Utility of circulating tumor DNA for detection and monitoring of endometrial cancer recurrence and progression ============================================================================================================== * Esther L. Moss * Diviya N. Gorsia * Anna Collins * Pavandeep Sandhu * Nalini Foreman * Anu Gore * Joey Wood * Christopher Kent * Elizabeth A. Stannard * Lee Silcock * David S. Guttery ## Abstract Despite the increasing incidence of endometrial cancer (EC) worldwide and the poor overall survival of patients who recur, no reliable biomarker exists for detecting and monitoring EC recurrence and progression during routine follow-up. Circulating tumor DNA (ctDNA), the tumor-derived fraction of cell-free DNA (cfDNA) holds promise as a sensitive method for monitoring cancer activity and stratifying patients that are likely to respond to chemotherapy. Here, we investigated the utility of ctDNA for detecting and monitoring EC recurrence and progression in 13 patients using a combination of highly-sensitive digital droplet PCR (ddPCR), targeted next-generation sequencing (tNGS) and personalized ctDNA assays. Using the Oncomine™ Pan-cancer cfDNA tNGS panel, at least 1 somatic mutation at a variant allele frequency (VAF) >20% was detected in 69% (9/13) of patient tumors. The four patients with no detectable tumor mutations at >20% VAF were whole exome sequenced, with all four harbouring mutations in genes not analyzed by the Oncomine™ Pan-cancer cfDNA panel. Analysis of matched and longitudinal plasma DNA revealed earlier detection of EC recurrence and progression compared to standard clinical imaging techniques and dynamic kinetics of ctDNA levels reflecting treatment response. We also detected, for the first time to our knowledge, acquired high microsatellite instability (MSI-H) in ctDNA from one patient whose primary tumor was MSI stable. Overall, our study suggests that ctDNA analysis, and in particular MSI analysis in ctDNA could become a useful biomarker for early detection and monitoring of EC recurrence and progression. Keywords * Endometrial cancer * Circulating tumor DNA * Digital droplet PCR * Ion Torrent * Whole exome sequencing ## Introduction Endometrial cancer (EC) is the second most common female cancer in the US, with over 60,000 women diagnosed each year1. In contrast to the high survival rate in patients with primary disease, the prognosis upon recurrence is poor, with a 5-year survival rate in patients with distant recurrence of 17%2, 3, and the majority of EC recurrences in high-risk cases occurring distally within the first 3 years post-treatment4. There is no specific blood protein biomarker that is recommended for use following an EC diagnosis; instead, regular clinical examination is advised with imaging. The ovarian cancer markers CA125 and HE4 have been shown to significantly correlate with prognosis; however, their detection rates in metastatic disease are low, 54% and 75% respectively, and even lower with local recurrence, 39% and 16%5. Hence, more effective biomarkers are required that can detect EC recurrence and disease progression earlier, as well as reliably reflecting the underlying kinetics of the disease. Cell-free DNA (cfDNA) can be extracted from plasma and the tumor-derived fraction of cfDNA, known as circulating tumor DNA (ctDNA) is rapidly becoming a quick and sensitive biomarker for tracking minimal residual disease and accurately reflecting therapeutic response in patients with actionable hotspot mutations6, 7, chromosomal rearrangements8, and gene amplifications9, 10. Although many studies have shown the ability of ctDNA for tracking and monitoring disease in cancers such as lung11, breast12 and colon cancer13, few studies have highlighted its promise in EC. CtDNA has been detected in 18% of primary ECs using next-generation sequencing14, as well as showing elevated levels 6 months before a rise in CA125 or radiological evidence of recurrence on CT imaging in gynecological cancers15. However, to date evidence is sparse highlighting the utility of ctDNA for detecting EC recurrence and monitoring treatment response during routine follow-up. Here, we used a combination of methods including targeted next-generation sequencing (tNGS), personalized ctDNA NGS analysis and digital droplet PCR (ddPCR) with the primary objective of detecting and monitoring EC recurrence and therapeutic response. ## Materials and Methods The study was approved by the Wales Research Ethics Committee 7 (17/WA/0342). All patients gave written informed consent prior to participation. Patients were recruited to the study if they were attending hospital follow-up appointments after an EC diagnosis, either after completion of primary treatment or after disease relapse. The reporting census date for this study was 15th September 2019. ### Extraction and quantitation of DNA Blood sampling and processing was performed as previously described16. Total cell-free DNA (cfDNA) was isolated from at least 3 mL of plasma using the QIAamp Circulating Nucleic Acid Kit (Qiagen) or using the MagMAX Cell-Free DNA Isolation Kit (Thermofisher) in combination with the Kingfisher Flex System (Thermofisher) in an automated manner according to manufacturer’s instructions. Isolation of DNA from lymphocytes and quantitation of total cfDNA was as described previously16. cfDNA levels (ng/mL) were converted to copies/mL plasma assuming 3.3 pg DNA per haploid genome. Formalin-fixed, paraffin-embedded (FFPE) tissue blocks with matching hematoxylin and eosin (H&E) for 13 patients from the study cohort (Table 1) were retrieved and reviewed by a histopathologist (EAS). Regions of tumor were removed from the block using a 1.5-mm tissue microarray needle. DNA was subsequently extracted from the FFPE tissue core using the Qiagen GeneRead Kit according to manufacturer’s instructions. View this table: [Table 1:](http://medrxiv.org/content/early/2020/03/06/2020.03.04.20030908/T1) Table 1: Tumor characteristics and outcome of study cohort (n=13) ### Oncomine targeted NGS Targeted NGS was performed on 20 ng of FFPE tumor DNA and lymphocyte DNA, and at least 5 ng of cfDNA (range 5 – 50 ng) using the Oncomine™ Pan-cancer cfDNA assay (Thermofisher – 52 genes covering >900 COSMIC mutations and 12 CNV regions). Temporary libraries were prepared using the Ion Chef and sequenced on Ion 540 chips using the Ion S5 XL. To achieve the highest coverage possible, a maximum of four samples were sequenced per chip achieving a minimum average read depth of 10,000x. Sequencing data was accessed through the Torrent Suite v5.6 and analyzed using Ion Reporter v5.6. For FFPE and lymphocyte DNA, the Oncomine TagSeq Pancan Tumor w1 workflow was used. For cfDNA, the Oncomine TagSeq Pancan Liquid Biopsy w1 workflow was used. ### Digital droplet PCR Droplet digital PCR (ddPCR) was used to analyse the *TP53* p.Y220C mutation (assay numbers dHsaCP2500536 and dHsaCP2500537; Bio-Rad Laboratories) in two longitudinal plasma samples according to manufacturers’ instructions. Thermal cycling conditions: 10-minute hold at 95 °C, 40 cycles of 95 °C for 15 seconds and then 55 °C for 60 seconds. Raw fluorescence amplitude was analyzed using the Quantasoft version 1.6.6.0320 software (Bio-Rad Laboratories) and used to obtain the fractional abundance17. ### Whole exome sequencing Illumina NextSeq500 was used to perform whole-exome sequencing on 150-200 ng of tumor DNA from each FFPE primary tumor block and 50ng of germline DNA (sequencing was performed using the Cell3 Target: Whole Exome kit, Nonacus Ltd, UK) at an average deduplicated on-target read depth of 200x for all four tumor DNA and 50x for matched lymphocyte samples. All WES data will be deposited in the European Genome–Phenome Archive on publication. ### Personalized cell free DNA library preparation For the four patients whose tumors were whole exome sequenced, a personalized ctDNA panel was developed for analysis of at least 2 SNVs per patient (Table S1). At least 5 ng of cfDNA (range 5 – 25 ng) underwent end-repair and A-tailing, facilitating direct ligation of Illumina sequencing adapters containing unique molecular identifiers. Individual adapter-ligated libraries were pooled in equal amounts and hybridized with DNA biotin-labelled probes (Cell3™ Target Custom Panel, Nonacus Ltd, Birmingham, UK). The captured libraries were sequenced using an Illumina MiSeq by Nonacus, UK, to a minimum depth of 30,000x. ### Bioinformatic analysis BAM files from whole exome sequencing or personalized ctDNA assays were prepared using an in-house pipeline to process sequencing output after demultiplexing. The BAM files were then aligned to human genome reference 38 (GRCh38). Somatic variants arising from WES were called using two variant callers, Platypus18 and Mutect219 whilst variants detected in cfDNA using the personalized cfDNA assay were called using solely Platypus. Variants were called using a minimum base quality and minimum mapping quality of 20, with a minimum of 1 read fitting these parameters for variants to be identified. Additionally, variants were filtered for those with an 11-base window around to the variant location to check for variants with a minimum quality < 15. The output VCF files were annotated using ANNOVAR20 to identify and annotate the subsets and variants. Microsatellite instability was analyzed using MSIsensor21 for detection of mononucleotide repeats at several genomic sites (BAT-25, BAT-26, NR-21, NR-24 and NR-27) in tumor alleles as determined by a shift in ≥3bp in each marker. MSI-high tumors were defined as those with ≥2 unstable markers, whilst samples with 1 unstable markers were classified as MSI low tumors and those with 0 unstable markers were classified as microsatellite stable (MSS). ## Results In total, a pilot cohort of 13 cases of mixed risk and mixed histology were analyzed. At the reporting census date (15th September 2019), six patients had recurred/progressed, three had died (two of their disease (patients 2 and 6) and one of a non-EC cause (patient 10)) and four patients (9, 11, 12, 13) were clinically well with no clinical or radiological sign of recurrence/progression (Table 1). ### Sequencing of primary tumors Initially, targeted NGS using the Oncomine™ Pan-Cancer cfDNA assay was conducted to identify the somatic mutations present in the primary tumor tissue in all cases; in two cases additional biopsies from metastatic and recurrent disease were also sequenced for patients 5 and 9 respectively. At least 1 somatic hotspot mutation at an arbitrary cut-off of ≥20% VAF was detected in 9/13 (69%) patient tumors that was not present in the matched lymphocytes (Table S2). In the other four cases, matched tumor and normal samples were sequenced using a whole exome (WES) approach, then used to guide development of a custom targeted cfDNA panel (Table S1). For details of the mutations detected in each patient and selected for custom ctDNA panel design, see Table S2. Two of the patients sequenced using WES (patients 1 and 2) were found to have an unusually high number of single nucleotide variants (SNVs) with 301 SNVs and 131 SNVs in 37MB respectively; using MSIsensor, patient 1 was identified as MSI-H and patient 2 harboured a *POLE* p.G541E mutation of unknown significance. ### Analysis of cfDNA Matched cfDNA for the nine patients with somatic mutations detected using the Oncomine Pan Cancer cfDNA assay was analyzed using the same assay. For patient 5, two additional cfDNA samples were analyzed using ddPCR. ctDNA was detected in at least one plasma sample for 7/9 (78%) patients using the Oncomine assay with additional mutations detected in ctDNA for three patients that were not present in the primary tumor (Figure 1, Table S2). Patient 11 and 12 did not have detectable ctDNA; however, at the time of analysis they were clinically well with no sign of disease recurrence. For the remaining four patients whose tumor underwent WES and subsequent analysis with personalized ctDNA assays, mutations detected in the primary tumor were also detected in cfDNA for two patients (Table S2). However, for two patients (1 and 3), the somatic mutations identified in ctDNA were not previously identified in the corresponding tumor tissue. Interestingly, these patients had local recurrence and the somatic mutations were all located in the mucin genes *MUC3A, MUC4* and *MUC16*. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/03/06/2020.03.04.20030908/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2020/03/06/2020.03.04.20030908/F1) Figure 1: Multiregional sampling from patient 4 alongside the respective variant allele frequencies of the major driver mutation, *TP53* p.Y220C, identified in the primary tumor sample (located in the endometrium), local metastatic lesion (located in the ovary) and the distal metastatic lesion (located in the omentum). The levels of cfDNA and ctDNA in pre-treatment and post-treatment plasma samples are shown in the accompanying left-hand graph, clearly demonstrating a rise in ctDNA levels 3 months prior to radiological relapse (right-hand). *samples analyzed by ddPCR. ### Comparison of primary and metastatic tissue with matched cfDNA To determine whether ctDNA could be representative of tumor heterogeneity, we analyzed primary and metastatic tissue and matched plasma DNA available for two patients using the Oncomine Pan-Cancer cfDNA assay. For Patient 5, a truncal *TP53* p.Y220C was detected in the primary tumor (90% variant allele frequency - VAF) and in the metastatic biopsies from an ovary (81% VAF) and the omentum (69% VAF), which was also detected in their cfDNA sample prior to treatment (Figure 1). For Patient 9, a truncal *PIK3CA* p.H1047R present in both the primary and relapse tumor sites was detected in cfDNA, along with additional *MET* p.T1010I, *FGFR2* p.S252W and *KRAS* p.G12A mutations that were not present in either biopsy. This suggests that ctDNA can be representative of the mutational profile of multiple tumor sites and give additional information regarding tumor evolution. ### Detection of recurrence and progression Four patients were diagnosed with stage I disease at diagnosis (patients 1, 2, 3 and 4) who subsequently recurred, ctDNA was detected in all four patients (100%) (Figure 2, Table S2). The lead time of ctDNA over radiological imaging was 10 months in one patient and there was 18 months between the first positive ctDNA and the patient developing symptoms (Figure 2). Two of these patients (1 and 2) had serial samples analyzed and disease progression was confirmed in patient 2 with increasing ctDNA levels in concordance with both clinical and imaging progression. Although somatic mutations were detected in the plasma for Patient 1, these mutations were not observed in the tumor tissue as previously mentioned. Two cases were recruited at the time of recurrence and therefore lead time could not be calculated. Four patients were stage IV at diagnosis (5, 6, 7 and 8) and had progressive disease on imaging; ctDNA was detected in all cases (100%). ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/03/06/2020.03.04.20030908/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2020/03/06/2020.03.04.20030908/F2) Figure 2: Summary of each patient’s results of clinical imaging and results of serial samples in terms of detection of ctDNA. ### Response to treatment Patient 5 underwent chemotherapy for distant recurrence and serial ctDNA levels mirrored the radiological response, even becoming undetectable during treatment, before rising again prior to radiological progression (Figure 1). An additional two patients (9 and 11) diagnosed with stage I EC received radiotherapy, one for a vaginal vault recurrence and the other as primary treatment, respectively. For patient 11, ctDNA was not detected in three plasma samples following primary radical radiotherapy, in keeping with clinical findings of complete response (Figure 2). ctDNA was detected following treatment of the vault recurrence (patient 9); however, became negative with time and remained negative, in keeping with clinical and radiological findings of complete resolution of the recurrence and no ongoing disease. ### Follow-up of high-risk cases In the three cases with high-risk EC at diagnosis (stage III non-endometrioid histology– patients 11, 12 and 13) ctDNA was detected in only one case (Figure 2). All patients are alive with no clinical or radiological sign of recurrence and continue to be followed-up. ### Tumor classification Microsatellite instability was also assessed for four patients who underwent WES analysis using the custom panel by determining the length of mononucleotide repeats at several genomic sites (BAT-25, BAT-26, NR-21, NR-24 and NR-27), indicative of defects in DNA repair. Only patient 1’s tumor was determined as MSI-H with all five markers displaying microsatellite instability, which was also detected in her last plasma sample taken at relapse, with three of the five MSI markers displaying microsatellite instability. Interestingly, analysis of longitudinal plasma DNA revealed patient 2 to have acquired MSI-H status, with two of the five markers displaying MSI in the last plasma sample taken post-recurrence and upon confirmation of progressive disease, despite her primary tumor being MSS. ## Discussion We have shown that ctDNA is highly sensitive in identifying endometrial cancer recurrence in different histological subtypes. In addition, we have shown that ctDNA is responsive following treatment, reflecting disease activity, and appears to have a lead time of up to 18 months over patient reported symptoms and up to 10 months compared to imaging. These results are in keeping with studies published in other tumor types and supports the clinical utility of ctDNA in early detection of recurrence12, 13. The use of different technologies for ctDNA detection has given further insight into the optimum methods that can be taken forward for use in a clinical trial determining the potential for this new development on patient management. Our results show a benefit in using WES to profile the tumor tissue and tracking mutations with a customised targeted cfDNA panel relative to using a driver gene approach which did not identify any mutations in four tumor samples, equating to approximately one third of our cohort who otherwise would not have been monitored. Additionally, the customised targeted panel has greater informative value in monitoring of tumor evolution, as exemplified by patients 1 and 3 for whom the targeted custom panel identified additional somatic variants not present in the primary tumor tissue. This further highlights the beneficial value of using a targeted customised panel approach over a driver gene approach as it may not elucidate the evolving genetic proportions as accurately as the personalized approach. Our results have also shown a putative utility in elucidating therapeutic response of patients and in early detection of therapeutic resistance as demonstrated in the cases of patient 5. The initial post-treatment plasma samples for patient 5 did not have somatic variants detected demonstrating the good therapeutic response seen in clinic; however, emergence of the clonal mutation consistent with therapeutic resistance was identified in the third post-treatment sample obtained, consequently resulting in associated clinical progression. In contrast to this, patients 11 and 12 both demonstrated a good therapeutic response clinically, corroborating the lack of mutations. Finally, our results have also shown an additional benefit of longitudinal monitoring of patients using personalized assays containing MSI markers with discovery of acquired MSI-H status through serial plasma sampling. To our knowledge, this is the first evidence of acquired MSI-H status in endometrial cancer and demonstrates that determination and monitoring of MSI status is also indicative of the underlying tumor biology - as demonstrated in the case of patient 2 - whose acquired MSI-H status was indicative of progression and corroborated by increasing ctDNA levels. Detection of MSI emergence in the final plasma sample may have been of clinical utility with the clinical likelihood of switching the therapeutic regimen, to immunotherapy to possibly bettering the clinical outcome of the patient22. Overall, we have shown that personalized ctDNA monitoring has utility in endometrial cancer; however, analysis in a much larger cohort is required and is currently ongoing. ## Data Availability All WES data will be deposited in the European Genome-Phenome Archive on publication. ## Footnotes * **Support:** This work was supported by a Hope Against Cancer/Leicester Precision Medicine Institute PhD studentship (DSG, DNG) in conjunction with the UK Department of Health on an Experimental Cancer Medicine Centre grant [C10604/A25151], and Medical Research Council (MRC) Proximity to Discovery scheme award [MCPC17194]. This research used the ALICE and SPECTRE High Performance Computing Facilities at the University of Leicester. * **Conflict-of-interest disclosure:** Lee Silcock is employed by Nonacus Limited. The remaining authors declare no potential conflicts of interest. * **Novelty and Impact:** This study demonstrates that detection of EC recurrence and progression can be detected and monitored using ctDNA, as well as acquired microsatellite instability which, in the future, could be used to guide management with early discontinuation of non-efficacious treatments that are not impacting on tumor activity. * Received March 4, 2020. * Revision received March 4, 2020. * Accepted March 6, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 1.Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, Alfano CM, Jemal A, Kramer JL, Siegel RL. Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin 2019;69: 363–85. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3322/caac.21565&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 2. 2.Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, et al., SEER Cancer Statistics Review, 1975-2016. National Cancer Institute, 2019. 3. 3.Makker V, Green AK, Wenham RM, Mutch D, Davidson B, Miller DS. New therapies for advanced, recurrent, and metastatic endometrial cancers. Gynecol Oncol Res Pract 2017;4: 19. 4. 4.de Boer SM, Powell ME, Mileshkin L, Katsaros D, Bessette P, Haie-Meder C, Ottevanger PB, Ledermann JA, Khaw P, D’Amico R, Fyles A, Baron MH, et al. Adjuvant chemoradiotherapy versus radiotherapy alone in women with high-risk endometrial cancer (PORTEC-3): patterns of recurrence and post-hoc survival analysis of a randomised phase 3 trial. Lancet Oncol 2019;20: 1273–85. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 5. 5.Abbink K, Zusterzeel PL, Geurts-Moespot AJ, Herwaarden AEV, Pijnenborg JM, Sweep FC, Massuger LF. HE4 is superior to CA125 in the detection of recurrent disease in highrisk endometrial cancer patients. Tumour Biol 2018;40: 1010428318757103. 6. 6.Garcia-Murillas I, Schiavon G, Weigelt B, Ng C, Hrebien S, Cutts RJ, Cheang M, Osin P, Nerurkar A, Kozarewa I, Garrido JA, Dowsett M, et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med 2015;7: 302ra133. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjE0OiI3LzMwMi8zMDJyYTEzMyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzAzLzA2LzIwMjAuMDMuMDQuMjAwMzA5MDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 7. 7.Guttery DS, Page K, Hills A, Woodley L, Marchese SD, Rghebi B, Hastings RK, Luo J, Pringle JH, Stebbing J, Coombes RC, Ali S, et al. Noninvasive detection of activating estrogen receptor 1 (ESR1) mutations in estrogen receptor-positive metastatic breast cancer. Clinical chemistry 2015;61: 974–82. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiY2xpbmNoZW0iO3M6NToicmVzaWQiO3M6ODoiNjEvNy85NzQiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wMy8wNi8yMDIwLjAzLjA0LjIwMDMwOTA4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 8. 8.Olsson E, Winter C, George A, Chen Y, Howlin J, Tang MH, Dahlgren M, Schulz R, Grabau D, van Westen D, Ferno M, Ingvar C, et al. Serial monitoring of circulating tumor DNA in patients with primary breast cancer for detection of occult metastatic disease. EMBO Mol Med 2015;7: 1034–47. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZW1ib21tIjtzOjU6InJlc2lkIjtzOjg6IjcvOC8xMDM0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDMvMDYvMjAyMC4wMy4wNC4yMDAzMDkwOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 9. 9.Page K, Guttery DS, Fernandez-Garcia D, Hills A, Hastings RK, Luo J, Goddard K, Shahin V, Woodley-Barker L, Rosales BM, Coombes RC, Stebbing J, et al. Next Generation Sequencing of Circulating Cell-Free DNA for Evaluating Mutations and Gene Amplification in Metastatic Breast Cancer. Clinical chemistry 2017;63: 532–41. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiY2xpbmNoZW0iO3M6NToicmVzaWQiO3M6ODoiNjMvMi81MzIiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wMy8wNi8yMDIwLjAzLjA0LjIwMDMwOTA4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 10. 10.Shaw JA, Page K, Blighe K, Hava N, Guttery D, Ward B, Brown J, Ruangpratheep C, Stebbing J, Payne R, Palmieri C, Cleator S, et al. Genomic analysis of circulating cell-free DNA infers breast cancer dormancy. Genome research 2012;22: 220–31. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ2Vub21lIjtzOjU6InJlc2lkIjtzOjg6IjIyLzIvMjIwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDMvMDYvMjAyMC4wMy4wNC4yMDAzMDkwOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 11. 11.Abbosh C, Birkbak NJ, Wilson GA, Jamal-Hanjani M, Constantin T, Salari R, Le Quesne J, Moore DA, Veeriah S, Rosenthal R, Marafioti T, Kirkizlar E, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545: 446–51. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature22364&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28445469&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 12. 12.Coombes RC, Page K, Salari R, Hastings RK, Armstrong A, Ahmed S, Ali S, Cleator S, Kenny L, Stebbing J, Rutherford M, Sethi H, et al. Personalized Detection of Circulating Tumor DNA Antedates Breast Cancer Metastatic Recurrence. Clinical cancer research : an official journal of the American Association for Cancer Research 2019;25: 4255–63. 13. 13.Reinert T, Henriksen TV, Christensen E, Sharma S, Salari R, Sethi H, Knudsen M, Nordentoft I, Wu HT, Tin AS, Heilskov Rasmussen M, Vang S, et al. Analysis of Plasma Cell-Free DNA by Ultradeep Sequencing in Patients With Stages I to III Colorectal Cancer. JAMA Oncol 2019. 14. 14.Bolivar AM, Luthra R, Mehrotra M, Chen W, Barkoh BA, Hu P, Zhang W, Broaddus RR. Targeted next-generation sequencing of endometrial cancer and matched circulating tumor DNA: identification of plasma-based, tumor-associated mutations in early stage patients. Mod Pathol 2019;32: 405–14. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 15. 15.Pereira E, Camacho-Vanegas O, Anand S, Sebra R, Catalina Camacho S, Garnar-Wortzel L, Nair N, Moshier E, Wooten M, Uzilov A, Chen R, Prasad-Hayes M, et al. Personalized Circulating Tumor DNA Biomarkers Dynamically Predict Treatment Response and Survival In Gynecologic Cancers. PloS one 2015;10: e0145754. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0145754&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26717006&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 16. 16.Page K, Guttery DS, Zahra N, Primrose L, Elshaw SR, Pringle JH, Blighe K, Marchese SD, Hills A, Woodley L, Stebbing J, Coombes RC, et al. Influence of plasma processing on recovery and analysis of circulating nucleic acids. PloS one 2013;8: e77963. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0077963&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24205045&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 17. 17.Shaw JA, Guttery DS, Hills A, Fernandez-Garcia D, Page K, Rosales BM, Goddard KS, Hastings RK, Luo J, Ogle O, Woodley L, Ali S, et al. Mutation Analysis of Cell-Free DNA and Single Circulating Tumor Cells in Metastatic Breast Cancer Patients with High Circulating Tumor Cell Counts. Clinical cancer research : an official journal of the American Association for Cancer Research 2017;23: 88–96. 18. 18.Rimmer A, Phan H, Mathieson I, Iqbal Z, Twigg SRF, Consortium WGS, Wilkie AOM, McVean G, Lunter G. Integrating mapping-, assembly-and haplotype-based approaches for calling variants in clinical sequencing applications. Nature genetics 2014;46: 912–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3036&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25017105&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 19. 19.Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C, Gabriel S, Meyerson M, Lander ES, Getz G. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 2013;31: 213–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nbt.2514&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23396013&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 20. 20.Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic acids research 2010;38: e164. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkq603&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20601685&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 21. 21.Niu B, Ye K, Zhang Q, Lu C, Xie M, McLellan MD, Wendl MC, Ding L. MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics 2014;30: 1015–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btt755&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24371154&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F03%2F06%2F2020.03.04.20030908.atom) 22. 22.Di Tucci C, Capone C, Galati G, Iacobelli V, Schiavi MC, Di Donato V, Muzii L, Panici PB. Immunotherapy in endometrial cancer: new scenarios on the horizon. J Gynecol Oncol 2019;30: e46.