Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 292
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 108(4): 656-668, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33770507

ABSTRACT

Genetic studies in underrepresented populations identify disproportionate numbers of novel associations. However, most genetic studies use genotyping arrays and sequenced reference panels that best capture variation most common in European ancestry populations. To compare data generation strategies best suited for underrepresented populations, we sequenced the whole genomes of 91 individuals to high coverage as part of the Neuropsychiatric Genetics of African Population-Psychosis (NeuroGAP-Psychosis) study with participants from Ethiopia, Kenya, South Africa, and Uganda. We used a downsampling approach to evaluate the quality of two cost-effective data generation strategies, GWAS arrays versus low-coverage sequencing, by calculating the concordance of imputed variants from these technologies with those from deep whole-genome sequencing data. We show that low-coverage sequencing at a depth of ≥4× captures variants of all frequencies more accurately than all commonly used GWAS arrays investigated and at a comparable cost. Lower depths of sequencing (0.5-1×) performed comparably to commonly used low-density GWAS arrays. Low-coverage sequencing is also sensitive to novel variation; 4× sequencing detects 45% of singletons and 95% of common variants identified in high-coverage African whole genomes. Low-coverage sequencing approaches surmount the problems induced by the ascertainment of common genotyping arrays, effectively identify novel variation particularly in underrepresented populations, and present opportunities to enhance variant discovery at a cost similar to traditional approaches.


Subject(s)
DNA Mutational Analysis/economics , DNA Mutational Analysis/standards , Genetic Variation/genetics , Genetics, Population/economics , Africa , DNA Mutational Analysis/methods , Genetics, Population/methods , Genome, Human/genetics , Genome-Wide Association Study , Health Equity , Humans , Microbiota , Whole Genome Sequencing/economics , Whole Genome Sequencing/standards
2.
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
3.
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
4.
Future Oncol ; 17(7): 853-864, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33263430

ABSTRACT

Background: Ongoing clinical trials are investigating PARP inhibitors to target the DNA damage repair (DDR) pathway in prostate cancer. DDR mutation screening will guide treatment strategy and assess eligibility for clinical trials. Materials & methods: This systematic review estimated the rate of DDR mutation testing or genetic counseling among men with or at risk of prostate cancer. Results: From 6856 records, one study fulfilled the inclusion criteria and described men undiagnosed with prostate cancer with a family history of BRCA1/2 mutation who received DDR mutation testing. Conclusion: With only one study included in this first systematic review of DDR mutation testing or genetic counseling in men with or at risk of prostate cancer, more research is warranted.


Subject(s)
DNA Mutational Analysis/statistics & numerical data , DNA Repair , Genetic Counseling/statistics & numerical data , Genetic Testing/statistics & numerical data , Prostatic Neoplasms/diagnosis , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Consensus , DNA Mutational Analysis/standards , Drug Resistance, Neoplasm/genetics , Genetic Counseling/standards , Genetic Testing/standards , Humans , Male , Medical History Taking , Mutation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Practice Guidelines as Topic , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics
5.
Future Oncol ; 16(28): 2177-2189, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32716216

ABSTRACT

Aim: To determine the concordance between plasma and tissue RAS mutation status in metastatic colorectal cancer patients to gauge whether blood-based testing is a viable alternative. We also evaluated the change in mutation status on progression. Materials/methods: RAS testing was performed on plasma from patients commencing first-line therapy (OncoBEAM™ RAS CEIVD kit). Results were then compared with formalin-fixed paraffin embedded tumor samples. Results: The overall percentage agreement (concordance) was 86.0% (86/100), which demonstrates that blood-based testing is an alternative to tissue-based testing. Reproducibility was 100% between three laboratories and 20% showed changes in their RAS mutational status on progression. Conclusion: These results show good concordance between tissue and plasma samples and suggest the need for longitudinal plasma testing during treatment to guide management decisions.


Subject(s)
Biomarkers, Tumor , Genes, ras , Mutation , Neoplasms/diagnosis , Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Alleles , Circulating Tumor DNA , DNA Mutational Analysis/methods , DNA Mutational Analysis/standards , Disease Progression , Female , Humans , Liquid Biopsy/methods , Liquid Biopsy/standards , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/blood , Neoplasms/therapy , Time-to-Treatment
6.
Nature ; 505(7485): 686-90, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24284629

ABSTRACT

RNA viruses exist as genetically diverse populations. It is thought that diversity and genetic structure of viral populations determine the rapid adaptation observed in RNA viruses and hence their pathogenesis. However, our understanding of the mechanisms underlying virus evolution has been limited by the inability to accurately describe the genetic structure of virus populations. Next-generation sequencing technologies generate data of sufficient depth to characterize virus populations, but are limited in their utility because most variants are present at very low frequencies and are thus indistinguishable from next-generation sequencing errors. Here we present an approach that reduces next-generation sequencing errors and allows the description of virus populations with unprecedented accuracy. Using this approach, we define the mutation rates of poliovirus and uncover the mutation landscape of the population. Furthermore, by monitoring changes in variant frequencies on serially passaged populations, we determined fitness values for thousands of mutations across the viral genome. Mapping of these fitness values onto three-dimensional structures of viral proteins offers a powerful approach for exploring structure-function relationships and potentially uncovering new functions. To our knowledge, our study provides the first single-nucleotide fitness landscape of an evolving RNA virus and establishes a general experimental platform for studying the genetic changes underlying the evolution of virus populations.


Subject(s)
DNA Mutational Analysis/methods , Genetic Fitness/genetics , Mutation/genetics , Poliovirus/genetics , DNA Mutational Analysis/standards , Evolution, Molecular , HeLa Cells , Humans , Models, Molecular , Mutation Rate , Poliovirus/chemistry , Poliovirus/growth & development , Quality Control , Structure-Activity Relationship
7.
J Clin Lab Anal ; 34(11): e23458, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32671901

ABSTRACT

OBJECTIVES: Droplet digital PCR (ddPCR) has been reported to have a superior validity over PCR with amplification-refractory mutation system (ARMS-PCR) for detecting the BRAF V600E mutation in thyroid nodule fine-needle aspiration (FNA) samples using cytological diagnosis as the reference. However, the added value of ddPCR on surgical decision-making remains to be illustrated when the technique is combined with FNA cytology. METHODS: A total of 277 consecutive patients with thyroid nodules were subjected to FNA cytology and BRAF V600E testing with ARMS-PCR. Within this patient cohort, 90 patients underwent surgical intervention with pathological diagnosis available. BRAF V600E testing with ddPCR was performed retrospectively using FNA frozen DNA specimens. The clinical validity and utility of ddPCR in comparison with ARMS-PCR were compared using surgical pathology as the reference. RESULTS: Overall, 101 BRAF V600E mutations were detected by ddPCR, including five ARMS negative patients, four of whom were confirmed to have papillary thyroid cancer (PTC) by surgical pathology. Of the 90 patients with surgical pathology, which is considered the gold standard, ddPCR BRAF V600E testing yielded a sensitivity of 91.3% and specificity of 100% for PTC diagnosis, higher than that of ARMS (sensitivity 83.1%, specificity 100%). However, ddPCR only identified one more candidate patient for surgical intervention than ARMS when the techniques were combined with cytology. CONCLUSIONS: This study highlighted the superior performance of ddPCR over ARMS in BRAF V600E detection from thyroid nodule FNA samples. Further studies are needed to evaluate the cost-effectiveness of replacing ARMS-PCR with ddPCR for surgical decision-making.


Subject(s)
DNA Mutational Analysis , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf/genetics , Thyroid Nodule/genetics , Thyroid Nodule/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy, Fine-Needle , DNA Mutational Analysis/methods , DNA Mutational Analysis/standards , Female , Humans , Male , Middle Aged , Mutation/genetics , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/standards , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies , Thyroid Gland/pathology , Thyroid Nodule/diagnosis , Thyroid Nodule/therapy , Young Adult
8.
Rev Neurol (Paris) ; 176(3): 166-169, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31932031

ABSTRACT

Pathophysiology of amyotrophic lateral sclerosis (ALS) remains partially understood even though it is accepted worldwide that motor neuron death results from a pluri-factorial process with a variable role of genetic factors. Although not distinguishable from a clinical point of view, familial forms of ALS (fALS, 10% of cases) and sporadic forms (sALS, 90% of cases) can be described. Since the identification of superoxide dismutase 1 gene (SOD1) mutations, more than 30 genes have been linked to fALS. Among these genes, five (C9ORF72, SOD1, TARDBP, FUS, TBK1) seem predominant with mutation frequencies of 40%, 20%, 5%, <5%, <5% in fALS and 6%, 3%, and <1% for the last three in sALS, respectively. The situation that classically leads to request genetic screening is the presence of a familial history of motor neuron disorders (MND) or fronto-temporal lobar dementia (FTLD). However, this dichotomy between fALS and sALS based on familial history can lead to mistakes since illegitimacy, ignorance of MND, FTD or psychiatric disorders within the family due to a familial censorship or lack of familial relationship, or a recessive autosomal inheritance could wrongly lead to failing to recognize a familial form. The significant development of genetic research and easier access to genetic tests in fALS increase the number of situations for which gene mutations are identified. The consequence is an increase in genetic requests from relatives of ALS patients who are eager to know their own genetic status and their own individual risk to develop ALS. Pre-symptomatic testing is thus becoming a daily issue in ALS Centers. This led us to propose a framework for such pre-symptomatic genetic testing for people at risk for developing ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/genetics , Genetic Testing/standards , Amyotrophic Lateral Sclerosis/epidemiology , Asymptomatic Diseases , Confidentiality/standards , DNA Mutational Analysis/methods , DNA Mutational Analysis/standards , Disclosure/standards , Early Diagnosis , Gene Frequency , Genetic Association Studies , Genetic Counseling/methods , Genetic Counseling/standards , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Molecular Diagnostic Techniques/methods , Molecular Diagnostic Techniques/standards , Prodromal Symptoms
9.
Oncologist ; 24(6): e318-e326, 2019 06.
Article in English | MEDLINE | ID: mdl-30846513

ABSTRACT

INTRODUCTION: Gefitinib, erlotinib, and afatinib represent the approved first-line options for epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC). Because pivotal trials frequently lack external validity, real-world data may help to depict the diagnostic-therapeutic pathway and treatment outcome in clinical practice. METHODS: MOST is a multicenter observational study promoted by the Veneto Oncology Network, aiming at monitoring the diagnostic-therapeutic pathway of patients with nonsquamous EGFR-mutant NSCLC. We reported treatment outcome in terms of median time to treatment failure (mTTF) and assessed the impact of each agent on the expense of the regional health system, comparing it with a prediction based on the pivotal trials. RESULTS: An EGFR mutation test was performed in 447 enrolled patients, of whom 124 had EGFR mutation and who received gefitinib (n = 69, 55%), erlotinib (n = 33, 27%), or afatinib (n = 22, 18%) as first-line treatment. Because erlotinib was administered within a clinical trial to 15 patients, final analysis was limited to 109 patients. mTTF was 15.3 months, regardless of the type of tyrosine kinase inhibitor (TKI) used. In the MOST study, the budget impact analysis showed a total expense of €3,238,602.17, whereas the cost estimation according to median progression-free survival from pivotal phase III trials was €1,813,557.88. CONCLUSION: Good regional adherence and compliance to the diagnostic-therapeutic pathway defined for patients with nonsquamous NSCLC was shown. mTTF did not significantly differ among the three targeted TKIs. Our budget impact analysis suggests the potential application of real-world data in the process of drug price negotiation. IMPLICATIONS FOR PRACTICE: The MOST study is a real-world data collection reporting a multicenter adherence and compliance to diagnostic-therapeutic pathways defined for patients with epidermal growth factor receptor-mutant non-small cell lung cancer. This represents an essential element of evidence-based medicine, providing information on patients and situations that may be challenging to assess using only data from randomized controlled trials, e.g., turn-around time of diagnostic tests, treatment compliance and persistence, guideline adherence, challenging-to-treat populations, drug safety, comparative effectiveness, and cost effectiveness. This study may be of interest to various stakeholders (patients, clinicians, and payers), providing a meaningful picture of the value of a given therapy in routine clinical practice.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Critical Pathways/statistics & numerical data , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Adult , Afatinib/economics , Afatinib/therapeutic use , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/economics , Carcinoma, Non-Small-Cell Lung/genetics , Cost-Benefit Analysis , Critical Pathways/standards , DNA Mutational Analysis/standards , DNA Mutational Analysis/statistics & numerical data , Disease Progression , Disease-Free Survival , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride/economics , Erlotinib Hydrochloride/therapeutic use , Female , Follow-Up Studies , Gefitinib/economics , Gefitinib/therapeutic use , Guideline Adherence/standards , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/economics , Lung Neoplasms/genetics , Male , Medication Adherence/statistics & numerical data , Middle Aged , Mutation , Practice Guidelines as Topic , Progression-Free Survival , Prospective Studies , Protein Kinase Inhibitors/economics , Time Factors , Treatment Failure
10.
Pediatr Nephrol ; 34(2): 195-210, 2019 02.
Article in English | MEDLINE | ID: mdl-29181713

ABSTRACT

Steroid-resistant nephrotic syndrome (SRNS) is a common cause of chronic kidney disease in childhood and has a significant risk of rapid progression to end-stage renal disease. The identification of over 50 monogenic causes of SRNS has revealed dysfunction in podocyte-associated proteins in the pathogenesis of proteinuria, highlighting their essential role in glomerular function. Recent technological advances in high-throughput sequencing have enabled indication-driven genetic panel testing for patients with SRNS. The availability of genetic testing, combined with the significant phenotypic variability of monogenic SRNS, poses unique challenges for clinicians when directing genetic testing. This highlights the need for clear clinical guidelines that provide a systematic approach for mutational screening in SRNS. The likelihood of identifying a causative mutation is inversely related to age at disease onset and is increased with a positive family history or the presence of extra-renal manifestations. An unequivocal molecular diagnosis could allow for a personalised treatment approach with weaning of immunosuppressive therapy, avoidance of renal biopsy and provision of accurate, well-informed genetic counselling. Identification of novel causative mutations will continue to unravel the pathogenic mechanisms of glomerular disease and provide new insights into podocyte biology and glomerular function.


Subject(s)
Drug Resistance/genetics , Genetic Testing/standards , Glucocorticoids/pharmacology , Nephrotic Syndrome/drug therapy , Patient Selection , Age Factors , Age of Onset , Biopsy , Child , DNA Mutational Analysis/standards , Glucocorticoids/therapeutic use , High-Throughput Nucleotide Sequencing/standards , Humans , Kidney Glomerulus/pathology , Medical History Taking , Mutation , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , Precision Medicine/methods , Treatment Outcome
12.
Clin Chem ; 64(12): 1732-1742, 2018 12.
Article in English | MEDLINE | ID: mdl-30237149

ABSTRACT

BACKGROUND: Droplet digital PCR (ddPCR) is an emerging technology for quantitative cell-free DNA oncology applications. However, assay performance criteria must be established in a standardized manner to harness this potential. We reasoned that standard protocols used in clinical chemistry assay validation should be able to fill this need. METHODS: We validated KRAS, EGFR, and BRAF quantitative ddPCR assays based on the Clinical Laboratory Improvement Act regulations for laboratory-developed tests in clinical chemistry and the matching Clinical and Laboratory Standards Institute guidelines. This included evaluation of limit of the blank (LOB), limit of detection (LOD), limit of quantification (LOQ), intraassay and interassay imprecision, analytical range, dilution linearity, accuracy (including comparison with orthogonal platforms), reference range study, interference, and stability studies. RESULTS: For the ddPCR assays, the LOB was 4 mutant copies, LODs were 12 to 22 copies, and LOQs were 35 to 64 copies. The upper limit of the dynamic range was 30000 copies, and dilutions were linear down to the LOQs with good accuracy of spike recovery of Horizon reference material. Method comparisons with next-generation sequencing and an alternative ddPCR platform showed complete qualitative agreement and quantitative concordance, with slopes of 0.73 to 0.97 and R 2s of 0.83 to 0.99. No substantial interferences were discovered. Wild-type copy numbers in plasma ranged from 462 to 6169/mL in healthy individuals. CONCLUSIONS: Standard clinical chemistry assay validation protocols can be applied to quantitative ddPCR assays. This should facilitate comparison of the performance of different assays and allow establishment of minimal significant change thresholds in monitoring applications.


Subject(s)
Chemistry, Clinical/standards , DNA Mutational Analysis/standards , Liquid Biopsy/standards , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/standards , Adult , Aged , Cell-Free Nucleic Acids , DNA Mutational Analysis/methods , ErbB Receptors/genetics , Female , Humans , Limit of Detection , Male , Middle Aged , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Reference Values
13.
J Natl Compr Canc Netw ; 16(5S): 601-604, 2018 05.
Article in English | MEDLINE | ID: mdl-29784736

ABSTRACT

Molecular testing is recommended for initial diagnosis in patients with non-small cell lung cancer (NSCLC), according to the updated NCCN Guidelines, because targeted therapies are available that can improve patient outcomes. Targeted therapies are currently approved for EGFR mutations, ALK and ROS1 gene rearrangements, and BRAF mutations, with the list of emerging "actionable" targets growing. The 2018 NCCN Guidelines for NSCLC incorporate new therapies, including the EGFR tyrosine kinase inhibitor osimertinib and the ALK inhibitor alectinib, as first-line preferences.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Practice Guidelines as Topic , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , DNA Mutational Analysis/standards , Gene Rearrangement , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Medical Oncology/standards , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/standards , Mutation , Precision Medicine/methods , Precision Medicine/standards , Progression-Free Survival , Protein Kinase Inhibitors/pharmacology , Randomized Controlled Trials as Topic , Societies, Medical/standards , United States/epidemiology
14.
Nucleic Acids Res ; 44(19): e146, 2016 Nov 02.
Article in English | MEDLINE | ID: mdl-27431322

ABSTRACT

Presence of excess unaltered, wild-type (WT) DNA providing no information of biological or clinical value often masks rare alterations containing diagnostic or therapeutic clues in cancer, prenatal diagnosis, infectious diseases or organ transplantation. With the surge of high-throughput technologies there is a growing demand for removing unaltered DNA over large pools-of-sequences. Here we present nuclease-assisted minor-allele enrichment with probe-overlap (NaME-PrO), a single-step approach with broad genome coverage that can remove WT-DNA from numerous sequences simultaneously, prior to genomic analysis. NaME-PrO employs a double-strand-DNA-specific nuclease and overlapping oligonucleotide-probes interrogating WT-DNA targets and guiding nuclease digestion to these sites. Mutation-containing DNA creates probe-DNA mismatches that inhibit digestion, thus subsequent DNA-amplification magnifies DNA-alterations at all selected targets. We demonstrate several-hundred-fold mutation enrichment in diverse human samples on multiple clinically relevant targets including tumor samples and circulating DNA in 50-plex reactions. Enrichment enables routine mutation detection at 0.01% abundance while by adjusting conditions it is possible to sequence mutations down to 0.00003% abundance, or to scan tumor-suppressor genes for rare mutations. NaME-PrO introduces a simple and highly parallel process to remove un-informative DNA sequences and unmask clinically and biologically useful alterations.


Subject(s)
Alleles , DNA Mutational Analysis , DNA/genetics , DNA/metabolism , Endonucleases/metabolism , Mutation , DNA Mutational Analysis/methods , DNA Mutational Analysis/standards , Humans , Male , Oligonucleotide Probes , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Reproducibility of Results , Sensitivity and Specificity , Workflow
15.
Cancer Sci ; 107(11): 1660-1666, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27575703

ABSTRACT

Next-generation sequencing (NGS) and digital PCR technologies allow analysis of the mutational profile of circulating cell-free DNA (cfDNA) in individuals with advanced lung cancer. We have now evaluated the feasibility of cfDNA sequencing for mutation detection in patients with non-small cell lung cancer at earlier stages. A total of 150 matched tumor and serum samples were collected from non-small cell lung cancer patients at stages IA-IIIA. Amplicon sequencing with DNA extracted from tumor tissue detected frequent mutations in EGFR (37% of patients), TP53 (39%), and KRAS (10%), consistent with previous findings. In contrast, NGS of cfDNA identified only EGFR, TP53, and PIK3CA mutations in three, five, and one patient, respectively, even though adequate amounts of cfDNA were extracted (median of 4936 copies/mL serum). Next-generation sequencing showed a high accuracy (98.8%) compared with droplet digital PCR for cfDNA mutation detection, suggesting that the low frequency of mutations in cfDNA was not due to a low assay sensitivity. Whereas the yield of cfDNA did not differ among tumor stages, the cfDNA mutations were detected in seven patients at stages IIA-IIIA and at T2b or T3. Tumor volume was significantly higher in the cfDNA mutation-positive patients than in the negative patients at stages T2b-T4 (159.1 ± 58.0 vs. 52.5 ± 9.9 cm3 , P = 0.014). Our results thus suggest that tumor volume is a determinant of the feasibility of mutation detection with cfDNA as the analyte.


Subject(s)
Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/genetics , DNA Mutational Analysis/methods , DNA, Neoplasm/blood , Lung Neoplasms/blood , Lung Neoplasms/genetics , Mutation/genetics , Tumor Burden , Carcinoma, Non-Small-Cell Lung/pathology , DNA Mutational Analysis/standards , Humans , Lung Neoplasms/pathology , Neoplasm Staging , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/standards
16.
Histopathology ; 69(4): 680-6, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27151331

ABSTRACT

BACKGROUND AND AIMS: BRAF or NRAS mutations occur in approximately 60% of cutaneous melanomas, and the identification of such mutations underpins the appropriate selection of patients who may benefit from BRAF and MEK inhibitor targeted therapies. The utility of immunohistochemistry (IHC) to detect NRAS(Q61L) mutations is currently unknown. This study sought to assess the sensitivity and specificity of anti-BRAF(V600E) (VE1), anti-NRAS(Q61R) (SP174) and anti-NRAS(Q61L) (26193) antibodies for mutation detection in a large series of cases. METHODS AND RESULTS: Mutation status was determined using the OncoCarta assay in 754 cutaneous melanomas. IHC with the anti-BRAF(V600E) antibody was performed in all cases, and the anti-NRAS(Q61R) and anti-NRAS(Q61L) antibodies were assessed in a subset of 302 samples utilizing tissue microarrays. The staining with the anti-BRAF(V600E) and anti-NRAS(Q61R) antibodies was diffuse, homogeneous and cytoplasmic. The anti-NRAS(Q61L) antibody displayed variable intensity staining, ranging from weak to strong in NRAS(Q61L) mutant tumours. The sensitivity and specificity for anti-BRAF(V600E) was 100 and 99.3%, anti-NRAS(Q61R) was 100 and 100% and anti-NRAS(Q61L) was 82.6 and 96.2%, respectively. CONCLUSIONS: The use of IHC is a fast, efficient and cost-effective method to identify single specific mutations in melanoma patients. BRAF(V600E) and NRAS(Q61R) antibodies have high sensitivity and specificity; however, the NRAS(Q61L) antibody appears less sensitive. IHC can help to facilitate the timely, appropriate selection and treatment of metastatic melanoma patients with targeted therapies. Detection of melanoma-associated mutations by IHC may also provide evidence for a diagnosis of melanoma in metastatic undifferentiated neoplasms lacking expression of melanoma antigens.


Subject(s)
DNA Mutational Analysis/methods , DNA Mutational Analysis/standards , GTP Phosphohydrolases/genetics , Melanoma/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry/methods , Immunohistochemistry/standards , Male , Melanoma/secondary , Middle Aged , Sensitivity and Specificity , Skin Neoplasms , Tissue Array Analysis , Melanoma, Cutaneous Malignant
17.
Genome Res ; 22(8): 1589-98, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22759861

ABSTRACT

Massively parallel sequencing technology and the associated rapidly decreasing sequencing costs have enabled systemic analyses of somatic mutations in large cohorts of cancer cases. Here we introduce a comprehensive mutational analysis pipeline that uses standardized sequence-based inputs along with multiple types of clinical data to establish correlations among mutation sites, affected genes and pathways, and to ultimately separate the commonly abundant passenger mutations from the truly significant events. In other words, we aim to determine the Mutational Significance in Cancer (MuSiC) for these large data sets. The integration of analytical operations in the MuSiC framework is widely applicable to a broad set of tumor types and offers the benefits of automation as well as standardization. Herein, we describe the computational structure and statistical underpinnings of the MuSiC pipeline and demonstrate its performance using 316 ovarian cancer samples from the TCGA ovarian cancer project. MuSiC correctly confirms many expected results, and identifies several potentially novel avenues for discovery.


Subject(s)
DNA Mutational Analysis/methods , Molecular Sequence Annotation/methods , Mutation , Ovarian Neoplasms/genetics , Software , Algorithms , BRCA1 Protein/genetics , Computational Biology/methods , DNA Mutational Analysis/standards , Female , Genes, Neoplasm , Humans , Reproducibility of Results
18.
Breast Cancer Res Treat ; 152(1): 129-136, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26022348

ABSTRACT

Multi-gene panels are used to identify genetic causes of hereditary breast and ovarian cancer (HBOC) in large patient cohorts. This study compares the diagnostic workflow in two centers and gives valuable insights into different next-generation sequencing (NGS) strategies. Moreover, we present data from 620 patients sequenced at both centers. Both sequencing centers are part of the German consortium for hereditary breast and ovarian cancer (GC-HBOC). All 620 patients included in this study were selected following standard BRCA1/2 testing guidelines. A set of 10 sequenced genes was analyzed per patient. Twelve samples were exchanged and sequenced at both centers. NGS results were highly concordant in 12 exchanged samples (205/206 variants = 99.51 %). One non-pathogenic variant was missed at center B due to a sequencing gap (no technical coverage). The custom enrichment at center B was optimized during this study; for example, the average number of missing bases was reduced by a factor of four (vers. 1: 1939.41, vers. 4: 506.01 bp). There were no sequencing gaps at center A, but four CCDS exons were not included in the enrichment. Pathogenic mutations were found in 12.10 % (75/620) of all patients: 4.84 % (30/620) in BRCA1, 4.35 % in BRCA2 (27/620), 0.97 % in CHEK2 (6/620), 0.65 % in ATM (4/620), 0.48 % in CDH1 (3/620), 0.32 % in PALB2 (2/620), 0.32 % in NBN (2/620), and 0.16 % in TP53 (1/620). NGS diagnostics for HBOC-related genes is robust, cost effective, and the method of choice for genetic testing in large cohorts. Adding 8 genes to standard BRCA1- and BRCA2-testing increased the mutation detection rate by one-third.


Subject(s)
Genetic Testing/methods , Genetic Testing/standards , Hereditary Breast and Ovarian Cancer Syndrome/diagnosis , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Computational Biology/methods , Computational Biology/standards , DNA Mutational Analysis/standards , DNA Mutational Analysis/trends , Female , Genomics/methods , Genomics/standards , High-Throughput Nucleotide Sequencing , Humans , Mutation , Reproducibility of Results
19.
J Transl Med ; 13: 287, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26335936

ABSTRACT

BACKGROUND: In 2014 the European Medicines Agency included exon 2, 3 and 4 KRAS and NRAS testing for the selection of metastatic colorectal cancer (mCRC) patients eligible for the therapy with anti-EGFR monoclonal antibodies. The Italian Association of Medical Oncology (AIOM) and the Italian Society of Pathology and Cytology (SIAPEC) organized an external quality assessment (EQA) scheme for CRC to evaluate inter-laboratory consistency and to ensure standardization of the results in the transition from KRAS to all-RAS testing. METHODS: Ten formalin fixed paraffin embedded specimens including KRAS/NRAS (exons 2, 3, 4) and BRAF (codon 600) mutations were validated by three referral laboratories and sent to 88 participant centers. Molecular pathology sample reports were also requested to each laboratory. A board of assessors from AIOM and SIAPEC evaluated the results according to a predefined scoring system. The scheme was composed of two rounds. RESULTS: In the first round 36% of the 88 participants failed, with 23 centers having at least one false positive or false negative while 9 centers did not meet the deadline. The genotyping error rate was higher when Sanger sequencing was employed for testing as compared with pyrosequencing (3 vs 1.3%; p = 0.01; Pearson Chi Square test). In the second round, the laboratories improved their performance, with 23/32 laboratories passing the round. Overall, 79/88 participants passed the RAS EQA scheme. Standardized Human Genome Variation Society nomenclature was incorrectly used to describe the mutations identified and relevant variations were noticed in the genotype specification. CONCLUSION: The results of the Italian RAS EQA scheme indicate that the mutational analyses are performed with good quality in many Italian centers, although significant differences in the methods used were highlighted. The relatively high number of centers failing the first round underlines the fundamental role in continued education covered by EQA schemes.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , DNA Mutational Analysis/methods , Laboratory Proficiency Testing , ras Proteins/genetics , Codon , DNA Mutational Analysis/standards , ErbB Receptors/metabolism , Europe , Exons , False Positive Reactions , Formaldehyde/chemistry , Genes, ras , Genotype , Humans , Italy , Models, Statistical , Mutation , Neoplasm Metastasis , Paraffin/chemistry , Quality Control , Reproducibility of Results
20.
Carcinogenesis ; 35(9): 2055-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24776219

ABSTRACT

DNA adducts are a measure of internal exposure to genotoxicants. However, the measurement of DNA adducts in molecular epidemiology studies often is precluded by the lack of fresh tissue. In contrast, formalin-fixed paraffin-embedded (FFPE) tissues frequently are accessible, although technical challenges remain in retrieval of high quality DNA suitable for biomonitoring of adducts. Aristolochic acids (AA) are human carcinogens found in Aristolochia plants, some of which have been used in the preparation of traditional Chinese herbal medicines. We previously established a method to measure DNA adducts of AA in FFPE tissue. In this study, we examine additional features of formalin fixation that could impact the quantity and quality of DNA and report on the recovery of AA-DNA adducts in mice exposed to AA. The yield of DNA isolated from tissues fixed with formalin decreased over 1 week; however, the levels of AA-DNA adducts were similar to those in fresh frozen tissue. Moreover, DNA from FFPE tissue served as a template for PCR amplification, yielding sequence data of comparable quality to DNA obtained from fresh frozen tissue. The estimates of AA-DNA adducts measured in freshly frozen tissue and matching FFPE tissue blocks of human kidney stored for 9 years showed good concordance. Thus, DNA isolated from FFPE tissues may be used to biomonitor DNA adducts and to amplify genes used for mutational analysis, providing clues regarding the origin of human cancers for which an environmental cause is suspected.


Subject(s)
Aristolochic Acids/metabolism , Carcinogens/metabolism , DNA Adducts/genetics , Animals , DNA Adducts/isolation & purification , DNA Adducts/metabolism , DNA Mutational Analysis/standards , Fixatives/chemistry , Formaldehyde/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Paraffin Embedding , Reference Standards , Spectrometry, Mass, Electrospray Ionization/standards , Tissue Fixation
SELECTION OF CITATIONS
SEARCH DETAIL