Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 629
Filter
1.
Nucleic Acids Res ; 52(1): 114-124, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38015437

ABSTRACT

Next-generation DNA sequencing (NGS) in short-read mode has recently been used for genetic testing in various clinical settings. NGS data accuracy is crucial in clinical settings, and several reports regarding quality control of NGS data, primarily focusing on establishing NGS sequence read accuracy, have been published thus far. Variant calling is another critical source of NGS errors that remains unexplored at the single-nucleotide level despite its established significance. In this study, we used a machine-learning-based method to establish an exome-wide benchmark of difficult-to-sequence regions at the nucleotide-residue resolution using 10 genome sequence features based on real-world NGS data accumulated in The Genome Aggregation Database (gnomAD) of the human reference genome sequence (GRCh38/hg38). The newly acquired metric, designated the 'UNMET score,' along with additional lines of structural information from the human genome, allowed us to assess the sequencing challenges within the exonic region of interest using conventional short-read NGS. Thus, the UNMET score could provide a basis for addressing potential sequential errors in protein-coding exons of the human reference genome sequence GRCh38/hg38 in clinical sequencing.


Subject(s)
Exome , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA , Humans , DNA , Exome/genetics , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/standards
2.
BMC Genomics ; 24(1): 117, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36927511

ABSTRACT

BACKGROUND: Generating the most contiguous, accurate genome assemblies given available sequencing technologies is a long-standing challenge in genome science. With the rise of long-read sequencing, assembly challenges have shifted from merely increasing contiguity to correctly assembling complex, repetitive regions of interest, ideally in a phased manner. At present, researchers largely choose between two types of long read data: longer, but less accurate sequences, or highly accurate, but shorter reads (i.e., >Q20 or 99% accurate). To better understand how these types of long-read data as well as scale of data (i.e., mean length and sequencing depth) influence genome assembly outcomes, we compared genome assemblies for a caddisfly, Hesperophylax magnus, generated with longer, but less accurate, Oxford Nanopore (ONT) R9.4.1 and highly accurate PacBio HiFi (HiFi) data. Next, we expanded this comparison to consider the influence of highly accurate long-read sequence data on genome assemblies across 6750 plant and animal genomes. For this broader comparison, we used HiFi data as a surrogate for highly accurate long-reads broadly as we could identify when they were used from GenBank metadata. RESULTS: HiFi reads outperformed ONT reads in all assembly metrics tested for the caddisfly data set and allowed for accurate assembly of the repetitive ~ 20 Kb H-fibroin gene. Across plants and animals, genome assemblies that incorporated HiFi reads were also more contiguous. For plants, the average HiFi assembly was 501% more contiguous (mean contig N50 = 20.5 Mb) than those generated with any other long-read data (mean contig N50 = 4.1 Mb). For animals, HiFi assemblies were 226% more contiguous (mean contig N50 = 20.9 Mb) versus other long-read assemblies (mean contig N50 = 9.3 Mb). In plants, we also found limited evidence that HiFi may offer a unique solution for overcoming genomic complexity that scales with assembly size. CONCLUSIONS: Highly accurate long-reads generated with HiFi or analogous technologies represent a key tool for maximizing genome assembly quality for a wide swath of plants and animals. This finding is particularly important when resources only allow for one type of sequencing data to be generated. Ultimately, to realize the promise of biodiversity genomics, we call for greater uptake of highly accurate long-reads in future studies.


Subject(s)
Biodiversity , Genomics , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA , Genomics/methods , Genomics/standards , Genomics/trends , Insecta/classification , Insecta/genetics , Fibroins/genetics , Contig Mapping , Genome, Insect/genetics , Animals , Databases, Nucleic Acid , Reproducibility of Results , Meta-Analysis as Topic , Datasets as Topic , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/standards , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , High-Throughput Nucleotide Sequencing/trends , Plants/genetics , Genome, Plant/genetics
3.
Eur J Med Genet ; 66(12): 104871, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38832911

ABSTRACT

Rare diseases encompass a diverse group of genetic disorders that affect a small proportion of the population. Identifying the underlying genetic causes of these conditions presents significant challenges due to their genetic heterogeneity and complexity. Conventional short-read sequencing (SRS) techniques have been widely used in diagnosing and investigating of rare diseases, with limitations due to the nature of short-read lengths. In recent years, long read sequencing (LRS) technologies have emerged as a valuable tool in overcoming these limitations. This minireview provides a concise overview of the applications of LRS in rare disease research and diagnosis, including the identification of disease-causing tandem repeat expansions, structural variations, and comprehensive analysis of pathogenic variants with LRS.


Subject(s)
High-Throughput Nucleotide Sequencing , Rare Diseases , Humans , Rare Diseases/genetics , Rare Diseases/diagnosis , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/standards
4.
Nature ; 611(7936): 519-531, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36261518

ABSTRACT

The current human reference genome, GRCh38, represents over 20 years of effort to generate a high-quality assembly, which has benefitted society1,2. However, it still has many gaps and errors, and does not represent a biological genome as it is a blend of multiple individuals3,4. Recently, a high-quality telomere-to-telomere reference, CHM13, was generated with the latest long-read technologies, but it was derived from a hydatidiform mole cell line with a nearly homozygous genome5. To address these limitations, the Human Pangenome Reference Consortium formed with the goal of creating high-quality, cost-effective, diploid genome assemblies for a pangenome reference that represents human genetic diversity6. Here, in our first scientific report, we determined which combination of current genome sequencing and assembly approaches yield the most complete and accurate diploid genome assembly with minimal manual curation. Approaches that used highly accurate long reads and parent-child data with graph-based haplotype phasing during assembly outperformed those that did not. Developing a combination of the top-performing methods, we generated our first high-quality diploid reference assembly, containing only approximately four gaps per chromosome on average, with most chromosomes within ±1% of the length of CHM13. Nearly 48% of protein-coding genes have non-synonymous amino acid changes between haplotypes, and centromeric regions showed the highest diversity. Our findings serve as a foundation for assembling near-complete diploid human genomes at scale for a pangenome reference to capture global genetic variation from single nucleotides to structural rearrangements.


Subject(s)
Chromosome Mapping , Diploidy , Genome, Human , Genomics , Humans , Chromosome Mapping/standards , Genome, Human/genetics , Haplotypes/genetics , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/standards , Reference Standards , Genomics/methods , Genomics/standards , Chromosomes, Human/genetics , Genetic Variation/genetics
5.
Gene ; 814: 146161, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-34995736

ABSTRACT

The patients with hepatic alveolar echinococcosis is poorly detected due to invasive and slow growth. Thus, early diagnosis of hepatic alveolar echinococcosis is so important for patients. Circular RNAs are crucial types of the non-coding RNA. Recent studies have provided serum-derived exosomal circRNAs as potential biomarkers for detection of various diseases. The clinical importance of exosomal circRNAs in hepatic alveolar echinococcosis have never been explored before. Here, we investigated the serum-derived exosomal circRNAs in the diagnosis of hepatic alveolar echinococcosis. Firstly, High-throughput Sequencing was performed using 9 hepatic alveolar echinococcosis and 9 control samples to detect hepatic alveolar echinococcosis related circRNAs. Afterwards, bioinformatic analyzes were performed to identify differentially expressed circRNAs and pathway analyzes were performed. Finally, validation of the determined circRNAs was performed using RT-PCR. The sequencing data indicated that 59 differentially expressed circRNAs; 31 up-regulated and 28 down-regulated circRNA in hepatic alveolar echinococcosis patients. The top 5 up-regulated and down-regulated circRNAs were selected for validation by RT-qPCR assay. As a result of the verification, circRNAs that were significantly up- and down-regulated showed an expression profile consistent with the results obtained. Importantly, our findings suggested that identified exosomal circRNAs could be a potential biomarker for the detection of hepatic alveolar echinococcosis serum and may help to understand the pathogenesis of hepatic alveolar echinococcosis.


Subject(s)
Echinococcosis, Hepatic/genetics , Exosomes/genetics , RNA, Circular/blood , Biomarkers/blood , Echinococcosis, Hepatic/blood , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing/standards , Humans , Quality Control , RNA-Seq/standards , Transcriptome
6.
Mol Biol Rep ; 49(1): 385-392, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34716505

ABSTRACT

BACKGROUND: High-throughput sequencing involves library preparation and amplification steps, which may induce contamination across samples or between samples and the environment. METHODS: We tested the effect of applying an inline-index strategy, in which DNA indices of 6 bp were added to both ends of the inserts at the ligation step of library prep for resolving the data contamination problem. RESULTS: Our results showed that the contamination ranged from 0.29 to 1.25% in one experiment and from 0.83 to 27.01% in the other. We also found that contamination could be environmental or from reagents besides cross-contamination between samples. CONCLUSIONS: Inline-index method is a useful experimental design to clean up the data and address the contamination problem which has been plaguing high-throughput sequencing data in many applications.


Subject(s)
DNA/analysis , Indicators and Reagents/chemistry , Sequence Analysis, DNA/standards , DNA/chemistry , DNA Contamination , Gene Library , High-Throughput Nucleotide Sequencing/standards
7.
Int J Lab Hematol ; 44(1): 118-126, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34585519

ABSTRACT

INTRODUCTION: Monitoring patients with acute myeloid leukemia can be implemented through various techniques such as multiparameter flow cytometry, real-time quantitative polymerase chain reaction, and next-generation sequencing. However, there is scarce studies when comparing the data of next-generation sequencing and flow cytometry for monitoring disease progression, particularly how they might supplement one another when used in tandem. METHODS: We investigated 107 patients via retrospective analysis using follow-up MFC and NGS data with a total of 717 MFC and 247 NGS studies to compare these methods in monitoring minimal/measurable residual disease. RESULTS: 197 instances were MFC+ /NGS+ , 3 were MFC- /NGS- , 44 were MFC- /NGS+ , and 3 are MFC+ /NGS- . The majority of the MFC- /NGS+ cases occurred within 6 months during the post-treatment phase (64%). Among 44 MFC- /NGS+ instances, 13 had similar NGS profiles to their original day 0 diagnosis. The remaining cases showed preleukemic clonal hematopoiesis mutations, "likely pathogenic mutations," or "variants of uncertain significance." CONCLUSION: Our findings show that flow cytometry has its advantages with comparable sensitivity in detecting minimal/measurable residual disease. Next-generation sequencing could be used in an increased and more regular capacity in conjunction with flow cytometry to achieve a more comprehensive surveillance of these patients, resulting in improved outcomes.


Subject(s)
Flow Cytometry/methods , Flow Cytometry/standards , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Leukemia, Myeloid, Acute/diagnosis , Neoplasm, Residual/diagnosis , Biomarkers, Tumor , Disease Management , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , Mutation , Reproducibility of Results , Retrospective Studies , Sensitivity and Specificity
8.
Br J Haematol ; 196(1): 19-30, 2022 01.
Article in English | MEDLINE | ID: mdl-34124782

ABSTRACT

With the focus of leukaemia management shifting to the implications of low-level disease burden, increasing attention is being paid on the development of highly sensitive methodologies required for detection. There are various techniques capable of identification of measurable residual disease (MRD) either evidencing as relevant mutation detection [e.g. nucleophosmin 1 (NPM1) mutation] or trace levels of leukaemic clonal populations. The vast majority of these methods only permit detection of a single clone or mutation. However, mass spectrometry and next-generation sequencing enable the interrogation of multiple genes simultaneously, facilitating a more complete genomic profile. In the present review, we explore the methodologies of both techniques in conjunction with the important advantages and limitations associated with each assay. We also highlight the evidence and the various instances where either technique has been used and propose future strategies for MRD detection.


Subject(s)
Biomarkers, Tumor , DNA Mutational Analysis/methods , Leukemia/diagnosis , Leukemia/etiology , Mutation , Neoplasm, Residual/diagnosis , Cost-Benefit Analysis , DNA Mutational Analysis/economics , DNA Mutational Analysis/standards , High-Throughput Nucleotide Sequencing/economics , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Mass Spectrometry/methods , Mass Spectrometry/standards , Mutation Rate , Reproducibility of Results , Sensitivity and Specificity
10.
J Clin Lab Anal ; 36(1): e24139, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34811797

ABSTRACT

BACKGROUND: Quality control materials are necessary for assay development, test validation, and proficiency testing in cancer mutation analysis. Most of the existing controls for somatic mutations only harbor a single variant and are derived from unstable cell lines. This study aimed to establish a method to create stable multianalyte controls in a defined background by genome editing in GM12878 cells, which also can be applied for the reference of next-generation sequencing. METHODS: GM12878 cells were electroporated with a donor plasmid containing a mutant DNA sequence and a Cas9/sgRNA expressing vector. The genome-edited GM12878 cell was validated with Sanger sequencing, amplification refractory mutation system (ARMS), and next-generation sequencing (NGS). RESULTS: We have successfully generated a mutant GM12878 cell line harboring the defined variants including single-nucleotide variants (SNVs), small insertions and deletions (indels), and structural variants (SVs). The introduction of intended mutations in GM12878 cell line was confirmed by both ARMS and sequencing methods. CONCLUSIONS: We developed a method for the preparation of the multiplexed controls for reference mutations in cancer gene by genome editing in GM12878 cells. This methodology can be used to generate other stable cancer reference materials with an unlimited supply.


Subject(s)
DNA Mutational Analysis , Gene Editing/methods , Neoplasms/genetics , CRISPR-Cas Systems/genetics , Cell Line, Tumor , DNA Mutational Analysis/methods , DNA Mutational Analysis/standards , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Mutation/genetics , Quality Control , Reference Standards
11.
Cancer Res Treat ; 54(1): 1-9, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34902959

ABSTRACT

Next-generation sequencing (NGS) is becoming essential in the fields of precision oncology. With implementation of NGS in daily clinic, the needs for continued education, facilitated interpretation of NGS results and optimal treatment delivery based on NGS results have been addressed. Molecular tumor board (MTB) is multidisciplinary approach to keep pace with the growing knowledge of complex molecular alterations in patients with advanced solid cancer. Although guidelines for NGS use and MTB have been developed in western countries, there is limitation for reflection of Korea's public health environment and daily clinical practice. These recommendations provide a critical guidance from NGS panel testing to final treatment decision based on MTB discussion.


Subject(s)
High-Throughput Nucleotide Sequencing/standards , Medical Oncology/standards , Neoplasms/therapy , Precision Medicine/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Neoplasms/diagnosis , Neoplasms/genetics , Practice Guidelines as Topic , Republic of Korea , Societies, Medical
12.
Br J Cancer ; 126(3): 514-520, 2022 02.
Article in English | MEDLINE | ID: mdl-34480094

ABSTRACT

BACKGROUND: Activating fusions of the NTRK1, NTRK2 and NTRK3 genes are drivers of carcinogenesis and proliferation across a broad range of tumour types in both adult and paediatric patients. Recently, the FDA granted tumour-agnostic approvals of TRK inhibitors, larotrectinib and entrectinib, based on significant and durable responses in multiple primary tumour types. Unfortunately, testing rates in clinical practice remain quite low. Adding plasma next-generation sequencing of circulating tumour DNA (ctDNA) to tissue-based testing increases the detection rate of oncogenic drivers and demonstrates high concordance with tissue genotyping. However, the clinical potential of ctDNA analysis to identify NTRK fusion-positive tumours has been largely unexplored. METHODS: We retrospectively reviewed a ctDNA database in advanced stage solid tumours for NTRK1 fusions. RESULTS: NTRK1 fusion events, with nine unique fusion partners, were identified in 37 patients. Of the cases for which clinical data were available, 44% had tissue testing for NTRK1 fusions; the NTRK1 fusion detected by ctDNA was confirmed in tissue in 88% of cases. Here, we report for the first time that minimally-invasive plasma NGS can detect NTRK fusions with a high positive predictive value. CONCLUSION: Plasma ctDNA represents a rapid, non-invasive screening method for this rare genomic target that may improve identification of patients who can benefit from TRK-targeted therapy and potentially identify subsequent on- and off-target resistance mechanisms.


Subject(s)
Biomarkers, Tumor/blood , Circulating Tumor DNA/genetics , High-Throughput Nucleotide Sequencing/methods , Neoplasms/pathology , Oncogene Proteins, Fusion , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Receptor, trkA/genetics , Benzamides/therapeutic use , Biomarkers, Tumor/genetics , Circulating Tumor DNA/blood , High-Throughput Nucleotide Sequencing/standards , Humans , Indazoles/therapeutic use , Neoplasm Staging , Neoplasms/blood , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use
13.
BMC Microbiol ; 21(1): 349, 2021 12 18.
Article in English | MEDLINE | ID: mdl-34922460

ABSTRACT

BACKGROUND: One limiting factor of short amplicon 16S rRNA gene sequencing approaches is the use of low DNA amounts in the amplicon generation step. Especially for low-biomass samples, insufficient or even commonly undetectable DNA amounts can limit or prohibit further analysis in standard protocols. RESULTS: Using a newly established protocol, very low DNA input amounts were found sufficient for reliable detection of bacteria using 16S rRNA gene sequencing compared to standard protocols. The improved protocol includes an optimized amplification strategy by using a digital droplet PCR. We demonstrate how PCR products are generated even when using very low concentrated DNA, unable to be detected by using a Qubit. Importantly, the use of different 16S rRNA gene primers had a greater effect on the resulting taxonomical profiles compared to using high or very low initial DNA amounts. CONCLUSION: Our improved protocol takes advantage of ddPCR and allows faithful amplification of very low amounts of template. With this, samples of low bacterial biomass become comparable to those with high amounts of bacteria, since the first and most biasing steps are the same. Besides, it is imperative to state DNA concentrations and volumes used and to include negative controls indicating possible shifts in taxonomical profiles. Despite this, results produced by using different primer pairs cannot be easily compared.


Subject(s)
Biomass , Polymerase Chain Reaction/methods , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA/methods , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bias , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Feces/microbiology , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Limit of Detection , Microbiota/genetics , Polymerase Chain Reaction/standards , Reproducibility of Results , Sequence Analysis, DNA/standards , Water Microbiology
14.
Eur Rev Med Pharmacol Sci ; 25(1 Suppl): 1-6, 2021 12.
Article in English | MEDLINE | ID: mdl-34890028

ABSTRACT

OBJECTIVE: While the bioinformatic workflow, from quality control to annotation, is quite standardized, the interpretation of variants is still a challenge. The decreasing cost of massively parallel NGS has produced hundreds of variants per patient to analyze and interpret. The ACMG "Standards and guidelines for the interpretation of sequence variants", widely adopted in clinical settings, assume that the clinician has a comprehensive knowledge of the literature and the disease. MATERIALS AND METHODS: To semi-automatize the application of the guidelines, we decided to develop an algorithm that exploits VarSome, a widely used platform that interprets variants on the basis of information from more than 70 genome databases. RESULTS: Here we explain how we integrated VarSome API into our existing clinical diagnostic pipeline for NGS data to obtain validated reproducible results as indicated by accuracy, sensitivity and specificity. CONCLUSIONS: We validated the automated pipeline to be sure that it was doing what we expected. We obtained 100% sensitivity, specificity and accuracy, confirming that it was suitable for use in a diagnostic setting.


Subject(s)
Algorithms , Genetic Variation/genetics , Genomics/standards , High-Throughput Nucleotide Sequencing/standards , Practice Guidelines as Topic/standards , Search Engine/standards , Computational Biology/methods , Computational Biology/standards , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Search Engine/methods , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/standards
15.
Am J Trop Med Hyg ; 106(2): 671-677, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34749306

ABSTRACT

For complex clinical cases where a parasitic infection is suspected, it can be difficult for clinicians to recommend an appropriate laboratory test. These tests are usually pathogen-specific and require a certain degree of suspicion for the precise etiology. A recently described assay, the universal parasite diagnostic (UPDx) can potentially provide a diagnosis of any parasite present in a specimen. Using primers that amplify DNA from all eukaryotes, UPDx differentiates several parasitic infections in blood by amplicon-based next-generation sequencing (NGS) of the 18S rDNA locus. As the state's public health reference laboratory, the Parasitology Laboratory at the Wadsworth Center (Albany, NY) receives specimens from patients who have potentially encountered a wide variety of parasites. As such, the ability to differentiate several blood parasites using a single assay is of interest. We assessed UPDx for its ability to confirm parasitic infections for 20 specimens that were previously identified by real-time PCR (RT-PCR). This included specimens positive for Babesia microti, Trypanosoma cruzi, Leishmania tropica, various Plasmodium species, and specimens comprising mixed Plasmodium sp. infections. Results obtained using UPDx were largely concordant with the RT-PCR assays. A T. cruzi positive specimen was negative by UPDx and for two mixed Plasmodium sp. infections only one species was detected. The results obtained for other specimens were concordant. We conclude that UPDx shows promise for the detection of blood parasites in diagnostic laboratories. As NGS becomes cheaper, assays like UPDx will become increasingly amenable to use in clinical settings.


Subject(s)
Blood-Borne Infections/diagnosis , High-Throughput Nucleotide Sequencing/standards , Laboratories , Molecular Diagnostic Techniques/standards , Parasitic Diseases/blood , Parasitic Diseases/diagnosis , Public Health , Blood-Borne Infections/parasitology , High-Throughput Nucleotide Sequencing/methods , Humans , Molecular Diagnostic Techniques/methods , Parasitic Diseases/classification , Parasitic Diseases/parasitology , RNA, Ribosomal, 18S/genetics , United States
17.
Nat Commun ; 12(1): 6123, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675197

ABSTRACT

Quantitation of rare somatic mutations is essential for basic research and translational clinical applications including minimal residual disease (MRD) detection. Though unique molecular identifier (UMI) has suppressed errors for rare mutation detection, the sequencing depth requirement is high. Here, we present Quantitative Blocker Displacement Amplification (QBDA) which integrates sequence-selective variant enrichment into UMI quantitation for accurate quantitation of mutations below 0.01% VAF at only 23,000X depth. Using a panel of 20 genes recurrently altered in acute myeloid leukemia, we demonstrate quantitation of various mutations including single base substitutions and indels down to 0.001% VAF at a single locus with less than 4 million sequencing reads, allowing sensitive MRD detection in patients during complete remission. In a pan-cancer panel and a melanoma hotspot panel, we detect mutations down to 0.1% VAF using only 1 million reads. QBDA provides a convenient and versatile method for sensitive mutation quantitation using low-depth sequencing.


Subject(s)
High-Throughput Nucleotide Sequencing/standards , Leukemia, Myeloid, Acute/genetics , Melanoma/genetics , Mutation , Neoplasm, Residual/genetics , Calibration , High-Throughput Nucleotide Sequencing/methods , Humans
18.
JCO Precis Oncol ; 52021 07.
Article in English | MEDLINE | ID: mdl-34632252

ABSTRACT

We conducted this systematic review to evaluate the clinical outcomes associated with molecular tumor board (MTB) review in patients with cancer. METHODS: A systematic search of PubMed was performed to identify studies reporting clinical outcomes in patients with cancer who were reviewed by an MTB. To be included, studies had to report clinical outcomes, including clinical benefit, response, progression-free survival, or overall survival. Two reviewers independently selected studies and assessed quality with the Quality Assessment Tool for Before-After (Pre-Post) Studies with No Control Group or the Quality Assessment Tool for Observational Cohort and Cross-Sectional Studies depending on the type of study being reviewed. RESULTS: Fourteen studies were included with a total of 3,328 patients with cancer. All studies included patients without standard-of-care treatment options and usually with multiple prior lines of therapy. In studies reporting response rates, patients receiving MTB-recommended therapy had overall response rates ranging from 0% to 67%. In the only trial powered on clinical outcome and including a control group, the group receiving MTB-recommended therapy had significantly improved rate of progression-free survival compared with those receiving conventional therapy. CONCLUSION: Although data quality is limited by a lack of prospective randomized controlled trials, MTBs appear to improve clinical outcomes for patients with cancer. Future research should concentrate on prospective trials and standardization of approach and outcomes.


Subject(s)
Biomarkers, Tumor/genetics , Medical Oncology/methods , Neoplasms/drug therapy , Patient Care Team/organization & administration , Precision Medicine/methods , Antineoplastic Agents/pharmacology , Clinical Decision-Making , DNA Mutational Analysis/standards , Genetic Testing/standards , Genetic Testing/trends , High-Throughput Nucleotide Sequencing/standards , Humans , Medical Oncology/organization & administration , Molecular Targeted Therapy , Mutation , Neoplasms/diagnosis , Neoplasms/genetics
19.
PLoS Comput Biol ; 17(10): e1008839, 2021 10.
Article in English | MEDLINE | ID: mdl-34634030

ABSTRACT

Hi-C is a sample preparation method that enables high-throughput sequencing to capture genome-wide spatial interactions between DNA molecules. The technique has been successfully applied to solve challenging problems such as 3D structural analysis of chromatin, scaffolding of large genome assemblies and more recently the accurate resolution of metagenome-assembled genomes (MAGs). Despite continued refinements, however, preparing a Hi-C library remains a complex laboratory protocol. To avoid costly failures and maximise the odds of successful outcomes, diligent quality management is recommended. Current wet-lab methods provide only a crude assay of Hi-C library quality, while key post-sequencing quality indicators used have-thus far-relied upon reference-based read-mapping. When a reference is accessible, this reliance introduces a concern for quality, where an incomplete or inexact reference skews the resulting quality indicators. We propose a new, reference-free approach that infers the total fraction of read-pairs that are a product of proximity ligation. This quantification of Hi-C library quality requires only a modest amount of sequencing data and is independent of other application-specific criteria. The algorithm builds upon the observation that proximity ligation events are likely to create k-mers that would not naturally occur in the sample. Our software tool (qc3C) is to our knowledge the first to implement a reference-free Hi-C QC tool, and also provides reference-based QC, enabling Hi-C to be more easily applied to non-model organisms and environmental samples. We characterise the accuracy of the new algorithm on simulated and real datasets and compare it to reference-based methods.


Subject(s)
Chromosome Mapping , Genomics , High-Throughput Nucleotide Sequencing , Quality Control , Software , Algorithms , Animals , Chromosome Mapping/methods , Chromosome Mapping/standards , DNA/chemistry , DNA/genetics , Gene Library , Genomics/methods , Genomics/standards , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Turtles
20.
Mol Biol Rep ; 48(12): 8203-8209, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34643925

ABSTRACT

Next generation sequencing (NGS) is a widespread molecular biology method integrated into clinical practice to detect genetic variants, for diagnostic and prognostic purposes. The scheduled external quality assessments (EQA) is integral part of clinical molecular laboratory quality assurance. The EQA provides an efficient system to compare analytic test performances among different laboratories, which is essential to evaluate consistency of molecular test. EQA failures demands targeted corrective action plans. In this context, the complexity of the NGS techniques requires careful and continuous quality control procedures. We report a tumor BRCA1/2 (tBRCA) testing benchmark discrepancy provided by the European Molecular Genetics Quality Network in our laboratory during a round of EQA for somatic mutation testing of BRCA genes in relation to ovarian cancer. The critical analysis emerging from the tBRCA EQA is presented. We underline that harmonization processes are still required for the EQA in the molecular biology field, especially if applied to the evaluation of methods characterized by high complexity.


Subject(s)
High-Throughput Nucleotide Sequencing/standards , Ovarian Neoplasms/genetics , BRCA1 Protein/analysis , BRCA1 Protein/genetics , BRCA2 Protein/analysis , BRCA2 Protein/genetics , Benchmarking/methods , Data Accuracy , Female , Genes, BRCA1 , Genes, BRCA2 , High-Throughput Nucleotide Sequencing/methods , Humans , Laboratories/standards , Quality Control , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...