51
|
Filis P, Kyrochristos I, Korakaki E, Baltagiannis EG, Thanos D, Roukos DH. Longitudinal ctDNA profiling in precision oncology and immunο-oncology. Drug Discov Today 2023; 28:103540. [PMID: 36822363 DOI: 10.1016/j.drudis.2023.103540] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/13/2022] [Accepted: 02/15/2023] [Indexed: 02/25/2023]
Abstract
Serial analysis of circulating tumor DNA (ctDNA) over the disease course is emerging as a prognostic, predictive and patient-monitoring biomarker. In the metastatic setting, several multigene ctDNA assays have been approved or recommended by regulatory organizations for personalized targeted therapy, especially for lung cancer. By contrast, in nonmetastatic disease, detection of ctDNA resulting from minimal residual disease (MRD) following multimodal treatment with curative intent presents major technical challenges. Several studies using tumor genotyping-informed serial ctDNA profiling have provided promising findings on the sensitivity and specificity of ctDNA in predicting the risk of recurrence. We discuss progress, limitations and future perspectives relating to the use of ctDNA as a biomarker to guide targeted therapy in metastatic disease, as well as the use of ctDNA MRD detection to guide adjuvant treatment in the nonmetastatic setting.
Collapse
Affiliation(s)
- Panagiotis Filis
- Centre for Biosystems and Genome Network Medicine, Ioannina University, 45110 Ioannina, Greece; Department of Medical Oncology, Medical School, University of Ioannina, 45110 Ioannina, Greece
| | - Ioannis Kyrochristos
- Centre for Biosystems and Genome Network Medicine, Ioannina University, 45110 Ioannina, Greece; Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, D-80539 Munich, Germany
| | - Efterpi Korakaki
- Centre for Biosystems and Genome Network Medicine, Ioannina University, 45110 Ioannina, Greece; Department of Physiology, Medical School, University of Ioannina, Ioannina 45110, Greece
| | - Evangelos G Baltagiannis
- Centre for Biosystems and Genome Network Medicine, Ioannina University, 45110 Ioannina, Greece; Department of Surgery, University Hospital of Ioannina, Ioannina 45500, Greece
| | - Dimitris Thanos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Dimitrios H Roukos
- Centre for Biosystems and Genome Network Medicine, Ioannina University, 45110 Ioannina, Greece; Department of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece.
| |
Collapse
|
52
|
de Abreu AR, Op de Beeck K, Laurent-Puig P, Taly V, Benhaim L. The Position of Circulating Tumor DNA in the Clinical Management of Colorectal Cancer. Cancers (Basel) 2023; 15:1284. [PMID: 36831626 PMCID: PMC9954551 DOI: 10.3390/cancers15041284] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer type worldwide, with over 1.9 million new cases and 935,000 related deaths in 2020. Within the next decade, the incidence of CRC is estimated to increase by 60% and the mortality by 80%. One of the underlying causes of poor prognosis is late detection, with 60 to 70% of the diagnoses occurring at advanced stages. Circulating cell-free DNA (ccfDNA) is probably the most promising tool for screening, diagnosis, prediction of therapeutic response, and prognosis. More specifically, the analysis of the tumor fraction within the ccfDNA (circulating tumor DNA, ctDNA) has great potential to improve the management of CRC. The present review provides an up-to-date and comprehensive overview of the various aspects related to ctDNA detection in CRC.
Collapse
Affiliation(s)
- Ana Regina de Abreu
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
- Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Ken Op de Beeck
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
- Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Pierre Laurent-Puig
- UMR-S1138, CNRS SNC5096, Équipe labélisée Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris, 75006 Paris, France
| | - Valerie Taly
- UMR-S1138, CNRS SNC5096, Équipe labélisée Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris, 75006 Paris, France
| | - Leonor Benhaim
- UMR-S1138, CNRS SNC5096, Équipe labélisée Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris, 75006 Paris, France
- Department of Visceral and Surgical Oncology, Gustave Roussy, Cancer Campus, 114 rue Edouard Vaillant, 94805 Villejuif, France
| |
Collapse
|
53
|
Puccini A, Martelli V, Pastorino A, Sciallero S, Sobrero A. ctDNA to Guide Treatment of Colorectal Cancer: Ready for Standard of Care? Curr Treat Options Oncol 2023; 24:76-92. [PMID: 36656505 DOI: 10.1007/s11864-022-01048-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2022] [Indexed: 01/20/2023]
Abstract
OPINION STATEMENT Circulating tumor DNA (ctDNA) has already shown clinically relevant results in early-stage colon cancer patient management. Its prognostic value is by far much stronger than that of the available clinico-pathological biomarkers, therefore, has the potential to personalize the treatment after radical surgery through intensifying or de-intensifying the adjuvant therapy. Further developments and improvements should be pursued by (a) optimizing ctDNA assays and (b) validating its clinical utility in the different stages of this disease. Two main avenues of ctDNA testing are being pursued: tumor-informed vs tumor-agnostic assays. Two main clinical trial designs are under study: ctDNA-based strategy and ctDNA-by-treatment interaction. The former needs large sample sizes to address the main questions of the studies; thus, the target delta benefit may be the main challenge in these trial designs. The latter may be challenged by unavoidable contamination bias. To date, several clinical trials are ongoing worldwide. We believe that this large number of trials may provide an excellent common database for the demonstration of surrogacy of ctDNA for the classical 3-year disease-free survival endpoint. This would mark a huge methodological improvement to speed up new drug testing and development in the adjuvant treatment of this disease.
Collapse
Affiliation(s)
- Alberto Puccini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, 16132, Genoa, Italy
| | - Valentino Martelli
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, 16132, Genoa, Italy
| | - Alessandro Pastorino
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Stefania Sciallero
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Alberto Sobrero
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy.
| |
Collapse
|
54
|
Vellanki PJ, Ghosh S, Pathak A, Fusco MJ, Bloomquist EW, Tang S, Singh H, Philip R, Pazdur R, Beaver JA. Regulatory implications of ctDNA in immuno-oncology for solid tumors. J Immunother Cancer 2023; 11:e005344. [PMID: 36796877 PMCID: PMC9936292 DOI: 10.1136/jitc-2022-005344] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 02/18/2023] Open
Abstract
In the era of precision oncology, use of circulating tumor DNA (ctDNA) is emerging as a minimally invasive approach for the diagnosis and management of patients with cancer and as an enrichment tool in clinical trials. In recent years, the US Food and Drug Administration has approved multiple ctDNA-based companion diagnostic assays for the safe and effective use of targeted therapies and ctDNA-based assays are also being developed for use with immuno-oncology-based therapies. For early-stage solid tumor cancers, ctDNA may be particularly important to detect molecular residual disease (MRD) to support early implementation of adjuvant or escalated therapy to prevent development of metastatic disease. Clinical trials are also increasingly using ctDNA MRD for patient selection and stratification, with an ultimate goal of improving trial efficiency through use of an enriched patient population. Standardization and harmonization of ctDNA assays and methodologies, along with further clinical validation of ctDNA as a prognostic and predictive biomarker, are necessary before ctDNA may be considered as an efficacy-response biomarker to support regulatory decision making.
Collapse
Affiliation(s)
- Paz J Vellanki
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Soma Ghosh
- Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Anand Pathak
- Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Michael J Fusco
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Erik W Bloomquist
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Shenghui Tang
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Harpreet Singh
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Reena Philip
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Richard Pazdur
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Julia A Beaver
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
55
|
Gao Z, Huang D, Chen H, Yang Y, An K, Ding C, Yuan Z, Zhai Z, Niu P, Gao Q, Cai J, Zeng Q, Wang Y, Hong Y, Rong W, Huang W, Lei F, Wang X, Chen S, Zhao X, Bai Y, Gu J. Development and validation of postoperative circulating tumor DNA combined with clinicopathological risk factors for recurrence prediction in patients with stage I-III colorectal cancer. J Transl Med 2023; 21:63. [PMID: 36717891 PMCID: PMC9887832 DOI: 10.1186/s12967-023-03884-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) detection following curative-intent surgery could directly reflect the presence of minimal residual disease, the ultimate cause of clinical recurrence. However, ctDNA is not postoperatively detected in ≥ 50% of patients with stage I-III colorectal cancer (CRC) who ultimately recur. Herein we sought to improve recurrence risk prediction by combining ctDNA with clinicopathological risk factors in stage I-III CRC. METHODS Two independent cohorts, both consisting of early-stage CRC patients who underwent curative surgery, were included: (i) the discovery cohort (N = 124) with tumor tissues and postoperative plasmas for ctDNA determination; and (ii) the external validation cohort (N = 125) with available ctDNA results. In the discovery cohort, somatic variations in tumor tissues and plasmas were determined via a 733-gene and 127-gene next-generation sequencing panel, respectively. RESULTS In the discovery cohort, 17 of 108 (15.7%) patients had detectable ctDNA. ctDNA-positive patients had a significantly high recurrence rate (76.5% vs. 16.5%, P < 0.001) and short recurrence-free survival (RFS; P < 0.001) versus ctDNA-negative patients. In addition to ctDNA status, the univariate Cox model identified pathologic stage, lymphovascular invasion, nerve invasion, and preoperative carcinoembryonic antigen level associated with RFS. We combined the ctDNA and clinicopathological risk factors (CTCP) to construct a model for recurrence prediction. A significantly higher recurrence rate (64.7% vs. 8.1%, P < 0.001) and worse RFS (P < 0.001) were seen in the high-risk patients classified by the CTCP model versus those in the low-risk patients. Receiver operating characteristic analysis demonstrated that the CTCP model outperformed ctDNA alone at recurrence prediction, which increased the sensitivity of 2 year RFS from 49.6% by ctDNA alone to 87.5%. Harrell's concordance index, calibration curve, and decision curve analysis also suggested that the CTCP model had good discrimination, consistency, and clinical utility. These results were reproduced in the validation cohort. CONCLUSION Combining postoperative ctDNA and clinical risk may better predict recurrence than ctDNA alone for developing a personalized postoperative management strategy for CRC.
Collapse
Affiliation(s)
- Zhaoya Gao
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Dandan Huang
- grid.452694.80000 0004 0644 5625Department of Oncology, Peking University Shougang Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Hui Chen
- Medical Affairs, 3D Medicines, Inc., Shanghai, China
| | - Yong Yang
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Ke An
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Changmin Ding
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Zheping Yuan
- Medical Affairs, 3D Medicines, Inc., Shanghai, China
| | - Zhichao Zhai
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Pengfei Niu
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Qingkun Gao
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Jinping Cai
- Medical Affairs, 3D Medicines, Inc., Shanghai, China
| | - Qingmin Zeng
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Yanzhao Wang
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Yuming Hong
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Wanshui Rong
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Wensheng Huang
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Fuming Lei
- grid.452694.80000 0004 0644 5625Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Xiaodong Wang
- grid.452694.80000 0004 0644 5625Department of Oncology, Peking University Shougang Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China
| | - Shiqing Chen
- Medical Affairs, 3D Medicines, Inc., Shanghai, China
| | - Xiaochen Zhao
- Medical Affairs, 3D Medicines, Inc., Shanghai, China
| | - Yuezong Bai
- Medical Affairs, 3D Medicines, Inc., Shanghai, China
| | - Jin Gu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No.9 Jinyuanzhuang Road, Shijingshan District, Beijing, China. .,Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center, Beijing, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China. .,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China. .,Peking University International Cancer Institute, Beijing, China.
| |
Collapse
|
56
|
Chan HT, Nagayama S, Otaki M, Chin YM, Fukunaga Y, Ueno M, Nakamura Y, Low SK. Tumor-informed or tumor-agnostic circulating tumor DNA as a biomarker for risk of recurrence in resected colorectal cancer patients. Front Oncol 2023; 12:1055968. [PMID: 36776372 PMCID: PMC9909342 DOI: 10.3389/fonc.2022.1055968] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/30/2022] [Indexed: 01/28/2023] Open
Abstract
Introduction Circulating tumor DNA (ctDNA) has been increasingly recognized as a promising minimally-invasive biomarker that could identify patients with minimal residual disease and a high risk of recurrence after definitive treatment. In this study, we've compared the clinical utility and sensitivity of 2 different approaches to ctDNA analyses: tumor-informed and tumor-agnostic in the management of colorectal (CRC) patients. The clinical benefits of a single timepoint ctDNA analysis compared to serial ctDNA monitoring after definitive treatment were also evaluated to uncover the ideal surveillance protocol. Methods Patient-paired resected tumor tissues, peripheral blood cells, and a total of 127 pre-operative and serial plasma cell-free DNA (cfDNA) samples after definitive treatment from 38 CRC patients that had undergone curative intent surgery were analyzed using a commercial NGS cfDNA panel. Results Up to 84% (32/38) of the recruited patients were detected with at least 1 genomic alteration from the tumor tissues that could be monitored using the tumor-informed ctDNA approach and none of the detected alterations were clonal hematopoiesis (CH) related. In contrast, 37% (14/38) of patients were detected with at least 1 monitoring alteration after exclusion of CH mutations using the tumor-agnostic approach. Serial plasma samples after definitive therapy were available for 31 patients. In the landmark ctDNA analysis, 24% (7/29) of patients had detectable ctDNA and were more likely to relapse than ctDNA-negative patients (p < 0.05). The landmark analysis sensitivity and specificity for recurrence were 67% and 87%, respectively. The incorporation of longitudinal ctDNA analysis at 6-months intervals improved the sensitivity to 100%. The median variant allele frequency (VAF) of the ctDNA mutations detected during surveillance was 0.028% (range: 0.018-0.783), where up to 80% (8/10) of the mutations were detected at VAF lower than the tumor-agnostic detection limit of 0.1%. Utilizing the tumor-agnostic approach reduced the recurrence detection sensitivity to 67% (4/6). Serial ctDNA analyses predicted disease recurrence at a median of 5 months ahead of radiological imaging. Conclusion Longitudinal monitoring using tumor-informed ctDNA testing shows high analytical sensitivity, low probability of false-positive results due to CH mutations, and improved sensitivity in detecting recurrence which may modify the clinical management of CRC.
Collapse
Affiliation(s)
- Hiu Ting Chan
- Project for Development of Liquid Biopsy Diagnosis, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Nagayama
- Department of Gastroenterological and Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan,Department of Surgery, Uji-Tokushukai Medical Center, Kyoto, Japan
| | - Masumi Otaki
- Department of Medical Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan,Department of Clinical Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoon Ming Chin
- Project for Development of Liquid Biopsy Diagnosis, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan,Department of Research and Development, Cancer Precision Medicine, Inc., Kawasaki, Japan
| | - Yosuke Fukunaga
- Department of Gastroenterological and Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masashi Ueno
- Department of Gastroenterological and Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yusuke Nakamura
- Project for Development of Liquid Biopsy Diagnosis, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan,National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Siew-Kee Low
- Project for Development of Liquid Biopsy Diagnosis, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan,*Correspondence: Siew-Kee Low,
| |
Collapse
|
57
|
Romesser PB, Sanchez-Vega F, Joshua Smith J. A methylation-based prognostic signature in stage II colorectal patients: Considerations for clinical adoption. J Natl Cancer Inst 2023; 115:8-11. [PMID: 36171662 PMCID: PMC9830476 DOI: 10.1093/jnci/djac184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 01/13/2023] Open
Affiliation(s)
- Paul B Romesser
- Colorectal Anal Cancer Service, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francisco Sanchez-Vega
- Colorectal Cancer Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - J Joshua Smith
- Colorectal Cancer Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
58
|
Wang Y, Fan X, Bao H, Xia F, Wan J, Shen L, Wang Y, Zhang H, Wei Y, Wu X, Shao Y, Li X, Xu Y, Cai S, Zhang Z. Utility of Circulating Free DNA Fragmentomics in the Prediction of Pathological Response after Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer. Clin Chem 2023; 69:88-99. [PMID: 36308331 DOI: 10.1093/clinchem/hvac173] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 08/25/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND A "Watch and Wait" (W&W) approach has become an alternative to surgery for locally advanced rectal cancer (LARC) after neoadjuvant chemoradiotherapy (nCRT). Precise prediction of pathological complete response (pCR) will improve patient selection for W&W. We investigated the utility of cell-free DNA (cfDNA) fragmentomics in predicting pCR. METHODS We recruited 119 LARC patients and evaluated nCRT response by pCR status and pathological or MRI tumor regression grade (mrTRG). Plasma samples before, during, and after nCRT were applied to deep targeted-panel sequencing, with 103 patients having complete samples. cfDNA fragment and 5'-end motif profiles were used to construct elastic-net logistic regression models to predict non-pCR. Predictive performance was measured by area under the receiver operator characteristic curve (AUC), sensitivity, and specificity. RESULTS In the training cohort, the model based on 5'-end motif profile plus mrTRG achieved the highest cross-validation AUC (0.92, 95% CI, 0.91-0.93). The AUC in a testing cohort was 0.96 (95% CI, 0.90-1.00). The models based on 5'-end motif profile alone or in combination with mrTRG both maintained good predictive ability for patients without detectable circulating tumor DNA (AUC 0.94, 95% CI, 0.93-0.95; AUC 0.95, 95% CI, 0.94-0.96). In an external validation cohort, the model trained with a local 5'-end motif profile obtained an AUC of 0.878 (95% CI, 0.801-0.956) in discriminating colorectal cancer from healthy subjects. CONCLUSIONS The combination of a 5'-end motif profile with mrTRG has the potential to predict the response to nCRT, and therefore may improve the patient selection for a W&W approach.
Collapse
Affiliation(s)
- Yaqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Xiaojun Fan
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Juefeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Yan Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Yulin Wei
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China.,School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| |
Collapse
|
59
|
Ko B, Hanna M, Yu M, Grady WM. Epigenetic Alterations in Colorectal Cancer. EPIGENETICS AND HUMAN HEALTH 2023:331-361. [DOI: 10.1007/978-3-031-42365-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
60
|
Dawood ZS, Alaimo L, Lima HA, Moazzam Z, Shaikh C, Ahmed AS, Munir MM, Endo Y, Pawlik TM. Circulating Tumor DNA, Imaging, and Carcinoembryonic Antigen: Comparison of Surveillance Strategies Among Patients Who Underwent Resection of Colorectal Cancer-A Systematic Review and Meta-analysis. Ann Surg Oncol 2023; 30:259-274. [PMID: 36219278 DOI: 10.1245/s10434-022-12641-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/22/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Almost one-third of colorectal cancer (CRC) patients experience recurrence after resection; nevertheless, follow-up strategies remain controversial. We sought to systematically assess and compare the accuracy of carcinoembryonic antigen (CEA), imaging [positron emission tomography (PET) and computed tomography (CT) scans], and circulating tumor DNA (CtDNA) as surveillance strategies. PATIENTS AND METHODS PubMed, Medline, Embase, Scopus, Cochrane, Web of Science, and CINAHL were systematically searched. The Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) was used to assess methodological quality. We performed a bivariate random-effects meta-analysis and reported pooled sensitivity, specificity, and diagnostic odds ratio (DOR) values for each surveillance strategy. RESULTS Thirty studies were included in the analysis. PET scans had the highest sensitivity to detect recurrence (0.95; 95%CI 0.91-0.97), followed by CT scans (0.77; 95%CI 0.67-0.85). CtDNA positivity had the highest specificity to detect recurrence (0.95; 95%CI 0.91-0.97), followed by increased CEA levels (0.88; 95%CI 0.82-0.92). Furthermore, PET scans had the highest DOR to detect recurrence (DOR 120.7; 95%CI 48.9-297.9) followed by CtDNA (DOR 37.6; 95%CI 20.8-68.0). CONCLUSION PET scans had the highest sensitivity and DOR to detect recurrence, while CtDNA had the highest specificity and second highest DOR. Combinations of traditional cross-sectional/functional imaging and newer platforms such as CtDNA may result in optimized surveillance of patients following resection of CRC.
Collapse
Affiliation(s)
- Zaiba Shafik Dawood
- Medical College, The Aga Khan University Hospital, Stadium Road, Karachi, 74800, Pakistan
| | - Laura Alaimo
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Henrique A Lima
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Zorays Moazzam
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Chanza Shaikh
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Muhammad Musaab Munir
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Yutaka Endo
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
61
|
Baraniskin A, Baba HA, Theegarten D, Mika T, Schroers R, Klein-Scory S. Liquid biopsy can cure early colorectal cancer recurrence - Case Report. Front Oncol 2023; 13:1141833. [PMID: 37207159 PMCID: PMC10188995 DOI: 10.3389/fonc.2023.1141833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023] Open
Abstract
In the context of colorectal cancer (CRC), circulating tumor DNA (ctDNA) is frequently used to monitor the minimal residual disease (MRD). ctDNA has become an excellent biomarker to predict which patients with CRC are likely to relapse due to the persistence of micrometastases. MRD diagnosis via analysis of ctDNA may allow much earlier detection of relapse compared with conventional diagnosis during follow-up. It should lead to an increased rate of curative-intended complete resection of an asymptomatic relapse. Besides, ctDNA can provide crucial information on whether and how intensively adjuvant or additive therapy should be administered. In the present case, analysis of ctDNA gave us a crucial hint to the use of more intensive diagnostics (MRI and Positron emission tomography-computed tomography PET-CT) which led to earlier detection of CRC relapse. Metastasis detected early are more likely to be completely resectable with curative intent.
Collapse
Affiliation(s)
- Alexander Baraniskin
- Department of Hematology and Oncology, Evangelisches Krankenhaus Hamm, Hamm, Germany
- Department of Medicine, Hematology and Oncology, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University of Bochum, Bochum, Germany
- *Correspondence: Alexander Baraniskin,
| | - Hideo A. Baba
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Dirk Theegarten
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thomas Mika
- Department of Medicine, Hematology and Oncology, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University of Bochum, Bochum, Germany
| | - Roland Schroers
- Department of Medicine, Hematology and Oncology, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University of Bochum, Bochum, Germany
| | - Susanne Klein-Scory
- Department of Medicine, Hematology and Oncology, University Hospital Knappschaftskrankenhaus Bochum GmbH, Ruhr University of Bochum, Bochum, Germany
| |
Collapse
|
62
|
Molecular residual disease and efficacy of adjuvant chemotherapy in patients with colorectal cancer. Nat Med 2023; 29:127-134. [PMID: 36646802 PMCID: PMC9873552 DOI: 10.1038/s41591-022-02115-4] [Citation(s) in RCA: 232] [Impact Index Per Article: 116.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/01/2022] [Indexed: 01/18/2023]
Abstract
Despite standard-of-care treatment, more than 30% of patients with resectable colorectal cancer (CRC) relapse. Circulating tumor DNA (ctDNA) analysis may enable postsurgical risk stratification and adjuvant chemotherapy (ACT) treatment decision-making. We report results from GALAXY, which is an observational arm of the ongoing CIRCULATE-Japan study (UMIN000039205) that analyzed presurgical and postsurgical ctDNA in patients with stage II-IV resectable CRC (n = 1,039). In this cohort, with a median follow-up of 16.74 months (range 0.49-24.83 months), postsurgical ctDNA positivity (at 4 weeks after surgery) was associated with higher recurrence risk (hazard ratio (HR) 10.0, P < 0.0001) and was the most significant prognostic factor associated with recurrence risk in patients with stage II or III CRC (HR 10.82, P < 0.001). Furthermore, postsurgical ctDNA positivity identified patients with stage II or III CRC who derived benefit from ACT (HR 6.59, P < 0.0001). The results of our study, a large and comprehensive prospective analysis of ctDNA in resectable CRC, support the use of ctDNA testing to identify patients who are at increased risk of recurrence and are likely to benefit from ACT.
Collapse
|
63
|
Solar Vasconcelos JP, Boutin M, Loree JM. Circulating tumor DNA in early-stage colon cancer: ready for prime time or needing refinement? Ther Adv Med Oncol 2022; 14:17588359221143975. [PMID: 36570410 PMCID: PMC9772953 DOI: 10.1177/17588359221143975] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022] Open
Abstract
Liquid biopsies are the detection of molecular information in fluids from patients with cancer. In colorectal cancer (CRC), the most promising liquid biopsy strategy is the use of circulating tumor DNA (ctDNA) from plasma. In early-stage CRC, the potential for ctDNA to impact care stems from the detection of minimal residual disease (MRD) to guide adjuvant therapy after curative intent treatment and in identifying recurrences during surveillance. As for any new diagnostic test, ctDNA assays must overcome pre-analytical and analytical challenges before clinical implementation. We will discuss important logistical and assay considerations that clinicians and patients should understand when assessing ctDNA assays. We will also delve into important concepts to aid in interpreting ctDNA results and potential incidental findings that may arise. Sequencing errors, germline variants, and clonal hematopoiesis of indeterminate potential (CHIP) must be addressed to properly interpret results. CHIP is also an important consideration that impacts patient prognosis through association with cardiovascular and hematologic diseases. With this background in place, we next review the best available evidence for the use of ctDNA in early-stage colon cancer. Observational cohorts have established MRD after surgery as a significant prognostic factor for recurrence in stage II and III colon cancer. It also has the ability to anticipate clinical recurrence before standard investigations when used in surveillance. The first and only interventional randomized trial to date evaluating ctDNA is DYNAMIC. The study demonstrated the noninferiority of a MRD detection-guided approach in selecting patients with stage II colon cancer for adjuvant treatment. Notwithstanding the important results, there are still important questions to be answered before ctDNA enters prime time in the clinic. However, future appears bright and ongoing trials will help clarify how to best use this technology in early-stage colon cancer.
Collapse
Affiliation(s)
| | - Melina Boutin
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
64
|
Alden SL, Dhani H, Palsuledesai CC, Krinshpun S, Jurdi A, Christenson E, Browner I, Rosner S. Post-operative ctDNA monitoring in stage I colon cancer: A case report. Front Oncol 2022; 12:1074786. [PMID: 36591529 PMCID: PMC9795219 DOI: 10.3389/fonc.2022.1074786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Circulating tumor DNA (ctDNA) level monitoring after surgery for colon cancer has been studied in stage II and III colon cancer to risk-stratify patients for adjuvant therapy. However, there is less data regarding the role of this diagnostic tool in the management of stage I disease, where current recommended surveillance is limited to screening colonoscopy at one year. In this report, we describe the case of a 57-year-old man with stage I colon cancer who underwent complete resection with adequate lymph node surgical sampling, normal preoperative CEA and no evidence of metastatic disease on initial imaging. The patient elected to undergo serial ctDNA monitoring after surgery. Rising ctDNA levels, five months after resection, prompted cross-sectional imaging which demonstrated metastatic disease to the liver. The patient subsequently received five cycles of leucovorin, 5-fluorouracil, oxaliplatin, and irinotecan with bevacizumab (FOLFOXIRI-Bev) and definitive microwave ablation to the liver metastases, with resulting undetectable ctDNA levels. The patient's imaging and colonoscopy one-year post-operatively showed no evidence of disease, with ctDNA levels remaining undetectable. This report highlights the value of ctDNA monitoring in patients with early-stage colon cancer and suggests that further, large-scale studies may be warranted to determine its appropriate clinical use.
Collapse
Affiliation(s)
- Stephanie L. Alden
- Johns Hopkins Hospital, Department of Medicine, Baltimore, MD, United States
| | | | | | | | | | - Eric Christenson
- Medical Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Ilene Browner
- Medical Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Samuel Rosner
- Medical Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
65
|
Mi J, Han X, Wang R, Ma R, Zhao D. Circulation tumour DNA in predicting recurrence and prognosis in operable colorectal cancer patients: A meta-analysis. Eur J Clin Invest 2022; 52:e13842. [PMID: 35856399 DOI: 10.1111/eci.13842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/14/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Selecting the appropriate patient for further treatment after surgery and adjuvant chemotherapy (ACT) for colorectal cancer (CRC) can improve the patient's prognosis. Circulating tumour DNA (ctDNA) has the potential to predict recurrence and prognosis after CRC surgery and ACT, but the results are still inconclusive. OBJECTIVES As the completed studies have small sample sizes and different experimental methods, a meta-analysis was conducted to assess the ctDNA on recurrence and prognosis after CRC surgery and ACT. METHODS PubMed, Embase, the Web of Science and the Cochrane Library were searched for potentially eligible studies published up to 6 March 2022. Pooled relative risk (RR) and pooled hazard ratio (HR) were calculated to evaluate recurrence and the prognosis of recurrence-free survival (RFS) following CRC surgery and ACT. RESULTS Fourteen studies published between 2014 and 2022 included 2393 patients, and 7189 serum samples were eventually included in the meta-analysis. The pooled revealed that ctDNA-positive patients were at high risk of recurrence after CRC surgery (RR = 4.43, 95% CI: 3.58-5.48, p < .05) and had a poorer prognosis for RFS (HR = 7.26, 95% CI: 5.48-9.62, p < .05). The pooled revealed that ctDNA-positive patients were at high risk of recurrence after ACT (RR = 5.77 95% CI: 4.33-7.69, p < .05) and had a poorer prognosis for RFS (HR = 13.96, 95% CI: 8.71-22.4, p < .05). CONCLUSION ctDNA-positive patients were at a high risk of recurrence after CRC surgery and ACT and had a poorer prognosis. Hence, ctDNA-positive patients required close follow-up and further treatments.
Collapse
Affiliation(s)
- Junjie Mi
- Digestive Endoscopy Center, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Xiaofang Han
- Reproductive Medicine, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Rong Wang
- Digestive Endoscopy Center, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Ruijun Ma
- Digestive Endoscopy Center, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Danyu Zhao
- Digestive Endoscopy Center, Shanxi Provincial People's Hospital, Taiyuan, China
| |
Collapse
|
66
|
Grancher A, Beaussire L, Manfredi S, Le Malicot K, Dutherage M, Verdier V, Mulot C, Bouché O, Phelip JM, Levaché CB, Deguiral P, Coutant S, Sefrioui D, Emile JF, Laurent-Puig P, Bibeau F, Michel P, Sarafan-Vasseur N, Lepage C, Di Fiore F. Postoperative circulating tumor DNA detection is associated with the risk of recurrence in patients resected for a stage II colorectal cancer. Front Oncol 2022; 12:973167. [PMID: 36439476 PMCID: PMC9685416 DOI: 10.3389/fonc.2022.973167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
Circulating tumor DNA (ctDNA) is reported to be promising in localized colorectal cancer (CRC). The present study aimed to retrospectively evaluate the impact of ctDNA in patients with a resected stage II CRC from the PROGIGE 13 trial with available paired tumor and blood samples. A group of recurrent patients were matched one-to-one with nonrecurrent patients according to sex, tumor location, treatment sequence, and blood collection timing. CtDNA was analyzed by digital PCR according to NGS of tumors. Disease-free survival (DFS) and overall survival (OS) were analyzed based on ctDNA, and the risks of recurrence and death were determined. A total of 134 patients were included, with 67 patients in each group. At least one alteration was identified in 115/134 tumors. Postoperative ctDNA was detected in 10/111 (9.0%) informative samples and was detected more frequently in the recurrent group (16.7% versus 1.8%; p = 0.02). The median DFS of ctDNA+ versus ctDNA- patients was 16.8 versus 54 months (p = 0.002), respectively, and the median OS was 51.3 versus 69.5 months (p = 0.03), respectively. CtDNA was associated with recurrence (ORa = 11.13, p = 0.03) and death (HRa = 3.15, p = 0.01). In conclusion, the presence of postoperative ctDNA is associated with both recurrence and survival in stage II CRC.
Collapse
Affiliation(s)
- Adrien Grancher
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
- *Correspondence: Adrien Grancher,
| | - Ludivine Beaussire
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
- Department of Medical Oncology, Henri Becquerel Centre, Rouen, Rouen, France
| | - Sylvain Manfredi
- Burgundy Digestive Cancer Registry, INSERM, Lipides, Nutrition, Cancers (LNC)-UMR1231, University Bourgogne Franche-Comté, Dijon, France
| | - Karine Le Malicot
- Burgundy Digestive Cancer Registry, INSERM, Lipides, Nutrition, Cancers (LNC)-UMR1231, University Bourgogne Franche-Comté, Dijon, France
| | - Marie Dutherage
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Vincent Verdier
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Claire Mulot
- Paris University, Biology Resources Center EPIGENETEC, Paris, France
| | - Olivier Bouché
- Department of Digestive Oncology, University Hospital of Reims, Reims, France
| | - Jean-Marc Phelip
- Department of Gastroenterology and Digestive Oncology, University Hospital of Saint Etienne, Saint Etienne, France
| | - Charles-Briac Levaché
- Department of Radiotherapy and Medical Oncology, Polyclinique Francheville, Périgueux, France
| | - Philippe Deguiral
- Department of Gastroenterology, St Nazaire Hospital, Saint-Nazaire, France
| | - Sophie Coutant
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - David Sefrioui
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Jean-François Emile
- Department of Pathology, Hôpital Ambroise-Paré, Boulogne-Billancourt, France
| | - Pierre Laurent-Puig
- Department of Biology, Georges Pompidou Hospital, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Frédéric Bibeau
- Department of Pathology, Caen University Hospital, Caen, France
| | - Pierre Michel
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Nasrin Sarafan-Vasseur
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Côme Lepage
- Burgundy Digestive Cancer Registry, INSERM, Lipides, Nutrition, Cancers (LNC)-UMR1231, University Bourgogne Franche-Comté, Dijon, France
| | - Frederic Di Fiore
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
- Department of Medical Oncology, Henri Becquerel Centre, Rouen, Rouen, France
| |
Collapse
|
67
|
Morris VK, Overman MJ, Lam M, Parseghian CM, Johnson B, Dasari A, Raghav K, Kee BK, Huey R, Wolff RA, Shen JP, Li J, Zorrilla I, Tzeng CWD, Tran Cao HS, Chun YS, Newhook TE, Vauthey N, Duose D, Luthra R, Haymaker C, Kopetz S. Bintrafusp alfa, an anti-PD-L1:TGF-β trap fusion protein, in patients with ctDNA-positive, liver-limited metastatic colorectal cancer. CANCER RESEARCH COMMUNICATIONS 2022; 2:979-986. [PMID: 36382087 PMCID: PMC9648419 DOI: 10.1158/2767-9764.crc-22-0194] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
BACKGROUND Identification of circulating tumor DNA (ctDNA) following curative intent therapies is a surrogate for microscopic residual disease for patients with metastatic colorectal cancer (mCRC). Preclinically, in micrometastatic microsatellite stable (MSS) CRC, increased TGF-β signaling results in exclusion of anti-tumor cytotoxic T cells from the tumor microenvironment. Bintrafusp alfa (BA) is a bifunctional fusion protein composed of the extracellular domain of the TGF-βRII receptor ("TGF-β trap") and anti-PD-L1 antibody. METHODS Patients with liver-limited, MSS mCRC and with detected ctDNA after complete resection of all known tumors and standard-of-care therapy were treated with 1200 mg of BA intravenously every 14 days for six doses. The primary endpoint was ctDNA clearance. Radiographic characteristics at recurrence were compared using independent t-tests to historical data from a similar cohort of patients with liver-limited mCRC who underwent observation. RESULTS Only 4 of 15 planned patients received BA before the study was stopped early for loss of equipoise. There was no grade ≥3 AE. None of the patients cleared ctDNA. All patients developed radiographic recurrence by the first planned restaging. Although not detectable at prior to treatment, TGFβ3 was found in circulation in all patients at cycle 2 day 1. Compared to a historical cohort, patients administered BA developed more metastases (15 versus 2, p=0.005) and greater tumor volumes (9 cm vs 2 cm, p=0.05). CONCLUSIONS Treatment with BA in patients with ctDNA-detected, liver-limited mCRC did not clear ctDNA and was associated with large-volume recurrence, highlighting the potential context-specific complexity of dual TGF-β and PD-L1 inhibition.
Collapse
Affiliation(s)
- Van K. Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Corresponding Author: Van K. Morris, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 426, Houston, TX 77030. Phone 713-792-2828. E-mail:
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Lam
- Department of Medical Oncology and Cancer Services, The University of Melbourne, Melbourne, Australia
| | - Christine M. Parseghian
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Benny Johnson
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bryan K. Kee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ryan Huey
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - June Li
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Isabel Zorrilla
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ching-Wei D. Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hop S. Tran Cao
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yun Shin Chun
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy E. Newhook
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dzifa Duose
- Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raja Luthra
- Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cara Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
68
|
Liu GY, Xia WX, Bi ZF, Lu N, Li WZ, Bei WX, Liang H, Xie JZ, Liu YM, Yao HR, Xiang YQ. Plasma Circulating Tumor Epstein-Barr Virus for the Surveillance of Cancer Progression in Bone-Only Metastatic Nasopharyngeal Carcinoma. Front Oncol 2022; 12:860700. [PMID: 35756638 PMCID: PMC9226300 DOI: 10.3389/fonc.2022.860700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/18/2022] [Indexed: 11/24/2022] Open
Abstract
Background Plasma Epstein–Barr virus DNA (EBV-DNA) is a sensitive and specific biomarker for nasopharyngeal carcinoma (NPC). We investigated whether longitudinal monitoring of EBV-DNA could accurately detect clinical disease progression in NPC patients with bone-only metastases. Methods In this retrospective study, a total of 105 patients with bone-only metastatic NPC who were treated with platinum-based first-line chemotherapy were enrolled. Undetectable EBV-DNA after first-line chemotherapy was defined as a biochemical complete response (BCR). The correlation of the EBV-DNA dynamic status with overall survival (OS) and progression-free survival (PFS) was determined by Cox regression. The correlation between non-normalized EBV-DNA period and PFS period was determined. Results After a median follow-up time of 53.4 months [Interquartile range (IQR): 42.8–80.6], 64 patients had disease progression. Thirty-nine of 105 patients (37.1%) had a BCR at all follow-up time points, and none of these 39 patients had disease progression, corresponding to a negative predictive value (NPV) of 100%. Sixty-six patients had a detectable EBV-DNA during surveillance, with 64 diagnosed as disease progression at the last follow-up, for a positive predictive value (PPV) of 97.0%. Actuarial 3-year OS rates were 45.0% for patients with detectable EBV-DNA during posttreatment surveillance and 100% for patients with undetectable EBV-DNA. Lastly, median lead time between non-normalized EBV-DNA and clinically proven progression was 5.87 ± 0.67 months. Conclusions Taken together, EBV-DNA provided predictive value for the bone-only metastatic NPC patients. The results should be validated in prospective randomized studies.
Collapse
Affiliation(s)
- Guo-Ying Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei-Xiong Xia
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhuo-Fei Bi
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nian Lu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wang-Zhong Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei-Xin Bei
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hu Liang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jun-Zhi Xie
- Richard Montgomery High School at Rockville of Maryland, Rockville, MD, United States
| | - Yi-Min Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - He-Rui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Qun Xiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
69
|
Chakrabarti S, Kasi AK, Parikh AR, Mahipal A. Finding Waldo: The Evolving Paradigm of Circulating Tumor DNA (ctDNA)-Guided Minimal Residual Disease (MRD) Assessment in Colorectal Cancer (CRC). Cancers (Basel) 2022; 14:3078. [PMID: 35804850 PMCID: PMC9265001 DOI: 10.3390/cancers14133078] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 11/23/2022] Open
Abstract
Circulating tumor DNA (ctDNA), the tumor-derived cell-free DNA fragments in the bloodstream carrying tumor-specific genetic and epigenetic alterations, represents an emerging novel tool for minimal residual disease (MRD) assessment in patients with resected colorectal cancer (CRC). For many decades, precise risk-stratification following curative-intent colorectal surgery has remained an enduring challenge. The current risk stratification strategy relies on clinicopathologic characteristics of the tumors that lacks precision and results in over-and undertreatment in a significant proportion of patients. Consequently, a biomarker that can reliably identify patients harboring MRD would be of critical importance in refining patient selection for adjuvant therapy. Several prospective cohort studies have provided compelling data suggesting that ctDNA could be a robust biomarker for MRD that outperforms all existing clinicopathologic criteria. Numerous clinical trials are currently underway to validate the ctDNA-guided MRD assessment and adjuvant treatment strategies. Once validated, the ctDNA technology will likely transform the adjuvant therapy paradigm of colorectal cancer, supporting ctDNA-guided treatment escalation and de-escalation. The current article presents a comprehensive overview of the published studies supporting the utility of ctDNA for MRD assessment in patients with CRC. We also discuss ongoing ctDNA-guided adjuvant clinical trials that will likely shape future adjuvant therapy strategies for patients with CRC.
Collapse
Affiliation(s)
- Sakti Chakrabarti
- Department of Hematology-Oncology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA
| | - Anup Kumar Kasi
- Division of Medical Oncology, University of Kansas, Kansas City, KS 66160, USA;
| | - Aparna R. Parikh
- Harvard Medical School, Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA;
| | - Amit Mahipal
- Mayo College of Medicine, Rochester, MN 54656, USA;
| |
Collapse
|
70
|
Mukherji R, Yin C, Hameed R, Alqahtani AZ, Kulasekaran M, He AR, Weinberg BA, Marshall JL, Hartley ML, Noel MS. The current state of molecular profiling in gastrointestinal malignancies. Biol Direct 2022; 17:15. [PMID: 35668531 PMCID: PMC9172079 DOI: 10.1186/s13062-022-00322-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/19/2022] [Indexed: 11/10/2022] Open
Abstract
This is a review of the current state of molecular profiling in gastrointestinal (GI) cancers and what to expect from this evolving field in the future. Individualized medicine is moving from broad panel testing of numerous genes or gene products in tumor biopsy samples, identifying biomarkers of prognosis and treatment response, to relatively noninvasive liquid biopsy assays, building on what we have learned in our tumor analysis and growing into its own evolving predictive and prognostic subspecialty. Hence, the field of GI precision oncology is exploding, and this review endeavors to summarize where we are now in preparation for the journey ahead.
Collapse
Affiliation(s)
- Reetu Mukherji
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Chao Yin
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Rumaisa Hameed
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Ali Z Alqahtani
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Monika Kulasekaran
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Aiwu R He
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Benjamin A Weinberg
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - John L Marshall
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Marion L Hartley
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA
| | - Marcus S Noel
- The Ruesch Center for the Cure of GI Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA.
- MedStar Georgetown University Hospital, 3800 Reservoir Rd. NW, Washington, DC, 20007, USA.
| |
Collapse
|
71
|
The History and Future of Basic and Translational Cell-Free DNA Research at a Glance. Diagnostics (Basel) 2022; 12:diagnostics12051192. [PMID: 35626347 PMCID: PMC9139999 DOI: 10.3390/diagnostics12051192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 11/30/2022] Open
Abstract
We discuss the early history of the structure of DNA and its involvement in gene structure as well as its mobility in and between cells and between tissues in the form of circulating cell-free DNA (cfDNA). This is followed by a view of the present status of the studies on cfDNA and clinical applications of circulating cell-free tumor DNA (ctDNA). The future developments and roles of ctDNA are also considered.
Collapse
|
72
|
Liu F, Dai Z, Cheng Q, Xu L, Huang L, Liu Z, Li X, Wang N, Wang G, Wang L, Wang Z. LncRNA-targeting bio-scaffold mediates triple immune effects for postoperative colorectal cancer immunotherapy. Biomaterials 2022; 284:121485. [DOI: 10.1016/j.biomaterials.2022.121485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/19/2022] [Accepted: 03/22/2022] [Indexed: 12/13/2022]
|
73
|
Hedtke M, Pessoa Rejas R, Froelich MF, Ast V, Duda A, Mirbach L, Costina V, Martens UM, Hofheinz R, Neumaier M, Haselmann V. Liquid profiling of circulating tumor DNA in colorectal cancer: steps needed to achieve its full clinical value as standard care. Mol Oncol 2022; 16:2042-2056. [PMID: 34873826 PMCID: PMC9120900 DOI: 10.1002/1878-0261.13156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/27/2021] [Accepted: 12/06/2021] [Indexed: 12/23/2022] Open
Abstract
The analysis of circulating tumor DNA (ctDNA) is at the threshold of implementation into standard care for colorectal cancer (CRC) patients. However, data about the clinical utility of liquid profiling (LP), its acceptance by clinicians, and its integration into clinical workflows in real-world settings remain limited. Here, LP tests requested as part of routine care since 2016 were retrospectively evaluated. Results show restrained request behavior that improved moderately over time, as well as reliable diagnostic performance comparable to translational studies, with an overall agreement of 91.7%. Extremely low ctDNA levels at < 0.1% in over 20% of cases, a high frequency of concomitant driver mutations (in up to 14% of cases), and ctDNA levels reflecting the clinical course of disease were revealed. However, certain limitations hampering successful translation of ctDNA into clinical practice were uncovered, including the lack of clinically relevant ctDNA thresholds, appropriate time points of LP requests, and integrative evaluation of ctDNA, imaging, and clinical findings. In conclusion, these results highlight the potential clinical value of LP for CRC patient management and demonstrate issues that need to be addressed for successful long-term implementation in clinical workflows.
Collapse
Affiliation(s)
- Maren Hedtke
- Institute of Clinical ChemistryMedical Faculty Mannheim of the University of HeidelbergUniversity Hospital MannheimGermany
| | - Rodrigo Pessoa Rejas
- Institute of Clinical ChemistryMedical Faculty Mannheim of the University of HeidelbergUniversity Hospital MannheimGermany
| | - Matthias F. Froelich
- Department of Radiology and Nuclear MedicineUniversity Medicine MannheimMedical Faculty Mannheim of the University of HeidelbergGermany
| | - Volker Ast
- Institute of Clinical ChemistryMedical Faculty Mannheim of the University of HeidelbergUniversity Hospital MannheimGermany
| | - Angelika Duda
- Institute of Clinical ChemistryMedical Faculty Mannheim of the University of HeidelbergUniversity Hospital MannheimGermany
| | - Laura Mirbach
- Institute of Clinical ChemistryMedical Faculty Mannheim of the University of HeidelbergUniversity Hospital MannheimGermany
| | - Victor Costina
- Institute of Clinical ChemistryMedical Faculty Mannheim of the University of HeidelbergUniversity Hospital MannheimGermany
| | - Uwe M. Martens
- Cancer Center Heilbronn‐FrankenSLK‐ClinicsMOLIT Institute for Personalized MedicineHeilbronnGermany
| | - Ralf‐Dieter Hofheinz
- Day Treatment Center (TTZ)Interdisciplinary Tumor Center Mannheim (ITM)III Medical ClinicMedical Faculty Mannheim of the University of HeidelbergUniversity Hospital MannheimGermany
| | - Michael Neumaier
- Institute of Clinical ChemistryMedical Faculty Mannheim of the University of HeidelbergUniversity Hospital MannheimGermany
| | - Verena Haselmann
- Institute of Clinical ChemistryMedical Faculty Mannheim of the University of HeidelbergUniversity Hospital MannheimGermany
| |
Collapse
|
74
|
Schraa SJ, van Rooijen KL, Koopman M, Vink GR, Fijneman RJA. Cell-Free Circulating (Tumor) DNA before Surgery as a Prognostic Factor in Non-Metastatic Colorectal Cancer: A Systematic Review. Cancers (Basel) 2022; 14:2218. [PMID: 35565347 PMCID: PMC9101623 DOI: 10.3390/cancers14092218] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/01/2023] Open
Abstract
Identification of non-metastatic colorectal cancer (CRC) patients with a high risk of recurrence after tumor resection is important to select patients who might benefit from adjuvant treatment. Cell-free DNA (cfDNA) and circulating tumor DNA (ctDNA) analyses after surgery are promising biomarkers to predict recurrence in these patients. However, these analyses face several challenges and do not allow guidance of neoadjuvant treatment, which might become a novel standard option in colon cancer treatment. The prognostic value of cfDNA/ctDNA before surgery is unclear. This systematic review aims to provide an overview of publications in which the prognostic value of presurgery cfDNA/ctDNA in non-metastatic CRC patients was studied and is performed according to PRISMA guidelines. A total of 29 out of 1233 articles were included and categorized into three groups that reflect the type of approach: measurement of cfDNA, ctDNA somatic alterations, and ctDNA methylation. Overall, a clear association between presurgery cfDNA/ctDNA and the outcome was not observed, but large studies that primarily focus on the prognostic value of presurgery cfDNA/ctDNA are lacking. Designing and performing studies that focus on the value of presurgery cfDNA/ctDNA is needed, in addition to standardization in the reporting of cfDNA/ctDNA results according to existing guidelines to improve comparability and interpretation among studies.
Collapse
Affiliation(s)
- Suzanna J. Schraa
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (S.J.S.); (K.L.v.R.); (M.K.); (G.R.V.)
| | - Karlijn L. van Rooijen
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (S.J.S.); (K.L.v.R.); (M.K.); (G.R.V.)
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (S.J.S.); (K.L.v.R.); (M.K.); (G.R.V.)
| | - Geraldine R. Vink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (S.J.S.); (K.L.v.R.); (M.K.); (G.R.V.)
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Godebaldkwartier 419, 3511 DT Utrecht, The Netherlands
| | - Remond J. A. Fijneman
- Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
75
|
Alese OB, Cook N, Ortega-Franco A, Ulanja MB, Tan L, Tie J. Circulating Tumor DNA: An Emerging Tool in Gastrointestinal Cancers. Am Soc Clin Oncol Educ Book 2022; 42:1-20. [PMID: 35471832 DOI: 10.1200/edbk_349143] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Circulating tumor DNA (ctDNA) is tumor-derived fragmented DNA in the bloodstream that has come from primary or metastatic cancer sites. Neoplasm-specific genetic and epigenetic abnormalities are increasingly being identified through liquid biopsy: a novel, minimally invasive technique used to isolate and analyze ctDNA in the peripheral circulation. Liquid biopsy and other emerging ctDNA technologies represent a paradigm shift in cancer diagnostics because they allow for the detection of minimal residual disease in patients with early-stage disease, improve risk stratification, capture tumor heterogeneity and genomic evolution, and enhance ctDNA-guided adjuvant and palliative cancer therapy. Moreover, ctDNA can be used to monitor the tumor response to neoadjuvant and postoperative therapy in patients with metastatic disease. Using clearance of ctDNA as an endpoint for escalation/de-escalation of adjuvant chemotherapy for patients considered to have high-risk disease has become an important area of research. The possibility of using ctDNA as a surrogate for treatment response-including for overall survival, progression-free survival, and disease-free survival-is an attractive concept; this surrogate will arguably reduce study duration and expedite the development of new therapies. In this review, we summarize the current evidence on the applications of ctDNA for the diagnosis and management of gastrointestinal tumors. Gastrointestinal cancers-including tumors of the esophagus, stomach, colon, liver, and pancreas-account for one-quarter of global cancer diagnoses and contribute to more than one-third of cancer-related deaths. Given the prevalence of gastrointestinal malignancies, ctDNA technology represents a powerful tool to reduce the global burden of disease.
Collapse
Affiliation(s)
- Olatunji B Alese
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Natalie Cook
- Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Manchester, United Kingdom.,Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Ana Ortega-Franco
- Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Mark B Ulanja
- Christus Ochsner St. Patrick Hospital, Lake Charles, LA
| | - Lavinia Tan
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Jeanne Tie
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Division of Personalized Oncology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| |
Collapse
|
76
|
Barsan V, Xia Y, Klein D, Gonzalez-Pena V, Youssef S, Inaba Y, Mahmud O, Natarajan S, Agarwal V, Pang Y, Autry R, Pui CH, Inaba H, Evans W, Gawad C. Simultaneous monitoring of disease and microbe dynamics through plasma DNA sequencing in pediatric patients with acute lymphoblastic leukemia. SCIENCE ADVANCES 2022; 8:eabj1360. [PMID: 35442732 PMCID: PMC9020671 DOI: 10.1126/sciadv.abj1360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 01/19/2022] [Indexed: 05/09/2023]
Abstract
Treatment of acute lymphoblastic leukemia (ALL) necessitates continuous risk assessment of leukemic disease burden and infections that arise in the setting of immunosuppression. This study was performed to assess the feasibility of a hybrid capture next-generation sequencing panel to longitudinally measure molecular leukemic disease clearance and microbial species abundance in 20 pediatric patients with ALL throughout induction chemotherapy. This proof of concept helps establish a technical and conceptual framework that we anticipate will be expanded and applied to additional patients with leukemia, as well as extended to additional cancer types. Molecular monitoring can help accelerate the attainment of insights into the temporal biology of host-microbe-leukemia interactions, including how those changes correlate with and alter anticancer therapy efficacy. We also anticipate that fewer invasive bone marrow examinations will be required, as these methods improve with standardization and are validated for clinical use.
Collapse
Affiliation(s)
- Valentin Barsan
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Yuntao Xia
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - David Klein
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Veronica Gonzalez-Pena
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Sarah Youssef
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Yuki Inaba
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Ousman Mahmud
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Sivaraman Natarajan
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Vibhu Agarwal
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Yakun Pang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Robert Autry
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - William Evans
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Charles Gawad
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94304, USA
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
77
|
Arisi MF, Dotan E, Fernandez SV. Circulating Tumor DNA in Precision Oncology and Its Applications in Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23084441. [PMID: 35457259 PMCID: PMC9024503 DOI: 10.3390/ijms23084441] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/29/2022] [Accepted: 04/14/2022] [Indexed: 02/06/2023] Open
Abstract
Circulating tumor DNA (ctDNA) is a component of cell-free DNA (cfDNA) that is shed by malignant tumors into the bloodstream and other bodily fluids. ctDNA can comprise up to 10% of a patient’s cfDNA depending on their tumor type and burden. The short half-life of ctDNA ensures that its detection captures tumor burden in real-time and offers a non-invasive method of repeatedly evaluating the genomic profile of a patient’s tumor. A challenge in ctDNA detection includes clonal hematopoiesis of indeterminate potential (CHIP), which can be distinguished from tumor variants using a paired whole-blood control. Most assays for ctDNA quantification rely on measurements of somatic variant allele frequency (VAF), which is a mutation-dependent method. Patients with certain types of solid tumors, including colorectal cancer (CRC), can have levels of cfDNA 50 times higher than healthy patients. ctDNA undergoes a precipitous drop shortly after tumor resection and therapy, and rising levels can foreshadow radiologic recurrence on the order of months. The amount of tumor bulk required for ctDNA detection is lower than that for computed tomography (CT) scan detection, with ctDNA detection preceding radiologic recurrence in many cases. cfDNA/ctDNA can be used for tumor molecular profiling to identify resistance mutations when tumor biopsy is not available, to detect minimal residual disease (MRD), to monitor therapy response, and for the detection of tumor relapse. Although ctDNA is not yet implemented in clinical practice, studies are ongoing to define the appropriate way to use it as a tool in the clinic. In this review article, we examine the general aspects of ctDNA, its status as a biomarker, and its role in the management of early (II–III) and late (IV; mCRC) stage colorectal cancer (CRC).
Collapse
Affiliation(s)
- Maria F. Arisi
- Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Efrat Dotan
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| | - Sandra V. Fernandez
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
- Correspondence:
| |
Collapse
|
78
|
Newhook TE, Vauthey JN. Colorectal liver metastases: state-of-the-art management and surgical approaches. Langenbecks Arch Surg 2022; 407:1765-1778. [DOI: 10.1007/s00423-022-02496-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023]
|
79
|
Liu W, Li Y, Tang Y, Song Q, Wang J, Li N, Chen S, Shi J, Wang S, Li Y, Jiao Y, Zeng Y, Jin J. Response prediction and risk stratification of patients with rectal cancer after neoadjuvant therapy through an analysis of circulating tumour DNA. EBioMedicine 2022; 78:103945. [PMID: 35306340 PMCID: PMC8933829 DOI: 10.1016/j.ebiom.2022.103945] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/21/2022] [Accepted: 03/03/2022] [Indexed: 11/11/2022] Open
Abstract
Background Multiple approaches based on cell-free DNA (cfDNA) have been applied to detect minimal residual disease (MRD) and to predict prognosis or recurrence. However, a comparison of the approaches used in different cohorts and studies is difficult. We aimed to compare multiple approaches for MRD analysis after neoadjuvant therapy (NAT) in patients with locally advanced rectal cancer (LARC). Methods Sixty patients with LARC from a multicentre, phase II/III randomized trial were included, with tissue and blood samples collected. For each cfDNA sample, we profiled MRD using 3 approaches: personalized assay targeting tumour-informed mutations, universal panel of genes frequently mutated in colorectal cancer (CRC), and low depth sequencing for copy number alterations (CNAs). Findings Positive MRD based on post-NAT personalized assay was significantly associated with an increased risk of recurrence (HR = 27.38; log-rank P < 0.0001). MRD analysis based on universal panel (HR = 5.18; log-rank P = 0.00086) and CNAs analysis (HR = 9.24; log-rank P = 0.00017) showed a compromised performance in predicting recurrence. Both the personalized assay and universal panel showed complementary pattern to CNAs analysis in detecting cases with recurrence and the combination of the two types of biomarkers may lead to better performance. Interpretation The combination of mutation profiling and CNA profiling can improve the detection of MRD, which may help optimize the treatment strategies for patients with LARC.
Collapse
|
80
|
Merk C, Martling A, Lindberg J, Benhaim L, Taieb J, Lind P. Circulating tumor DNA (ctDNA) in adjuvant therapy of early stage colon cancer: current status and future perspectives. Acta Oncol 2022; 61:523-530. [PMID: 35139729 DOI: 10.1080/0284186x.2022.2033831] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND This article reviews the current knowledge on circulating tumor DNA (ctDNA) in early stage colon cancer and ongoing trials on ctDNA-guided treatment in the adjuvant setting. METHODS A literature search of Pubmed was performed to identify studies on ctDNA in early stage colon cancer and neoadjuvant or adjuvant treatment. For ongoing trials, we searched clinicaltrials.gov and the Australian New Zealand Clinical Trials Registry (ANZCTR). RESULTS Several studies show that ctDNA is a strong predictor for recurrence and survival after surgery and adjuvant chemotherapy. The specificity of this marker is extremely high, and the sensitivity is increasing with the development of technology. Recurrences can be detected very early and the analysis can potentially be used to guide neoadjuvant and adjuvant treatment. Ongoing and planned studies are now looking into escalation and de-escalation of therapy according to ctDNA-status after surgery. CONCLUSION Serial measurement of ctDNA shows great promise as a marker for both prognosis and response to treatment in early colon cancer. Future studies will show whether we can use this analysis for tailoring treatment for patients in the adjuvant and neoadjuvant setting. With improved technology, ctDNA has the potential of becoming a 'game-changer' in the treatment of early stage colon cancers.
Collapse
Affiliation(s)
- Cecilia Merk
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Martling
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Léonor Benhaim
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, Paris, France
- Department of Visceral and Surgical Oncology, Villejuif, France
| | - Julien Taieb
- Department of Gastroenterology and Gastrointestinal Oncology, Hôpital Européen Georges-Pompidou, AP-HP, Université de Paris, Paris, France
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, Université de Paris, USPC, Equipe labellisée Ligue Nationale Contre le Cancer, SIRIC CARPEM, Paris, France
| | - Pehr Lind
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Sweden
| |
Collapse
|
81
|
Hayashi T, Yoshida Y, Yamada T, Tanaka K, Shimaoka H, Kajitani R, Munechika T, Nagano H, Matsumoto Y, Komono A, Sakamoto R, Aisu N, Yoshimatsu G, Yoshimura F, Hasegawa S. Relationship between perioperative oncological evaluation and recurrence using circulating tumor DNA with KRAS mutation in patients with colorectal cancer. Cancer Med 2022; 11:3126-3135. [PMID: 35312176 PMCID: PMC9385586 DOI: 10.1002/cam4.4677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 12/29/2022] Open
Abstract
Background The detection of circulating tumor DNA (ctDNA) in colorectal cancer (CRC) by liquid biopsy may have prognostic information. In this perioperative study, we evaluate if there is a relationship between mutant allele frequency (MAF) of Kirsten rat sarcoma viral oncogene homolog (KRAS) and tumor recurrence and how that could be useful in the early detection of recurrence. Methods Among 304 cases of colorectal cancer surgery, ctDNA was sampled from the perioperative blood of 84 patients with CRC with KRAS mutation (exon 4 p.A146T, exon 4 p.A146V, exon 2 p.G12A, exon 2 p.G12C, exon 2 p.G12D, exon 2 p.G12S, exon 2 p.G12V, exon 2 p.G13D, exon 3 p.Q61H) and analyzed using the digital polymerase chain reaction system. The median observation period was 26 months. Results Although the relationship between the perioperative MAF of KRAS and recurrence was not proved, tumor diameter, tumor depth, and stage were correlated with the preoperative MAF of KRAS (p = 0.034, p = 0.002, p = 0.008). However, tumor diameter, tumor depth, and stage did not correlate with MAF of KRAS at postoperative day 30. Conclusions In this study, pathological tumor size, tumor depth, and stage were correlated with preoperative MAF of KRAS, but it was unreliable to predict recurrence by detection of ctDNA with KRAS mutation in the perioperative period of colorectal surgery.
Collapse
Affiliation(s)
- Takaomi Hayashi
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoichiro Yoshida
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Teppei Yamada
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Keita Tanaka
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Hideki Shimaoka
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Ryuji Kajitani
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Taro Munechika
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Hideki Nagano
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshiko Matsumoto
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Akira Komono
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Ryohei Sakamoto
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Naoya Aisu
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Gumpei Yoshimatsu
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Fumihiro Yoshimura
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Suguru Hasegawa
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| |
Collapse
|
82
|
Haselmann V, Hedtke M, Neumaier M. Liquid Profiling for Cancer Patient Stratification in Precision Medicine—Current Status and Challenges for Successful Implementation in Standard Care. Diagnostics (Basel) 2022; 12:diagnostics12030748. [PMID: 35328301 PMCID: PMC8947441 DOI: 10.3390/diagnostics12030748] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor DNA (ctDNA), accurately described by the term liquid profiling (LP), enables real-time assessment of the tumor mutational profile as a minimally invasive test and has therefore rapidly gained traction, particular for the management of cancer patients. By LP, tumor-specific genetic alterations can be determined as part of companion diagnostics to guide selection of appropriate targeted therapeutics. Because LP facilitates longitudinal monitoring of cancer patients, it can be used to detect acquired resistant mechanisms or as a personalized biomarker for earlier detection of disease recurrence, among other applications. However, LP is not yet integrated into routine care to the extent that might be expected. This is due to the lack of harmonization and standardization of preanalytical and analytical workflows, the lack of proper quality controls, limited evidence of its clinical utility, heterogeneous study results, the uncertainty of clinicians regarding the value and appropriate indications for LP and its interpretation, and finally, the lack of reimbursement for most LP tests. In this review, the value proposition of LP for cancer patient management and treatment optimization, the current status of implementation in standard care, and the main challenges that need to be overcome are discussed in detail.
Collapse
|
83
|
Kasi PM, Fehringer G, Taniguchi H, Starling N, Nakamura Y, Kotani D, Powles T, Li BT, Pusztai L, Aushev VN, Kalashnikova E, Sharma S, Malhotra M, Demko ZP, Aleshin A, Rodriguez A, Billings PR, Grothey A, Taieb J, Cunningham D, Yoshino T, Kopetz S. Impact of Circulating Tumor DNA-Based Detection of Molecular Residual Disease on the Conduct and Design of Clinical Trials for Solid Tumors. JCO Precis Oncol 2022; 6:e2100181. [PMID: 35263168 PMCID: PMC8926064 DOI: 10.1200/po.21.00181] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Earlier detection of cancer recurrence using circulating tumor DNA (ctDNA) to detect molecular residual disease (MRD) has the potential to dramatically affect cancer management. We review evidence supporting the use of ctDNA as a biomarker for detection of MRD and highlight the potential impact that ctDNA testing could have on the conduct of clinical trials.
Collapse
Affiliation(s)
- Pashtoon M Kasi
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA
| | | | - Hiroya Taniguchi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Naureen Starling
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Daisuke Kotani
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Thomas Powles
- Barts Cancer Institute, Queen Mary University of London ECMC, Barts Health, London, United Kingdom
| | - Bob T Li
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY
| | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | | | | | | | | | | | | | | | | | - Axel Grothey
- West Cancer Center and Research Institute, Germantown, TN
| | - Julien Taieb
- Georges Pompidou European Hospital, SIRIC-CARPEM, Université de Paris, Paris, France
| | - David Cunningham
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
84
|
Garinet S, Wang P, Mansuet-Lupo A, Fournel L, Wislez M, Blons H. Updated Prognostic Factors in Localized NSCLC. Cancers (Basel) 2022; 14:cancers14061400. [PMID: 35326552 PMCID: PMC8945995 DOI: 10.3390/cancers14061400] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the most common cause of cancer mortality worldwide, and non-small cell lung cancer (NSCLC) represents 80% of lung cancer subtypes. Patients with localized non-small cell lung cancer may be considered for upfront surgical treatment. However, the overall 5-year survival rate is 59%. To improve survival, adjuvant chemotherapy (ACT) was largely explored and showed an overall benefit of survival at 5 years < 7%. The evaluation of recurrence risk and subsequent need for ACT is only based on tumor stage (TNM classification); however, more than 25% of patients with stage IA/B tumors will relapse. Recently, adjuvant targeted therapy has been approved for EGFR-mutated resected NSCLC and trials are evaluating other targeted therapies and immunotherapies in adjuvant settings. Costs, treatment duration, emergence of resistant clones and side effects stress the need for a better selection of patients. The identification and validation of prognostic and theranostic markers to better stratify patients who could benefit from adjuvant therapies are needed. In this review, we report current validated clinical, pathological and molecular prognosis biomarkers that influence outcome in resected NSCLC, and we also describe molecular biomarkers under evaluation that could be available in daily practice to drive ACT in resected NSCLC.
Collapse
Affiliation(s)
- Simon Garinet
- Pharmacogenomics and Molecular Oncology Unit, Biochemistry Department, Assistance Publique—Hopitaux de Paris, Hôpital Européen Georges Pompidou, 75015 Paris, France;
- Centre de Recherche des Cordeliers, INSERM UMRS-1138, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Pascal Wang
- Oncology Thoracic Unit, Pulmonology Department, Assistance Publique—Hopitaux de Paris, Hôpital Cochin, 75014 Paris, France; (P.W.); (M.W.)
| | - Audrey Mansuet-Lupo
- Pathology Department, Assistance Publique—Hopitaux de Paris, Hôpital Cochin, 75014 Paris, France;
| | - Ludovic Fournel
- Thoracic Surgery Department, Assistance Publique—Hopitaux de Paris, Hôpital Cochin, 75014 Paris, France;
| | - Marie Wislez
- Oncology Thoracic Unit, Pulmonology Department, Assistance Publique—Hopitaux de Paris, Hôpital Cochin, 75014 Paris, France; (P.W.); (M.W.)
| | - Hélène Blons
- Pharmacogenomics and Molecular Oncology Unit, Biochemistry Department, Assistance Publique—Hopitaux de Paris, Hôpital Européen Georges Pompidou, 75015 Paris, France;
- Centre de Recherche des Cordeliers, INSERM UMRS-1138, Sorbonne Université, Université de Paris, 75006 Paris, France
- Correspondence:
| |
Collapse
|
85
|
Clinical Validity of Circulating Tumor DNA as Prognostic and Predictive Marker for Personalized Colorectal Cancer Patient Management. Cancers (Basel) 2022; 14:cancers14030851. [PMID: 35159118 PMCID: PMC8834623 DOI: 10.3390/cancers14030851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/21/2022] [Accepted: 02/07/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary CtDNA analysis is a promising tool in liquid biopsy for the detection of tumor recurrence and progression, and is increasingly adopted into clinical practice. Still, guidelines for the accurate clinical interpretation of ctDNA analysis results are largely lacking, especially for tumor mutant variants detected at very low frequencies. Here, we show that cutoff determination for the detection and quantification of low-frequency mutant variants enables the accurate prediction of residual disease, tumor recurrence and progression, even before clinical evidence. CtDNA analysis using these cutoffs outperformed cfDNA and CEA level measurements. With these findings, we highlight the need to thoroughly validate each liquid biopsy assay and define the assay-specific limit of blanks (LOB) and limit of quantifications (LOQ) of BRAF p.V600E and KRAS p.G12/p.G13 assays for clinical interpretation. Our approach enables accurate clinical interpretation to support clinical decision making. Abstract Circulating tumor DNA (ctDNA) is a promising liquid biopsy (LB) marker to support clinical decisions in precision medicine. For implementation into routine clinical practice, clinicians need precise ctDNA level cutoffs for reporting residual disease and monitoring tumor burden changes during therapy. We clinically validated the limit of blank (LOB) and the limit of quantification (LOQ) of assays for the clinically most relevant somatic variants BRAF p.V600E and KRAS p.G12/p.G13 in colorectal cancer (CRC) in a study cohort encompassing a total of 212 plasma samples. We prove that residual disease detection using the LOB as a clinically verified cutoff for ctDNA positivity is in concordance with clinical evidence of metastasis or recurrence. We further show that tumor burden changes during chemotherapy and the course of disease are correctly predicted using the LOQ as a cutoff for quantitative ctDNA changes. The high potential of LB using ctDNA for accurately predicting the course of disease was proven by direct comparison to the routinely used carcinoembryonic antigen (CEA) as well as the circulating free DNA (cfDNA) concentration. Our results show that LB using validated ctDNA assays outperforms CEA and cfDNA for residual disease detection and the prediction of tumor burden changes.
Collapse
|
86
|
Stasik S, Mende M, Schuster C, Mahler S, Aust D, Tannapfel A, Reinacher-Schick A, Baretton G, Krippendorf C, Bornhäuser M, Ehninger G, Folprecht G, Thiede C. Sensitive Quantification of Cell-Free Tumor DNA for Early Detection of Recurrence in Colorectal Cancer. Front Genet 2022; 12:811291. [PMID: 35069704 PMCID: PMC8766716 DOI: 10.3389/fgene.2021.811291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
The detection of plasma cell–free tumor DNA (ctDNA) is prognostic in colorectal cancer (CRC) and has potential for early prediction of disease recurrence. In clinical routine, ctDNA-based diagnostics are limited by the low concentration of ctDNA and error rates of standard next-generation sequencing (NGS) approaches. We evaluated the potential to increase the stability and yield of plasma cell–free DNA (cfDNA) for routine diagnostic purposes using different blood collection tubes and various manual or automated cfDNA extraction protocols. Sensitivity for low-level ctDNA was measured in KRAS-mutant cfDNA using an error-reduced NGS procedure. To test the applicability of rapid evaluation of ctDNA persistence in clinical routine, we prospectively analyzed postoperative samples of 67 CRC (stage II) patients. ctDNA detection was linear between 0.0045 and 45%, with high sensitivity (94%) and specificity (100%) for mutations at 0.1% VAF. The stability and yield of cfDNA were superior when using Streck BCT tubes and a protocol by Zymo Research. Sensitivity for ctDNA increased 1.5-fold by the integration of variant reads from triplicate PCRs and with PCR template concentration. In clinical samples, ctDNA persistence was found in ∼9% of samples, drawn 2 weeks after surgery. Moreover, in a retrospective analysis of 14 CRC patients with relapse during adjuvant therapy, we successfully detected ctDNA (median 0.38% VAF; range 0.18–5.04% VAF) in 92.85% of patients significantly prior (median 112 days) to imaging-based surveillance. Using optimized pre-analytical conditions, the detection of postoperative ctDNA is feasible with excellent sensitivity and allows the prediction of CRC recurrence in routine oncology testing.
Collapse
Affiliation(s)
- Sebastian Stasik
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Heidelberg, Germany
| | - Marika Mende
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | | | - Sandra Mahler
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Daniela Aust
- Institute of Pathology, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | | | - Anke Reinacher-Schick
- Department of Hematology, Oncology and Palliative Care, St. Josef Hospital, Ruhr University, Bochum, Germany
| | - Gustavo Baretton
- Institute of Pathology, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | | | - Martin Bornhäuser
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Heidelberg, Germany
| | - Gerhard Ehninger
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Gunnar Folprecht
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Christian Thiede
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| |
Collapse
|
87
|
Pu X, Li Z, Wang X, Jiang H. Ascites and Serial Plasma Circulating Tumor DNA for Predicting the Effectiveness of Hyperthermic Intraperitoneal Chemotherapy in Patients With Peritoneal Carcinomatosis. Front Oncol 2022; 12:791418. [PMID: 35145914 PMCID: PMC8821810 DOI: 10.3389/fonc.2022.791418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeWe investigated the value of ascites and serial plasma circulating tumor DNA (ctDNA) for predicting response to hyperthermic intraperitoneal chemotherapy (HIPEC), monitoring tumor burden, and predicting prognosis in patients with peritoneal carcinomatosis (PC).Experimental DesignIn this observational study, 19 patients with PC were enrolled. Serial plasma ctDNA was analyzed using next-generation sequencing. The molecular tumor burden index (mTBI) was used to detect ctDNA, and concurrent changes in the dominant clone variant allele frequency (VAF) and common tumor markers were used as controls. The correlation between ascites and plasma ctDNA comutated genes was expressed by VAF. The overall response rate (complete response + partial response) after HIPEC was determined. Ascites progression-free survival (PFS) and overall survival (OS) were determined, and potential correlations between these outcomes and change in mTBI (△mTBI), change in sum-VAF (△sum-VAF), dominant close VAF, and tumor markers were assessed.ResultsThe overall response rate at 1 month after HIPEC was 100%. The △mTBI (r = 0.673; P = 0.023) and △sum-VAF (r = 0.945; P <0.001) were significantly positively correlated with ascites PFS; these correlations were stronger than those of the dominant clone VAF (r = 0.588; P = 0.057) and tumor markers in the same period (r =0.091; P = 0.790). Patients with a low baseline mTBI (<0.67) demonstrated significantly longer ascites PFS (P = 0.003; HR = 0.157; 95% CI: 0.046–0.540) and OS (P = 0.017; HR = 0.296; 95% CI: 0.109–0.804) than those with a high baseline mTBI (≥0.67). Consistent mutations were detected in plasma and ascites (r = 0.794; P = 0.001).ConclusionA real-time serial plasma ctDNA assay accurately reflected tumor burden. The △mTBI and △sum-VAF can be used as predictors of HIPEC efficacy in patients with PC. A high baseline mTBI may be a negative risk factor for prognosis.
Collapse
Affiliation(s)
- Xiaolin Pu
- Department of Oncology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Zongyuan Li
- Department of Oncology, Graduate School of Dalian Medical University, Dalian, China
| | - Xiaoying Wang
- Department of Oncology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Hua Jiang
- Department of Oncology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
- *Correspondence: Hua Jiang,
| |
Collapse
|
88
|
Wang SQ, Chai CX, Wang BR, Zhu F, Shang D, Li M. Liquid biopsy: Precise diagnosis and therapy for cholangiocarcinoma. World J Gastrointest Oncol 2022; 14:362-365. [PMID: 35116122 PMCID: PMC8790414 DOI: 10.4251/wjgo.v14.i1.362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/19/2021] [Accepted: 11/26/2021] [Indexed: 02/06/2023] Open
Abstract
The following letter to the editor highlights the review titled "Liquid biopsy in cholangiocarcinoma: Current status and future perspective" in World J Gastrointest Oncol 2021; 13: 332-350. It is necessary to realize individualized therapy to improve the clinical prognosis of patients with cholangiocarcinoma.
Collapse
Affiliation(s)
- Si-Qin Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Chu-Xing Chai
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Bi-Rong Wang
- Department of Breast and Thyroid Surgery, Wuhan Fourth Hospital; Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Feng Zhu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Dan Shang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
89
|
Popp J, Weinberg DS, Enns E, Nyman JA, Beck JR, Kuntz KM. Reevaluating the Evidence for Intensive Postoperative Extracolonic Surveillance for Nonmetastatic Colorectal Cancer. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2022; 25:36-46. [PMID: 35031098 PMCID: PMC9186065 DOI: 10.1016/j.jval.2021.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 07/19/2021] [Accepted: 07/31/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVES The FACS, GILDA, and COLOFOL trials have cast doubt on the value of intensive extracolonic surveillance for resected nonmetastatic colorectal cancer and by extension metastasectomy. We reexamined this pessimistic interpretation. We evaluate an alternative explanation: insufficient power to detect a realistically sized survival benefit that may be clinically meaningful. METHODS A microsimulation model of postdiagnosis colorectal cancer was constructed assuming an empirically plausible efficacy for metastasectomy and thus surveillance. The model was used to predict the large-sample mortality reduction expected for each trial and the implied statistical power. A potential recurrence imbalance in the FACS trial was investigated. Goodness of fit between model predictions and trial results were evaluated. Downstream life expectancy was estimated and power calculations performed for future trials evaluating surveillance and metastasectomy. RESULTS For all 3 trials, the model predicted a mortality reduction of ≤5% and power of <10%. The FACS recurrence imbalance likely led to a large relative bias (>2.5) in the hazard ratio for overall survival favoring control. After adjustment, both COLOFOL and FACS results were consistent with model predictions (P>.5). A 2.6 (95% credible interval 0.5-5.1) and 3.6 (95% credible interval 0.8-7.0) month increase in life expectancy is predicted comparing intensive extracolonic surveillance-routine computed tomography scans and carcinoembryonic antigen assays-with 1 computed tomography scan at 12 months or no surveillance, respectively. An adequately sized surveillance trial is not feasible. A metastasectomy trial should randomize at least 200 to 300 patients. CONCLUSIONS Recent trial results do not warrant de novo skepticism of metastasectomy nor targeted extracolonic surveillance. Given the potential for clinically meaningful life-expectancy gain and significant uncertainty, a trial of metastasectomy is needed.
Collapse
Affiliation(s)
- Jonah Popp
- Center for Evidence Synthesis in Health, Department of Health Services, Policy, and Practice, School of Public Health, Brown University, Providence, RI, USA.
| | - David S Weinberg
- Department of Medicine, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Eva Enns
- Division of Health Policy and Management, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - John A Nyman
- Division of Health Policy and Management, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - J Robert Beck
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Karen M Kuntz
- Division of Health Policy and Management, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
90
|
Callesen LB, Takacova T, Hamfjord J, Würschmidt F, Oldhafer KJ, Brüning R, Arnold D, Spindler KLG. Circulating DNA in patients undergoing loco-regional treatment of colorectal cancer metastases: a systematic review and meta-analysis. Ther Adv Med Oncol 2022; 14:17588359221133171. [PMID: 36339929 PMCID: PMC9634210 DOI: 10.1177/17588359221133171] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022] Open
Abstract
Background Loco-regional treatment strategies of colorectal cancer (CRC) metastases are evolving, but biological markers that can benefit patients and assist physicians in clinical decisions are lacking. The primary objective of this systematic review and meta-analysis is to investigate the current knowledge on circulating DNA and its clinical utility in predicting outcomes in patients undergoing loco-regional treatment of CRC metastases. Methods A systematic search of PubMed, Embase, and Cochrane Central Register of Controlled Trials was conducted on March 22, 2022. We included studies on patients undergoing loco-regional treatment of CRC metastases reporting the predictive or prognostic value of circulating DNA in the blood. Hazard ratios (HR) were pooled in separate random-effects meta-analyses to investigate if pre- or post-ablation measurements of circulating DNA were associated with survival. The risk of bias was assessed according to the Quality in Prognosis Studies tool. Results Twenty-eight studies with 2868 patients were included, of which 16 studies were eligible for meta-analyses. As expected in this new research field, a majority of included studies (n = 21/28) had a high risk of bias in at least one domain. Circulating DNA above the cutoff in a plasma sample taken before loco-regional treatment was associated with a short recurrence-free survival [pooled HR = 2.8, 95% confidence interval (CI) 1.4-5.7, n = 162] and overall survival (pooled HR = 4.7, 95% CI 1.1-20.6, n = 105). Circulating DNA above the cutoff in a plasma sample taken after loco-regional treatment was associated with a short recurrence-free survival (pooled HR = 4.5, 95% CI 3.4-6.1, n = 569) and overall survival (pooled HR = 7.5, 95% CI 2.0-27.3, n = 161). There was limited data on the association between dynamics in circulating DNA and outcome. Conclusions Measurements of circulating DNA can be valuable when selecting and monitoring patients undergoing loco-regional treatment of CRC metastases. Studies designed to investigate the true clinical utility of circulating DNA in the context of various ablation modalities are warranted.The review has been registered at PROSPERO (ID: CRD42022320032).
Collapse
Affiliation(s)
| | - Tana Takacova
- Asklepios Tumorzentrum Hamburg, Hamburg,
Germany
- Department of Oncology and Palliative Care with
Sections Hematology and Rheumatology, AK Altona, Hamburg, Germany
| | - Julian Hamfjord
- Department of Oncology, Oslo University
Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for
Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of
Medicine, University of Oslo, Oslo, Norway
| | | | - Karl J. Oldhafer
- Asklepios Tumorzentrum Hamburg, Hamburg,
Germany
- Department of Surgery, Division of
Hepato-biliary and Pancreatic (HBP) Surgery, AK Barmbek, Hamburg,
Germany
- Faculty of Medicine, Semmelweis University
Budapest, Asklepios Campus Hamburg, Hamburg, Germany
| | - Roland Brüning
- Asklepios Tumorzentrum Hamburg, Hamburg,
Germany
- Department of Radiology and Neuroradiology, AK
Barmbek, Hamburg, Germany
| | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg, Hamburg,
Germany
- Department of Oncology and Palliative Care
with Sections Hematology and Rheumatology, AK Altona, Hamburg, Germany
| | - Karen-Lise G. Spindler
- Department of Experimental Clinical Oncology,
Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus
University, Aarhus Denmark
- Asklepios Tumorzentrum Hamburg, Hamburg,
Germany
| |
Collapse
|
91
|
Circulating tumor DNA as a prognostic indicator of colorectal cancer recurrence-a systematic review and meta-analysis. Int J Colorectal Dis 2022; 37:1021-1027. [PMID: 35384496 PMCID: PMC8983807 DOI: 10.1007/s00384-022-04144-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE Colorectal cancer (CRC) is one of the leading causes of cancer-related death worldwide. After resection, patients need extensive follow-up to detect asymptomatic recurrences as early as possible to obtain optimal treatment. This study evaluated the prognostic value of circulating tumor DNA (ctDNA) for CRC recurrence. METHODS Two investigators independently conducted a systematic literature search of peer-reviewed studies that investigated the prognostic value of ctDNA in CRC. Fixed effects or random effects models were applied for all analyses based on the assessment of heterogeneity. RESULTS A total of 189 studies were initially retrieved from all databases; ultimately, eight studies with 879 CRC patients were included in this analysis. The pooled median recurrence-free survival was 11.36 months for ctDNA-positive patients. Meta-analysis of hazard ratio (HR) suggested that postoperative ctDNA-positive patients were more likely to experience cancer recurrence than ctDNA-negative patients (pooled HR: 5.41; 95% confidence interval (CI): 2.37-8.45). CONCLUSIONS Successive monitoring of ctDNA status and follow-up with postoperative computed tomography (CT)/magnetic resonance imaging (MRI) are useful tools to detect early recurrence in postoperative ctDNA-positive patients.
Collapse
|
92
|
Yang F, Tang J, Zhao Z, Zhao C, Xiang Y. Circulating tumor DNA: a noninvasive biomarker for tracking ovarian cancer. Reprod Biol Endocrinol 2021; 19:178. [PMID: 34861867 PMCID: PMC8641226 DOI: 10.1186/s12958-021-00860-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 11/11/2021] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer is the fifth leading cause of cancer-related mortality in women worldwide. Despite the development of technologies over decades to improve the diagnosis and treatment of patients with ovarian cancer, the survival rate remains dismal, mainly because most patients are diagnosed at a late stage. Traditional treatment methods and biomarkers such as cancer antigen-125 as a cancer screening tool lack specificity and cannot offer personalized combinatorial therapy schemes. Circulating tumor DNA (ctDNA) is a promising biomarker for ovarian cancer and can be detected using a noninvasive liquid biopsy. A wide variety of ctDNA applications are being elucidated in multiple studies for tracking ovarian carcinoma during diagnostic and prognostic evaluations of patients and are being integrated into clinical trials to evaluate the disease. Furthermore, ctDNA analysis may be used in combination with multiple "omic" techniques to analyze proteins, epigenetics, RNA, nucleosomes, exosomes, and associated immune markers to promote early detection. However, several technical and biological hurdles impede the application of ctDNA analysis. Certain intrinsic features of ctDNA that may enhance its utility as a biomarker are problematic for its detection, including ctDNA lengths, copy number variations, and methylation. Before the development of ctDNA assays for integration in the clinic, such issues are required to be resolved since these assays have substantial potential as a test for cancer screening. This review focuses on studies concerning the potential clinical applications of ctDNA in ovarian cancer diagnosis and discusses our perspective on the clinical research aimed to treat this daunting form of cancer.
Collapse
Affiliation(s)
- Fang Yang
- Department of Physiology, Basic Medical College, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jun Tang
- Department of Physiology, Basic Medical College, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Zihao Zhao
- Department of Physiology, Basic Medical College, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Chunling Zhao
- Department of Physiology, Basic Medical College, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yuancai Xiang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
93
|
Moding EJ, Nabet BY, Alizadeh AA, Diehn M. Detecting Liquid Remnants of Solid Tumors: Circulating Tumor DNA Minimal Residual Disease. Cancer Discov 2021; 11:2968-2986. [PMID: 34785539 PMCID: PMC8976700 DOI: 10.1158/2159-8290.cd-21-0634] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/24/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
Growing evidence demonstrates that circulating tumor DNA (ctDNA) minimal residual disease (MRD) following treatment for solid tumors predicts relapse. These results suggest that ctDNA MRD could identify candidates for adjuvant therapy and measure response to such treatment. Importantly, factors such as assay type, amount of ctDNA release, and technical and biological background can affect ctDNA MRD results. Furthermore, the clinical utility of ctDNA MRD for treatment personalization remains to be fully established. Here, we review the evidence supporting the value of ctDNA MRD in solid cancers and highlight key considerations in the application of this potentially transformative biomarker. SIGNIFICANCE ctDNA analysis enables detection of MRD and predicts relapse after definitive treatment for solid cancers, thereby promising to revolutionize personalization of adjuvant and consolidation therapies.
Collapse
Affiliation(s)
- Everett J. Moding
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Barzin Y. Nabet
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Current address: Department of Oncology Biomarker Development, Genentech, South San Francisco, CA 94080, USA
| | - Ash A. Alizadeh
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California, USA
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
94
|
Bruscaggin A, di Bergamo LT, Spina V, Hodkinson B, Forestieri G, Bonfiglio F, Condoluci A, Wu W, Pirosa MC, Faderl MR, Koch R, Schaffer M, Alvarez JD, Fourneau N, Gerber B, Stussi G, Zucca E, Balasubramanian S, Rossi D. Circulating tumor DNA for comprehensive noninvasive monitoring of lymphoma treated with ibrutinib plus nivolumab. Blood Adv 2021; 5:4674-4685. [PMID: 34500472 PMCID: PMC8759132 DOI: 10.1182/bloodadvances.2021004528] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/04/2021] [Indexed: 11/20/2022] Open
Abstract
To advance the use of circulating tumor DNA (ctDNA) applications, their broad clinical validity must be tested in different treatment settings, including targeted therapies. Using the prespecified longitudinal systematic collection of plasma samples in the phase 1/2a LYM1002 trial (registered on www.clinicaltrials.gov as NCT02329847), we tested the clinical validity of ctDNA for baseline mutation profiling, residual tumor load quantification, and acquisition of resistance mutations in patients with lymphoma treated with ibrutinib+nivolumab. Inclusion criterion for this ancillary biological study was the availability of blood collected at baseline and cycle 3, day 1. Overall, 172 ctDNA samples from 67 patients were analyzed by the LyV4.0 ctDNA Cancer Personalized Profiling Deep Sequencing Assay. Among baseline variants in ctDNA, only TP53 mutations (detected in 25.4% of patients) were associated with shorter progression-free survival; clones harboring baseline TP53 mutations did not disappear during treatment. Molecular response, defined as a >2-log reduction in ctDNA levels after 2 cycles of therapy (28 days), was achieved in 28.6% of patients with relapsed diffuse large B-cell lymphoma who had ≥1 baseline variant and was associated with best response and improved progression-free survival. Clonal evolution occurred frequently during treatment, and 10.3% new mutations were identified after 2 treatment cycles in nonresponders. PLCG2 was the topmost among genes that acquired new mutations. No patients acquired the C481S BTK mutation implicated in resistance to ibrutinib in CLL. Collectively, our results provide the proof of concept that ctDNA is useful for noninvasive monitoring of lymphoma treated with targeted agents in the clinical trial setting.
Collapse
Affiliation(s)
- Alessio Bruscaggin
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | | | - Valeria Spina
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | | | - Gabriela Forestieri
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Ferdinando Bonfiglio
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Adalgisa Condoluci
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Wei Wu
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Maria C. Pirosa
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Martin R. Faderl
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Ricardo Koch
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | | | | | | | - Bernhard Gerber
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Georg Stussi
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | | | | | - Davide Rossi
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
95
|
Peng Y, Mei W, Ma K, Zeng C. Circulating Tumor DNA and Minimal Residual Disease (MRD) in Solid Tumors: Current Horizons and Future Perspectives. Front Oncol 2021; 11:763790. [PMID: 34868984 PMCID: PMC8637327 DOI: 10.3389/fonc.2021.763790] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
Circulating tumor DNA (ctDNA) is cell-free DNA (cfDNA) fragment in the bloodstream that originates from malignant tumors or circulating tumor cells. Recently, ctDNA has emerged as a promising non-invasive biomarker in clinical oncology. Analysis of ctDNA opens up new avenues for individualized cancer diagnosis and therapy in various types of tumors. Evidence suggests that minimum residual disease (MRD) is closely associated with disease recurrence, thus identifying specific genetic and molecular alterations as novel MRD detection targets using ctDNA has been a research focus. MRD is considered a promising prognostic marker to identify individuals at increased risk of recurrence and who may benefit from treatment. This review summarizes the current knowledge of ctDNA and MRD in solid tumors, focusing on the potential clinical applications and challenges. We describe the current state of ctDNA detection methods and the milestones of ctDNA development and discuss how ctDNA analysis may be an alternative for tissue biopsy. Additionally, we evaluate the clinical utility of ctDNA analysis in solid tumors, such as recurrence risk assessment, monitoring response, and resistance mechanism analysis. MRD detection aids in assessing treatment response, patient prognosis, and risk of recurrence. Moreover, this review highlights current advancements in utilizing ctDNA to monitor the MRD of solid tumors such as lung cancer, breast cancer, and colon cancer. Overall, the clinical application of ctDNA-based MRD detection can assist clinical decision-making and improve patient outcomes in malignant tumors.
Collapse
Affiliation(s)
- Yan Peng
- Department of Obstetrics, Longhua District Central Hospital, Shenzhen, China
| | - Wuxuan Mei
- Clinical Medical College, Hubei University of Science and Technology, Xianning, China
| | - Kaidong Ma
- Department of Obstetrics, Longhua District Central Hospital, Shenzhen, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, China
| |
Collapse
|
96
|
Li N, Wang B, Li J, Shao Y, Li M, Li J, Kuang P, Liu Z, Sun T, Wu H, Ou W, Wang S. Perioperative circulating tumor DNA as a potential prognostic marker for operable stage I to IIIA non–small cell lung cancer. Cancer 2021; 128:708-718. [DOI: 10.1002/cncr.33985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/06/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022]
Affiliation(s)
- Ning Li
- Department of Breast Oncology Sun Yat‐Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou China
| | - Bao‐Xiao Wang
- Department of Otolaryngology, Head and Neck Surgery Sun Yat‐Sen Memorial HospitalSun Yat‐Sen University Guangzhou China
| | - Jian Li
- Department of Diagnostic and Interventional Ultrasound Sun Yat‐Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou China
| | - Yang Shao
- Nanjing Geneseeq Technology, Inc Nanjing China
- School of Public Health Nanjing Medical University Nanjing China
| | - Ming‐Tian Li
- Department of Operating Center Sun Yat‐Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou China
| | - Jian‐Jun Li
- Department of Endoscopy Sun Yat‐Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou China
| | - Peng‐Peng Kuang
- Otorhinolaryngology HospitalFirst Affiliated HospitalSun Yat‐Sen University Guangzhou China
| | - Zui Liu
- Division of Cardiac Surgery Heart Center First Affiliated HospitalSun Yat‐Sen University Guangzhou China
| | - Tian‐Yu Sun
- Department of Thoracic Surgery Sun Yat‐Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou China
| | - Hui‐Qi Wu
- Department of Thoracic Surgery Sun Yat‐Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou China
| | - Wei Ou
- Department of Thoracic Surgery Sun Yat‐Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou China
| | - Si‐Yu Wang
- Department of Thoracic Surgery Sun Yat‐Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou China
| |
Collapse
|
97
|
Imyanitov E, Kuligina E. Molecular testing for colorectal cancer: Clinical applications. World J Gastrointest Oncol 2021; 13:1288-1301. [PMID: 34721767 PMCID: PMC8529925 DOI: 10.4251/wjgo.v13.i10.1288] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/19/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
Molecular genetic analysis is an integral part of colorectal cancer (CRC) management. The choice of systemic therapy for CRC is largely based on the results of tumor molecular testing. Evaluation of the KRAS and NRAS gene status is mandatory for consideration of anti-epidermal growth factor receptor (EGFR) therapy. Tumors with the BRAF V600E substitution are characterized by aggressive behaviour, may require intensified cytotoxic regimens and benefit from combined BRAF and EGFR inhibition. The inactivation of DNA mismatch repair (MMR), or MUTYH gene, or DNA polymerase epsilon results in excessive tumor mutational burden; these CRCs are highly antigenic and therefore sensitive to immune checkpoint inhibitors. Some CRCs are characterized by overexpression of the HER2 oncogene and respond to the appropriate targeted therapy. There are CRCs with clinical signs of hereditary predisposition to this disease, which require germline genetic testing. Liquid biopsy is an emerging technology that has the potential to assist CRC screening, control the efficacy of surgical intervention and guide disease monitoring. The landscape of CRC molecular diagnosis is currently undergoing profound changes due to the increasing use of next generation sequencing.
Collapse
Affiliation(s)
- Evgeny Imyanitov
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, Saint-Petersburg 194100, Russia
- Department of Oncology, I.I. Mechnikov North-Western Medical University, Saint-Petersburg 191015, Russia
| | - Ekaterina Kuligina
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
| |
Collapse
|
98
|
Clinical Applications of Minimal Residual Disease Assessments by Tumor-Informed and Tumor-Uninformed Circulating Tumor DNA in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13184547. [PMID: 34572774 PMCID: PMC8471730 DOI: 10.3390/cancers13184547] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Circulating tumor DNA, or ctDNA, are fragments of tumor DNA that can be detected in the blood of patients with colorectal cancer. Measuring ctDNA levels in the blood has shown the potential to provide important information that can be helpful in the clinical care of patients with colorectal cancer. For example, in patients with colon cancer that has been removed by surgery, measuring ctDNA in the blood can predict the likelihood of cancer recurrence, while in those with metastatic colorectal cancer, measuring ctDNA can inform the clinician whether chemotherapy is effective at earlier timepoints than currently available tests. In this review, we discuss the results from ongoing studies describing the utility of ctDNA measurements across all stages of colorectal cancer. We also discuss the various clinical scenarios that ctDNA may have the most immediate impact in colorectal cancer management. Abstract Emerging data suggest that circulating tumor DNA (ctDNA) can detect colorectal cancer (CRC)-specific signals across both non-metastatic and metastatic settings. With the development of multiple platforms, including tumor-informed and tumor-agnostic ctDNA assays and demonstration of their provocative analytic performance to detect minimal residual disease, there are now ongoing, phase III randomized clinical trials to evaluate their role in the management paradigm of CRC. In this review, we highlight landmark studies that have formed the basis for ongoing studies on the clinically applicability of plasma ctDNA assays in resected, stage I–III CRC and metastatic CRC. We discuss clinical settings by which ctDNA may have the most immediate impact in routine clinical practice. These include the potential for ctDNA to (1) guide surveillance and intensification or de-intensification strategies of adjuvant therapy in resected, stage I–III CRC, (2) predict treatment response to neoadjuvant therapy in locally advanced rectal cancer inclusive of total neoadjuvant therapy (TNT), and (3) predict response to systemic and surgical therapies in metastatic disease. We end by considering clinical variables that can influence our ability to reliably interpret ctDNA dynamics in the clinic.
Collapse
|
99
|
Postoperative Circulating Tumor DNA Can Predict High Risk Patients with Colorectal Cancer Based on Next-Generation Sequencing. Cancers (Basel) 2021; 13:cancers13164190. [PMID: 34439344 PMCID: PMC8391973 DOI: 10.3390/cancers13164190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Circulating tumor DNA (ctDNA) is a minimally invasive biomarker useful for monitoring minimum residual disease, recurrence, and treatment response in colorectal cancer (CRC). We analyzed circulating tumor DNA from patients with CRC to evaluate analytical and clinical performances using next-generation sequencing (NGS). It is clear that postoperative circulating tumor DNA detection provides valuable information to determine whether a patient might at high risk of disease recurrence or have a persistent tumor lesion. The NGS assay not only showed excellent analytical performance, but also shows a state-of-art diagnostic option in patient-oriented precision medicine. Abstract The objective of this study was to characterize circulating tumor DNA (ctDNA) mutations in colorectal cancer (CRC) patients and evaluate their prognostic values during treatment. Forty-nine patients with CRC planned for operation were enrolled. A total of 115 plasma samples were collected pre-operation, post-operation, and post-chemotherapy. ctDNA analysis was performed using next-generation sequencing (NGS) including 14 genes. In 22 (44.9%) out of 49 patients, at least one mutation (40 total mutations) was detected in the initial plasma sample. The median sum of variant allele frequency was 0.74% (range: 0.10–29.57%). TP53 mutations were the most frequent (17 of 49 patients, 34.7%), followed by APC (18.4%), KRAS (12.2%), FBXW7 (8.2%), NRAS (2.0%), PIK3CA (2.0%), and SMAD4 (2.0%). After surgery, five (14.3%) out of 35 patients harbored ctDNA mutation. All five patients experienced relapse or metastasis during follow-up. It was noteworthy that all three patients with persistent ctDNA relapsed after R0 resection. After chemotherapy, ctDNA analysis was performed for 31 patients, all of which were ctDNA-negative. Analytical and clinical performances of NGS to utilize ctDNA in CRC were determined. Results revealed that postoperative ctDNA might serve as a marker for identifying risk of recurrence, thus contributing to patient-oriented treatment strategies.
Collapse
|
100
|
Tarazona N, Gimeno-Valiente F, Gambardella V, Huerta M, Roselló S, Zuniga S, Calon A, Carbonell-Asins JA, Fontana E, Martinez-Ciarpaglini C, Eason K, Rentero-Garrido P, Fleitas T, Papaccio F, Moro-Valdezate D, Nyamundanda G, Castillo J, Espí A, Sadanandam A, Roda D, Cervantes A. Detection of postoperative plasma circulating tumour DNA and lack of CDX2 expression as markers of recurrence in patients with localised colon cancer. ESMO Open 2021; 5:e000847. [PMID: 32967918 PMCID: PMC7513635 DOI: 10.1136/esmoopen-2020-000847] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/18/2020] [Accepted: 08/10/2020] [Indexed: 01/05/2023] Open
Abstract
Background Colon cancer (CC) is a heterogeneous disease. Novel prognostic factors beyond pathological staging are required to accurately identify patients at higher risk of relapse. Integrating these new biological factors, such as plasma circulating tumour DNA (ctDNA), CDX2 staining, inflammation-associated cytokines and transcriptomic consensus molecular subtypes (CMS) classification, into a multimodal approach may improve our accuracy in determining risk of recurrence. Methods One hundred and fifty patients consecutively diagnosed with localised CC were prospectively enrolled in our study. ctDNA was tracked to detect minimal residual disease by droplet digital PCR. CDX2 expression was analysed by immunostaining. Plasma levels of cytokines potentially involved in disease progression were measured using ELISAs. A 96 custom gene panel for nCounter assay was used to classify CC into colorectal cancer assigner and CMS. Results Most patients were classified into CMS4 (37%) and CMS2 (28%), followed by CMS1 (20%) and CMS3 (15%) groups. CDX2-negative tumours were enriched in CMS1 and CMS4 subtypes. In univariable analysis, prognosis was influenced by primary tumour location, stage, vascular and perineural invasion together with high interleukin-6 plasma levels at baseline, tumours belonging to CMS 1 vs CMS2 +CMS3, ctDNA presence in plasma and CDX2 loss. However, only positive ctDNA in plasma samples (HR 13.64; p=0.002) and lack of CDX2 expression (HR 23.12; p=0.001) were found to be independent prognostic factors for disease-free survival in the multivariable model. Conclusions ctDNA detection after surgery and lack of CDX2 expression identified patients at very high risk of recurrence in localised CC.
Collapse
Affiliation(s)
- Noelia Tarazona
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CIBERONC, University of Valencia, Valencia, Spain
| | - Francisco Gimeno-Valiente
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - Valentina Gambardella
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CIBERONC, University of Valencia, Valencia, Spain
| | - Marisol Huerta
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - Susana Roselló
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CIBERONC, University of Valencia, Valencia, Spain
| | | | - Alexandre Calon
- Cancer Research Programme, Hospital del Mar Research Institute (IMIM), Barcelona, Catalunya, Spain
| | | | - Elisa Fontana
- Division of Molecular Pathology, Institute of cancer Research, London, UK
| | | | - Katherine Eason
- Division of Molecular Pathology, Institute of cancer Research, London, UK
| | | | - Tania Fleitas
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CIBERONC, University of Valencia, Valencia, Spain
| | - Federica Papaccio
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - David Moro-Valdezate
- Department of Surgery, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - Gift Nyamundanda
- Division of Molecular Pathology, Institute of cancer Research, London, UK
| | - Josefa Castillo
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CIBERONC, University of Valencia, Valencia, Spain
| | - Alejandro Espí
- Department of Surgery, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - Anguraj Sadanandam
- Division of Molecular Pathology, Institute of cancer Research, London, UK
| | - Desamparados Roda
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CIBERONC, University of Valencia, Valencia, Spain
| | - Andrés Cervantes
- Department of Medical Oncology, Biomedical Research Institute INCLIVA, CIBERONC, University of Valencia, Valencia, Spain.
| |
Collapse
|