Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
BMC Infect Dis ; 24(1): 1139, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39390446

ABSTRACT

We investigate the emergence, mutation profile, and dissemination of SARS-CoV-2 lineage B.1.214.2, first identified in Belgium in January 2021. This variant, featuring a 3-amino acid insertion in the spike protein similar to the Omicron variant, was speculated to enhance transmissibility or immune evasion. Initially detected in international travelers, it substantially transmitted in Central Africa, Belgium, Switzerland, and France, peaking in April 2021. Our travel-aware phylogeographic analysis, incorporating travel history, estimated the origin to the Republic of the Congo, with primary European entry through France and Belgium, and multiple smaller introductions during the epidemic. We correlate its spread with human travel patterns and air passenger data. Further, upon reviewing national reports of SARS-CoV-2 outbreaks in Belgian nursing homes, we found this strain caused moderately severe outcomes (8.7% case fatality ratio). A distinct nasopharyngeal immune response was observed in elderly patients, characterized by 80% unique signatures, higher B- and T-cell activation, increased type I IFN signaling, and reduced NK, Th17, and complement system activation, compared to similar outbreaks. This unique immune response may explain the variant's epidemiological behavior and underscores the need for nasal vaccine strategies against emerging variants.


Subject(s)
COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , SARS-CoV-2/genetics , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/virology , COVID-19/epidemiology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Aged , Male , Travel , Belgium/epidemiology , Middle Aged , Female , Adult , Phylogeography , Nasopharynx/virology
2.
Sci Rep ; 14(1): 18191, 2024 08 06.
Article in English | MEDLINE | ID: mdl-39107402

ABSTRACT

Cobas EGFR mutation Test v2 was FDA-approved as qualitative liquid biopsy for actionable EGFR variants in non-small cell lung cancer (NSCLC). It generates semiquantitative index (SQI) values that correlate with mutant allele levels, but decision thresholds for clinical use in NSCLC surveillance are lacking. We conducted long-term ctDNA monitoring in 20 subjects with EGFR-mutated NSCLC; resulting in a 155 on-treatment samples. We defined optimal SQI intervals to predict/rule-out progression within 12 weeks from sampling and performed orthogonal calibration versus deep-sequencing and digital PCR. SQI showed significant diagnostic power (AUC 0.848, 95% CI 0.782-0.901). SQI below 5 (63% of samples) had 93% (95% CI 87-96%) NPV, while SQI above 10 (25% of samples) had 69% (95% CI 56-80%) PPV. Cobas EGFR showed perfect agreement with sequencing (Kappa 0.860; 95% CI 0.674-1.00) and digital PCR. SQI values strongly (r: 0.910, 95% 0.821-0.956) correlated to mutant allele concentrations with SQI of 5 and 10 corresponding to 6-9 (0.2-0.3%) and 64-105 (1.1-1.6%) mutant allele copies/mL (VAF) respectively. Our dual-threshold classifier of SQI 0/5/10 yielded informative results in 88% of blood draws with high NPV and good overall clinical utility for patient-centric surveillance of metastatic NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Lung Neoplasms , Mutation , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Female , Middle Aged , Aged , Liquid Biopsy/methods , Circulating Tumor DNA/genetics , Circulating Tumor DNA/blood , DNA Mutational Analysis/methods , Neoplasm Metastasis
3.
Clin Chim Acta ; 562: 119854, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38977169

ABSTRACT

BACKGROUND AND AIMS: We aimed to develop an easily deployable artificial intelligence (AI)-driven model for rapid prediction of urine culture test results. MATERIAL AND METHODS: We utilized a training dataset (n = 34,584 urine samples) and two separate, unseen test sets (n = 10,083 and 9,289 samples). Various machine learning models were compared for diagnostic performance. Predictive parameters included urinalysis results (dipstick and flow cytometry), patient demographics (age and gender), and sample collection method. RESULTS: Although more complex models achieved the highest AUCs for predicting positive cultures (highest: multilayer perceptron (MLP) with AUC of 0.884, 95% CI 0.878-0.89), multiple logistic regression (MLR) using only flow cytometry parameters achieved a very good AUC (0.858, 95% CI 0.852-0.865). To aid interpretation, prediction results of the MLP and MLR models were categorized based on likelihood ratio (LR) for positivity: highly unlikely (LR 0.1), unlikely (LR 0.3), grey zone (LR 0.9), likely (LR 5.0), and highly likely (LR 40). This resulted in 17%, 28%, 34%, 9%, and 13% of samples falling into each respective category for the MLR model and 20%, 26%, 31%, 7%, and 16% for the MLP model. CONCLUSIONS: In conclusion, this robust model has the potential to assist clinicians in their decision-making process by providing insights prior to the availability of urine culture results in a significant portion of samples (∼2/3rd).


Subject(s)
Artificial Intelligence , Urinalysis , Humans , Urinalysis/methods , Male , Female , Adult , Middle Aged , Adolescent , Aged , Young Adult , Machine Learning , Urine/chemistry , Urine/microbiology , Child
4.
Nat Commun ; 15(1): 5114, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879593

ABSTRACT

The global scientific response to COVID 19 highlighted the urgent need for increased throughput and capacity in bioanalytical laboratories, especially for the precise quantification of proteins that pertain to health and disease. Acoustic ejection mass spectrometry (AEMS) represents a much-needed paradigm shift for ultra-fast biomarker screening. Here, a quantitative AEMS assays is presented, employing peptide immunocapture to enrich (i) 10 acute phase response (APR) protein markers from plasma, and (ii) SARS-CoV-2 NCAP peptides from nasopharyngeal swabs. The APR proteins were quantified in 267 plasma samples, in triplicate in 4.8 h, with %CV from 4.2% to 10.5%. SARS-CoV-2 peptides were quantified in triplicate from 145 viral swabs in 10 min. This assay represents a 15-fold speed improvement over LC-MS, with instrument stability demonstrated across 10,000 peptide measurements. The combination of speed from AEMS and selectivity from peptide immunocapture enables ultra-high throughput, reproducible quantitative biomarker screening in very large cohorts.


Subject(s)
Biomarkers , COVID-19 , Mass Spectrometry , SARS-CoV-2 , Humans , Biomarkers/blood , COVID-19/diagnosis , COVID-19/virology , COVID-19/blood , SARS-CoV-2/immunology , Mass Spectrometry/methods , Peptides , Coronavirus Nucleocapsid Proteins/analysis , Phosphoproteins
6.
Genes (Basel) ; 14(3)2023 03 11.
Article in English | MEDLINE | ID: mdl-36980965

ABSTRACT

Hereditary tyrosinemia type 1 (HT1) is a genetic disorder of the tyrosine degradation pathway (TIMD) with unmet therapeutic needs. HT1 patients are unable to fully break down the amino acid tyrosine due to a deficient fumarylacetoacetate hydrolase (FAH) enzyme and, therefore, accumulate toxic tyrosine intermediates. If left untreated, they experience hepatic failure with comorbidities involving the renal and neurological system and the development of hepatocellular carcinoma (HCC). Nitisinone (NTBC), a potent inhibitor of the 4-hydroxyphenylpyruvate dioxygenase (HPD) enzyme, rescues HT1 patients from severe illness and death. However, despite its demonstrated benefits, HT1 patients under continuous NTBC therapy are at risk to develop HCC and adverse reactions in the eye, blood and lymphatic system, the mechanism of which is poorly understood. Moreover, NTBC does not restore the enzymatic defects inflicted by the disease nor does it cure HT1. Here, the changes in molecular pathways associated to the development and progression of HT1-driven liver disease that remains uncorrected under NTBC therapy were investigated using whole transcriptome analyses on the livers of Fah- and Hgd-deficient mice under continuous NTBC therapy and after seven days of NTBC therapy discontinuation. Alkaptonuria (AKU) was used as a tyrosine-inherited metabolic disorder reference disease with non-hepatic manifestations. The differentially expressed genes were enriched in toxicological gene classes related to liver disease, liver damage, liver regeneration and liver cancer, in particular HCC. Most importantly, a set of 25 genes related to liver disease and HCC development was identified that was differentially regulated in HT1 vs. AKU mouse livers under NTBC therapy. Some of those were further modulated upon NTBC therapy discontinuation in HT1 but not in AKU livers. Altogether, our data indicate that NTBC therapy does not completely resolves HT1-driven liver disease and supports the sustained risk to develop HCC over time as different HCC markers, including Moxd1, Saa, Mt, Dbp and Cxcl1, were significantly increased under NTBC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Tyrosinemias , Mice , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Tyrosinemias/drug therapy , Tyrosinemias/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Phenotype , Tyrosine/genetics
7.
BMC Bioinformatics ; 24(1): 73, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36859168

ABSTRACT

BACKGROUND: DNA mismatch repair deficiency (dMMR) testing is crucial for detection of microsatellite unstable (MSI) tumors. MSI is detected by aberrant indel length distributions of microsatellite markers, either by visual inspection of PCR-fragment length profiles or by automated bioinformatic scoring on next-generation sequencing (NGS) data. The former is time-consuming and low-throughput while the latter typically relies on simplified binary scoring of a single parameter of the indel distribution. The purpose of this study was to use machine learning to process the full complexity of indel distributions and integrate it into a robust script for screening of dMMR on small gene panel-based NGS data of clinical tumor samples without paired normal tissue. METHODS: Scikit-learn was used to train 7 models on normalized read depth data of 36 microsatellite loci in a cohort of 133 MMR proficient (pMMR) and 46 dMMR tumor samples, taking loss of MLH1/MSH2/PMS2/MSH6 protein expression as reference method. After selection of the optimal model and microsatellite panel the two top-performing models per locus (logistic regression and support vector machine) were integrated into a novel script (DeltaMSI) for combined prediction of MSI status on 28 marker loci at sample level. Diagnostic performance of DeltaMSI was compared to that of mSINGS, a widely used script for MSI detection on unpaired tumor samples. The robustness of DeltaMSI was evaluated on 1072 unselected, consecutive solid tumor samples in a real-world setting sequenced using capture chemistry, and 116 solid tumor samples sequenced by amplicon chemistry. Likelihood ratios were used to select result intervals with clinical validity. RESULTS: DeltaMSI achieved higher robustness at equal diagnostic power (AUC = 0.950; 95% CI 0.910-0.975) as compared to mSINGS (AUC = 0.876; 95% CI 0.823-0.918). Its sensitivity of 90% at 100% specificity indicated its clinical potential for high-throughput MSI screening in all tumor types. Clinical Trial Number/IRB B1172020000040, Ethical Committee, AZ Delta General Hospital.


Subject(s)
Artificial Intelligence , Microsatellite Instability , Humans , Microsatellite Repeats , High-Throughput Nucleotide Sequencing , Machine Learning
8.
Anal Chem ; 94(50): 17379-17387, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36490367

ABSTRACT

The pandemic readiness toolbox needs to be extended, targeting different biomolecules, using orthogonal experimental set-ups. Here, we build on our Cov-MS effort using LC-MS, adding SISCAPA technology to enrich proteotypic peptides of the SARS-CoV-2 nucleocapsid (N) protein from trypsin-digested patient samples. The Cov2MS assay is compatible with most matrices including nasopharyngeal swabs, saliva, and plasma and has increased sensitivity into the attomole range, a 1000-fold improvement compared to direct detection in a matrix. A strong positive correlation was observed with qPCR detection beyond a quantification cycle of 30-31, the level where no live virus can be cultured. The automatable sample preparation and reduced LC dependency allow analysis of up to 500 samples per day per instrument. Importantly, peptide enrichment allows detection of the N protein in pooled samples without sensitivity loss. Easily multiplexed, we detect variants and propose targets for Influenza A and B detection. Thus, the Cov2MS assay can be adapted to test for many different pathogens in pooled samples, providing longitudinal epidemiological monitoring of large numbers of pathogens within a population as an early warning system.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19 Testing , Clinical Laboratory Techniques/methods , Mass Spectrometry/methods , Peptides , Sensitivity and Specificity
9.
Viruses ; 14(10)2022 10 20.
Article in English | MEDLINE | ID: mdl-36298856

ABSTRACT

An adequate SARS-CoV-2 genomic surveillance strategy has proven to be essential for countries to obtain a thorough understanding of the variants and lineages being imported and successfully established within their borders. During 2020, genomic surveillance in Belgium was not structurally implemented but performed by individual research laboratories that had to acquire the necessary funds themselves to perform this important task. At the start of 2021, a nationwide genomic surveillance consortium was established in Belgium to markedly increase the country's genomic sequencing efforts (both in terms of intensity and representativeness), to perform quality control among participating laboratories, and to enable coordination and collaboration of research projects and publications. We here discuss the genomic surveillance efforts in Belgium before and after the establishment of its genomic sequencing consortium, provide an overview of the specifics of the consortium, and explore more details regarding the scientific studies that have been published as a result of the increased number of Belgian SARS-CoV-2 genomes that have become available.


Subject(s)
COVID-19 , Pandemics , Humans , Belgium/epidemiology , COVID-19/epidemiology , Genome, Viral , Genomics , SARS-CoV-2/genetics , High-Throughput Nucleotide Sequencing
10.
J Clin Virol Plus ; 2(3): 100090, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35693461

ABSTRACT

Background: Real-time polymerase chain reaction (RT-PCR) testing on a nasopharyngeal swab is the current standard for SARS-CoV-2 virus detection. Since collection of this sample type is experienced uncomfortable by patients, saliva- and oropharyngeal swab collections should be considered as alternative specimens. Objectives: Evaluation of the relative performance of oropharyngeal swab, nasopharyngeal swab and saliva for the RT-PCR based SARS-CoV-2 Delta (B.1.617.2) and Omicron (B.1.1.529) variant detection. Study design: Nasopharyngeal swab, oropharyngeal swab and saliva were collected from 246 adult patients who presented for SARS-CoV-2 testing at the screening centre in Ypres (Belgium). RT-PCR SARS-CoV-2 detection was performed on all three sample types separately. Variant type was determined for each positive patient using whole genome sequencing or Allplex SARS-CoV-2 variants I and II Assay. Results and conclusions: Saliva is superior compared to nasopharyngeal swab for the detection of the Omicron variant. For the detection of the Delta variant, nasopharyngeal swab and saliva can be considered equivalent specimens. Oropharyngeal swab is the least sensitive sample type and shows little added value when collected in addition to a single nasopharyngeal swab.

11.
Infect Dis (Lond) ; 54(10): 731-737, 2022 10.
Article in English | MEDLINE | ID: mdl-35638185

ABSTRACT

BACKGROUND: Current method for diagnosis of SARS-CoV-2 infection is an RT-PCR test on the nasopharyngeal or oropharyngeal swab. Rapid diagnosis is essential for containing viral spread and triage of symptomatic patients presenting to hospital ER departments. As a faster alternative to RT-PCR, we evaluated a SARS-Cov-2 Rapid Antigen test in symptomatic patients presenting to hospital ER departments. METHODS: We evaluated the diagnostic performance of the Roche SARS-CoV-2 Rapid Antigen test (SD Biosensor) for detection of SARS-CoV-2 compared to RT-PCR. RESULTS: Our study showed inferior performance of the SARS-CoV-2 Rapid Antigen test for detection of SARS-CoV-2. Firstly, because of the lack of specificity, which is potentially life-threatening due to the association of nosocomial-acquired SARS-CoV-2 infection. Secondly, with a sensitivity of 45.5%, it is impossible to rule out SARS-CoV-2 infection, resulting in reflex PCR-testing. Comparison of viral load in RT-PCR positive samples with corresponding antigen results showed a significant difference between antigen positive and negative samples. COVID-19 infection will not be detected in patients admitted to the hospital in an early or late phase, typically associated with low viral loads. Sensitivity increases when testing within 5-7 symptomatic days, but the implementation of this cut-off is impractical in ER settings. However, diagnostic performance is better to detect high viral load (> = 5 log10 copies/mL) linked with contagiousness. CONCLUSION: Our study showed inferior performance of the Roche SARS-CoV-2 Rapid Antigen test (SD Biosensor) for detection of SARS-CoV-2 which limits its use as a diagnostic gatekeeper in ER departments, but is able to differentiate contagious individuals.


Subject(s)
COVID-19 Serological Testing , COVID-19 , Antigens, Viral , COVID-19/diagnosis , Emergency Service, Hospital , Humans , SARS-CoV-2 , Sensitivity and Specificity
12.
NPJ Vaccines ; 7(1): 35, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35260578

ABSTRACT

We report the levels of neutralising antibodies against Wuhan, Delta and Omicron variants in unimmunized infected (group 1), immunised and boosted (group 2) and infected immunised and boosted (group 3) adult individuals. Our observations support the rapid administration of a booster vaccine dose to prevent infection and disease caused by Omicron.

13.
Front Immunol ; 13: 1062136, 2022.
Article in English | MEDLINE | ID: mdl-36618384

ABSTRACT

Background: Patients with cancer, especially hematological cancer, are at increased risk for breakthrough COVID-19 infection. So far, a predictive biomarker that can assess compromised vaccine-induced anti-SARS-CoV-2 immunity in cancer patients has not been proposed. Methods: We employed machine learning approaches to identify a biomarker signature based on blood cytokines, chemokines, and immune- and non-immune-related growth factors linked to vaccine immunogenicity in 199 cancer patients receiving the BNT162b2 vaccine. Results: C-reactive protein (general marker of inflammation), interleukin (IL)-15 (a pro-inflammatory cytokine), IL-18 (interferon-gamma inducing factor), and placental growth factor (an angiogenic cytokine) correctly classified patients with a diminished vaccine response assessed at day 49 with >80% accuracy. Amongst these, CRP showed the highest predictive value for poor response to vaccine administration. Importantly, this unique signature of vaccine response was present at different studied timepoints both before and after vaccination and was not majorly affected by different anti-cancer treatments. Conclusion: We propose a blood-based signature of cytokines and growth factors that can be employed in identifying cancer patients at persistent high risk of COVID-19 despite vaccination with BNT162b2. Our data also suggest that such a signature may reflect the inherent immunological constitution of some cancer patients who are refractive to immunotherapy.


Subject(s)
BNT162 Vaccine , COVID-19 , Cytokines , Neoplasms , Humans , BNT162 Vaccine/immunology , COVID-19/prevention & control , Cytokines/blood , Intercellular Signaling Peptides and Proteins
14.
Am J Clin Pathol ; 157(5): 731-741, 2022 05 04.
Article in English | MEDLINE | ID: mdl-34724038

ABSTRACT

BACKGROUND: Detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern associated with immune escape is important to safeguard vaccination efficacy. We describe the potential of delayed N gene amplification in the Allplex SARS-CoV-2 Assay (Seegene) for screening of the B.1.351 (20H/501.V2, variant of concern 2 [VOC.V2], South African SARS-CoV-2 variant) lineage. METHODS: In a study cohort of 397 consecutive polymerase chain reaction-positive samples genotyped by whole-genome sequencing, amplification curves of E/N/S-RdRP targets indicated delayedN vs E gene amplification characteristic of B.1.351. Logistic regression was used to calculate a VOC.V2 probability score that was evaluated as a separate screening test in an independent validation cohort vs sequencing. RESULTS: B.1.351 showed a proportionally delayed amplification of the  N vs E gene. In logistic regression, only N and E gene cycle thresholds independently contributed to B.1.351 prediction, allowing calculation of a VOC.V2 probability score with an area under the curve of 0.94. At an optimal dichotomous cutoff point of 0.12, the VOC.V2 probability score achieved 98.7% sensitivity at 79.9% specificity, resulting in a negative predictive value (NPV) of 99.6% and a positive predictive value of 54.6%. The probability of B.1.351 increased with an increasing VOC.V2 probability score, achieving a likelihood ratio of 12.01 above 0.5. A near-maximal NPV was confirmed in 153 consecutive validation samples. CONCLUSIONS: Delayed N vs E gene amplification in the Allplex SARS-CoV-2 Assay can be used for fast and highly sensitive screening of B.1.351.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/diagnosis , Humans , Probability , SARS-CoV-2/genetics , Whole Genome Sequencing
16.
Cells ; 10(7)2021 07 04.
Article in English | MEDLINE | ID: mdl-34359863

ABSTRACT

Ongoing beta cell death in type 1 diabetes (T1D) can be detected using biomarkers selectively discharged by dying beta cells into plasma. microRNA-375 (miR-375) ranks among the top biomarkers based on studies in animal models and human islet transplantation. Our objective was to identify additional microRNAs that are co-released with miR-375 proportionate to the amount of beta cell destruction. RT-PCR profiling of 733 microRNAs in a discovery cohort of T1D patients 1 h before/after islet transplantation indicated increased plasma levels of 22 microRNAs. Sub-selection for beta cell selectivity resulted in 15 microRNAs that were subjected to double-blinded multicenter analysis. This led to the identification of eight microRNAs that were consistently increased during early graft destruction: besides miR-375, these included miR-132/204/410/200a/429/125b, microRNAs with known function and enrichment in beta cells. Their potential clinical translation was investigated in a third independent cohort of 46 transplant patients by correlating post-transplant microRNA levels to C-peptide levels 2 months later. Only miR-375 and miR-132 had prognostic potential for graft outcome, and none of the newly identified microRNAs outperformed miR-375 in multiple regression. In conclusion, this study reveals multiple beta cell-enriched microRNAs that are co-released with miR-375 and can be used as complementary biomarkers of beta cell death.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans Transplantation , MicroRNAs/genetics , Biomarkers/metabolism , Cell Count , Cohort Studies , Gene Expression Profiling , Gene Expression Regulation , Humans , MicroRNAs/metabolism , ROC Curve , Reproducibility of Results , Tropism
17.
Sci Rep ; 11(1): 12880, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145315

ABSTRACT

DNA mismatch repair deficiency (dMMR) testing is crucial for diagnosing Lynch syndrome and detection of microsatellite unstable (MSI) tumors eligible for immunotherapy. The aim of this study was to compare the relative diagnostic performance of three molecular MSI assays: polymerase chain reaction (PCR), MSI testing by Idylla and next-generation-sequencing (NGS) on 49 tumor samples (28 colorectal and 21 endometrial adenocarcinomas) versus immunohistochemistry (IHC). Discrepancies were investigated by MLH1 methylation analysis and integrated with germline results if available. Overall, the molecular assays achieved equivalent diagnostic performance for MSI detection with area under the ROC curves (AUC) of respectively 0.91 for Idylla and PCR, and 0.93 for NGS. In colorectal cancers with tumor cell percentages ≥ 30% all three molecular assays achieved 100% sensitivity and specificity (AUC = 1) versus IHC. Also, in endometrial cancers, all three molecular assays showed equivalent diagnostic performance, albeit at a clearly lower sensitivity ranging from 58% for Idylla to 75% for NGS, corresponding to negative predictive values from 78 to 86%. PCR, Idylla and NGS show similar diagnostic performance for dMMR detection in colorectal and endometrial cancers. Molecular MSI analysis has lower sensitivity for dMMR detection in endometrial cancer indicating that combined use of both IHC and molecular methods is recommended.Clinical Trial Number/IRB: B1172020000040, Ethical Committee, AZ Delta General Hospital.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Genetic Predisposition to Disease , Microsatellite Instability , Biomarkers, Tumor , Clinical Decision-Making , Computational Biology/methods , Disease Management , Female , Genetic Association Studies/methods , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry/methods , Male , Molecular Diagnostic Techniques , ROC Curve , Reproducibility of Results
18.
Orphanet J Rare Dis ; 16(1): 89, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33596965

ABSTRACT

BACKGROUND: One objective of the Belgian Rare Diseases plan is to improve patients' management using phenotypic tests and, more specifically, the access to those tests by identifying the biochemical analyses used for rare diseases, developing new financing conditions and establishing reference laboratories. METHODS: A feasibility study was performed from May 2015 until August 2016 in order to select the financeable biochemical analyses, and, among them, those that should be performed by reference laboratories. This selection was based on an inventory of analyses used for rare diseases and a survey addressed to the Belgian laboratories of clinical pathology (investigating the annual analytical costs, volumes, turnaround times and the tests unavailable in Belgium and outsourced abroad). A proposal of financeable analyses, financing modalities, reference laboratories' scope and budget estimation was developed and submitted to the Belgian healthcare authorities. After its approval in December 2016, the implementation phase took place from January 2017 until December 2019. RESULTS: In 2019, new reimbursement conditions have been published for 46 analyses and eighteen reference laboratories have been recognized. Collaborations have also been developed with 5 foreign laboratories in order to organize the outsourcing and financing of 9 analyses unavailable in Belgium. CONCLUSIONS: In the context of clinical pathology and rare diseases, this initiative enabled to identify unreimbursed analyses and to meet the most crucial financial needs. It also contributed to improve patients' management by establishing Belgian reference laboratories and foreign referral laboratories for highly-specific analyses and a permanent surveillance, quality and financing framework for those tests.


Subject(s)
Diagnostic Tests, Routine , Rare Diseases , Belgium , Budgets , Humans , Laboratories , Rare Diseases/diagnosis
19.
Radiology ; 298(1): E30-E37, 2021 01.
Article in English | MEDLINE | ID: mdl-32776832

ABSTRACT

Background The use of chest CT for coronavirus disease 2019 (COVID-19) diagnosis or triage in health care settings with limited severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) polymerase chain reaction (PCR) capacity is controversial. COVID-19 Reporting and Data System (CO-RADS) categorization of the level of COVID-19 suspicion might improve diagnostic performance. Purpose To investigate the value of chest CT with CO-RADS classification to screen for asymptomatic SARS-CoV-2 infections and to determine its diagnostic performance in individuals with COVID-19 symptoms during the exponential phase of viral spread. Materials and Methods In this secondary analysis of a prospective trial, from March 2020 to April 2020, parallel SARS-CoV-2 PCR and CT with categorization of COVID-19 suspicion was performed with CO-RADS for individuals with COVID-19 symptoms and control participants without COVID-19 symptoms admitted to the hospital for medical emergencies unrelated to COVID-19. CT with CO-RADS was categorized on a five-point scale from 1 (very low suspicion) to 5 (very high suspicion). Area under the receiver operating curve (AUC) was calculated in symptomatic versus asymptomatic individuals to predict positive SARS-CoV-2 PCR, and likelihood ratios for each CO-RADS score were used for rational selection of diagnostic thresholds. Results A total of 859 individuals (median age, 70 years; interquartile range, 52-81 years; 443 men) with COVID-19 symptoms and 1138 control participants (median age, 68 years; interquartile range, 52-81 years; 588 men) were evaluated. CT with CO-RADS had good diagnostic performance (P < .001) in both symptomatic (AUC, 0.89) and asymptomatic (AUC, 0.70) individuals. In symptomatic individuals (42% PCR positive), CO-RADS 3 or greater detected positive PCR with high sensitivity (89%, 319 of 358) and specificity of 73%. In asymptomatic individuals (5% PCR positive), a CO-RADS score of 3 or greater detected SARS-CoV-2 infection with low sensitivity (45%, 27 of 60) but high specificity (89%). Conclusion CT with Coronavirus Disease 2019 Reporting and Data System (CO-RADS) had good diagnostic performance in symptomatic individuals, supporting its application for triage. Sensitivity in asymptomatic individuals was insufficient to justify its use as a first-line screening approach. Incidental detection of CO-RADS 3 or greater in asymptomatic individuals should trigger testing for respiratory pathogens. © RSNA, 2020 Online supplemental material is available for this article.


Subject(s)
COVID-19/diagnostic imaging , Tomography, X-Ray Computed , Aged , Aged, 80 and over , Asymptomatic Infections , Female , Humans , Male , Middle Aged , Prospective Studies , Thorax/diagnostic imaging
20.
Am J Clin Pathol ; 155(3): 381-388, 2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33236114

ABSTRACT

OBJECTIVES: Vitamin D deficiency was previously correlated with incidence and severity of coronavirus disease 2019 (COVID-19). We investigated the association between serum 25-hydroxyvitamin D (25(OH)D) level on admission and radiologic stage and outcome of COVID-19 pneumonia. METHODS: A retrospective observational trial was done on 186 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected individuals hospitalized from March 1, 2020, to April 7, 2020, with combined chest computed tomography (CT) and 25(OH)D measurement on admission. Multivariate regression analysis was performed to study if vitamin D deficiency (25(OH)D <20 ng/mL) correlates with survival independently of confounding comorbidities. RESULTS: Of the patients with COVID-19, 59% were vitamin D deficient on admission: 47% of females and 67% of males. In particular, male patients with COVID-19 showed progressively lower 25(OH)D with advancing radiologic stage, with deficiency rates increasing from 55% in stage 1 to 74% in stage 3. Vitamin D deficiency on admission was not confounded by age, ethnicity, chronic lung disease, coronary artery disease/hypertension, or diabetes and was associated with mortality (odds ratio [OR], 3.87; 95% confidence interval [CI], 1.30-11.55), independent of age (OR, 1.09; 95% CI, 1.03-1.14), chronic lung disease (OR, 3.61; 95% CI, 1.18-11.09), and extent of lung damage expressed by chest CT severity score (OR, 1.12; 95% CI, 1.01-1.25). CONCLUSIONS: Low 25(OH)D levels on admission are associated with COVID-19 disease stage and mortality.


Subject(s)
COVID-19/blood , COVID-19/mortality , COVID-19/pathology , Vitamin D Deficiency/complications , Vitamin D/analogs & derivatives , Aged , Aged, 80 and over , Female , Hospital Mortality , Humans , Incidence , Male , Middle Aged , Retrospective Studies , Risk Factors , SARS-CoV-2 , Vitamin D/blood , Vitamin D Deficiency/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL