forcibly. But this will always ask you to accept/refuse cookies when you visit our site again.

We fully respect if you want to refuse cookies, but to avoid asking you each time again to kindly allow us to store a cookie for that purpose. You are always free to unsubscribe or other cookies to get a better experience. If you refuse cookies, we will delete all cookies set in our domain.

We provide you with a list of cookies stored on your computer in our domain, so that you can check what we have stored. For security reasons, we cannot display or modify cookies from other domains. You can check these in your browser's security settings.

.

Google Analytics Cookies

These cookies collect information that is used in aggregate form to help us understand how our website is used or how effective our marketing campaigns are, or to help us customise our website and application for you to improve your experience.

If you do not want us to track your visit to our site, you can disable this in your browser here:

.

Other external services

We also use various external services such as Google Webfonts, Google Maps and external video providers. Since these providers may collect personal data such as your IP address, you can block them here. Please note that this may significantly reduce the functionality and appearance of our site. Changes will only be effective once you reload the page

Google Webfont Settings:

Google Maps Settings:

Google reCaptcha settings:

Vimeo and Youtube videos embedding:

.

Privacy Beleid

U kunt meer lezen over onze cookies en privacy-instellingen op onze Privacybeleid-pagina.

Privacy policy
Accept settingsHide notification only

Subscribe now!

Become a reader.

Free subscription