Genetic Alterations Detected by Targeted Next-generation Sequencing and Their Clinical Implications in Neuroblastoma

2020 ◽  
Vol 40 (12) ◽  
pp. 7057-7065
Author(s):  
KYUNG-NAM KOH ◽  
JI-YOUNG LEE ◽  
JINYEONG LIM ◽  
JUHEE SHIN ◽  
SUNG HAN KANG ◽  
...  
2012 ◽  
Vol 6 (S6) ◽  
Author(s):  
A Rose Brannon ◽  
Efsevia Vakiani ◽  
Sasinya Scott ◽  
Brooke Sylvester ◽  
Krishan Kania ◽  
...  

Author(s):  
In Ae Kim ◽  
Jae Young Hur ◽  
Hee Joung Kim ◽  
Jung Hoon Park ◽  
Jae Joon Hwang ◽  
...  

Abstract Background Despite surgical resection, early lung adenocarcinoma has a recurrence rate of 20–50%. No clear predictive markers for recurrence of early lung adenocarcinoma are available. Targeted next-generation sequencing (NGS) is rarely used to identify recurrence-related genes. We aimed to identify genetic alterations that can predict recurrence, by comparing the molecular profiles of patient groups with and without recurrence. Methods Tissues from 230 patients with resected stage I–II lung adenocarcinoma (median follow-up: 49 months) were analyzed via targeted NGS for 207 cancer-related genes. The recurrence-free survival according to the number and type of mutation was estimated using the Kaplan–Meier method. Independent predictive biomarkers related to recurrence were identified using the Cox proportional hazards model. Results Recurrence was observed in 64 patients (27.8%). In multivariate analysis adjusted for age, sex, smoking history, stage, surgical mode, and visceral pleural invasion, the CTNNB1 mutation and fusion genes (ALK, ROS1, RET) were negative prognostic factors for recurrence in early-stage lung adenocarcinoma (HR 4.47, p = 0.001; HR 2.73, p = 0.009). EGFR mutation was a favorable factor (HR 0.51, p = 0.016), but the CTNNB1/EGFR co-mutations were negative predictors (HR 19.2, p < 0.001). TP53 mutation was a negative predictor compared with EGFR mutation for recurrence (HR 5.24, p = 0.02). Conclusions: Targeted NGS can provide valuable information to predict recurrence and identify patients at high recurrence risk, facilitating selection of the treatment strategy among close monitoring and adjuvant-targeted therapy. Larger datasets are required to validate these findings.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi104-vi104
Author(s):  
Jeanette K Petersen ◽  
Henning B Boldt ◽  
Mia Sorensen ◽  
Rikke H Dahlrot ◽  
Steinbjørn Hansen ◽  
...  

Abstract INTRODUCTION Targeted next-generation sequencing (NGS) offers promising diagnostic perspectives by making it possible to detect genetic alterations with high accuracy in multiple genes as part of the daily diagnostic work-up. The importance of genetic alterations is reflected in the 2016 WHO classification of CNS tumors, where specific alterations are incorporated in the definition of certain entities. The aim of this study was to assess the diagnostic potential of a customized targeted glioma-tailored 20-gene panel to identify molecular entities of gliomas and explore their distinct mutational profiles. Moreover, the prognostic impact of molecular diagnostics was explored. METHODS We used a setup with NGS, immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) on a series of 756 glioma from both a retro- and prospective cohort. NGS data were obtained for 347 gliomas (225 analyzed prospectively, 122 analyzed retrospectively). RESULTS Three major clusters were identified by unsupervised hierarchial cluster analysis: 1) isocitrate dehydrogenase gene (IDH) 1 or 2 mutant astrocytomas with frequent mutations in TP53 (83%) and ATRX (69%), 2) IDH-mutant and 1p/19q-codeleted oligodendrogliomas with frequent mutations in TERT (65%) and CIC (52%), and 3) IDH-wildtype astrocytic gliomas/glioblastomas with frequent mutations in TERT (32%), PTEN (28%) and TP53 (22%). Among the IHC IDH1-R132H negative gliomas, NGS detected other IDH mutations in 16% of the cases. Moreover, rare but diagnostically important mutations such as BRAF V600E and H3F3A K27M were detected. Survival analysis of the reclassified gliomas showed a clear difference in survival between patients with WHO 2007 and 2016 diagnoses. Successful prospective NGS analyses were obtained for 98% of the gliomas in daily diagnostics. CONCLUSION Application of NGS panel sequencing improves diagnostic accuracy and is feasible in daily diagnostics. NGS is ready to become part of standard diagnostics in the field of gliomas.


Mediastinum ◽  
2021 ◽  
Vol 5 ◽  
pp. AB006-AB006
Author(s):  
Adam Szpechcinski ◽  
Malgorzata Szolkowska ◽  
Sebastian Winiarski ◽  
Urszula Lechowicz ◽  
Piotr Wisniewski ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5609-5609
Author(s):  
Hiroshi Ikeda ◽  
Yasushi Sasaki ◽  
Soushi Ibata ◽  
Masahiro Yoshida ◽  
Ayumi Tatekoshi ◽  
...  

Abstract Introduction 2016 NCCN guideline recommended that induction therapy used conventional chemotherapy such as proteasome inhibitors and immunomodulatory drugs for newly diagnosis Multiple Myeloma (MM) patients. Now, HDAC inhibitor, CS-1 Antibody agents, and novel proteasome inhibitor were available for the treatment of MM. Thereby, the outcome of MM was significantly improved regarding response rates and overall survival. However, relapse is inevitable in almost all patients and the cure of myeloma is difficult even now. Recurrence of myeloma is typically more aggressive with each relapse, leading to the development of treatment-refractory disease. For each treatment, we have to choose appropriate new agents. However, we do not know how to choose these new agents Therefore, we try to use next generation sequencer as a tool of drug choosing system. The aim of this study is to identify genetic alterations in MM cells by next generation sequencer for determining the optimal drug and predicting drug resistance in the future. Patients and Methods: At first, we reviewed newly diagnosed 11 patients with MM (Male: Female 6:5) The median age was 62.27 years (range, 37-78 ). Patients received novel agents including bortezomib in Sapporo Medical University Hospital. Patient plasma cell DNA was extracted from magnetic bead-enriched bone marrow CD138-positive fraction.CD138-negative cells and peripheral lymphocyte were used as matched non-tumor cells. Next, forty nanograms of DNA were used for multiplex PCR amplification with an Ion Ampliseq Comprehensive Cancer Panel. This cancer panel offers targeted coverage of all exons in 409 tumor suppressor genes and oncogenes frequently cited and frequently mutated in human cancers. (covered regions: 95.4% of total). We sequenced 15,992 regions which obtained more than 1.5 megabases of target sequence. Result: Each sample underwent on average 8.3 million sequencing reads after quality filtering. The mean read depths were 539x, and >95% of targeted bases were represented by at least 20 reads. The average number of synonymous mutations detected per patient was 5.8 (range 0-11). We also detected copy number variations which segments of genome can be deleted from sequencing data. We found genetic alterations that associated with poor prognosis and refractory to chemotherapy of MM patients. We detected mutationin several patients such as EGFR,IKBKB,ERBB3,MYH11,CYLD,TP53,CDH2. These genomes are mainly involved in several important pathways, including cell cycle regulation, RTK-MAPK-PI3K and NF-kB. And then, we can detect main mutation pathway of cancer cells and choose the pathway blockable agents. Conclusion: We performed targeted next-generation sequencing gene analysis of malignant plasma cells from patients with MM. Next generation sequencing analysis of Myeloma cells can detect mutation and copy number variations. These data predict of drug resistance and facilitate improvements in the treatment of MM patients. We can use targeted next-generation sequencing as tool of drug choosing system. This method is useful for determining the optimal drug for patients with MM in the future. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document