151
|
He Y, Ma X, Chen K, Liu F, Cai S, Han-Zhang H, Hou T, Xiang J, Peng J. Perioperative Circulating Tumor DNA in Colorectal Liver Metastases: Concordance with Metastatic Tissue and Predictive Value for Tumor Burden and Prognosis. Cancer Manag Res 2020; 12:1621-1630. [PMID: 32184665 PMCID: PMC7061429 DOI: 10.2147/cmar.s240869] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/08/2020] [Indexed: 12/18/2022] Open
Abstract
Background The surgical resection of colorectal cancer with liver metastases (CLM) has proven to be the most important modality for long-term survival, while effective biomarkers for outcome prediction or postoperative surveillance are still lacking. Currently, circulating biomarkers obtained from a liquid biopsy are widely used to assess the treatment response, disease recurrence and progression. In this study, we analyzed the value of the liquid biopsy, which includes circulating tumor DNA (ctDNA) and cell-free DNA (cfDNA), in patients with CLM. Methods Capture-based targeted deep sequencing was performed on matched pre-surgery, post-surgery and liver metastatic tissues of 20 CRC patients who underwent the resection of liver metastases between May and September 2017 using a panel consisting of 41 genes. Mutation landscapes obtained from pre-surgery plasma samples and metastatic tissue samples were compared. Results Collectively, we identified 47 mutations from 17 pre-surgery plasma samples (85%), and the remaining 3 patients had no mutation detected from the panel. We revealed a high by-variant concordance rate of 82.14% between pre-surgery plasma samples and liver metastatic tissue samples. We further analyzed the correlation between ctDNA, cfDNA, CEA and tumor burden and revealed a positive correlation between ctDNA and tumor burden (R=0.69, p=0.002). As of the date for data cutoff, 8/20 patients experienced relapse. Our study also demonstrated that pre-surgery ctDNA (p<0.001), cfDNA (p=0.001) and CEA (p=0.012) levels had predictive value for relapse. Patients with low pre-surgery ctDNA (p<0.001), cfDNA (p=0.001) or CEA (p=0.012) levels were more likely to experience prolonged progression-free survival. Conclusion Our data demonstrate that the genomic profile obtained from ctDNA is comparable with the genomic profile obtained from metastatic liver tumors. Furthermore, our study also show that pre-surgery ctDNA levels are positively correlated with tumor burden. In addition, pre-surgery ctDNA, cfDNA and CEA levels have predictive value for relapse.
Collapse
Affiliation(s)
- Yiping He
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xiaoji Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Ke Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Department of Endoscopy, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China
| | - Fangqi Liu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Han Han-Zhang
- Burning Rock Biotech, Guangzhou, Guangdong, People's Republic of China
| | - Ting Hou
- Burning Rock Biotech, Guangzhou, Guangdong, People's Republic of China
| | - Jianxing Xiang
- Burning Rock Biotech, Guangzhou, Guangdong, People's Republic of China
| | - Junjie Peng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
152
|
To YH, Lee M, Gibbs P, Tie J. Circulating tumour DNA in early stage colorectal cancer: can blood tell all? ANNALS OF TRANSLATIONAL MEDICINE 2020; 7:S358. [PMID: 32016076 DOI: 10.21037/atm.2019.09.55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yat Hang To
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Margaret Lee
- Division of Systems Biology and Personalised Medicine, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Oncology, Western Health, Victoria, Australia.,Department of Medical Oncology, Eastern Health, Victoria, Australia
| | - Peter Gibbs
- Division of Systems Biology and Personalised Medicine, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Oncology, Western Health, Victoria, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria, Australia
| | - Jeanne Tie
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Division of Systems Biology and Personalised Medicine, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Oncology, Western Health, Victoria, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria, Australia
| |
Collapse
|
153
|
Chera BS, Kumar S, Shen C, Amdur R, Dagan R, Green R, Goldman E, Weiss J, Grilley-Olson J, Patel S, Zanation A, Hackman T, Blumberg J, Patel S, Thorp B, Weissler M, Yarbrough W, Sheets N, Mendenhall W, Tan XM, Gupta GP. Plasma Circulating Tumor HPV DNA for the Surveillance of Cancer Recurrence in HPV-Associated Oropharyngeal Cancer. J Clin Oncol 2020; 38:1050-1058. [PMID: 32017652 PMCID: PMC7106982 DOI: 10.1200/jco.19.02444] [Citation(s) in RCA: 244] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Plasma circulating tumor human papillomavirus DNA (ctHPVDNA) is a sensitive and specific biomarker of human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC). We investigated whether longitudinal monitoring of ctHPVDNA during post-treatment surveillance could accurately detect clinical disease recurrence. METHODS AND MATERIALS A prospective biomarker clinical trial was conducted among patients with nonmetastatic HPV-associated (p16-positive) OPSCC. All patients were treated with curative-intent chemoradiotherapy (CRT). Patients underwent a 3-month post-CRT positron emission tomography/computed tomography scan and were thereafter clinically evaluated every 2-4 months (years 1-2), then every 6 months (years 3-5). Chest imaging was performed every 6 months. Blood specimens were collected every 6-9 months for analysis of plasma ctHPVDNA using a multianalyte digital polymerase chain reaction assay. The primary endpoint was to estimate the negative predictive value (NPV) and positive predictive value (PPV) of ctHPVDNA surveillance. RESULTS One hundred fifteen patients were enrolled, and 1,006 blood samples were analyzed. After a median follow-up time of 23 months (range, 6.1-54.7 months), 15 patients (13%) developed disease recurrence. Eighty-seven patients had undetectable ctHPVDNA at all post-treatment time points, and none developed recurrence (NPV, 100%; 95% CI, 96% to 100%). Twenty-eight patients developed a positive ctHPVDNA during post-treatment surveillance, 15 of whom were diagnosed with biopsy-proven recurrence. Sixteen patients had 2 consecutively positive ctHPVDNA blood tests, 15 of whom developed biopsy-proven recurrence. Two consecutively positive ctHPVDNA blood tests had a PPV of 94% (95% CI, 70% to 99%). Median lead time between ctHPVDNA positivity and biopsy-proven recurrence was 3.9 months (range, 0.37-12.9 months). CONCLUSION Detection of ctHPVDNA in two consecutive plasma samples during post-treatment surveillance has high PPV and NPV for identifying disease recurrence in patients with HPV-associated oropharyngeal cancer and may facilitate earlier initiation of salvage therapy.
Collapse
Affiliation(s)
- Bhishamjit S Chera
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, NC.,Lineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC
| | - Sunil Kumar
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, NC.,Lineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC
| | - Colette Shen
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, NC.,Lineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC
| | - Robert Amdur
- Department of Radiation Oncology, University of Florida Hospitals, Gainesville, FL
| | - Roi Dagan
- Department of Radiation Oncology, University of Florida Hospitals, Gainesville, FL
| | - Rebecca Green
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Emily Goldman
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jared Weiss
- Lineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC.,Department of Medicine, Division of Hematology Oncology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Juneko Grilley-Olson
- Lineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC.,Department of Medicine, Division of Hematology Oncology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Shetal Patel
- Lineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC.,Department of Medicine, Division of Hematology Oncology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Adam Zanation
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Trevor Hackman
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Jeff Blumberg
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Samip Patel
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Brian Thorp
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Mark Weissler
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Wendell Yarbrough
- Department of Medicine, Division of Hematology Oncology, University of North Carolina School of Medicine, Chapel Hill, NC.,Department of Pathology, University of North Carolina School of Medicine, Chapel Hill, NC
| | | | - William Mendenhall
- Department of Medicine, Division of Hematology Oncology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Xianming M Tan
- Lineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC
| | - Gaorav P Gupta
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, NC.,Lineberger Comprehensive Cancer Center, University of North Carolina Hospitals, Chapel Hill, NC
| |
Collapse
|
154
|
Osterman E, Glimelius B. ASO Author Reflections: Emerging Risk Factors in Colon Cancer-End of the Line for Clinomics? Ann Surg Oncol 2019; 27:1046-1047. [PMID: 31884560 PMCID: PMC7060154 DOI: 10.1245/s10434-019-08149-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Indexed: 11/18/2022]
Affiliation(s)
- Erik Osterman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden. .,Department of Surgery, Gävle Hospital, Gävle, Sweden.
| | - Bengt Glimelius
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
155
|
Xia S, Ye J, Chen Y, Lizaso A, Huang L, Shi L, Su J, Han-Zhang H, Chuai S, Li L, Chen Y. Parallel serial assessment of somatic mutation and methylation profile from circulating tumor DNA predicts treatment response and impending disease progression in osimertinib-treated lung adenocarcinoma patients. Transl Lung Cancer Res 2019; 8:1016-1028. [PMID: 32010579 DOI: 10.21037/tlcr.2019.12.09] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Circulating tumor DNA (ctDNA) harboring tumor-specific genetic and epigenetic aberrations allows for early detection and real-time monitoring of tumor dynamics. In this study, we aimed to evaluate the potential of parallel serial assessment of somatic mutation and methylation profile in monitoring the response to osimertinib of epidermal growth factor receptor (EGFR) T790M-positive advanced lung adenocarcinoma patients. Methods Parallel somatic mutation and DNA methylation profiling was performed on a total of 85 longitudinal plasma samples obtained from 8 stage IV osimertinib-treated EGFR T790M-positive lung adenocarcinoma patients. Results Our results revealed a significant correlation between the by-patient methylation level with the maximum allele fraction (maxAF, P=0.0002). The methylation levels were significantly higher in the plasma samples of patients with detectable somatic mutations than patients without somatic mutations (P=0.0003) and healthy controls (P=0.0018). Moreover, analysis of both the DNA methylation level and maxAF revealed four trends of treatment response. Collectively, the decrease in methylation level and maxAF reflected treatment efficacy, while the gradual increase reflected impending disease progression (PD). Elevated methylation levels and maxAF were observed in 6 and 5 patients in an average lead-time of 3.0 and 1.9 months, respectively, prior to evaluation of PD using radiological imaging. Conclusions DNA methylation profiling has the potential to predict disease relapse prior to evaluation through radiological modalities, suggesting that serial assessment of methylation level in combination with somatic mutation profiling are reliable methods for treatment monitoring. These methods should thus be incorporated with imaging modalities for a more comprehensive work-up of treatment response, particularly for patients treated with targeted therapies.
Collapse
Affiliation(s)
- Shu Xia
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Affiliated Hospital of Qinghai University, Xining 810000, China
| | - Junyi Ye
- Burning Rock Biotech, Guangzhou 510300, China
| | - Yu Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | | | - Le Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lei Shi
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Su
- Burning Rock Biotech, Guangzhou 510300, China
| | | | | | - Lingling Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuan Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
156
|
Liu Z, Xie Z, Zhao S, Ye D, Cai X, Cheng B, Li C, Xiong S, Li J, Liang H, Chen Z, Liang P, Liu J, He J, Liang W. Presence of allele frequency heterogeneity defined by ctDNA profiling predicts unfavorable overall survival of NSCLC. Transl Lung Cancer Res 2019; 8:1045-1050. [PMID: 32010581 DOI: 10.21037/tlcr.2019.12.10] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background The generation of subclonal (low-frequency) mutations is driven by tumor mutations and the relationship between the heterogeneity of tumor mutation abundance and non-small cell lung cancer (NSCLC) remains unknown. We investigate the role of allele frequency heterogeneity (AFH) defined by circulating tumor DNA (ctDNA) profiling in predicting prognosis in advanced NSCLC patients. Methods Publicly available data set of POPLAR (N=211) and OAK (N=642) trials were used for analyzing. A low ratio of allele frequency (AF) of a mutation to the maximum-somatic-allele-frequency (MSAF) was used to define the presence of AFH. The prognostic value of AF/MSAF ratio that was below a defined cutoff point in overall survival (OS) was evaluated using Cox-proportional hazards regression; and the structural break point was determined by LOESS regression and Chow test. The derived AFH was also explored in an independent cohort (N=259) of advanced NSCLC receiving first-line EGFR-TKIs from the First Affiliated Hospital of Guangzhou Medical University. Results In the POPLAR and OAK cohort, low AF/MSAF ratio was found to be significantly associated with unfavorable OS in univariate and multivariate analysis. The structural break point analysis demonstrated that AF/MSAF <10% could yield the optimal value to stratify patients with poor OS, which was applied for defining the presence of AFH. The presence of AFH significantly correlated with unfavorable OS in advanced NSCLC regardless of treatment arms (overall: HR 1.52, immunotherapy: HR 1.81, chemotherapy: HR 1.32, all P<0.05). In the exploratory EGFR-TKIs cohort, the presence of AFH was also significantly associated with shorter OS (HR 1.72, P=0.039). Conclusions Our results demonstrate that the presence of AFH predict unfavorable prognosis in advanced NSCLC despite which drug the patients used. The presence of AFH defined by ctDNA might provide an easily-accessible biomarker for risk stratification in the current clinical practice.
Collapse
Affiliation(s)
- Zhichao Liu
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China.,Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhanhong Xie
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Shen Zhao
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Dawei Ye
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiuyu Cai
- Department of General Internal Medicine, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Bo Cheng
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Caichen Li
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Shan Xiong
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Jianfu Li
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Hengrui Liang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Zisheng Chen
- Department of Respiratory Medicine, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| | - Peng Liang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Jun Liu
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| |
Collapse
|
157
|
Burke HB, Kopetz S. Is the Patient Cured? JAMA Oncol 2019; 5:1695-1697. [PMID: 31621795 DOI: 10.1001/jamaoncol.2019.3612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Harry B Burke
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
158
|
Tarazona N, Gimeno-Valiente F, Gambardella V, Zuñiga S, Rentero-Garrido P, Huerta M, Roselló S, Martinez-Ciarpaglini C, Carbonell-Asins JA, Carrasco F, Ferrer-Martínez A, Bruixola G, Fleitas T, Martín J, Tébar-Martínez R, Moro D, Castillo J, Espí A, Roda D, Cervantes A. Targeted next-generation sequencing of circulating-tumor DNA for tracking minimal residual disease in localized colon cancer. Ann Oncol 2019; 30:1804-1812. [PMID: 31562764 DOI: 10.1093/annonc/mdz390] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND A high percentage of patients diagnosed with localized colon cancer (CC) will relapse after curative treatment. Although pathological staging currently guides our treatment decisions, there are no biomarkers determining minimal residual disease (MRD) and patients are at risk of being undertreated or even overtreated with chemotherapy in this setting. Circulating-tumor DNA (ctDNA) can to be a useful tool to better detect risk of relapse. PATIENTS AND METHODS One hundred and fifty patients diagnosed with localized CC were prospectively enrolled in our study. Tumor tissue from those patients was sequenced by a custom-targeted next-generation sequencing (NGS) panel to characterize somatic mutations. A minimum variant allele frequency (VAF) of 5% was applied for variant filtering. Orthogonal droplet digital PCR (ddPCR) validation was carried out. We selected known variants with higher VAF to track ctDNA in the plasma samples by ddPCR. RESULTS NGS found known pathological mutations in 132 (88%) primary tumors. ddPCR showed high concordance with NGS (r = 0.77) for VAF in primary tumors. Detection of ctDNA after surgery and in serial plasma samples during follow-up were associated with poorer disease-free survival (DFS) [hazard ratio (HR), 17.56; log-rank P = 0.0014 and HR, 11.33; log-rank P = 0.0001, respectively]. Tracking at least two variants in plasma increased the ability to identify MRD to 87.5%. ctDNA was the only significantly independent predictor of DFS in multivariable analysis. In patients treated with adjuvant chemotherapy, presence of ctDNA after therapy was associated with early relapse (HR 10.02; log-rank P < 0.0001). Detection of ctDNA at follow-up preceded radiological recurrence with a median lead time of 11.5 months. CONCLUSIONS Plasma postoperative ctDNA detected MRD and identified patients at high risk of relapse in localized CC. Mutation tracking with more than one variant in serial plasma samples improved our accuracy in predicting MRD.
Collapse
Affiliation(s)
- N Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - F Gimeno-Valiente
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - V Gambardella
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - S Zuñiga
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Precision Medicine Unit, Valencia, Spain; Bioinformatics and Biostatistics Unit, Valencia, Spain
| | - P Rentero-Garrido
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Precision Medicine Unit, Valencia, Spain
| | - M Huerta
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - S Roselló
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - C Martinez-Ciarpaglini
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain; Departments of Pathology, Valencia, Spain
| | - J A Carbonell-Asins
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Precision Medicine Unit, Valencia, Spain; Bioinformatics and Biostatistics Unit, Valencia, Spain
| | - F Carrasco
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain; Precision Medicine Unit, Valencia, Spain
| | - A Ferrer-Martínez
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain; Precision Medicine Unit, Valencia, Spain
| | - G Bruixola
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - T Fleitas
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - J Martín
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain; Bioinformatics and Biostatistics Unit, Valencia, Spain
| | - R Tébar-Martínez
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Precision Medicine Unit, Valencia, Spain
| | - D Moro
- Departments of Surgery, INCLIVA Biomedical Research Institute, Valencia, Spain
| | - J Castillo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - A Espí
- Departments of Surgery, INCLIVA Biomedical Research Institute, Valencia, Spain
| | - D Roda
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - A Cervantes
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain.
| |
Collapse
|
159
|
Mas L, Bachet JB, Taly V, Bouché O, Taieb J, Cohen R, Meurisse A, Normand C, Gornet JM, Artru P, Louafi S, Thirot-Bidault A, Baumgaertner I, Coriat R, Tougeron D, Lecomte T, Mary F, Aparicio T, Marthey L, Blons H, Vernerey D, Laurent-Puig P. BRAF Mutation Status in Circulating Tumor DNA from Patients with Metastatic Colorectal Cancer: Extended Mutation Analysis from the AGEO RASANC Study. Cancers (Basel) 2019; 11:998. [PMID: 31319569 PMCID: PMC6679157 DOI: 10.3390/cancers11070998] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 01/25/2023] Open
Abstract
In patients with metastatic colorectal cancer (mCRC), RAS and BRAF mutations are currently determined by tumor sample analysis. Here, we report BRAF mutation status analysis in paired tumor tissue and plasma samples of mCRC patients included in the AGEO RASANC prospective cohort study. Four hundred and twenty-five patients were enrolled. Plasma samples were analyzed by next-generation sequencing (NGS). When no mutation was identified, we used two methylated specific biomarkers (digital droplet PCR) to determine the presence or absence of circulating tumor DNA (ctDNA). Patients with conclusive ctDNA results were defined as those with at least one mutation or one methylated biomarker. The kappa coefficient and accuracy were 0.79 (95% CI: 0.67-0.91) and 97.3% (95% CI: 95.2-98.6%) between the BRAF status in plasma and tissue for patients with available paired samples (n = 405), and 0.89 (95% CI: 0.80-0.99) and 98.5% (95% CI: 96.4-99.5%) for those with conclusive ctDNA (n = 323). The absence of liver metastasis was the main factor associated to inconclusive ctDNA results. In patients with liver metastasis, the kappa coefficient was 0.91 (95% CI, 0.81-1.00) and accuracy was 98.6% (95% CI, 96.5-99.6%). We demonstrate satisfying concordance between tissue and plasma BRAF mutation detection, especially in patients with liver metastasis, arguing for plasma ctDNA testing for routine BRAF mutation analysis in these patients.
Collapse
Affiliation(s)
- Leo Mas
- Department of Hepato-Gastroenterology, Groupe Hospitalier Pitié Salpêtrière, 75013 Paris, France
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
| | - Jean-Baptiste Bachet
- Department of Hepato-Gastroenterology, Groupe Hospitalier Pitié Salpêtrière, 75013 Paris, France
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Sorbonne Université, UPMC Université, 75006 Paris, France
- Université Sorbonne Paris Cité, INSERM UMR-S1147 MEPPOT, CNRS SNC5014, Centre Universitaire des Saints-Pères, Equipe labellisée Ligue Nationale Contre le Cancer, 75006 Paris, France
| | - Valerie Taly
- Université Sorbonne Paris Cité, INSERM UMR-S1147 MEPPOT, CNRS SNC5014, Centre Universitaire des Saints-Pères, Equipe labellisée Ligue Nationale Contre le Cancer, 75006 Paris, France
| | - Olivier Bouché
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Hepato-Gastroenterology, Hôpital Robert Debré, 51100 Reims, France
| | - Julien Taieb
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Oncology Digestive, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Romain Cohen
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Sorbonne Université, UPMC Université, 75006 Paris, France
- Department of Oncology, Hôpital Saint-Antoine, 75012 Paris, France
| | - Aurelia Meurisse
- Department of Methodology and Quality of Life in Oncology, INSERM UMR 1098, Hôpital Universitaire de Besancon, 25000 Besancon, France
| | - Corinne Normand
- Université Sorbonne Paris Cité, INSERM UMR-S1147 MEPPOT, CNRS SNC5014, Centre Universitaire des Saints-Pères, Equipe labellisée Ligue Nationale Contre le Cancer, 75006 Paris, France
| | - Jean-Marc Gornet
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Gastroenterology, Hôpital Saint-Louis, 75010 Paris, France
| | - Pascal Artru
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Gastroenterology, Hôpital Privé Jean Mermoz, 69008 Lyon, France
| | - Samy Louafi
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Gastroenterology, Centre Hospitalier Sud Francilien, 91100 Corbeil-Essonnes, France
- Department of Gastroenterology, Groupe Hospitalier Nord Essonne, 91160 Longjumeau, France
| | - Anne Thirot-Bidault
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Gastroenterology, Hôpital Kremlin Bicêtre, 94043 Le Kremlin-Bicêtre, France
| | - Isabelle Baumgaertner
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Oncology, Hôpital Henri Mondor, 94000 Créteil, France
| | - Romain Coriat
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Gastroenterology, Hôpital Cochin, 75014 Paris, France
| | - David Tougeron
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Depatment of Gastroenterology, Centre Hospitalo-Universitaire de Poitiers, 86000 Poitiers, France
| | - Thierry Lecomte
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Gastroenterology, Centre Hospitalo-Universitaire de Tours, 37000 Tours, France
| | - Florence Mary
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Gastroenterology, Hôpital Avicenne, 93000 Bobigny, France
| | - Thomas Aparicio
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Department of Gastroenterology, Hôpital Saint-Louis, 75010 Paris, France
- Department of Gastroenterology, Hôpital Avicenne, 93000 Bobigny, France
| | - Lysiane Marthey
- AGEO (Association des Gastroentérologues Oncologues), 75013 Paris, France
- Depatment of Gastroenterology, Hôpital Antoine Béclère, 92023 Clamart, France
| | - Helene Blons
- Université Sorbonne Paris Cité, INSERM UMR-S1147 MEPPOT, CNRS SNC5014, Centre Universitaire des Saints-Pères, Equipe labellisée Ligue Nationale Contre le Cancer, 75006 Paris, France
- Department of Biochemistry, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Dewi Vernerey
- Department of Methodology and Quality of Life in Oncology, INSERM UMR 1098, Hôpital Universitaire de Besancon, 25000 Besancon, France
| | - Pierre Laurent-Puig
- Université Sorbonne Paris Cité, INSERM UMR-S1147 MEPPOT, CNRS SNC5014, Centre Universitaire des Saints-Pères, Equipe labellisée Ligue Nationale Contre le Cancer, 75006 Paris, France.
| |
Collapse
|