Modeling clonal structure over narrow time frames via circulating tumor DNA in metastatic breast cancer

Zachary T Weber, Katharine A Collier, David Tallman, Juliet Forman, Sachet Shukla, Sarah Asad, Justin Rhoades, Samuel Freeman, Heather A Parsons, Nicole O Williams, Romualdo Barroso-Sousa, Elizabeth H Stover, Haider Mahdi, Carrie Cibulskis, Niall J Lennon, Gavin Ha, Viktor A Adalsteinsson, Sara M Tolaney, Daniel G Stover, Zachary T Weber, Katharine A Collier, David Tallman, Juliet Forman, Sachet Shukla, Sarah Asad, Justin Rhoades, Samuel Freeman, Heather A Parsons, Nicole O Williams, Romualdo Barroso-Sousa, Elizabeth H Stover, Haider Mahdi, Carrie Cibulskis, Niall J Lennon, Gavin Ha, Viktor A Adalsteinsson, Sara M Tolaney, Daniel G Stover

Abstract

Background: Circulating tumor DNA (ctDNA) offers minimally invasive means to repeatedly interrogate tumor genomes, providing opportunities to monitor clonal dynamics induced by metastasis and therapeutic selective pressures. In metastatic cancers, ctDNA profiling allows for simultaneous analysis of both local and distant sites of recurrence. Despite the promise of ctDNA sampling, its utility in real-time genetic monitoring remains largely unexplored.

Methods: In this exploratory analysis, we characterize high-frequency ctDNA sample series collected over narrow time frames from seven patients with metastatic triple-negative breast cancer, each undergoing treatment with Cabozantinib, a multi-tyrosine kinase inhibitor (NCT01738438, https://ichgcp.net/clinical-trials-registry/NCT01738438 ). Applying orthogonal whole exome sequencing, ultra-low pass whole genome sequencing, and 396-gene targeted panel sequencing, we analyzed 42 plasma-derived ctDNA libraries, representing 4-8 samples per patient with 6-42 days between samples. Integrating tumor fraction, copy number, and somatic variant information, we model tumor clonal dynamics, predict neoantigens, and evaluate consistency of genomic information from orthogonal assays.

Results: We measured considerable variation in ctDNA tumor faction in each patient, often conflicting with RECIST imaging response metrics. In orthogonal sequencing, we found high concordance between targeted panel and whole exome sequencing in both variant detection and variant allele frequency estimation (specificity = 95.5%, VAF correlation, r = 0.949), Copy number remained generally stable, despite resolution limitations posed by low tumor fraction. Through modeling, we inferred and tracked distinct clonal populations specific to each patient and built phylogenetic trees revealing alterations in hallmark breast cancer drivers, including TP53, PIK3CA, CDK4, and PTEN. Our modeling revealed varied responses to therapy, with some individuals displaying stable clonal profiles, while others showed signs of substantial expansion or reduction in prevalence, with characteristic alterations of varied literature annotation in relation to the study drug. Finally, we predicted and tracked neoantigen-producing alterations across time, exposing translationally relevant detection patterns.

Conclusions: Despite technical challenges arising from low tumor content, metastatic ctDNA monitoring can aid our understanding of response and progression, while minimizing patient risk and discomfort. In this study, we demonstrate the potential for high-frequency monitoring of evolving genomic features, providing an important step toward scalable, translational genomics for clinical decision making.

Keywords: Circulating tumor DNA; Liquid biopsy; Neoantigens; Serial sequencing; Targeted panel sequencing; Tumor evolution; Ultra-low pass whole genome sequencing; ctDNA.

Conflict of interest statement

S.A.S. reported nonfinancial support from Bristol-Myers Squibb outside the submitted work. S.A.S. previously advised and has received consulting fees from Neon Therapeutics. S.A.S. reported nonfinancial support from Bristol-Myers Squibb, and equity in Agenus Inc., Agios Pharmaceuticals, Breakbio Corp., Bristol-Myers Squibb, Indiscine and Lumos Pharma, outside the submitted work. R.B.S. reported consulting Fees (e.g., advisory boards); Author; Roche, Merck, Eli Lilly. Fees for Non-CME Services Received Directly from Commercial Interest or their Agents (e.g., speakers’ bureaus); Author; Eli Lilly, Libbs, Novartis, Pfizer, ROCHE, Bristol-Myers Squibb. G. Ha: Receipt of Intellectual Property Rights/Patent Holder; Broad Institute. VAA reported advisory boards for AGCT GmbH and BerMs Inc. S.M.T. reported consulting fees (e.g., advisory boards); AstraZeneca, Lilly, Merck, Nektar, Novartis, Pfizer, Genentech/Roche, Immunomedics, Bristol-Myers Squibb, Eisai, Nanostring, Puma, Sanofi, Celldex, Odonate, Seattle Genetics, Daiichi Sankyo, Silverback Therapeutics, Abbvie, Athenex, OncoPep, Kyowa Kirin Pharmaceuticals, Samsung BioepsiVs. The remaining authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Study design and sampling dynamics. a Schematic diagram of the analysis workflow from patient selection, sample capture, and sequencing to downstream analyses. We leveraged the Terra Genomics/FireCloud platform for data storage and high-performance computing tasks. b Schematic representation of sampling density for each of the seven cohort members on study, also specifying whether whole exome sequencing and/or targeted panel sequencing was performed on that sample. All samples received ultra-low-pass whole genome sequencing. c Tumor fraction dynamics colored by individual. Tumor fraction was measured on study using ultra-low-pass whole genome sequencing and the ichorCNA algorithm. d Tumor fraction dynamics recolored by RECIST v1.1 response by imaging categories. RECIST v1.1 bucket response type into several categories: complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD)
Fig. 2
Fig. 2
Orthogonal ctDNA sequencing approaches are highly concordant. Somatic SNV and INDEL calling of whole exome sequencing (WES; average depth 150X) and targeted panel sequencing (TPS; nominal sequencing depth 10,000X) were completed on the Terra/Firecloud platform using gatk-Mutect2 pipelines (McKenna et al., 2010). a Variant recall assessment of TPS on somatic variants discovered in one or more WES assays. Only variants intersecting theoretical capture regions of TPS were considered. Variants used in assessment were those called in WES at any point, which also overlapped in genomic position with target or bait regions included in the TPS. X’s indicate a lack of adequate sequencing depth in the TPS. Center and right panels compare variant allele frequency (VAF) data from each assay. b Scatter plot comparing estimated VAF in TPS and WES sequencing across all individuals and time points. 1:1 line drawn for reference. c WES and ULP-WGS based algorithmic estimates of sample purity (a.k.a. tumor fraction) across samples and time points with high tumor fraction (TFx > 10%). d Algorithm estimation of ploidy (averaged copy number state across genome) across WES and ULP-WGS-based methods at time points with high tumor fraction. ABSOLUTE Soln.1 and Soln.2 represent the top two proposed solutions by model likelihood (Included here, as ABSOLUTE often suggests manual curation and/or override of the top solution)
Fig. 3.
Fig. 3.
Copy number profiles are stable. Ultra-low pass whole genome sequencing (ULP-WGS) was performed on all 42 ctDNA samples and tumor fraction and copy number data derived using ichorCNA. a Genome-wide copy profile of patient RP-466, derived from ULP-WGS on liquid biopsy ctDNA, showing changes in focal event resolution resulting from shifts in tumor fraction. Dark green segments represent a copy number of 1; blue represent neutral or 2 copies, brown and red represent 3 and 4+, respectively. b Scatter plot of computed log-ratios in ULP-WGS, compared to those derived from WES or TPS data using binned read-count of on and off target bins. c Discrete copy number confusion matrix for ULP-WGS based calls at first and last time points. All samples had tumor fraction ≥10%. Genomic positions assayed between first and last time points were uniformly and randomly sampled, and discrete copy number states were capped between one and seven during initial ichorCNA analyses
Fig. 4
Fig. 4
Tumor subclonal dynamics vary across patients. Models of clonal and subclonal populations which make up the cancers of metastatic patients, derived using PyClone [34]. Variant inputs include union of filter-passing alterations from each sampled time point delivered by the commercially available liquid-biopsy targeted panel-sequencing pipeline at the Broad Institute. Copy number information and purity were derived from ichorCNA. a, b Clonal prevalence dynamics, clustering, and inferred phylogenetic tree structure for patient RP-466, revealing generally unchanging populations in the tumor, with important drivers occupying early positions in cell lineages. c, d RP-527 clonal dynamics profile and inferred tree structure showing statistically significant clonal expansion of cell lineage marked by non-synonymous DDR2 and RNF43 variants. e, f RP-557 profile and tree showing the opposite trend as RP-527, with a decreasing cell population marked by RB1 mutation
Fig. 5
Fig. 5
Whole exome sequencing uncovers driver mutations and allows neoantigen prediction. Whole exome sequencing results from 31 total samples with tumor fraction ≥10% using short variant and INDEL calling tools from gatk-Mutect2 pipelines (McKenna et al., 2010), with subsequent neoantigen binding predictions for known MHC molecules from NetMHCpan 4.0 (Reynisson et al., 2020). a Driver mutations found via whole exome sequencing across time points. Variant data visualized are those whose genes have been previously annotated in literature as breast cancer drivers or pan cancer drivers. b Trends in predicted neoantigens among cohort members. Strong binders are denoted as those peptide sequences with NetMHCpan ranks <0.5%, and weak binders are those with ranks <2%. Neoantigen Generating sSNV are alterations whose changes to peptide structure are predicted to produce neoantigens capable of strong or weak binding to known MHC molecules. c, d Neoantigen dynamics from patient RP-527 and RP-535, showing proportions of detected neoantigens and dropout over time. Strong, weak, and ND labels correspond to binding affinity of predicted neoantigens, as well as a non-detected category to capture dropout. Threads are colored by their state at the final sequencing time point

References

    1. Stroun M, Anker P, Lyautey J, Lederrey C, Maurice PA. Isolation and characterization of DNA from the plasma of cancer patients. Eur J Cancer Clin Oncol. 1987;23(6):707–712. doi: 10.1016/0277-5379(87)90266-5.
    1. Stroun M, Anker P, Maurice P, Lyautey J, Lederrey C, Beljanski M. Neoplastic characteristics of the DNA found in the plasma of cancer patients. Oncology. 1989;46(5):318–322. doi: 10.1159/000226740.
    1. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61(4):1659–1665.
    1. Adalsteinsson VA, Ha G, Freeman SS, Choudhury AD, Stover DG, Parsons HA, Gydush G, Reed SC, Rotem D, Rhoades J, Loginov D, Livitz D, Rosebrock D, Leshchiner I, Kim J, Stewart C, Rosenberg M, Francis JM, Zhang CZ, Cohen O, Oh C, Ding H, Polak P, Lloyd M, Mahmud S, Helvie K, Merrill MS, Santiago RA, O’Connor EP, Jeong SH, Leeson R, Barry RM, Kramkowski JF, Zhang Z, Polacek L, Lohr JG, Schleicher M, Lipscomb E, Saltzman A, Oliver NM, Marini L, Waks AG, Harshman LC, Tolaney SM, van Allen EM, Winer EP, Lin NU, Nakabayashi M, Taplin ME, Johannessen CM, Garraway LA, Golub TR, Boehm JS, Wagle N, Getz G, Love JC, Meyerson M. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun. 2017;8(1):1324. doi: 10.1038/s41467-017-00965-y.
    1. Dawson SJ, Tsui DW, Murtaza M, Biggs H, Rueda OM, Chin SF, Dunning MJ, Gale D, Forshew T, Mahler-Araujo B, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368(13):1199–1209. doi: 10.1056/NEJMoa1213261.
    1. Schiavon G, Hrebien S, Garcia-Murillas I, Cutts RJ, Pearson A, Tarazona N, Fenwick K, Kozarewa I, Lopez-Knowles E, Ribas R, et al. Analysis of <em>ESR1</em> mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Sci Transl Med. 2015;7(313):313ra182. doi: 10.1126/scitranslmed.aac7551.
    1. Lohr JG, Adalsteinsson VA, Cibulskis K, Choudhury AD, Rosenberg M, Cruz-Gordillo P, Francis JM, Zhang CZ, Shalek AK, Satija R, Trombetta JJ, Lu D, Tallapragada N, Tahirova N, Kim S, Blumenstiel B, Sougnez C, Lowe A, Wong B, Auclair D, van Allen EM, Nakabayashi M, Lis RT, Lee GSM, Li T, Chabot MS, Ly A, Taplin ME, Clancy TE, Loda M, Regev A, Meyerson M, Hahn WC, Kantoff PW, Golub TR, Getz G, Boehm JS, Love JC. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat Biotechnol. 2014;32(5):479–484. doi: 10.1038/nbt.2892.
    1. Wyatt AW, Annala M, Aggarwal R, Beja K, Feng F, Youngren J, Foye A, Lloyd P, Nykter M, Beer TM, et al. Concordance of circulating tumor dna and matched metastatic tissue biopsy in prostate cancer. J Natl Cancer Inst. 2017;109(12):djx118. doi: 10.1093/jnci/djx118.
    1. Manier S, Park J, Capelletti M, Bustoros M, Freeman SS, Ha G, Rhoades J, Liu CJ, Huynh D, Reed SC, Gydush G, Salem KZ, Rotem D, Freymond C, Yosef A, Perilla-Glen A, Garderet L, van Allen EM, Kumar S, Love JC, Getz G, Adalsteinsson VA, Ghobrial IM. Whole-exome sequencing of cell-free DNA and circulating tumor cells in multiple myeloma. Nat Commun. 2018;9(1):1691. doi: 10.1038/s41467-018-04001-5.
    1. Manier S, Park J, Freeman S, Ha G, Capelletti M, Reed S, Gydush G, Rotem D, Rhoades J, Salem K, Freymond C, Huynh D, Sacco A, Leblebjian H, Perilla Glen A, Yosef A, Palumbo A, Garderet L, Kumar S, Roccaro AM, Facon T, van Allen E, Love JC, Getz G, Adalsteinsson V, Ghobrial IM. Whole-exome sequencing and targeted deep sequencing of cfDNA enables a comprehensive mutational profiling of multiple myeloma. Blood. 2016;128(22):197. doi: 10.1182/blood.V128.22.197.197.
    1. Stover DG, Parsons HA, Ha G, Freeman SS, Barry WT, Guo H, Choudhury AD, Gydush G, Reed SC, Rhoades J, Rotem D, Hughes ME, Dillon DA, Partridge AH, Wagle N, Krop IE, Getz G, Golub TR, Love JC, Winer EP, Tolaney SM, Lin NU, Adalsteinsson VA. Association of cell-free DNA tumor fraction and somatic copy number alterations with survival in metastatic triple-negative breast cancer. J Clin Oncol. 2018;36(6):543–553. doi: 10.1200/JCO.2017.76.0033.
    1. Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, Liu CL, Neal JW, Wakelee HA, Merritt RE, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20(5):548–554. doi: 10.1038/nm.3519.
    1. Paruchuri A, Chen H-Z, Bonneville R, Reeser JW, Wing MR, Krook MA, Samorodnitsky E, Miya J, Dao T, Smith A, et al. Research autopsy demonstrates polyclonal acquired resistance in a patient with metastatic gi stromal tumor. JCO Precision Oncol. 2020;4:131–138. doi: 10.1200/PO.19.00328.
    1. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6(224):224ra224. doi: 10.1126/scitranslmed.3007094.
    1. Siravegna G, Mussolin B, Buscarino M, Corti G, Cassingena A, Crisafulli G, Ponzetti A, Cremolini C, Amatu A, Lauricella C, Lamba S, Hobor S, Avallone A, Valtorta E, Rospo G, Medico E, Motta V, Antoniotti C, Tatangelo F, Bellosillo B, Veronese S, Budillon A, Montagut C, Racca P, Marsoni S, Falcone A, Corcoran RB, di Nicolantonio F, Loupakis F, Siena S, Sartore-Bianchi A, Bardelli A. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med. 2015;21(7):795–801. doi: 10.1038/nm.3870.
    1. Heidary M, Auer M, Ulz P, Heitzer E, Petru E, Gasch C, Riethdorf S, Mauermann O, Lafer I, Pristauz G, Lax S, Pantel K, Geigl JB, Speicher MR. The dynamic range of circulating tumor DNA in metastatic breast cancer. Breast Cancer Res. 2014;16(4):421. doi: 10.1186/s13058-014-0421-y.
    1. Fribbens C, O'Leary B, Kilburn L, Hrebien S, Garcia-Murillas I, Beaney M, Cristofanilli M, Andre F, Loi S, Loibl S, et al. Plasma ESR1 mutations and the treatment of estrogen receptor-positive advanced breast cancer. J Clin Oncol. 2016;34(25):2961–2968. doi: 10.1200/JCO.2016.67.3061.
    1. O'Leary B, Hrebien S, Morden JP, Beaney M, Fribbens C, Huang X, Liu Y, Bartlett CH, Koehler M, Cristofanilli M, et al. Early circulating tumor DNA dynamics and clonal selection with palbociclib and fulvestrant for breast cancer. Nat Commun. 2018;9(1):896. doi: 10.1038/s41467-018-03215-x.
    1. Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, Douville C, Javed AA, Wong F, Mattox A, Hruban RH, Wolfgang CL, Goggins MG, Dal Molin M, Wang TL, Roden R, Klein AP, Ptak J, Dobbyn L, Schaefer J, Silliman N, Popoli M, Vogelstein JT, Browne JD, Schoen RE, Brand RE, Tie J, Gibbs P, Wong HL, Mansfield AS, Jen J, Hanash SM, Falconi M, Allen PJ, Zhou S, Bettegowda C, Diaz LA, Jr, Tomasetti C, Kinzler KW, Vogelstein B, Lennon AM, Papadopoulos N. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359(6378):926–930. doi: 10.1126/science.aar3247.
    1. Garcia-Murillas I, Schiavon G, Weigelt B, Ng C, Hrebien S, Cutts RJ, Cheang M, Osin P, Nerurkar A, Kozarewa I, et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med. 2015;7(302):302ra133. doi: 10.1126/scitranslmed.aab0021.
    1. Garcia-Murillas I, Chopra N, Comino-Méndez I, Beaney M, Tovey H, Cutts RJ, Swift C, Kriplani D, Afentakis M, Hrebien S, Walsh-Crestani G, Barry P, Johnston SRD, Ring A, Bliss J, Russell S, Evans A, Skene A, Wheatley D, Dowsett M, Smith IE, Turner NC. Assessment of molecular relapse detection in early-stage breast cancer. JAMA Oncol. 2019;5(10):1473–1478. doi: 10.1001/jamaoncol.2019.1838.
    1. Parsons HA, Rhoades J, Reed SC, Gydush G, Ram P, Exman P, Xiong K, Lo CC, Li T, Fleharty M, Kirkner GJ, Rotem D, Cohen O, Yu F, Fitarelli-Kiehl M, Leong KW, Hughes ME, Rosenberg SM, Collins LC, Miller KD, Blumenstiel B, Trippa L, Cibulskis C, Neuberg DS, DeFelice M, Freeman SS, Lennon NJ, Wagle N, Ha G, Stover DG, Choudhury AD, Getz G, Winer EP, Meyerson M, Lin NU, Krop I, Love JC, Makrigiorgos GM, Partridge AH, Mayer EL, Golub TR, Adalsteinsson VA. Sensitive detection of minimal residual disease in patients treated for early-stage breast cancer. Clin Cancer Res. 2020;26(11):2556–2564. doi: 10.1158/1078-0432.CCR-19-3005.
    1. Radovich M, Jiang G, Hancock BA, Chitambar C, Nanda R, Falkson C, Lynce FC, Gallagher C, Isaacs C, Blaya M, Paplomata E, Walling R, Daily K, Mahtani R, Thompson MA, Graham R, Cooper ME, Pavlick DC, Albacker LA, Gregg J, Solzak JP, Chen YH, Bales CL, Cantor E, Shen F, Storniolo AMV, Badve S, Ballinger TJ, Chang CL, Zhong Y, Savran C, Miller KD, Schneider BP. Association of circulating tumor DNA and circulating tumor cells after neoadjuvant chemotherapy with disease recurrence in patients with triple-negative breast cancer: preplanned secondary analysis of the BRE12-158 randomized clinical trial. JAMA Oncol. 2020;6(9):1410–1415. doi: 10.1001/jamaoncol.2020.2295.
    1. Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer Registry. Cancer. 2007;109(9):1721–1728. doi: 10.1002/cncr.22618.
    1. Lin NU, Claus E, Sohl J, Razzak AR, Arnaout A, Winer EP. Sites of distant recurrence and clinical outcomes in patients with metastatic triple-negative breast cancer: high incidence of central nervous system metastases. Cancer. 2008;113(10):2638–2645. doi: 10.1002/cncr.23930.
    1. Lin NU, Vanderplas A, Hughes ME, Theriault RL, Edge SB, Wong YN, Blayney DW, Niland JC, Winer EP, Weeks JC. Clinicopathologic features, patterns of recurrence, and survival among women with triple-negative breast cancer in the National Comprehensive Cancer Network. Cancer. 2012;118(22):5463–5472. doi: 10.1002/cncr.27581.
    1. Tolaney SM, Ziehr DR, Guo H, Ng MR, Barry WT, Higgins MJ, Isakoff SJ, Brock JE, Ivanova EV, Paweletz CP, Demeo MK, Ramaiya NH, Overmoyer BA, Jain RK, Winer EP, Duda DG. Phase II and biomarker study of cabozantinib in metastatic triple-negative breast cancer patients. Oncologist. 2017;22(1):25–32. doi: 10.1634/theoncologist.2016-0229.
    1. Stover DG, Collier KA, Tallman D, Forman J, Shukla S, Asad S, Rhoades J, Freeman S, Cherian M, Sardesai S, et al. Abstract PD9-08: modeling clonal structure over narrow time frames via circulating tumor DNA in metastatic breast cancer. Cancer Res. 2021;81(4 Supplement):PD9-08-PD09-08.
    1. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M, DePristo MA. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20(9):1297–1303. doi: 10.1101/gr.107524.110.
    1. Birger C, Stewart C, Leshchiner I, Egalina L, Getz G. CGA WES Characterization Pipeline. 2021.
    1. Carter SL, Cibulskis K, Helman E, McKenna A, Shen H, Zack T, Laird PW, Onofrio RC, Winckler W, Weir BA, Beroukhim R, Pellman D, Levine DA, Lander ES, Meyerson M, Getz G. Absolute quantification of somatic DNA alterations in human cancer. Nat Biotechnol. 2012;30(5):413–421. doi: 10.1038/nbt.2203.
    1. Shen R, Seshan VE. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 2016;44(16):e131. doi: 10.1093/nar/gkw520.
    1. Talevich E, Shain AH, Botton T, Bastian BC. CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput Biol. 2016;12(4):e1004873. doi: 10.1371/journal.pcbi.1004873.
    1. Roth A, Khattra J, Yap D, Wan A, Laks E, Biele J, Ha G, Aparicio S, Bouchard-Côté A, Shah SP. PyClone: statistical inference of clonal population structure in cancer. Nat Methods. 2014;11(4):396–398. doi: 10.1038/nmeth.2883.
    1. Malikic S, McPherson AW, Donmez N, Sahinalp CS. Clonality inference in multiple tumor samples using phylogeny. Bioinformatics. 2015;31(9):1349–1356. doi: 10.1093/bioinformatics/btv003.
    1. Reynisson B, Alvarez B, Paul S, Peters B, Nielsen M. NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res. 2020;48(W1):W449–w454. doi: 10.1093/nar/gkaa379.
    1. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32(18):2847–2849. doi: 10.1093/bioinformatics/btw313.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45(2):228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Choi H, Charnsangavej C, Faria SC, Macapinlac HA, Burgess MA, Patel SR, Chen LL, Podoloff DA, Benjamin RS. Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: proposal of new computed tomography response criteria. J Clin Oncol. 2007;25(13):1753–1759. doi: 10.1200/JCO.2006.07.3049.
    1. Kuderer NM, Burton KA, Blau S, Rose AL, Parker S, Lyman GH, Blau CA. Comparison of 2 commercially available next-generation sequencing platforms in oncology. JAMA Oncol. 2017;3(7):996–998. doi: 10.1001/jamaoncol.2016.4983.
    1. Shih C-S, Blakeley J, Clapp DW, Armstrong AE, Wolters P, Dombi E, Cutter G, Ullrich NJ, Allen J, Packer R, et al. Abstract CT233: treatment of neurofibromatosis type 1 (NF1)-related plexiform neurofibromas (PN) with cabozantinib (XL184): a neurofibromatosis clinical trials consortium phase ii trial. Cancer Res. 2019;79(13 Supplement):CT233.
    1. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, Nik-Zainal S, Martin S, Varela I, Bignell GR, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486(7403):400–404. doi: 10.1038/nature11017.
    1. Sanchez-Garcia F, Villagrasa P, Matsui J, Kotliar D, Castro V, Akavia UD, Chen BJ, Saucedo-Cuevas L, Rodriguez Barrueco R, Llobet-Navas D, Silva JM, Pe’er D. Integration of genomic data enables selective discovery of breast cancer drivers. Cell. 2014;159(6):1461–1475. doi: 10.1016/j.cell.2014.10.048.
    1. I.C.G.C. TPCAaGC Pan-cancer analysis of whole genomes. Nature. 2020;578(7793):82–93. doi: 10.1038/s41586-020-1969-6.
    1. Chakravarty D, Gao J, Phillips SM, Kundra R, Zhang H, Wang J, et al. OncoKB: a precision oncology knowledge base. JCO Precis Oncol. 2017;2017:PO.17.00011. 10.1200/PO.17.00011. Epub 2017 May 16.
    1. Amir E, Miller N, Geddie W, Freedman O, Kassam F, Simmons C, Oldfield M, Dranitsaris G, Tomlinson G, Laupacis A, Tannock IF, Clemons M. Prospective study evaluating the impact of tissue confirmation of metastatic disease in patients with breast cancer. J Clin Oncol. 2012;30(6):587–592. doi: 10.1200/JCO.2010.33.5232.
    1. Chikarmane SA, Tirumani SH, Howard SA, Jagannathan JP, DiPiro PJ. Metastatic patterns of breast cancer subtypes: what radiologists should know in the era of personalized cancer medicine. Clin Radiol. 2015;70(1):1–10. doi: 10.1016/j.crad.2014.08.015.
    1. Yates LR, Knappskog S, Wedge D, Farmery JHR, Gonzalez S, Martincorena I, Alexandrov LB, Van Loo P, Haugland HK, Lilleng PK, et al. Genomic evolution of breast cancer metastasis and relapse. Cancer Cell. 2017;32(2):169–184 e167. doi: 10.1016/j.ccell.2017.07.005.
    1. Kim C, Gao R, Sei E, Brandt R, Hartman J, Hatschek T, Crosetto N, Foukakis T, Navin NE. Chemoresistance evolution in triple-negative breast cancer delineated by single-cell sequencing. Cell. 2018;173(4):879–893.e813. doi: 10.1016/j.cell.2018.03.041.
    1. Gao R, Davis A, McDonald TO, Sei E, Shi X, Wang Y, Tsai PC, Casasent A, Waters J, Zhang H, et al. Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat Genet. 2016;48(10):1119–1130. doi: 10.1038/ng.3641.
    1. McDonald BR, Contente-Cuomo T, Sammut SJ, Odenheimer-Bergman A, Ernst B, Perdigones N, et al. Personalized circulating tumor DNA analysis to detect residual disease after neoadjuvant therapy in breast cancer. Sci Transl Med. 2019;11(504).
    1. Thress KS, Brant R, Carr TH, Dearden S, Jenkins S, Brown H, Hammett T, Cantarini M, Barrett JC. EGFR mutation detection in ctDNA from NSCLC patient plasma: a cross-platform comparison of leading technologies to support the clinical development of AZD9291. Lung Cancer. 2015;90(3):509–515. doi: 10.1016/j.lungcan.2015.10.004.
    1. Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, et al. Circulating tumor DNA Analysis in patients with cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J Clin Oncol. 2018;36(16):1631–41. 10.1200/JCO.2017.76.8671. Epub 2018 Mar 5.
    1. Weber Z, Collier KA, Tallman D, Stover DG. Modeling clonal structure over narrow time frames via circulating tumor DNA in metastatic breast cancer. GitLab. 2021; .

Source: PubMed

3
Abonnieren