Next-Generation Sequencing–Based Companion Diagnostics: From Biomarker Discovery to Clinical Implementation

Author(s):  
Saumya Pant ◽  
Manjunath Ramarao ◽  
George A. Green ◽  
Michael C. Montalto
Cancers ◽  
2019 ◽  
Vol 11 (11) ◽  
pp. 1641 ◽  
Author(s):  
Caterina Fumagalli ◽  
Federica Tomao ◽  
Ilaria Betella ◽  
Alessandra Rappa ◽  
Mariarosaria Calvello ◽  
...  

The PARP inhibitor olaparib has been approved in the maintenance setting of platinum-sensitive epithelial ovarian cancer patients with germline or somatic BRCA1/2 mutation. Therefore, the availability of a tumor BRCA test has become a clinical need. We report the results of the clinical implementation of a tumor BRCA test within the frame of an institutional workflow for the management of patients with nonmucinous and nonborderline epithelial ovarian cancer. In total, 223 patients with epithelial ovarian cancer were prospectively analyzed. BRCA1/2 status was evaluated on formalin-fixed, paraffin-embedded tumor specimens using next-generation sequencing technology. The tumor BRCA test had a success rate of 99.1% (221 of 223 successfully analyzed cases) and a median turnaround time of 17 calendar days. Among the 221 cases, BRCA1 or BRCA2 pathogenic/likely pathogenic mutations were found in 62 (28.1%) cases and variants of uncertain significance in 25 (11.3%) cases. The concordance rate between tumor BRCA test results and germline BRCA1/2 status was 87%, with five cases harboring pathogenic/likely pathogenic somatic-only mutations. The next-generation, sequencing-based tumor BRCA test showed a high success rate and a turnaround time compatible with clinical purposes. The tumor BRCA test could be implemented in a molecular diagnostic setting and it may guide the clinical management of patients with epithelial ovarian cancer.


2015 ◽  
Vol 8 (1) ◽  
Author(s):  
Juan Pablo Lopez ◽  
Alpha Diallo ◽  
Cristiana Cruceanu ◽  
Laura M. Fiori ◽  
Sylvie Laboissiere ◽  
...  

2021 ◽  
Author(s):  
Kikuya Kato ◽  
Jiro Okami ◽  
Harumi Nakamura ◽  
Keiichiro Honma ◽  
Yoshiharu Sato ◽  
...  

Companion diagnostics, which predict the efficacy of molecular targeted agents based on genetic information, are indispensable for the treatment of advanced non-small cell lung carcinoma. Recent increase in the number of molecular targeted agents and the corresponding target genes have led to the demand for the simultaneous testing of multiple genes. Although gene panels using next-generation sequencing (NGS panels) are ideal for this purpose, conventional panels require high tumor content, and biopsy samples often do not meet this requirement. We developed a new NGS panel called a compact panel, to accommodate biopsy samples without the restriction of tumor content. The compact panel is characterized by high sensitivity, with detection limits for mutations of 0.14%, 0.20%, 0.48%, 0.24%, and 0.20% for EGFR exon 19 deletion, L858R, T790M, BRAF V600E, and KRAS G12C, respectively. Mutation detection also has a high quantitative ability, with correlation coefficients ranging from 0.966 to 0.992. The panel detected fusions in samples whose tumor cell content was as low as 1%. The compact panel exhibited good concordance with approved tests as follows: EGFR positive, 100.0 (95% confidence interval 95.5-100); EGFR negative, 90.9 (82.2-96.3); ALK positive, 96.7 (83.8-99.9); ALK negative, 98.4 (97.2-99.2); ROS1 positive, 100 (66.4-100); ROS1 negative, 99.0 (97.1-99.2); MET positive, 98.0 (89.0-100); MET negative 100 (92.8-100). The analytical performance demonstrated that the compact panel can handle various types of biopsy samples obtained by routine clinical practice, without requiring strict pathological monitoring like in case of conventional NGS panels.


Cancers ◽  
2019 ◽  
Vol 11 (9) ◽  
pp. 1229 ◽  
Author(s):  
Maria Gabriela O. Fernandes ◽  
Maria Jacob ◽  
Natália Martins ◽  
Conceição Souto Moura ◽  
Susana Guimarães ◽  
...  

Identification of targetable molecular changes is essential for selecting appropriate treatment in patients with advanced lung adenocarcinoma. Methods: In this study, a Sanger sequencing plus Fluorescence In Situ Hybridization (FISH) sequential approach was compared with a Next-Generation Sequencing (NGS)-based approach for the detection of actionable genomic mutations in an experimental cohort (EC) of 117 patients with advanced lung adenocarcinoma. Its applicability was assessed in small biopsies and cytology specimens previously tested for epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) mutational status, comparing the molecular changes identified and the impact on clinical outcomes. Subsequently, an NGS-based approach was applied and tested in an implementation cohort (IC) in clinical practice. Using Sanger and FISH, patients were classified as EGFR-mutated (n = 22, 18.8%), ALK-mutated (n = 9, 7.7%), and unclassifiable (UC) (n = 86, 73.5%). Retesting the EC with NGS led to the identification of at least one gene variant in 56 (47.9%) patients, totaling 68 variants among all samples. Still, in the EC, combining NGS plus FISH for ALK, patients were classified as 23 (19.7%) EGFR; 20 (17.1%) KRAS; five (4.3%) B-Raf proto-oncogene (BRAF); one (0.9%) Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2); one (0.9%) STK11; one (0.9%) TP53, and nine (7.7%) ALK mutated. Only 57 (48.7%) remained genomically UC, reducing the UC rate by 24.8%. Fourteen (12.0%) patients presented synchronous alterations. Concordance between NGS and Sanger for EGFR status was very high (κ = 0.972; 99.1%). In the IC, a combined DNA and RNA NGS panel was used in 123 patients. Genomic variants were found in 79 (64.2%). In addition, eight (6.3%) EML4-ALK, four (3.1%), KIF5B-RET, four (3.1%) CD74-ROS1, one (0.8%) TPM3-NTRK translocations and three (2.4%) exon 14 skipping MET Proto-Oncogene (MET) mutations were detected, and 36% were treatable alterations. Conclusions: This study supports the use of NGS as the first-line test for genomic profiling of patients with advanced lung adenocarcinoma.


2013 ◽  
Vol 2013 ◽  
pp. 1-8 ◽  
Author(s):  
O. M. Vanakker ◽  
A. De Paepe

Pharmacogenetics is considered as a prime example of how personalized medicine nowadays can be put into practice. However, genotyping to guide pharmacological treatment is relatively uncommon in the routine clinical practice. Several reasons can be found why the application of pharmacogenetics is less than initially anticipated, which include the contradictory results obtained for certain variants and the lack of guidelines for clinical implementation. However, more reproducible results are being generated, and efforts have been made to establish working groups focussing on evidence-based clinical guidelines. For another pharmacogenetic hurdle, the speed by which a pharmacogenetic profile for a certain drug can be obtained in an individual patient, there has been a revolution in molecular genetics through the introduction of next generation sequencing (NGS), making it possible to sequence a large number of genes up to the complete genome in a single reaction. Besides the enthusiasm due to the tremendous increase of our sequencing capacities, several considerations need to be made regarding quality and interpretation of the sequence data as well as ethical aspects of this technology. This paper will focus on the different NGS applications that may be useful for pharmacogenomics in children and the challenges that they bring on.


Sign in / Sign up

Export Citation Format

Share Document