Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 5354-5354
    Abstract: Solid tumor profiling assays need to deliver accurate and consistent results in the face of decreased quality and quantity of nucleic acids extracted from FFPE samples. Understanding the performance of a particular solid tumor profiling assay with FFPE tissue is critical, but with limited and non-renewable samples available to most assay-developers, the sample number used to understand this performance can be small. TruSight® Tumor 1701 is an Illumina-developed comprehensive solid tumor profiling panel targeting 170 genes using DNA and RNA from FFPE samples. In order to confirm the robustness of the assay with FFPE tissue, 2310 FFPE samples were brought in-house and evaluated. Quantity of both DNA and RNA extraction were determined by various methods, including AccuClear™, Qubit™ and Quantifluor® fluourometric assays. Overall, & gt;95% of the samples achieved the minimum concentrations required for the TruSight® Tumor 170 assay. As a surrogate for DNA quality, we measured the amplification potential of the nucleic acid by assessing a ΔCq value using quantitative PCR after normalization to a fixed input mass. To assess RNA quality, we used the DV200 metric, which measures the percentage of RNA fragments & gt;200 nucleotides in length. We examined ΔCq and DV200 values across different tissues and didn’t find a significant difference between tissues. Finally, we assessed the ability of samples to pass the sample quality control (QC) metrics in the TruSight® Tumor 170 assay. These QC metrics ensure accurate variant calling, with a sensitivity and specificity of ≥95%. We found that samples that had a ΔCq value of ≤5 and a DV200 value of ≥20 achieved a QC success rate above 95%. This data highlights the need for further investigation into the methods for extraction, quantification and quality assessment of nucleic acids for solid tumor profiling and underscores the robustness of TruSight® Tumor 170 with FFPE samples. 1 For Research Use Only. Not for use in diagnostic procedures. Citation Format: Jennifer S. LoCoco, Li Teng, Danny Chou, Xiao Chen, Byron Luo, Jennifer Sayne, Ashley Adams, Naseem Ajili, Cody Chivers, Beena Murthy, Laurel Ball, Allan Castaneda, Katie Clark, Brian Crain, Anthony Daulo, Manh Do, Tingting Du, Sarah Dumm, Yonmee Han, Michael Havern, Chia-Ling Hsieh, Tingting Jiang, Suzanne Johansen, Scott Lang, Rachel Liang, Jaime McLean, Yousef Nassiri, Austin Purdy, Jason Rostron, Jennifer Silhavy, June Snedecor, Natasha Talago, Li Teng, Kevin Wu, Chen Zhao, Clare Zlatkov, Ali Kuraishy, Karen Gutekunst, Sohela De Rozieres, Matthew Friedenberg, Han-Yu Chuang, Anne C. Jager. Evaluation of quantity, quality and performance with the TruSight® Tumor 170 solid tumor profiling assay of nucleic acids extracted from formalin-fixed paraffin-embedded (FFPE) tissue sections [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5354. doi:10.1158/1538-7445.AM2017-5354
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Genome Biology, Springer Science and Business Media LLC, Vol. 22, No. 1 ( 2021-12)
    Abstract: Targeted sequencing using oncopanels requires comprehensive assessments of accuracy and detection sensitivity to ensure analytical validity. By employing reference materials characterized by the U.S. Food and Drug Administration-led SEquence Quality Control project phase2 (SEQC2) effort, we perform a cross-platform multi-lab evaluation of eight Pan-Cancer panels to assess best practices for oncopanel sequencing. Results All panels demonstrate high sensitivity across targeted high-confidence coding regions and variant types for the variants previously verified to have variant allele frequency (VAF) in the 5–20% range. Sensitivity is reduced by utilizing VAF thresholds due to inherent variability in VAF measurements. Enforcing a VAF threshold for reporting has a positive impact on reducing false positive calls. Importantly, the false positive rate is found to be significantly higher outside the high-confidence coding regions, resulting in lower reproducibility. Thus, region restriction and VAF thresholds lead to low relative technical variability in estimating promising biomarkers and tumor mutational burden. Conclusion This comprehensive study provides actionable guidelines for oncopanel sequencing and clear evidence that supports a simplified approach to assess the analytical performance of oncopanels. It will facilitate the rapid implementation, validation, and quality control of oncopanels in clinical use.
    Type of Medium: Online Resource
    ISSN: 1474-760X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2040529-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 565-565
    Abstract: Recent studies have highlighted the importance of gene fusions and splice variants in solid tumor profiling1. Next-generation sequencing can be an effective means of detecting these alterations in FFPE samples using RNA rather than DNA, as a single chimeric RNA transcript could result from numerous alterations in DNA2. To that end, Illumina developed TruSight® Tumor 1703, a comprehensive, hybrid capture-based NGS assay targeting 170 key cancer genes. Along with a DNA workflow, the assay includes a RNA workflow for the identification of splice variants and gene fusions. Following sequencing on the NextSeq® or HiSeq® instruments, TruSight® Tumor 170 offers an analytical pipeline which initiates variant calling. These algorithms were first optimized against the simulated read data from & gt;350 fusions and splice variants reported in the RNA content of the gene panel. A hybrid approach of read alignment and assembly was used to enhance the fusion calling sensitivity. Deliberate filters were designed to reduce false positive calling from sequence homologs, polymerase read-through, or FFPE artifacts. For splice variant calling, a panel of FFPE non-cancerous samples were used to capture false positive mutation calls. With endogenous RNA splicing in cellular physiology, exon-boundary probes were added in the hybrid capture to enhance enrichment efficiency. To the best of our knowledge, there is not yet a standard definition for the limit of detection (LoD) in detecting gene fusions and splice variants from NGS data. We propose to define the LoD of a fusion calling and splice variant NGS panel as the lowest molecule count of a chimeric transcript that could be reliably detected with a sufficient number of supporting sequencing reads. To determine the LoD of TruSight® Tumor 170 using this definition, we mixed cell lines expressing a panel of known fusions and splice variants to measure the copy number of each chimeric transcript. Using these samples we examined the ability of the assay to confidently detect the alterations using 40 ng of RNA input. To demonstrate the analytical sensitivity and specificity of this NGS based assay, we compiled a panel of 49 mixed samples and validated the molecule count to be near the LoD of 5 copies per ng RNA input by PCR. The sensitivity was & gt;98% for fusions and 100% for splice variants. For understanding the limit of blank (LoB) of the assay, another panel of 40 samples not harboring fusions and splice variants was also assessed by TruSight® Tumor 170. These samples demonstrated a ~97% specificity for fusion calling and & gt;95% specificity for splice variant calling. These results indicate that the TruSight® Tumor 170 panel analysis can identify lowly expressed fusions and splice variants from a small amount of compromised RNA from solid tumor samples at high analytical sensitivity and specificity. 1 Klijn et al. (2015) 2 Maher et al. (2009) 3 For Research Use Only. Citation Format: Tingting Du, June Snedecor, Jennifer S. LoCoco, Xiao Chen, Laurel Ball, Allan Castaneda, Danny Chou, Katie Clark, Brian Crain, Anthony Daulo, Manh Do, Sarah Dumm, Yonmee Han, Mike Havern, Chia-Ling Hsieh, Tingting Jiang, Suzanne Johansen, Scott Lang, Rachel Liang, Jaime McLean, Yousef Nassiri, Austin Purdy, Jason Rostron, Jennifer Silhavy, Natasha Talago, Li Teng, Kevin Wu, Clare Zlatkov, Chen Zhao, Ali Kuraishy, Karen Gutekunst, Sohela De Rozieres, Matthew Friedenberg, Anne C. Jager, Han-Yu Chuang. Analytical performance of TruSight® Tumor 170 in the detection of gene fusions and splice variants using RNA from formalin-fixed, paraffin-embedded (FFPE) solid tumor samples [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 565. doi:10.1158/1538-7445.AM2017-565
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. 3047-3047
    Abstract: 3047 Background: The National Cancer Institute Molecular Analysis for Therapy Choice (NCI-MATCH) multi-arm phase II clinical trial tested biopsies from patients with advanced refractory cancer to assign treatment based on tumor molecular profile. Enrollment criteria for treatment with the mTOR1/2 inhibitor sapanisertib in Arm M, or the Hedgehog pathway inhibitor vismodegib in Arm T, included detection of mutations in TSC1/TSC2 or PTCH1/SMO in tissue, respectively. For a subset of patients enrolled on these two arms, matched plasma was also collected at time of enrollment or on treatment to evaluate ctDNA. Methods: Screening for NCI-MATCH was based on tissue sequencing that was initially performed centrally using the 143-gene Oncomine Comprehensive Assay version 2 (OCAv2) and later referred to NCI-MATCH Designated Laboratory (DL) Network to perform sequencing for patient referral. Cell-free DNA (cfDNA) extracted from plasma collected in Streck tubes was input at 10 – 30 ng into a modified version of the Illumina TruSight Oncology 500 assay (TSO 500). Following error correction and read collapsing post-sequencing, variant calls were made and concordance between cfDNA libraries meeting prespecified QC criteria and tissue was evaluated. Results: Plasma, representing multiple collection timepoints, was available for 30/49 Arm M and 21/34 Arm T enrolled patients. A total of 95 cfDNA libraries, including replicates, were constructed and sequenced. Unique patient samples were tested across various timepoints (n = 35 pretreatment; n = 35 Cycle 2 Day 1; n = 8 at progression) plus replicates, with 90 (94.7%) libraries passing all prespecified QC criteria. Among Arm M patients there were 3 patients whose enrollment variant did not overlap with the TSO 500 gene panel. In total, 14 of 27 (51.8%) patients in Arm M and 18 of 21 (85.7%) patients in Arm T were concordant for enrollment mutation for at least one collection timepoint. In Arm T, 85.7% (n=38) of patient samples had at least one oncogenic or likely oncogenic (O/LO) mutation in the tumor sample at enrollment, with 73.6% (n = 28) of those also being detected by cfDNA. Interestingly, within Arm M and Arm T, a total of 7 patient samples were detected as MSI-H, with corresponding bTMB scores ranging from ~6 to 〉 100 Mut/Mb (median = 86.7). Conclusions: Detection of the enrollment mutations in treatment arms was observed in 51.8% - 85.7% of patients tested by ctDNA with overall variant concordance between tissue and ctDNA of at least 73.6%. These findings support the use of liquid biopsy as a tool for understanding genomic profiles of cancer patients both at diagnosis and progression, less invasively than standard tissue biopsies. In addition, 7 patients were identified as MSI-H, suggesting that blood-based testing could complement tissue testing for identifying patients who may benefit from targeted therapy.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Genome Biology, Springer Science and Business Media LLC, Vol. 23, No. 1 ( 2022-12)
    Abstract: Clinical laboratories routinely use formalin-fixed paraffin-embedded (FFPE) tissue or cell block cytology samples in oncology panel sequencing to identify mutations that can predict patient response to targeted therapy. To understand the technical error due to FFPE processing, a robustly characterized diploid cell line was used to create FFPE samples with four different pre-tissue processing formalin fixation times. A total of 96 FFPE sections were then distributed to different laboratories for targeted sequencing analysis by four oncopanels, and variants resulting from technical error were identified. Results Tissue sections that fail more frequently show low cellularity, lower than recommended library preparation DNA input, or target sequencing depth. Importantly, sections from block surfaces are more likely to show FFPE-specific errors, akin to “edge effects” seen in histology, while the inner samples display no quality degradation related to fixation time. Conclusions To assure reliable results, we recommend avoiding the block surface portion and restricting mutation detection to genomic regions of high confidence.
    Type of Medium: Online Resource
    ISSN: 1474-760X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2040529-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. 2560-2560
    Abstract: 2560 Background: NCI-MATCH was the largest precision medicine initiative conducted to date. Arm Z1D was a study of nivolumab in patients (pts) whose tumors were mismatch repair deficient (MMRd) by immunohistochemistry (IHC). Plasma was available for analysis of circulating tumor DNA (ctDNA), a component of cell-free DNA (cfDNA), in 39 of 47 pts enrolled to treatment. Large ctDNA gene panels that interrogate microsatellite instability (MSI), tumor mutational burden (TMB) and other complex biomarkers may provide an important complement to tumor-based assessments. Methods: 10 – 30 ng of cfDNA extracted from Streck tubes was tested with a modified version of the 523-gene TruSightä Oncology 500 ctDNA RUO assay (Illumina, San Diego, CA). Blood MSI (bMSI) status was calculated by interrogating ~2400 microsatellite sites and applying an experimentally validated cut-off. Blood TMB (bTMB) was calculated as total number of nonsynonymous and synonymous SNVs and Indels identified per Mb. Tumor sequencing was performed with the 143-gene Oncomine Comprehensive Assay v2. Overlap of tumor and plasma gene panels was evaluated for concordance. MMRd was evaluated by MLH1 and MSH2 IHC expression. MSI PCR testing was performed on 7 microsatellite sites. Clinical response was assessed by RECIST v1.1 criteria. Results: ORR was 31% (12/39) for 39 MMRd patients with plasma samples. Among these, 34 pts (87%) were classified as MSI-high (MSI-H) by ctDNA at baseline. The 5 MSS cases included 1 CR, 2 PRs and 2 unevaluable for response. The median bTMB was 56.4 mut/Mb (range: 1.65 – 435 mut/Mb). All pts had at least one oncogenic/likely oncogenic alteration identified in plasma. Of 136 of these mutations identified in tumor, 107 (78.6%) were detected in plasma. The most frequently mutated genes in plasma included TP53 (n = 26 pts), RNF43 (n = 16 pts, all G659Vfs*41) and PIK3CA (n = 16 pts). MLH1 (n = 5) and MSH2 (n = 5) mutations were also identified. RNF43 G659Vfs*41, which was not interrogated by the targeted tumor panel, is known to be strongly associated with MSI-H status. In this heavily pre-treated cohort (median prior therapies = 3), variants in genes commonly associated with clonal hematopoiesis of indeterminant potential were identified in all pts. Changes in cfDNA tumor fraction tracked with clinical response in 4 of 4 pts where on-treatment blood was collected for TSO 500 assessment. Conclusions: 78.6% of oncogenic/likely oncogenic alterations found in tumor where also identified in plasma. 87% of MMRd pts by tumor IHC were defined as MSI-H by baseline ctDNA, and median bTMB was 56.4 mut/Mb. These data suggest blood-based MSI and TMB should be further explored as biomarkers for the non-invasive identification of patients who may be candidates for immune checkpoint blockade, particularly when tissue is limiting.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 40, No. 16_suppl ( 2022-06-01), p. 3035-3035
    Abstract: 3035 Background: The National Cancer Institute Molecular Analysis for Therapy Choice (NCI-MATCH) multi-arm phase II clinical trial tested tumor tissue from 5,954 patients with advanced refractory cancer to assign treatment based on the molecular profile. Molecular profiling was successful for 93% of patients. For 267 of the patients who were not enrolled because molecular profiling was not successful, plasma cfDNA was evaluated to provide insight into the potential utility of blood-based testing in a broad spectrum of histologies when tissue is not evaluable. Methods: Cell-free DNA was extracted from plasma collected from Streck blood tubes and quantitated. Libraries were constructed using ³ 15 ng cfDNA into the Illumina TruSight Oncology 500 ctDNA RUO Assay, including unique molecular identifiers and duplex barcodes for error correction. Libraries were sequenced on the NovaSeq 6000 with S4 XP flow cells. Results: Of the 267 samples, 250 samples (94%) were evaluable, representing 72 histologies, including colorectal cancer (N = 36), lung adenocarcinoma (N = 15), pancreatic adenocarcinoma (N = 14), and invasive breast carcinoma (N = 12). Of these, 231 (92%) had ³ 1 OncoKB annotated mutation, with 208 patients (83%) having putative somatic mutations detected in genes not commonly associated with clonal hematopoiesis. The most common somatic mutations were in TP53, KRAS, APC, and PIK3CA, reported in 51%, 20%, 12%, and 12% of patients respectively. A total of 109 patients (44%) had ³ 1 actionable mutation of interest (aMOI) reported that could have been used for treatment assignment in the NCI-MATCH clinical trial. After applying histology and molecular exclusions, 75 patients (30%) had ³ 1 aMOI. The most common assignable treatment arms were Z1B/Z1BX1 (palbociclib with CCND1/2/3, N = 13), Z1F (copanlisib with PIK3CA Mutations, N = 13), S1/S1X1 (trametinib with NF1 mutation, N = 12), and Z1C/Z1CX1 (palbociclib with CDK4/CDK6 Amplification and Rb Expression by IHC, N = 10). Mutations in genes commonly associated with clonal hematopoiesis (CH) were prevalent in this population. Along with the expected high frequency of DNMT3A (21% of patients) and TET2 (11%) mutations, PPM1D mutations were the highest amongst CH genes, with 61 patients (24%) having ³ 1 PPM1D mutation, likely due to the heavily pre-treated nature of these patients. Conclusions: Variants observed in the blood are consistent with what is reported in the tissue. Using liquid biopsy when tissue is not evaluable can expand the ability of patients to obtain mutation information that can inform treatment compared to using tumor tissue only. Cell-free DNA provided valuable mutation information for these patients and could have resulted in up to an additional 75 patients being eligible for treatment selection based on their mutation profile. These results indicate that blood-based screening could be a tool for future NCI-sponsored clinical studies.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2022
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 3732-3732
    Abstract: Expanding the paradigm of solid tumor profiling from single-gene testing to comprehensive panels presents many challenges. One such challenges is the ability of these panels to detect genetic alterations from FFPE samples, where the DNA is of low abundance and often heavily compromised. Despite these challenges, next-generation sequencing (NGS) offers the ability to assess multiple variants simultaneously in an ever-expanding list of relevant tumor genes. To that end, Illumina developed a comprehensive, hybrid capture-based NGS assay targeting 170 key cancer genes that is FFPE optimized. The assay consists of a DNA workflow for the identification of single and multiple nucleotide variants (SNVs, MNVs), small insertions and deletions (indels), gene amplifications, as well as a RNA workflow for the identification of splice variants and gene fusions. Following sequencing on the NextSeq® or HiSeq® instruments, the analytical pipeline initiates variant calling. The DNA aligner and variant callers were first optimized against the simulated read data from & gt;40,000 COSMIC[1] mutations reported in the exons of the 170 genes. To reduce false positive variant calling due to systematic errors, each variant call was evaluated against its locus specific background error distribution. This distribution was compiled from a panel of FFPE normal samples and was also used to normalize against systematic bias in read coverage to increase the accuracy of amplification calling. Furthermore, gene amplification calling was improved by the addition of enhancer probes to the hybrid capture pool. The analytical sensitivity and specificity of TruSight® Tumor 170* was assessed on a large collection of FFPE samples and reference material. A panel of 72 cancer samples, including multiple tissue types, reference standards, and cell line and FFPE mixes were used to evaluate the limit of detection. The samples contained 533 SNVs, 80 indels including deletions up to 30 base pairs and insertions up to 31 base pairs, 4 MNVs, and 31 gene amplifications, characterized by orthogonal testing methods. Using 40 ng DNA input, detection sensitivity of the & gt;1000 variants (including replicates) tested at variant allele frequencies down to ~5% was at 99.6%, while detection sensitivity of gene amplifications as low as 1.45x to 2.2x was at 98%. For limit of blank samples, a panel of 24 normal samples was used. Again using 40 ng DNA input, we show & gt;99% specificity for small variant calling and & gt;95% specificity for gene amplification calling. These data demonstrates the TruSight® Tumor 170 is able to detect multiple variant types within a single sample at low nucleic acid input, while exhibiting high analytical sensitivity and specificity for low allele fraction detection. [1] Forbes, et al. (2015) *For Research Use Only. Not for use in diagnostic procedures. Citation Format: Danny Chou, Xiao Chen, Austin Purdy, Li Teng, Byron Luo, Chen Zhao, Laurel Ball, Allan Castaneda, Katie Clark, Brian Crain, Anthony Daulo, Manh Do, Tingting Du, Sarah Dumm, Yonmee Han, Michael Havern, Chia-Ling Hsieh, Tingting Jiang, Suzanne Johansen, Scott Lang, Rachel Liang, Jennifer S. LoCoco, Jaime McLean, Yousef Nassiri, Jason Rostron, Jennifer Silhavy, June Snedecor, Natasha Talago, Kevin Wu, Clare Zlatkov, Ali Kuraishy, Karen Gutekunst, Sohela De Rozieres, Matthew Friedenberg, Han-Yu Chuang, Anne C. Jager. Analytical performance of TruSight® Tumor 170 on small nucleotide variations and gene amplifications using DNA from formalin-fixed, paraffin-embedded (FFPE) solid tumor samples [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3732. doi:10.1158/1538-7445.AM2017-3732
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 3607-3607
    Abstract: Gene fusions have long been considered strong drivers of cellular transformation, making the accurate and precise assessment of these variants a necessity for any tumor profiling assay. Recent studies have indicated the utility of next-generation sequencing (NGS) for tumor profiling due to increasing data output and decreasing costs of the technology. Unfortunately, because a critical facet of NGS is the evaluation of short DNA fragments, sufficiently covering all possible breakpoint regions (many of which are intronic) has proven difficult and costly. Recent studies have indicated that NGS may prove better at detecting gene fusions using RNA instead of DNA, given the higher probability of breakpoint-spanning reads. This allows for de-novo discovery of fusion partners without knowing the precise breakpoint and guarantees expression of the fusion transcript. To that end, Illumina is developing a novel method for simultaneous library preparation from low input amounts of degraded DNA and RNA from a single FFPE tumor sample. With a turnaround time from nucleic acid to data of less than 4 days, this enrichment-based assay surveys 170 genes for single nucleotide variants and small indels, 57 genes for gene amplifications, 55 genes for fusions and four genes for splice variants. To determine the limit of detection for gene fusions, a panel of different synthetic RNA transcripts were prepared in vitro, pooled at equal molar amounts, and spiked into 20ng of cell line RNA (MCF-7). Fusions were detected over several orders of magnitude down to 1×10-8 picomoles, equivalent to 3 to 15 fusion transcripts per cell. In addition, a similar range of fusion detection was observed when RNA from two different cell lines were mixed, as when RNA from a cell line with high expression of an FGFR2-COL14A1 fusion was mixed in proportional amounts with RNA from a different cell line where FGFR2 is minimally expressed. Importantly, our method allowed for fusion detection from as little as 100 picograms of cell line RNA. We then tested our new method on previously characterized FFPE solid tumor samples harboring known gene rearrangements identified by FISH and other methods. Not only was the NGS method able to detect the majority of previously characterized variants, including EML4-ALK and SDC4-ROS1, it also identified the gene fusions and their uncharacterized fusions partners by combining the non-targeted sequence information gained from using an enrichment-based assay with novel fusion calling algorithms. From this information, we were able to glean new insights into the structure of the rearrangements and how the gene fusions may be involved in tumorigenesis. These results indicate that NGS can identify fusions from the low amounts of degraded RNA from solid tumor samples, identify fusion partners not uncovered by current technologies, and further emphasizes the advantage of NGS in solid tumor profiling. Citation Format: Julianna Tdr Parks, Luo Byron, Brian Crain, Snedecor June, Zhao Chen, Tingting Du, Gabriel L. Sica, Taofee K. Owonikoko, Stewart G. Neill, Scott Newman, Debra F. Saxe, Jennifer S. LoCoco, Han-Yu Chuang, Charles Lin, Kathryn M. Stephens, Michael R. Rossi, Matthew C. Friedenberg. An evaluation of NGS to identify gene fusions using RNA from FFPE solid tumor samples. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3607.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: SSRN Electronic Journal, Elsevier BV
    Type of Medium: Online Resource
    ISSN: 1556-5068
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages