Show simple item record

dc.contributor.authorGarofalo, Andreaen_US
dc.contributor.authorSholl, Lynetteen_US
dc.contributor.authorReardon, Brendanen_US
dc.contributor.authorTaylor-Weiner, Amaroen_US
dc.contributor.authorAmin-Mansour, Alien_US
dc.contributor.authorMiao, Dianaen_US
dc.contributor.authorLiu, Daviden_US
dc.contributor.authorOliver, Nellyen_US
dc.contributor.authorMacConaill, Lauraen_US
dc.contributor.authorDucar, Matthewen_US
dc.contributor.authorRojas-Rudilla, Vanesaen_US
dc.contributor.authorGiannakis, Mariosen_US
dc.contributor.authorGhazani, Arezouen_US
dc.contributor.authorGray, Stacyen_US
dc.contributor.authorJanne, Pasien_US
dc.contributor.authorGarber, Judyen_US
dc.contributor.authorJoffe, Steveen_US
dc.contributor.authorLindeman, Nealen_US
dc.contributor.authorWagle, Nikhilen_US
dc.contributor.authorGarraway, Levi A.en_US
dc.contributor.authorVan Allen, Eliezer M.en_US
dc.date.accessioned2016-08-09T14:52:08Z
dc.date.issued2016en_US
dc.identifier.citationGarofalo, A., L. Sholl, B. Reardon, A. Taylor-Weiner, A. Amin-Mansour, D. Miao, D. Liu, et al. 2016. “The impact of tumor profiling approaches and genomic data strategies for cancer precision medicine.” Genome Medicine 8 (1): 79. doi:10.1186/s13073-016-0333-9. http://dx.doi.org/10.1186/s13073-016-0333-9.en
dc.identifier.issn1756-994Xen
dc.identifier.urihttp://nrs.harvard.edu/urn-3:HUL.InstRepos:27822081
dc.description.abstractBackground: The diversity of clinical tumor profiling approaches (small panels to whole exomes with matched or unmatched germline analysis) may engender uncertainty about their benefits and liabilities, particularly in light of reported germline false positives in tumor-only profiling and use of global mutational and/or neoantigen data. The goal of this study was to determine the impact of genomic analysis strategies on error rates and data interpretation across contexts and ancestries. Methods: We modeled common tumor profiling modalities—large (n = 300 genes), medium (n = 48 genes), and small (n = 15 genes) panels—using clinical whole exomes (WES) from 157 patients with lung or colon adenocarcinoma. We created a tumor-only analysis algorithm to assess germline false positive rates, the impact of patient ancestry on tumor-only results, and neoantigen detection. Results: After optimizing a germline filtering strategy, the germline false positive rate with tumor-only large panel sequencing was 14 % (144/1012 variants). For patients whose tumor-only results underwent molecular pathologist review (n = 91), 50/54 (93 %) false positives were correctly interpreted as uncertain variants. Increased germline false positives were observed in tumor-only sequencing of non-European compared with European ancestry patients (p < 0.001; Fisher’s exact) when basic germline filtering approaches were used; however, the ExAC database (60,706 germline exomes) mitigated this disparity (p = 0.53). Matched and unmatched large panel mutational load correlated with WES mutational load (r2 = 0.99 and 0.93, respectively; p < 0.001). Neoantigen load also correlated (r2 = 0.80; p < 0.001), though WES identified a broader spectrum of neoantigens. Small panels did not predict mutational or neoantigen load. Conclusions: Large tumor-only targeted panels are sufficient for most somatic variant identification and mutational load prediction if paired with expanded germline analysis strategies and molecular pathologist review. Paired germline sequencing reduced overall false positive mutation calls and WES provided the most neoantigens. Without patient-matched germline data, large germline databases are needed to minimize false positive mutation calling and mitigate ethnic disparities. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0333-9) contains supplementary material, which is available to authorized users.en
dc.language.isoen_USen
dc.publisherBioMed Centralen
dc.relation.isversionofdoi:10.1186/s13073-016-0333-9en
dc.relation.hasversionhttp://www.ncbi.nlm.nih.gov/pmc/articles/PMC4962446/pdf/en
dash.licenseLAAen_US
dc.subjectGenomicsen
dc.subjectPrecision medicineen
dc.subjectDisparitiesen
dc.subjectImmuno-oncologyen
dc.subjectNeoantigensen
dc.subjectPanel testingen
dc.titleThe impact of tumor profiling approaches and genomic data strategies for cancer precision medicineen
dc.typeJournal Articleen_US
dc.description.versionVersion of Recorden
dc.relation.journalGenome Medicineen
dash.depositing.authorSholl, Lynetteen_US
dc.date.available2016-08-09T14:52:08Z
dc.identifier.doi10.1186/s13073-016-0333-9*
dash.authorsorderedfalse
dash.contributor.affiliatedGiannakis, Marios
dash.contributor.affiliatedGarraway, Levi
dash.contributor.affiliatedGarber, Judy
dash.contributor.affiliatedGhazani, Arezou
dash.contributor.affiliatedMiao, Diana
dash.contributor.affiliatedGray, Stacy
dash.contributor.affiliatedLindeman, Neal
dash.contributor.affiliatedJanne, Pasi
dash.contributor.affiliatedMacConaill, Laura
dash.contributor.affiliatedSholl, Lynette
dash.contributor.affiliatedVan Allen, Eliezer
dash.contributor.affiliatedWagle, Nikhil
dash.contributor.affiliatedLiu, David


Files in this item

Thumbnail

This item appears in the following Collection(s)

Show simple item record