Circulating Tumor DNA Analysis for Detection of Minimal Residual Disease After Chemoradiotherapy for Localized Esophageal Cancer

Tej D Azad, Aadel A Chaudhuri, Penny Fang, Yawei Qiao, Mohammad S Esfahani, Jacob J Chabon, Emily G Hamilton, Yi D Yang, Alex Lovejoy, Aaron M Newman, David M Kurtz, Michael Jin, Joseph Schroers-Martin, Henning Stehr, Chih Long Liu, Angela Bik-Yu Hui, Viren Patel, Dipen Maru, Steven H Lin, Ash A Alizadeh, Maximilian Diehn, Tej D Azad, Aadel A Chaudhuri, Penny Fang, Yawei Qiao, Mohammad S Esfahani, Jacob J Chabon, Emily G Hamilton, Yi D Yang, Alex Lovejoy, Aaron M Newman, David M Kurtz, Michael Jin, Joseph Schroers-Martin, Henning Stehr, Chih Long Liu, Angela Bik-Yu Hui, Viren Patel, Dipen Maru, Steven H Lin, Ash A Alizadeh, Maximilian Diehn

Abstract

Background & aims: Biomarkers are needed to risk stratify after chemoradiotherapy for localized esophageal cancer. These could improve identification of patients at risk for cancer progression and selection of additional therapy.

Methods: We performed deep sequencing (CAncer Personalized Profiling by deep Sequencing, [CAPP-Seq]) analyses of plasma cell-free DNA collected from 45 patients before and after chemoradiotherapy for esophageal cancer, as well as DNA from leukocytes and fixed esophageal tumor biopsy samples collected during esophagogastroduodenoscopy. Patients were treated from May 2010 through October 2015; 23 patients subsequently underwent esophagectomy, and 22 did not undergo surgery. We also sequenced DNA from blood samples from 40 healthy control individuals. We analyzed 802 regions of 607 genes for single-nucleotide variants previously associated with esophageal adenocarcinoma or squamous cell carcinoma. Patients underwent imaging analyses 6-8 weeks after chemoradiotherapy and were followed for 5 years. Our primary aim was to determine whether detection of circulating tumor DNA (ctDNA) after chemoradiotherapy is associated with risk of tumor progression (growth of local, regional, or distant tumors, detected by imaging or biopsy).

Results: The median proportion of tumor-derived DNA in total cell-free DNA before treatment was 0.07%, indicating that ultrasensitive assays are needed for quantification and analysis of ctDNA from localized esophageal tumors. Detection of ctDNA after chemoradiotherapy was associated with tumor progression (hazard ratio, 18.7; P < .0001), formation of distant metastases (hazard ratio, 32.1; P < .0001), and shorter disease-specific survival times (hazard ratio, 23.1; P < .0001). A higher proportion of patients with tumor progression had new mutations detected in plasma samples collected after chemoradiotherapy than patients without progression (P = .03). Detection of ctDNA after chemoradiotherapy preceded radiographic evidence of tumor progression by an average of 2.8 months. Among patients who received chemoradiotherapy without surgery, combined ctDNA and metabolic imaging analysis predicted progression in 100% of patients with tumor progression, compared with 71% for only ctDNA detection and 57% for only metabolic imaging analysis (P < .001 for comparison of either technique to combined analysis).

Conclusions: In an analysis of cell-free DNA in blood samples from patients who underwent chemoradiotherapy for esophageal cancer, detection of ctDNA was associated with tumor progression, metastasis, and disease-specific survival. Analysis of ctDNA might be used to identify patients at highest risk for tumor progression.

Keywords: Chemoradiotherapy; Genetics; Polymorphism; SNP.

Copyright © 2020 AGA Institute. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.. Pre-CRT assessment of ctDNA in…
Figure 1.. Pre-CRT assessment of ctDNA in patients with locally advanced ESCA.
(A) Study overview of forty-five patients with EGD-proven locally advanced ESCA enrolled in the study. Plasma samples were collected before and after chemoradiotherapy (CRT). Following CRT, patients either underwent esophagectomy (n=23) or no further treatment (n=22). (B) Clinical characteristics and tumor genotyping results. Pre-CRT ctDNA detection status and clinical characteristics, where each column is a patient and each row is a parameter (e.g. histology). Similarly, the associated co-mutation plot depicts patient-level mutational profiles of 45 tumors from patients with esophageal cancer genotyped by our ESCA-specific NGS panel. Genes mutated in at least 5% of the patients in our cohort are depicted. The fraction of tumors with mutations in each gene is denoted on the left. (C) Comparison of absolute ctDNA concentration between Stage III esophageal adenocarcinoma (EAC, n=16) and Stage III lung adenocarcinoma (LUAD, n=10). P value calculated by the Mann-Whitney test. (D) Pre-treatment ctDNA concentration in esophageal adenocarcinoma (EAC, n= 23) and esophageal squamous cell carcinoma (ESCC, n=7) patients. Patients were included if tumor-informed mutations were detected either pre- or post-CRT. Data represent mean + SEM. P value was calculated by the Mann-Whitney test. (E) Correlation of ctDNA concentration (haploid genome equivalents per mL, hGE/mL) with pre-CRT metabolic tumor volume (MTV) measured by PET-CT, stratified by histology. P value and ρ were calculated by Pearson correlation.
Figure 2.. Detection of tumor-informed mutations following…
Figure 2.. Detection of tumor-informed mutations following chemoradiotherapy is strongly prognostic.
(A) Tumor variant allele fraction (VAF), stratified by whether and at which time point a patient had detectable ctDNA (x-axis categories) and further stratified by whether a given mutation was detectable in plasma (column colors). P value calculated by a 2-way ANOVA and was significant for difference based on if a mutation was detected in plasma (P = .01), but not for if a patient was detected pre- or post-CRT. (B) Tumor mutations detected in plasma pre- and post-CRT. Denominator is the total number of patients with detectable ctDNA, pre- and post-CRT (n=27 and n=5, respectively). Genes depicted were mutated in more than 5% of tumors in cBioportal ESCA datasets. (C-E) Kaplan–Meier analyses comparing patients with detectable and undetectable ctDNA in the post-CRT sample using tumor-informed ctDNA detection for (C) freedom from progression (P<.0001, HR = 18.7 (95%CI, 1.1–316.5)), (D) distant metastasis-free survival (P<.0001, HR = 32.1 (95%CI, 1.8–559.2)) and (E) disease-specific survival (P < .0001, HR = 23.1 (95%CI, 2.0–273.5)). ctDNA negative (n =26) versus ctDNA positive (n = 5). Time (days) was measured from the landmark (post-CRT blood draw). One patient was excluded from this analysis due to an event that occurred prior to the post-CRT blood collection. P values and hazard ratios were calculated from the log-rank test.
Figure 3.. Putative emergent mutations following CRT…
Figure 3.. Putative emergent mutations following CRT may be associated with disease progression.
(A) In five patients we detected six new nonsynonymous mutations in post-CRT plasma that were absent pre-CRT. Column dot plots indicates allele fractions of both tumor-informed (circles) and emergent (squares) mutations. These mutations were absent in pre-CRT plasma and matched germline, tumor, and in 20 healthy controls. (B) Presence of putative emergent mutations in patients who developed disease progression (progressors; n = 10) versus those who did not (non-progressors; n = 20). P value derived from Fisher’s exact test. (C) Patient (EP32) with non-FDG avid stage II EAC treated with CRT-alone who developed an emergent ARID1A G696V mutation following CRT. This patient went on to develop distant metastasis (para-aortic lymph node). (D) Kaplan–Meier analysis of freedom from progression (P < .0001, HR = 13.2 (95%CI, 2.3–75.1)) comparing patients with or without detectable ctDNA post-CRT (n=8 and 23, respectively). Detectable ctDNA is defined as detection of tumor-informed or putative emergent mutations following CRT. Time (days) was measured from the landmark (post-CRT blood draw). P values and hazard ratios (HR) were calculated from the log-rank test. AF, allele fraction; CRT, chemoradiotherapy; IMRT, intensity modulated radiotherapy; EAC, esophageal adenocarcinoma.
Figure 4.. ctDNA allows earlier and more…
Figure 4.. ctDNA allows earlier and more robust detection of recurrence compared to PET-CT imaging.
(A) Kaplan–Meier analysis for event-free survival (P = .0026), comparing ctDNA detection at the post-CRT time point with standard-of-care PET-CT imaging (n=10). Time to event (days) was measured from end of CRT. P values and hazard ratios (HR) were calculated from the log-rank test. (B) Column dot plot of the lead time to radiographic recognition of recurrence for ctDNA detection (n=7), with the bars representing mean (114.9) and standard error of the mean (32.9). (C) Patient (EP58) with stage IIIA EAC with partial response to CRT on surveillance imaging and a decrease in post-CRT MTV but had increased levels of ctDNA post-CRT and experiences distant liver metastasis. (D) Patient (EP29) with stage IIB EAC with equivocal surveillance imaging, increased post-CRT MTV, and undetectable post-CRT ctDNA who achieves long-term survival.
Figure 5.. Integrating ctDNA and ΔTLG enables…
Figure 5.. Integrating ctDNA and ΔTLG enables improves detection of recurrence among patients receiving CRT alone.
Kaplan–Meier analysis for (A) freedom from progression (P=.0093, HR 6.4 (95% CI 1.2–33.1)) and (B) disease-specific survival (P=.015, HR 9.1 (95% CI 1.4–60.8)) stratified by post-CRT ctDNA detection among patients receiving CRT alone (n=12). Time to event (days) was measured from post-CRT PET imaging. P values and hazard ratios (HR) were calculated from the log-rank test. (C)Mean sensitivity and specificity of ctDNA, ΔTLG, and both, for any recurrence detection (n=12). ΔTLG is defined as the percent change in TLG in response to CRT. Mean and standard deviation calculated using leave-one-out approach. P values calculated by the Mann-Whitney test. Kaplan–Meier analysis for (D) freedom from progression (P=.0011) and (E) disease-specific survival (P=.026) stratified by integration of ctDNA detection and ΔTLG at the post-CRT time point (n=12). Patients in the red curve (n=7) either had detectable ctDNA following CRT or had ΔTLG ≥ −48.5% while patients in the blue curve (n=5) had undetectable ctDNA after CRT and had ΔTLG < −48.5%. Time to event (days) was measured from post-CRT PET imaging. P values were calculated from the log-rank test. Hazard ratios are not reported as they are undefined given the absence of any events in the ctDNA and ΔTLG negative group. (F) Patient (EP34) with stage IIA ESCC treated with CRT-alone who achieved long-term survival with no evidence of disease. Patient had detectable ctDNA pre-treatment and remains undetectable following CRT.

References

    1. van Hagen P, Hulshof MC, van Lanschot JJ, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 2012;366:2074–84.
    1. Shapiro J, van Lanschot JJB, Hulshof M, et al. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol 2015;16:1090–1098.
    1. Noordman BJ, Spaander MCW, Valkema R, et al. Detection of residual disease after neoadjuvant chemoradiotherapy for oesophageal cancer (preSANO): a prospective multicentre, diagnostic cohort study. Lancet Oncol 2018.
    1. Donahue JM, Nichols FC, Li Z, et al. Complete pathologic response after neoadjuvant chemoradiotherapy for esophageal cancer is associated with enhanced survival. Ann Thorac Surg 2009;87:392–8; discussion 398–9.
    1. Bedenne L, Michel P, Bouche O, et al. Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol 2007;25:1160–8.
    1. Stahl M, Stuschke M, Lehmann N, et al. Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 2005;23:2310–7.
    1. Berger AC, Farma J, Scott WJ, et al. Complete response to neoadjuvant chemoradiotherapy in esophageal carcinoma is associated with significantly improved survival. J Clin Oncol 2005;23:4330–7.
    1. Schneider PM, Metzger R, Schaefer H, et al. Response evaluation by endoscopy, rebiopsy, and endoscopic ultrasound does not accurately predict histopathologic regression after neoadjuvant chemoradiation for esophageal cancer. Ann Surg 2008;248:902–8.
    1. Westerterp M, van Westreenen HL, Reitsma JB, et al. Esophageal cancer: CT, endoscopic US, and FDG PET for assessment of response to neoadjuvant therapy--systematic review. Radiology 2005;236:841–51.
    1. Garcia-Murillas I, Schiavon G, Weigelt B, et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med 2015;7:302ra133.
    1. Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med 2016;8:346ra92.
    1. Chaudhuri AA, Chabon JJ, Lovejoy AF, et al. Early Detection of Molecular Residual Disease in Localized Lung Cancer by Circulating Tumor DNA Profiling. Cancer Discov 2017;7:1394–1403.
    1. Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545:446–451.
    1. Luo H, Li H, Hu Z, et al. Noninvasive diagnosis and monitoring of mutations by deep sequencing of circulating tumor DNA in esophageal squamous cell carcinoma. Biochem Biophys Res Commun 2016;471:596–602.
    1. Hsieh CC, Hsu HS, Chang SC, et al. Circulating Cell-Free DNA Levels Could Predict Oncological Outcomes of Patients Undergoing Esophagectomy for Esophageal Squamous Cell Carcinoma. Int J Mol Sci 2016;17.
    1. Newman AM, Lovejoy AF, Klass DM, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol 2016;34:547–555.
    1. Newman AM, Bratman SV, To J, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med 2014;20:548–54.
    1. Chabon JJ, Simmons AD, Lovejoy AF, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun 2016;7:11815.
    1. Giobbie-Hurder A, Gelber RD, Regan MM. Challenges of guarantee-time bias. J Clin Oncol 2013;31:2963–9.
    1. Dulak AM, Stojanov P, Peng S, et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat Genet 2013;45:478–86.
    1. Agrawal N, Jiao Y, Bettegowda C, et al. Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. Cancer Discov 2012;2:899–905.
    1. Cancer Genome Atlas Research N, Analysis Working Group: Asan U, Agency BCC, et al. Integrated genomic characterization of oesophageal carcinoma. Nature 2017;541:169–175.
    1. Song Y, Li L, Ou Y, et al. Identification of genomic alterations in oesophageal squamous cell cancer. Nature 2014;509:91–5.
    1. Sausen M, Phallen J, Adleff V, et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat Commun 2015;6:7686.
    1. Tie J, Cohen JD, Wang Y, et al. Serial circulating tumour DNA analysis during multimodality treatment of locally advanced rectal cancer: a prospective biomarker study. Gut 2018.
    1. Bailey M, Tokheim C, Porta-Pardo E, et al. Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell 2018;173:371–385.
    1. Findlay JM, Castro-Giner F, Makino S, et al. Differential clonal evolution in oesophageal cancers in response to neo-adjuvant chemotherapy. Nat Commun 2016;7:11111.
    1. Genovese G, Kahler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014;371:2477–87.
    1. Xie M, Lu C, Wang J, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014;20:1472–8.
    1. Park IH, Kim JY. Surveillance or resection after chemoradiation in esophageal cancer. Ann Transl Med 2018;6:82.
    1. Cronin-Fenton DP, Sharp L, Carsin AE, et al. Patterns of care and effects on mortality for cancers of the oesophagus and gastric cardia: a population-based study. Eur J Cancer 2007;43:565–75.
    1. Jayachandran P, Pai RK, Quon A, et al. Postchemoradiotherapy positron emission tomography predicts pathologic response and survival in patients with esophageal cancer. Int J Radiat Oncol Biol Phys 2012;84:471–7.
    1. Monjazeb AM, Riedlinger G, Aklilu M, et al. Outcomes of patients with esophageal cancer staged with [(1)(8)F]fluorodeoxyglucose positron emission tomography (FDG-PET): can postchemoradiotherapy FDG-PET predict the utility of resection? J Clin Oncol 2010;28:4714–21.
    1. Roedl JB, Colen RR, Holalkere NS, et al. Adenocarcinomas of the esophagus: response to chemoradiotherapy is associated with decrease of metabolic tumor volume as measured on PET-CT. Comparison to histopathologic and clinical response evaluation. Radiother Oncol 2008;89:278–86.
    1. Alghamedi A, Buduhan G, Tan L, et al. Quality of life assessment in esophagectomy patients. Ann Transl Med 2018;6:84.
    1. Vellayappan BA, Soon YY, Ku GY, et al. Chemoradiotherapy versus chemoradiotherapy plus surgery for esophageal cancer. Cochrane Database Syst Rev 2017;8:CD010511.
    1. Lopez A, Harada K, Mizrak Kaya D, et al. Liquid biopsies in gastrointestinal malignancies: when is the big day? Expert Rev Anticancer Ther 2018;18:19–38.
    1. Creemers A, Krausz S, Strijker M, et al. Clinical value of ctDNA in upper-GI cancers: A systematic review and meta-analysis. Biochim Biophys Acta 2017;1868:394–403.
    1. Openshaw MR, Richards CJ, Guttery DS, et al. The genetics of gastroesophageal adenocarcinoma and the use of circulating cell free DNA for disease detection and monitoring. Expert Rev Mol Diagn 2017;17:459–470.
    1. Andolfo I, Petrosino G, Vecchione L, et al. Detection of erbB2 copy number variations in plasma of patients with esophageal carcinoma. BMC Cancer 2011;11:126.
    1. Komatsu S, Ichikawa D, Hirajima S, et al. Clinical impact of predicting CCND1 amplification using plasma DNA in superficial esophageal squamous cell carcinoma. Dig Dis Sci 2014;59:1152–9.
    1. Ueda M, Iguchi T, Masuda T, et al. Somatic mutations in plasma cell-free DNA are diagnostic markers for esophageal squamous cell carcinoma recurrence. Oncotarget 2016;7:62280–62291.

Source: PubMed

3
Iratkozz fel