scholarly journals Systematic discovery of gene fusions in pediatric cancer by integrating RNA-seq and WGS

2021 ◽  
Author(s):  
Ianthe A.E.M. van Belzen ◽  
Casey Cai ◽  
Marc van Tuil ◽  
Shashi Badloe ◽  
Eric Strengman ◽  
...  

Background Gene fusions are important cancer drivers in pediatric cancer and their accurate detection is essential for diagnosis and treatment. Clinical decision-making requires high confidence and precision of detection. Recent developments show RNA sequencing (RNA-seq) is promising for genome-wide detection of fusion products, but hindered by many false positives that require extensive manual curation and impede discovery of pathogenic fusions. Results We developed Fusion-sq to detect tumor-specific gene fusions by integrating and 'fusing' evidence from RNA-seq and whole genome sequencing (WGS) using intron-exon gene structure. In a pediatric pan-cancer cohort of 130 patients, we identified 165 high confidence tumor-specific gene fusions and their underlying structural variants (SVs). This includes all clinically relevant fusions known to be present in this cohort (30 patients). Fusion-sq distinguishes healthy-occurring from tumor-specific fusions, and resolves fusions in amplified regions and copy number unstable genomes. A high gene fusion burden is associated with copy number instability. We identified 27 potentially pathogenic fusions involving oncogenes or tumor-suppressor genes characterised by underlying SVs or expression changes indicative of activating or disruptive effects. Conclusions Our results indicate how clinically relevant and potentially pathogenic gene fusions can be identified and their functional effects investigated by combining WGS and RNA-seq. Integrating RNA fusion predictions with underlying SVs advances fusion detection beyond extensive manual filtering. Taken together, we developed a method for identifying candidate fusions that is suitable for precision oncology applications. Our method provides multi-omics evidence for assessing the pathogenicity of tumor-specific fusions for future clinical decision making.

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 11035-11035
Author(s):  
Kristen Marrone ◽  
Jessica Tao ◽  
Jenna VanLiere Canzoniero ◽  
Paola Ghanem ◽  
Emily Nizialek ◽  
...  

11035 Background: The accelerated impact of next generation sequencing (NGS) in clinical decision making requires the integration of cancer genomics and precision oncology focused training into medical oncology education. The Johns Hopkins Molecular Tumor Board (JH MTB) is a multi-disciplinary effort focused on integration of NGS findings with critical evidence interpretation to generate personalized recommendations tailored to the genetic footprint of individual patients. Methods: The JH MTB and the Medical Oncology Fellowship Program have developed a 3-month precision oncology elective for fellows in their research years. Commencing fall of 2020, the goals of this elective are to enhance the understanding of NGS platforms and findings, advance the interpretation and characterization of molecular assay outputs by use of mutation annotators and knowledgebases and ultimately master the art of matching NGS findings with available therapies. Fellow integration into the MTB focuses on mentored case-based learning in mutation characterization and ranking by levels of evidence for actionability, with culmination in form of verbal presentations and written summary reports of final MTB recommendations. A mixed methods questionnaire was administered to evaluate progress since elective initiation. Results: Three learners who have participated as of February 2021 were included. Of the two who had completed the MTB elective, each have presented at least 10 cases, with at least 1 scholarly publication planned. All indicated strong agreement that MTB elective had increased their comfort with interpreting clinical NGS reports as well as the use of knowledgebases and variant annotators. Exposure to experts in the field of molecular precision oncology, identification of resources necessary to interpret clinical NGS reports, development of ability to critically assess various NGS platforms, and gained familiarity with computational analyses relevant to clinical decision making were noted as strengths of the MTB elective. Areas of improvement included ongoing initiatives that involve streamlining variant annotation and transcription of information for written reports. Conclusions: A longitudinal elective in the JHU MTB has been found to be preliminarily effective in promoting knowledge mastery and creating academic opportunities related to the clinical application of precision medicine. Future directions will include leveraging of the MTB infrastructure for research projects, learner integration into computational laboratory meetings, and expansion of the MTB curriculum to include different levels of learners from multiple medical education programs. Continued elective participation will be key to understanding how best to facilitate adaptive expertise in assigning clinical relevance to genomic findings, ultimately improving precision medicine delivery in patient care and trial development.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 1568-1568
Author(s):  
Navdeep Dehar ◽  
Tasnima Abedin ◽  
Patricia A. Tang ◽  
D. Gwyn Bebb ◽  
Winson Y. Cheung

1568 Background: With the increasing number and frequency of biomarker and genetic tests that are offered to patients with cancer, it is important to ensure that they fully understand the implications of these tests. In this survey study, we aimed to compare the attitudes and expectations of patients and cancer physicians about the role of biomarker and genetic testing in clinical decision-making. Methods: Two separate, complimentary, self-administered questionnaires for cancer patients and their physicians, respectively, were collected in Calgary, Alberta, Canada. Survey responses from patients were subsequently matched with those of their corresponding oncologists to form patient–oncologist dyads. We determined the concordance rates between responses of patients and those of their oncologists. Results: A total of 113 patients and 15 physicians participated in the study from July to September 2019. Patients demonstrated good understanding of general cancer biology (79%) and diagnostic processes (91%) associated with precision oncology. About 70% patients were willing to undergo minor procedures, and participate in research involving biomarker or genetic testing; however, this was over-estimated by their physicians in 82% of cases. Many patients felt that their tumor should be tested to guide treatment (70%) and were not bothered by potential delays in treatment due to testing (23%). These views from patients were largely shared by their oncologists (concordance 64%). While only 28% patients thought that they had enough knowledge to make informed decisions, majority (68%) said that they needed more information. Importantly, knowledge and expectations regarding the applications of biomarker or genetic test results on actual diagnosis and prognosis were grossly discrepant between patients and their oncologists (concordance 26% and 36%, respectively). Conclusions: Patients and cancer physicians tend to be aware of the advances in precision oncology and are willing to participate in biomarker and genetic testing and research. However, they do not consistently agree about the roles and applications of these tests, which may result in misplaced expectations. Strategies to improve education and communication are needed to align these expectations and improve the quality of clinical decision-making.


2020 ◽  
Vol 26 (7) ◽  
pp. 992-994 ◽  
Author(s):  
David Tamborero ◽  
◽  
Rodrigo Dienstmann ◽  
Maan Haj Rachid ◽  
Jorrit Boekel ◽  
...  

2021 ◽  
pp. 1-3
Author(s):  
Ira Seibel

In the era of precision oncology, major strides are being made to use individual tumor information for clinical decision-making. Differing from traditional biopsy methods, the emerging practice of liquid biopsy provides a minimally invasive way of obtaining tumor cells and derived molecules. Liquid biopsy provides a means to detect and monitor disease progression, recurrence, and treatment response in a noninvasive way, and to potentially complement classical biopsy. Uveal melanoma (UM) is a unique malignancy, with diagnosis heavily reliant on imaging, few repeat biopsies, and a high rate of metastasis, which occurs hematogenously and often many years after diagnosis. In this disease setting, a noninvasive biomarker to detect, monitor, and study the disease in real time could lead to better disease understanding and patient care. While advances have been made in the detection of tumor-disseminated components, sensitivity and specificity remain important challenges. Ambiguity remains in how to interpret current findings and in how liquid biopsy can have a place in clinical practice. Related publications in UM are few compared to other cancers, but with further studies we may be able to uncover more about the biology of disseminated molecules and the mechanisms involved in the progression to metastasis.


2018 ◽  
Author(s):  
Nicholas G. Smith ◽  
Rekha Gyanchandani ◽  
Grzegorz Gurda ◽  
Peter C. Lucas ◽  
Ryan J. Hartmaier ◽  
...  

AbstractBackgroundBreast cancer is the most common invasive cancer among women worldwide. Next-generation sequencing (NGS) has revolutionized the study of cancer across research labs around the globe, however genomic testing in clinical settings remain limited. Advances in sequencing reliability, pipeline analysis, accumulation of relevant data, and the reduction of costs are rapidly increasing the feasibility of NGS-based clinical decision making.MethodsWe report the development of MammaSeq, a breast cancer specific NGS panel, targeting 79 genes and 1369 mutations, optimized for use in primary and metastatic breast cancer. To validate the panel, 46 solid tumor and 14 plasma circulating-free cfDNA samples were sequenced to a mean depth of 2311X and 1820 X respectively. Variants were called using Ion Torrent Suite 4.0 and annotated with cravat CHASM. CNVKit was used to call copy number variants in the solid tumor cohort. The oncoKB Precision Oncology Database was used to identify clinically actionable variants. ddPCR was used to validate select cfDNA mutations.ResultsIn cohorts of 46 solid tumors and 14 cfDNA samples from patients with advanced breast cancer we identified 592 and 43 protein coding mutations. Mutations per sample in the solid tumor cohort ranged from 1 to 128 (median 3) and the cfDNA cohort ranged from 0 to 26 (median 2.5). Copy number analysis in the solid tumor cohort identified 46 amplifications and 35 deletions. We identified 26 clinically actionable variants (levels 1-3) annotated by OncoKB, distributed across 20 out of 46 cases (40%), in the solid tumor cohort. Allele frequencies of ESR1 and FOXA1 mutations correlated with CA.27.29 levels in patient matched blood draws.ConclusionsIn solid tumors biopsies and cfDNA, MammaSeq detects clinicaly actionable mutations (oncoKB levels 1-3) in 22/46 (48%) solid tumors and in 4/14 (29%) of cfDNA samples. MammaSeq is a targeted panel suitable for clinically actionable mutation detection in breast cancer.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 3128-3128
Author(s):  
Meena Sadaps ◽  
Kathryn Demski ◽  
Ying Ni ◽  
Vicky Konig ◽  
Brandie Leach ◽  
...  

3128 Background: Multidisciplinary molecular tumor boards were first established with the onset of precision oncology (PO), as many clinicians were unfamiliar with the interpretation and incorporation of the information into clinical practice. PO has since rapidly evolved and integrated itself into standard of care practices for most cancer patients, yet molecular tumor boards have not grown accordingly and in fact some have been discontinued. There remains a paucity of data in regards to the value and impact of molecular tumor board discussions themselves. We previously reported on our longitudinal experiences in PO ( Sadaps et al, 2018), focusing on the therapeutic impact of matched therapy. Here, we report on the utility of our molecular tumor board in clinical decision making. Methods: We conducted a retrospective review of patients seen at Cleveland Clinic with a solid tumor malignancy who had large panel, next-generation-sequencing (NGS) performed via any commercial platform from November 2019-January 2021. Cases were filtered through a local therapeutic algorithm and then reviewed individually. Initial review was performed by a core genomics committee comprised of 2 oncologists and 2 genetic counselors. Interesting and/or complex cases were flagged for discussion at our bimonthly molecular tumor board, which is regularly attended by medical oncologists, pathologists, genetic counselors, bioinformaticians, and patient care coordinators. Data analyzed included categorization of treatment recommendations and the percentage of cases for which initial recommendations were changed based on tumor board discussion. Results: Of 782 total cases, 575 (73.5%) had a clinically relevant genomics tumor board (GTB) recommendation as compared to 51.7% from our previously reported study. 16.7% of patients had on label recommendation(s) and 86.4% had off label/ clinical trial recommendation(s). 179 (22.9%) patients were recommended for genetic counseling (GC). During our bimonthly GTB, we discussed 173 (22.1%) of these cases. Of the discussed cases, the most common tumor types were hepatobiliary (18.5%), lower gastrointestinal (17.3%), and breast (16.2%). Topics of discussion at GTB included such things as pathologic/histologic/molecular testing, prioritization of available trials, appropriateness of an off label therapy, and need for a genetics consult. Discussion at GTB resulted in a change in treatment recommendation in 63 (36.4%) cases. Conclusions: Discussions from multidisciplinary molecular tumor board impacted treatment decisions for our patients. Referral to GC was also common and should be considered an integral part of somatic sequencing review. Molecular tumor boards remain a crucial platform for treatment guidance and clinical management, especially given the increase in “actionability” over the years due to newly discovered targets and targeted therapies in this rapidly evolving field.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e15085-e15085
Author(s):  
Katerina Nuzhdina ◽  
Yaroslav Lozinsky ◽  
Svetlana Podsvirova ◽  
Arthur Baisangurov ◽  
Kelley Morgan ◽  
...  

e15085 Background: Analysis of the genetic and transcriptomic profile of solid tumors via next-generation sequencing (NGS) assays is fundamental to propel precision medicine into clinical practice. NGS technology applied to tumor analysis allows for the characterization of somatic alterations, clonality, altered gene expression, and other parameters using a small amount of tissue. Therefore, to uncover cancer-promoting and suppressing activity of the tumor and the tumor microenvironment (TME), we developed the BostonGene TUMOR PORTRAIT assay, integrating whole-exome sequencing (WES) and mRNA sequencing (RNA-seq). Here, we demonstrate the analytical and clinical validity of the assay. Methods: The accuracy, reproducibility, and robustness of the BostonGene assay were evaluated using reference genomic DNA, reference RNA, well-characterized cell lines, and fresh frozen (FF) tumor and normal tissue containing known single nucleotide variants (SNVs), indels, copy number alterations (CNAs), gene fusions and a reference RNA Spike-In mix containing known levels of specific transcripts. The analysis was performed using the BostonGene automated pipeline. Additionally, we demonstrated high concordance of gene expression measured using two orthogonal techniques, RNA-seq and RT-PCR. Results: The BostonGene TUMOR PORTRAIT assay demonstrated high specificity (>98.1%, >99.8%, >96.9%) and sensitivity (>98.3%, >99.2%, >97.1%) for the detection of SNVs, indels, and CNAs, respectively, with low false-positive and false-negative rates. The assay demonstrated a 100% concordance in the mutation (SNV/indel) call rate across all replicates, and a 100% concordance in the mutation (SNV/indel) and copy number variation (CNV) call rate across all runs. The measurement of gene expression from RNA-seq was achieved with high accuracy (>0.96%) and low variation across genes (<6.0%), demonstrating the ability of the assay to provide transcriptomic information. Furthermore, gene fusions were detected in RNA-seq data with high sensitivity (>95.8%) and specificity (>99%) in a reproducible manner. Using the integrated pipeline that utilizes both WES and RNA-seq, we were able to accurately compute all disease-relevant molecular parameters including tumor genomics, tumor transcriptome phenotype, expression of clinically actionable therapeutic targets, tumor microenvironment composition, and tumor clonality within the single BostonGene TUMOR PORTRAIT report. Conclusions: Analytical and clinical validation results show that the BostonGene TUMOR PORTRAIT assay, based on the sequencing of DNA and RNA, provides a comprehensive and accurate view of the tumor molecular profile, identifying all clinically actionable genetic, transcriptomic, and TME targets. Validation of the BostonGene TUMOR PORTRAIT assay provides a solid foundation for the future development of precision oncology.


Author(s):  
Jia Zeng ◽  
Md Abu Shufean

The rapid growth and decreasing cost of Next-generation sequencing (NGS) technologies have made it possible to conduct routine large panel genomic sequencing in many disease settings, especially in the oncology domain. Furthermore, it is now known that optimal disease management of patients depends on individualized cancer treatment guided by comprehensive molecular testing. However, translating results from molecular sequencing reports into actionable clinical insights remains a challenge to most clinicians. In this review, we discuss about some representative systems that leverage artificial intelligence (AI) to facilitate some processes of clinicians’ decision making based upon molecular data, focusing on their application in precision oncology. Some limitations and pitfalls of the current application of AI in clinical decision making are also discussed.


2011 ◽  
Vol 20 (4) ◽  
pp. 121-123
Author(s):  
Jeri A. Logemann

Evidence-based practice requires astute clinicians to blend our best clinical judgment with the best available external evidence and the patient's own values and expectations. Sometimes, we value one more than another during clinical decision-making, though it is never wise to do so, and sometimes other factors that we are unaware of produce unanticipated clinical outcomes. Sometimes, we feel very strongly about one clinical method or another, and hopefully that belief is founded in evidence. Some beliefs, however, are not founded in evidence. The sound use of evidence is the best way to navigate the debates within our field of practice.


Sign in / Sign up

Export Citation Format

Share Document