scholarly journals Prospective Next-Generation Sequencing Molecular Profiling of Myeloid Malignancies: Assessment of Information Benefit and Impact on Patient Care

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3848-3848
Author(s):  
Suzanne Kamel-Reid ◽  
Mariam Thomas ◽  
Mahadeo A. Sukhai ◽  
Dwayne L. Barber ◽  
Swati Garg ◽  
...  

Abstract Introduction. Recent genome profiling studies have increased our understanding of the mutation landscapes of myeloid malignancies. Molecular testing of AMLs (NPM1, FLT3-ITD, KIT) and MPNs (JAK2, CALR) constitute current diagnostic standard-of-care. Evidence for the diagnostic, prognostic and/or therapeutic impact of a growing set of genes and variants in myeloid malignancies allows for more accurate patient stratification and enhanced patient management. This has led to consideration of next-generation sequencing (NGS) approaches to simultaneously detect multiple variants in myeloid malignancies for use in the clinical diagnostic setting, to supplant single-gene molecular assays. We designed the Princess Margaret Advanced Genomics in Leukemia (AGILE) trial to prospectively assess the utility of NGS molecular profiling in the management of patients with myeloid malignancies. Methods. Patients for the AGILE trial are consented at the time of diagnosis using an REB approved written consent. Bone marrow or peripheral blood samples are collected at consent, accessioned within CoPath, and DNA extracted for NGS testing. NGS molecular profiling was performed using the TruSight Myeloid Sequencing Panel (TMSP; Illumina) on the MiSeq benchtop genome sequencer (Illumina) by the University Health Network Advanced Molecular Diagnostics Laboratory. The TMSP enables profiling of 54 genes (39 hotspot region; 15 complete coding region coverage) using amplicon-based library preparation and sequencing by synthesis. The TMSP detects the CALR 52 base pair deletion relevant to myelofibrosis, but not FLT3 internal tandem duplications greater than 30 base pair in size. Data were analyzed by NextGENe (v.2.3.1, SoftGenetics) and MiSeq Reporter v2.4.60. A specific script enabling alignment and calling of CALR deletions was added to the analysis to ensure there were no false negative calls. Additional testing and verification of CEBPA variants was performed by Sanger sequencing. Variants were interpreted according to Sukhai et al (Genetics in Medicine, 2015), reviewed by lab directors and reported in the Electronic Patient Record. Impact on patient care was defined as: potential for post-consolidation clinical trials; changes to frequency of monitoring; and, changes to transplant management. Cases were discussed in an interdisciplinary Genomic Tumor Board setting, at which NGS profiling data were reviewed in the context of all other diagnostic information for the patient, to determine impact on patient care. Results. Between February 11 and July 24, 2015, 162 patients were consented for AGILE; 148/162 were profiled by NGS, and to date 124/148 have been reviewed and interpreted. 62/124 (50%) of interpreted cases had a diagnosis of acute myeloid leukemia (AML); 21/124 (20%) with myeloproliferative neoplasms (MPNs); 13/124 (10%) with myelodysplastic syndromes (MDS); 6/124 (5%) with MDS/MPN; and, 15% with other hematologic malignancies. 90% of all cases profiled were informative for at least one variant (range 1-9 variants, average 3.1 variants/case). AML, MDS and MDS/MPN cases exhibited slightly more variants (3.4-4.4 variants/case) than did MPN cases (2.6 variants/case). Overall, 69% of variants were potentially actionable (Sukhai et al, 2015: 23% class 1; 8% class 2; 38% class 3), with a large fraction of cases (90/124, 72.6%) demonstrating at least one class 1 or class 3 variant. Additionally, 73/124 (58.9%) of patients exhibited actionable, class 1, variants not currently being identified by routine molecular diagnostics. In AMLs and MPNs, 88-90% of cases exhibited at least one potentially actionable variant; NGS profiling was more informative in AMLs (62% of cases exhibiting potentially actionable variants not profiled in standard of care testing, compared to 12% of MPN cases). Conclusions. We report the results of a prospective analysis of integrated NGS profiling in the context of diagnosis and management of patients with myeloid malignancies. Using a targeted NGS panel, molecular profiling of patients yielded significant information benefit over current standard approaches in 58.9% of cases analyzed, enabling potential impact on patient management. These data highlight the utility of NGS profiling to complement the initial diagnostic evaluation of myeloid malignancies. Disclosures Gupta: Incyte: Honoraria, Research Funding; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees.

2018 ◽  
Author(s):  
Angela Abicht ◽  
Teresa Neuhann ◽  
Stefanie Balg ◽  
Daniela Gonzalez-Fassreiner ◽  
Verena Steinke-Lange ◽  
...  

2018 ◽  
Vol 56 (9) ◽  
Author(s):  
Patricia J. Simner ◽  
Heather B. Miller ◽  
Florian P. Breitwieser ◽  
Gabriel Pinilla Monsalve ◽  
Carlos A. Pardo ◽  
...  

ABSTRACT The purpose of this study was to develop and optimize different processing, extraction, amplification, and sequencing methods for metagenomic next-generation sequencing (mNGS) of cerebrospinal fluid (CSF) specimens. We applied mNGS to 10 CSF samples with known standard-of-care testing (SoC) results (8 positive and 2 negative). Each sample was subjected to nine different methods by varying the sample processing protocols (supernatant, pellet, neat CSF), sample pretreatment (with or without bead beating), and the requirement of nucleic acid amplification steps using DNA sequencing (DNASeq) (with or without whole-genome amplification [WGA]) and RNA sequencing (RNASeq) methods. Negative extraction controls (NECs) were used for each method variation (4/CSF sample). Host depletion (HD) was performed on a subset of samples. We correctly determined the pathogen in 7 of 8 positive samples by mNGS compared to SoC. The two negative samples were correctly interpreted as negative. The processing protocol applied to neat CSF specimens was found to be the most successful technique for all pathogen types. While bead beating introduced bias, we found it increased the detection yield of certain organism groups. WGA prior to DNASeq was beneficial for defining pathogens at the positive threshold, and a combined DNA and RNA approach yielded results with a higher confidence when detected by both methods. HD was required for detection of a low-level-positive enterovirus sample. We demonstrate that NECs are required for interpretation of these complex results and that it is important to understand the common contaminants introduced during mNGS. Optimizing mNGS requires the use of a combination of techniques to achieve the most sensitive, agnostic approach that nonetheless may be less sensitive than SoC tools.


2011 ◽  
Vol 11 (3) ◽  
pp. 333-343 ◽  
Author(s):  
Zhenqiang Su ◽  
Baitang Ning ◽  
Hong Fang ◽  
Huixiao Hong ◽  
Roger Perkins ◽  
...  

Blood ◽  
2021 ◽  
Author(s):  
Hsuan-Ting Huang ◽  
Maria Eugenia Figueroa

Epigenetic deregulation is now a well-recognized -though not yet fully understood- mechanism that contributes to the development and progression of myeloid malignancies. In the past 15 years, next generation sequencing studies have revealed patterns of aberrant DNA methylation, altered chromatin states, and mutations in chromatin modifiers across the spectrum of myeloid malignancies. Studies into the mechanisms that drive these diseases through mouse modeling have helped identify new avenues for therapeutic interventions, from initial treatment to resistant, relapsed disease. This is particularly significant when chemotherapy with cytotoxic agents remains the general standard of care. In this review, we will discuss some of the recent findings of epigenetic mechanisms and how these are informing the development of more targeted strategies for therapeutic intervention in myeloid malignancies.


Author(s):  
Saskia Biskup

Next-Generation-Sequencing (NGS) techniques are currently on the rise. This is seen as a revolution by (most) geneticists. The wealth of data stemming from Next-Generation-Sequencing will without a doubt lead to significant advances in the field of molecular diagnostics. On the clinical side, this will be higher detection rates of the genetic causes of particular diseases in patients. On the scientific side, NGS techniques will lead to the discovering of genes related to certain diseases (see, for example, Mardis, et al., 2009; Haack, et al., 2010; Lupski, et al., 2010). However, these advances come at a price: geneticists will be confronted with different and new ICT issues related to NGS. Because of the so far unknown amount of data stemming from NGS, these ICT issues need to be taken seriously. The purpose of this chapter is to give an overview on the different ICT aspects that come with the introduction of Next-Generation-Sequencing in molecular diagnostics.


2020 ◽  
Vol 10 (4) ◽  
pp. 206
Author(s):  
Georgia Ι. Grigoriadou ◽  
Stepan M. Esagian ◽  
Han Suk Ryu ◽  
Ilias P. Nikas

Malignant pleural effusions (MPEs) often develop in advanced cancer patients and confer significant morbidity and mortality. In this review, we evaluated whether molecular profiling of MPEs with next generation sequencing (NGS) could have a role in cancer management, focusing on lung cancer. We reviewed and compared the diagnostic performance of pleural fluid liquid biopsy with other types of samples. When applied in MPEs, NGS may have comparable performance with corresponding tissue biopsies, yield higher DNA amount, and detect more genetic aberrations than blood-derived liquid biopsies. NGS in MPEs may also be preferable to plasma liquid biopsy in advanced cancer patients with a MPE and a paucicellular or difficult to obtain tissue/fine-needle aspiration biopsy. Of interest, post-centrifuge supernatant NGS may exhibit superior results compared to cell pellet, cell block or other materials. NGS in MPEs can also guide clinicians in tailoring established therapies and identifying therapy resistance. Evidence is still premature regarding the role of NGS in MPEs from patients with cancers other than lung. We concluded that MPE processing could provide useful prognostic and theranostic information, besides its diagnostic role.


2018 ◽  
Vol 71 (9) ◽  
pp. 767-773 ◽  
Author(s):  
Caterina Fumagalli ◽  
Davide Vacirca ◽  
Alessandra Rappa ◽  
Antonio Passaro ◽  
Juliana Guarize ◽  
...  

BackgroundMolecular profiling of advanced non-small cell lung cancers (NSCLC) is essential to identify patients who may benefit from targeted treatments. In the last years, the number of potentially actionable molecular alterations has rapidly increased. Next-generation sequencing allows for the analysis of multiple genes simultaneously.AimsTo evaluate the feasibility and the throughput of next-generation sequencing in clinical molecular diagnostics of advanced NSCLC.MethodsA single-institution cohort of 535 non-squamous NSCLC was profiled using a next-generation sequencing panel targeting 22 actionable and cancer-related genes.Results441 non-squamous NSCLC (82.4%) harboured at least one gene alteration, including 340 cases (63.6%) with clinically relevant molecular aberrations. Mutations have been detected in all but one gene (FGFR1) of the panel. Recurrent alterations were observed in KRAS, TP53, EGFR, STK11 and MET genes, whereas the remaining genes were mutated in <5% of the cases. Concurrent mutations were detected in 183 tumours (34.2%), mostly impairing KRAS or EGFR in association with TP53 alterations.ConclusionsThe study highlights the feasibility of targeted next-generation sequencing in clinical setting. The majority of NSCLC harboured mutations in clinically relevant genes, thus identifying patients who might benefit from different targeted therapies.


Sign in / Sign up

Export Citation Format

Share Document