Clinical validation of the tempus xT next-generation targeted oncology sequencing assay

Nike Beaubier, Robert Tell, Denise Lau, Jerod R Parsons, Stephen Bush, Jason Perera, Shelly Sorrells, Timothy Baker, Alan Chang, Jackson Michuda, Catherine Iguartua, Shelley MacNeil, Kaanan Shah, Philip Ellis, Kimberly Yeatts, Brett Mahon, Timothy Taxter, Martin Bontrager, Aly Khan, Robert Huether, Eric Lefkofsky, Kevin P White, Nike Beaubier, Robert Tell, Denise Lau, Jerod R Parsons, Stephen Bush, Jason Perera, Shelly Sorrells, Timothy Baker, Alan Chang, Jackson Michuda, Catherine Iguartua, Shelley MacNeil, Kaanan Shah, Philip Ellis, Kimberly Yeatts, Brett Mahon, Timothy Taxter, Martin Bontrager, Aly Khan, Robert Huether, Eric Lefkofsky, Kevin P White

Abstract

We developed and clinically validated a hybrid capture next generation sequencing assay to detect somatic alterations and microsatellite instability in solid tumors and hematologic malignancies. This targeted oncology assay utilizes tumor-normal matched samples for highly accurate somatic alteration calling and whole transcriptome RNA sequencing for unbiased identification of gene fusion events. The assay was validated with a combination of clinical specimens and cell lines, and recorded a sensitivity of 99.1% for single nucleotide variants, 98.1% for indels, 99.9% for gene rearrangements, 98.4% for copy number variations, and 99.9% for microsatellite instability detection. This assay presents a wide array of data for clinical management and clinical trial enrollment while conserving limited tissue.

Keywords: next-generation sequencing assay validation; tumor profiling.

Conflict of interest statement

CONFLICTS OF INTEREST All authors have a financial relationship as employees of Tempus Labs, Inc.

Figures

Figure 1. Performance of SNV and Indel…
Figure 1. Performance of SNV and Indel Detection by the Tempus xT assay
(A) Precision of indel detection by VAF. Precision was calculated for each bin of variants with allele fractions rounded to the nearest 5 percent. The vertical black line corresponds to the LOD. (B) Precision of SNV detection by VAF, as in A. (C) Correlation of xT assay indel VAFs to xO assay indel VAFs. (D) Correlation of xT assay SNV VAFs to xO assay SNV VAFs. (E) VAFs of indels and SNVs detected on chromosome 17 in four samples with serial 1:1 dilutions of the xT assay. Dark blue lines indicate best fit of a linear model. (F) Positive control detection. Boxplots of the VAF of three SNVs in a positive control sample run on every xT assay over a period of 5 months. The single point marked in black is an AKT1 p.E17K variant which failed filtering criteria.
Figure 2. Analysis of rearrangement detection performance…
Figure 2. Analysis of rearrangement detection performance by the Tempus pipeline and retrospective analysis of recurrent fusions
(A) Fusions detected by cancer type. (B) Positive control fusion samples (ROS1-SLC34A2) processed by the xT assay over the course of 4 months. The fusion was expected at 5% VAF in the control and was consistently detected across flow cells and instruments. (C) Limit of detection analysis for two serially diluted fusions (ALK-EML4 and RET-ANKRD26). Both fusions were detected down to 3–5% simulated VAF by the Tempus xT Assay. (D) Functional characterization of fusions detected by the xT assay. Domains, regions, and sites are highlighted for orientation along the amino acid sequence for each protein involved in the rearrangement event. (E) Analysis of the recurrent TMPRSS2-ERG fusion found in 13 prostate cancers detected by the xT assay. The xT assay consistently localized breakpoints to the expected functional domains resulting in the TMPRSS2 promoter and replacing the first several exons of ERG by a chromosomal deletion. This results in the functional domains of ERG being largely intact, but under the control of the TMPRSS2 promoter.
Figure 3. Analysis of performance of the…
Figure 3. Analysis of performance of the xT assay in the detection of copy number variations
(A) Detection comparison of Tempus copy number calls against validated ERBB2 control ratios. (B) Plot of reported CNVs by cancer type for the xT patient cohort sequenced at Tempus. (C) A representative 1p-19q co-deletion detected commonly in oligodendrogliomas. The blue dots represent genomic regions showing one copy loss while the grey points represent neutral segments.
Figure 4. Survey of immunotherapy markers across…
Figure 4. Survey of immunotherapy markers across diverse cancer types
(A) The distribution of TMB for each cancer type plotted on a log2 scale and ordered by the median TMB. Outliers (data points beyond 1.5x interquartile range) are shown as individual points. (B) Analysis of samples in the top 10th percentile of TMB. The inset shows the distribution of TMB across all samples included in the study, with the vertical bar marking the 90th percentile of TMB. The bar chart shows the proportion of predicted antigenic mutations of the non-synonymous mutations detected. MSI-H samples are highlighted in red. The color-coded matrices show the MMR gene mutations detected by mutation type (top), predicted MLH1 methylation status (middle), and DNA repair gene mutations detected (bottom).
Figure 5. Mutational landscape across all cancer…
Figure 5. Mutational landscape across all cancer types
(A) Plot of a subset of patients within the Tempus xT cohort containing at least one biologically relevant alteration, stratified by alteration prevalence. Patients were clustered by the mutational profile similarity of genes (y-axis) with at least 5 actionable alterations detected within the cohort. Patient cancer type is displayed by the colored bar below the matrix. (B) Lollipop plot of TP53, KRAS, and PIK3CA alterations detected by the xT assay across all cancer types.

References

    1. Kumar-Sinha C, Chinnaiyan AM. Precision oncology in the age of integrative genomics. Nat Biotechnol. 2018;36:46–60.
    1. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–2465.
    1. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–723.
    1. Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, Powles T, Donskov F, Neiman V, et al. CheckMate 214 Investigators Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N Engl J Med. 2018;378:1277–1290.
    1. Tran E, Robbins PF, Lu YC, Prickett TD, Gartner JJ, Jia L, Pasetto A, Zheng Z, Ray S, Groh EM, Kriley IR, Rosenberg SA. T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. N Engl J Med. 2016;375:2255–2262.
    1. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, Qayed M, De Moerloose B, Hiramatsu H, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med. 2018;378:439–448.
    1. Bommareddy PK, Patel A, Hossain S, Kaufman HL. Talimogene Laherparepvec (T-VEC) and Other Oncolytic Viruses for the Treatment of Melanoma. AM J Clin Dermatol. 2017;18:1–15.
    1. Beaubier N, Tell R, Huether R, Bontrager M, Bush S, Parsons J, Shah K, Baker T, Selkov G, Taxter T, Thomas A, Bettis S, Khan A, et al. Clinical validation of the Tempus xO assay. Oncotarget. 2018;9:25826–32. doi: 10.18632/oncotarget.25381.
    1. Olshen A, Venkatraman ES, Lucito R, Wigler M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics. 2004;5:557–572.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–60.
    1. Layer RM, Chiang C, Quinlan AR, Hall IM. LUMPY: a probabilistic framework for structural variant discovery. Genome Biol. 2014;15:R84.
    1. Chiang C, Layer RM, Faust GG, Lindberg MR, Rose DB, Garrison EP, Marth GT, Quinlan AR, Hall IM. SpeedSeq: ultra-fast personal genome analysis and interpretation. Nat Methods. 2015;12:966–968.
    1. Buell CR, Yuan Q, Ouyang S, Liu J, Zhu W, Wang A, Maiti R, Haas B, Wortman J, Pertea M, Jones KM, Kim M, Overton L, et al. Sequence, annotation, and analysis of synteny between rice chromosome 3 and diverged grass species. Genome Res. 2005;15:1284–1291.
    1. Dienstmann R, Dong F, Borger D, Dias-Santagata D, Ellisen LW, Le LP, Iafrate AJ. Standardized decision support in next generation sequencing reports of somatic cancer variants. Mol Oncol. 2014;8:859–873.
    1. Ritter DI, Roychowdhury S, Roy A, Rao S, Landrum MJ, Sonin D, Shekar M, Davis CF, Hart RK, Micheel C, Weaver M, Van Allen EM, Parsons DW, et al. Somatic cancer variant curation and harmonization through consensus minimum variant level data. Genome Med. 2016;8:117.
    1. Li MM, Datto M, Duncavage EJ, Kulkkarni S, Lindeman NI, Roy S, Tsimberidou AM, Vnecak-Jones CL, Wolff DJ, Younes A, Nikiforova MN. Standards and guidelines for the interpretation and reporting of sequence variants in cancer: a joint consensus recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists. J Mol Diagn. 2017;19:4–23.
    1. Kalia SS, Adelman K, Bale SJ, Chung WK, Eng C, Evans JP, Herman GE, Hufnagel SB, Klein TE, Korf BR, McKevley KD, Ormond KE, Richards CS, et al. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics. Genet Med. 2017;19:249–255.
    1. Szolek A, Schubert B, Mohr C, Sturm M, Feldhahn M, Kohlbacher O. OptiType: precision HLA typing from next-generation sequencing data. Bioinformatics. 2014;30:3310–3316.
    1. Rubinsteyn A, O'Donnell T, Damaraju N, Hammerbacher J. Predicting peptide-MHC binding affinities with imputed training data. bioRxiv. 2016
    1. Chiang DY, Getz G, Jaffe DB, O'Kelly MJ, Zhao X, Carter SL, Russ C, Nusbaum C, Meyerson M, Lander ES. High-resolution mapping of copy-number alterations with massively parallel sequencing. Nat Methods. 2009;6:99–103.
    1. Zhao M, Wang Q, Wang Q, Jia P, Zhao Z. Computational tools for copy number variation (CNV) detection using next-generation sequencing data: features and perspectives. BMC Bioinformatics. 2013;14:S1.
    1. Wang K, Li M, Hadley D, Liu R, Glessner J, Grant SF, Hakonarson H, Bucan M. PennCNV: An integrated hidden Markov model designed for high-resolution copy number variation detection in whole genome SNP genotyping data. Genome Res. 2007;17:1665–1674.
    1. Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C, Stuart JM, Cancer Genome Atlas Research Network The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet. 2013;45:1113–1120.
    1. Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, Srinivasan P, Gao J, Chakravarty D, Devlin SM, Hellmann MD, Barron DA, Schram AM, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23:703–713.
    1. Assarzadegan N, Montgomery E, Anders RA. Immune checkpoint inhibitor colitis: the flip side of the wonder drugs. Virchows Arch. 2018;472:125–133.
    1. Scott ES, Long GV, Guminski A, Clifton-Bligh RJ, Menzies AM, Tsang VH. The spectrum, incidence, kinetics and management of endocrinopathies with immune checkpoint inhibitors for metastatic melanoma. Eur J Endocrinol. 2018;178:173–80.
    1. Wei W, Luo Z. Risk of gastrointestinal toxicities with PD-1 inhibitors in cancer patients: A meta-analysis of randomized clinical trials. Medicine. 2017;96:e8931.
    1. Makarious D, Horwood K, Coward JIG. Myasthenia gravis: An emerging toxicity of immune checkpoint inhibitors. Eur J Cancer. 2017;82:128–136.
    1. Goodman A, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, Stephens PJ, Daniels GA, Kurzrock R. Tumor Mutational Burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16:2598–2608.
    1. Schwaederle M, Zhao M, Lee JJ, Lazar V, Leyland-Jones B, Schilsky RL, Mendelsohn J, Kurzrock R. Association of Biomarker-Based Treatment Strategies with Response Rates and Progression-Free Survival in Refractory Malignant Neoplasms: A Meta-analysis. JAMA Oncol. 2016;2:1452–1459.
    1. Schwaederle M, Zhao M, Lee JJ, Eggermont AM, Schilsky RL, Mendelsohn J, Lazar V, Kurzrock R. Impact of Precision Medicine in Diverse Cancers: A Meta-Analysis of Phase II Clinical Trials. J Clin Oncol. 2015;33:3817–25.
    1. Wheler JJ, Janku F, Naing A, Li Y, Stephen B, Zinner R, Subbiah V, Fu S, Karp D, Falchook GS, Tsimeridou AM, Piha-Paul S, Anderson R, et al. Cancer Therapy Directed by Comprehensive Genomic Profiling: A Single Center Study. Cancer Res. 2016;76:3690–3701.
    1. Coyne GO, Takebe N, Chen AP. Defining precision: The precision medicine initiative trials NCI-MPACT and NCI-MATCH. Curr Probl Cancer. 2017;41:182–193.
    1. Markman M. Maurie Markman on the Groundbreaking TAPUR Trial. Oncology (Williston Park) 2017;31:158–68. 168.
    1. Hirshfield KM, Tolkkunov D, Zhong H, Ali SM, Stein MN, Murphy S, Vig H, Vazquez A, Glod J, Moss RA, Belyi V, Chan CS, Chen S, et al. Clinical Actionability of Comprehensive Genomic Profiling for Management of Rare or Refractory Cancers. Oncologist. 2016;21:1315–1325.
    1. Groisberg R, Hong DS, Holla V, Janku F, Piha-Paul S, Ravi V, Benjamin R, Kumar Patel S, Somaiah N, Conley A, Ali SM, Schrock AB, Ross JS, et al. Clinical genomic profiling to identify actionable alterations for investigational therapies in patients with diverse sarcomas. Oncotarget. 2017;8:39254–67. doi: 10.18632/oncotarget.16845.
    1. Ross JS, Wang K, Gay L, Otto GA, White E, Iwanik K, Palmer G, Yelensky R, Lipson DM, Chmielecki J, Erlich RL, Rankin AN, Ali SM, et al. Comprehensive Genomic Profiling of Carcinoma of Unknown Primary Site: New Routes to Targeted Therapies. JAMA Oncol. 2015;1:40–49.
    1. Ross JS, Badve S, Wang K, Sheehan CE, Boguniewicz AB, Otto GA, Yelensky R, Lipson D, Ali S, Morosini D, Chliemlecki J, Elvin JA, Miller VA, Stephens PJ. Genomic profiling of advanced-stage, metaplastic breast carcinoma by next-generation sequencing reveals frequent, targetable genomic abnormalities and potential new treatment options. Arch Pathol Lab Med. 2015;139:642–649.
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.
    1. Faust GG, Hall IM. SAMBLASTER: fast duplicate marking and structural variant read extraction. Bioinformatics. 2014;30:2503–2505.
    1. Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia S, Chakravarty D, Daian F, Gao Q, et al. Cancer Genome Atlas Research Network Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell. 2018;173:321–337.e10.

Source: PubMed

3
Se inscrever