1
|
Rosenlund L, Guldbrandsen K, Ahlborn LB, Bloch M, Skougaard K, Albrecht-Beste E, Nellemann HM, Krakauer M, Gørtz PM, Fledelius J, Nielsen AL, Holdgaard PC, Nielsen SS, Grüner JM, Højsgaard A, Petersen RH, Møller LB, Dahl M, Frank MS, Ehlers JH, Saghir Z, Pøhl M, Borissova S, Land LH, Kristiansen C, McCulloch T, Mortensen LS, Christophersen MS, Hilberg O, Rasmussen TL, Simonsen Schwaner SH, Laursen CB, Bodtger U, Lonsdale MN, Meyer CN, Gerke O, Mortensen J, Rasmussen TR, Hjorthaug K, Larsen KR, Meldgaard P, Fischer BM, Sorensen BS. ctDNA can detect minimal residual disease in curative treated non-small cell lung cancer patients using a tumor agnostic approach. Lung Cancer 2025; 203:108528. [PMID: 40220718 DOI: 10.1016/j.lungcan.2025.108528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) has the potential to become a reliable biomarker for identifying minimal residual disease (MRD) and predicting recurrence in patients with non-small cell lung cancer (NSCLC) following curative treatment. However, there is a lack of studies that investigate the clinical validity of ctDNA using a tumor-agnostic approach, which can provide significant clinical benefits. METHODS We analyzed samples from 45 NSCLC patients recruited in a prospective national multicenter study, all of whom had undergone curative treatment. A total of 38 pre-treatment plasma samples and 76 post-treatment plasma samples were examined using a commercially available cancer personalized profiling by deep sequencing (CAPP-seq) strategy, and a tumor-agnostic approach. Post-treatment samples were collected at two distinct landmark time points: Follow-up 1 (0.5-4.5 months post-treatment) and Follow-up 2 (4.5-7.5 months post-treatment). RESULTS Detectable ctDNA post-treatment was significantly associated with increased risk of tumor recurrence and shorter recurrence-free survival (RFS). Using only a single blood sample taken from Follow-up 2, we correctly identified MRD in 50% of the patients who later experienced recurrence. However, subgroup analysis further revealed that in patients treated with radiotherapy or chemoradiotherapy (CRT), ctDNA detection was significantly linked to shorter RFS in the MRD analysis from Follow-up 2, but not in the MRD analysis from Follow-up 1. CONCLUSION These findings suggest that post-treatment ctDNA, detected using a tumor-agnostic approach, is a reliable biomarker for predicting recurrence in NSCLC patients following curative treatment. However, the optimal timing for blood sampling to detect MRD appears to depend on the type of curative treatment received.
Collapse
Affiliation(s)
- Lærke Rosenlund
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Kasper Guldbrandsen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Lise Barlebo Ahlborn
- Department of Genomic Medicine, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Martin Bloch
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Kristin Skougaard
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark; Department of Oncology, University Hospital of Southern Denmark - Roskilde, Denmark
| | - Elisabeth Albrecht-Beste
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Martin Krakauer
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Peter Michael Gørtz
- Department of Nuclear Medicine, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| | - Joan Fledelius
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | | | - Paw Christian Holdgaard
- Department of Nuclear Medicine, University Hospital of Southern Denmark, Lillebaelt Hospital - Vejle, Denmark
| | - Søren Steen Nielsen
- Department of Nuclear Medicine, Aalborg University Hospital, Aalborg, Denmark
| | - Julie Marie Grüner
- Department of Clinical Physiology and Nuclear Medicine, Zealand University Hospital - Køge, Denmark
| | - Anette Højsgaard
- Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Rene Horsleben Petersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Cardiothoracic Surgery, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Morten Dahl
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Biochemistry, Zealand University Hospital - Køge, Denmark
| | - Malene Støchkel Frank
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Oncology and Palliative Care, Zealand University Hospital, Denmark
| | - Jeanette Haar Ehlers
- Department of Oncology, University Hospital of Southern Denmark - Roskilde, Denmark; Medicin 2, Holbæk Hospital, Holbæk, Denmark
| | - Zaigham Saghir
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Medicine, Section of Pulmonary Medicine, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| | - Mette Pøhl
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Svetlana Borissova
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Lotte Holm Land
- Department of Oncology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Charlotte Kristiansen
- Department of Oncology, University Hospital of Southern Denmark, Lillebaelt Hospital - Vejle, Denmark
| | - Tine McCulloch
- Department of Oncology, Aalborg University Hospital, Aalborg Denmark; Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | | | - Malene Søby Christophersen
- Department of Respiratory Disease, University Hospital of Southern Denmark, Lillebaelt Hospital - Vejle, Denmark; Department of Emergency Medicine, Regional Hospital Horsens, Horsens, Denmark
| | - Ole Hilberg
- Department of Respiratory Disease, University Hospital of Southern Denmark, Lillebaelt Hospital - Vejle, Denmark; Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Thor Lind Rasmussen
- Department of Respiratory Diseases, Aalborg University Hospital, Aalborg, Denmark
| | - Signe Høyer Simonsen Schwaner
- Department of Respiratory Medicine, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Christian B Laursen
- Department of Respiratory Medicine, Odense University Hospital, Odense, Denmark; Odense Respiratory Research Unit (ODIN), Department of Clinical Medicine, University of Southern Denmark, Odense, Denmark
| | - Uffe Bodtger
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark; Respiratory Research Unit PLUZ, Department of Respiratory Medicine, Zealand University Hospital - Næstved, Denmark
| | - Markus Nowak Lonsdale
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Christian Niels Meyer
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Medicine, Zealand University Hospital - Roskilde, Denmark
| | - Oke Gerke
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jann Mortensen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Riis Rasmussen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Respiratory Medicine and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | - Karin Hjorthaug
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Klaus Richter Larsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Respiratory Medicine, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Peter Meldgaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Barbara Malene Fischer
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
2
|
van der Wilk BJ, Eyck BM, Wijnhoven BPL, Lagarde SM, Rosman C, Noordman BJ, Valkema MJ, Bisseling TM, Coene PPLO, van Det MJ, Dekker JWT, van Dieren JM, Doukas M, van Esser S, Fiets WE, Hartgrink HH, Heisterkamp J, Holster IL, Klarenbeek B, van Klaveren D, Kouw E, Kouwenhoven EA, Luyer MD, Mostert B, Nieuwenhuijzen GAP, Oostenbrug LE, Pierie JP, van Sandick JW, Sosef MN, Spaander MCW, Valkema R, van der Zaag ES, Steyerberg EW, van Lanschot JJB. Neoadjuvant chemoradiotherapy followed by active surveillance versus standard surgery for oesophageal cancer (SANO trial): a multicentre, stepped-wedge, cluster-randomised, non-inferiority, phase 3 trial. Lancet Oncol 2025; 26:425-436. [PMID: 40112851 DOI: 10.1016/s1470-2045(25)00027-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 01/03/2025] [Accepted: 01/20/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND A substantial proportion of individuals with oesophageal cancer have a pathological complete response after neoadjuvant chemoradiotherapy and oesophagectomy. We aimed to investigate whether active surveillance could be an alternative for individuals with a clinical complete response after neoadjuvant chemoradiotherapy. METHODS We performed a multicentre, stepped-wedge, cluster-randomised, non-inferiority, phase 3 trial in 12 Dutch hospitals. Individuals with locally advanced oesophageal cancer and a clinical complete response after neoadjuvant chemoradiotherapy (ie, no tumour detected with endoscopic biopsies, ultrasound, and PET-CT) underwent active surveillance or standard surgery (ie, oesophagectomy within 2 weeks after reaching clinical complete response). There were no inclusion restrictions regarding comorbidities or performance status, but participants had carcinoma, were age 18 years or older, and were treated with curative intent. Randomisation of hospitals was performed using computer-generated sequences without stratification methods, after an initial phase of all hospitals performing standard surgery. The primary endpoint was overall survival, analysed according to a modified intention-to-treat principle (allowing crossover at time of clinical complete response) and an intention-to-treat principle. Non-inferiority was defined as 2-year survival rate for active surveillance of 15% or less below that for standard surgery. The trial was registered within the Netherlands Trial Register, NTR-6803, and the inclusion phase has been completed. FINDINGS Between Nov 8, 2017, and Jan 17, 2021, 1115 individuals were screened, of whom 309 were included. 198 underwent active surveillance and 111 underwent standard surgery. 242 (78%) participants were male and 67 (22%) were female. Median follow-up was 38 months (IQR 32-48). 2-year overall survival for active surveillance (74% [95% CI 69-78]) was non-inferior to standard surgery (71% [62-78]) after modified intention-to-treat analysis (one-sided 95% boundary: 7% lower). It remained non-inferior in the intention-to-treat analysis (75% [68-80] vs 70% [63-77], one-sided 95% boundary: 6% lower). There were no significant differences in overall survival according to modified intention-to-treat analysis (hazard ratio 1·14, two-sided 95% CI 0·74-1·78) or intention-to-treat analysis (0·83, 0·53-1·31). The frequency of postoperative complications and postoperative mortality after standard surgery or postponed surgery after active surveillance was similar between groups. INTERPRETATION Overall survival after active surveillance for oesophageal cancer was non-inferior compared with standard surgery after 2 years. For the long-term efficacy of active surveillance, extended follow-up is required. The results of the present trial could be used for patient counselling and shared decision making. FUNDING Dutch Cancer Society (KWF) and Netherlands Organisation for Health Research and Development (ZonMw).
Collapse
Affiliation(s)
- Berend J van der Wilk
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands.
| | - Ben M Eyck
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | - Bas P L Wijnhoven
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | - Sjoerd M Lagarde
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | - Camiel Rosman
- Department of Surgery, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Bo J Noordman
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | - Maria J Valkema
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | - Tanya M Bisseling
- Department of Gastroenterology and Hepatology, Radboud University Medical Centre, Nijmegen, Netherlands
| | | | - Marc J van Det
- Department of Surgery, ZGT Hospital, Almelo, Netherlands
| | | | - Jolanda M van Dieren
- Department of Gastrointestinal Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | - Stijn van Esser
- Department of Surgery, Reinier de Graaf Gasthuis, Delft, Netherlands
| | - W Edward Fiets
- Department of Medical Oncology, Medical Centre Leeuwarden, Leeuwarden, Netherlands
| | - Henk H Hartgrink
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - Joos Heisterkamp
- Department of Surgery, Elisabeth Tweesteden Hospital, Tilburg, Netherlands
| | - I Lisanne Holster
- Department of Gastroenterology, Maasstad Hospital, Rotterdam, Netherlands
| | - Bastiaan Klarenbeek
- Department of Surgery, Radboud University Medical Centre, Nijmegen, Netherlands
| | - David van Klaveren
- Department of Public Health, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | - Eva Kouw
- Department of Gastroenterology and Hepatology, Gelre Hospital, Apeldoorn, Netherlands
| | | | - Misha D Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | | | | | - Jean-Pierre Pierie
- Department of Surgery, Medical Centre Leeuwarden, Leeuwarden, Netherlands
| | - Johanna W van Sandick
- Department of Surgical Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Meindert N Sosef
- Department of Surgery, Zuyderland Medical Centre, Heerlen, Netherlands
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | - Roelf Valkema
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | | | - Ewout W Steyerberg
- Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
| | - J Jan B van Lanschot
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| |
Collapse
|
3
|
Kobayashi S, Nakamura Y, Hashimoto T, Bando H, Oki E, Karasaki T, Horinouchi H, Ozaki Y, Iwata H, Kato T, Miyake H, Ohba A, Ikeda M, Chiyoda T, Hasegawa K, Fujisawa T, Matsuura K, Namikawa K, Yajima S, Yoshino T, Hasegawa K. Japan society of clinical oncology position paper on appropriate clinical use of molecular residual disease (MRD) testing. Int J Clin Oncol 2025; 30:605-654. [PMID: 39920551 PMCID: PMC11946966 DOI: 10.1007/s10147-024-02683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 12/09/2024] [Indexed: 02/09/2025]
Abstract
Although the 5-year relative survival rates for resectable solid tumors have improved over the past few years, the risk of postoperative recurrence necessitates effective monitoring strategies. Recent advancements in molecular residual disease (MRD) testing based on circulating tumor DNA (ctDNA) analysis have shown considerable promise in the context of predicting recurrence; however, significant barriers to widespread clinical implementation remain-mainly, low awareness among healthcare professionals, high costs, and lack of standardized assays and comprehensive evidence. This position paper, led by the Japan Society of Clinical Oncology, aims to establish a common framework for the appropriate clinical use of MRD testing in a tumor type-agnostic manner. It synthesizes currently available evidence, reviews region-specific clinical trends, addresses critical clinical questions related to MRD testing, and offers recommendations to guide healthcare professionals, biotechnology and pharmaceutical companies, and regulatory authorities. These recommendations were developed based on a voting process involving 15 expert members, ensuring a consensus-driven approach. These findings underscore the importance of collaborative efforts among various stakeholders in enhancing the clinical utility of MRD testing. This project aimed to foster consensus and provide clear guidelines to support the advancement of precision medicine in oncology and improve patient outcomes in the context of perioperative care.
Collapse
Affiliation(s)
- Shin Kobayashi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
- Perioperative Treatment Development Promotion Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
| | - Yoshiaki Nakamura
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
- Translational Research Support Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
- International Research Promotion Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
| | - Tadayoshi Hashimoto
- Perioperative Treatment Development Promotion Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Translational Research Support Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Hideaki Bando
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Translational Research Support Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Karasaki
- Department of Thoracic Surgery, Respiratory Center, Toranomon Hospital, Tokyo, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yukinori Ozaki
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroji Iwata
- Department of Advanced Clinical Research and Development, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Taigo Kato
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hideaki Miyake
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akihiro Ohba
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Takao Fujisawa
- Translational Research Support Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kazuto Matsuura
- Department of Head and Neck Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shugo Yajima
- Department of Urology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Kiyoshi Hasegawa
- Department of Surgery, Graduate School of Medicine, Hepato-Biliary-Pancreatic Surgery Division, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Mukherjee S. Non-surgical approach to operable oesophageal cancer: is it prime time yet? Lancet Oncol 2025; 26:400-401. [PMID: 40112847 DOI: 10.1016/s1470-2045(25)00072-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/22/2025]
Affiliation(s)
- Somnath Mukherjee
- Oxford Cancer Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7LE, UK.
| |
Collapse
|
5
|
Jiang W, Zhang B, Xu J, Xue L, Wang L. Current status and perspectives of esophageal cancer: a comprehensive review. Cancer Commun (Lond) 2025; 45:281-331. [PMID: 39723635 PMCID: PMC11947622 DOI: 10.1002/cac2.12645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Esophageal cancer (EC) continues to be a significant global health concern, with two main subtypes: esophageal squamous cell carcinoma and esophageal adenocarcinoma. Prevention and changes in etiology, improvements in early detection, and refinements in the treatment have led to remarkable progress in the outcomes of EC patients in the past two decades. This seminar provides an in-depth analysis of advances in the epidemiology, disease biology, screening, diagnosis, and treatment landscape of esophageal cancer, focusing on the ongoing debate surrounding multimodality therapy. Despite significant advancements, EC remains a deadly disease, underscoring the need for continued research into early detection methods, understanding the molecular mechanisms, and developing effective treatments.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Radiation OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenGuangdongP. R. China
| | - Bo Zhang
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Jiaqi Xu
- Department of PathologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Liyan Xue
- Department of PathologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Luhua Wang
- Department of Radiation OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenGuangdongP. R. China
| |
Collapse
|
6
|
Huizer TJ, Lagarde SM, Nuyttens JJ, Oudijk L, Spaander MC, Valkema R, Mostert B, Wijnhoven BP. Active surveillance in patients with a complete clinical response after neoadjuvant chemoradiotherapy for esophageal- and gastroesophageal junction cancer. Innov Surg Sci 2025; 10:11-19. [PMID: 40144783 PMCID: PMC11934941 DOI: 10.1515/iss-2023-0010] [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: 03/02/2023] [Accepted: 07/30/2024] [Indexed: 03/28/2025] Open
Abstract
Neoadjuvant chemoradiotherapy in patients with esophageal- and gastroesophageal junction cancer induces tumor regression. In approximately one fourth of patients, this leads to a pathological complete response in the resection specimen. Hence, active surveillance may be an alternative strategy in patients without residual disease after neoadjuvant chemoradiotherapy. Previous studies have shown that the combination of esophagogastroduodenoscopy with bite-on-bite biopsies, endoscopic ultrasound with fine needle aspiration of suspected lymph nodes, and a PET-CT-scan can be considered adequate for the detection of residual disease. So far, it has been unclear whether active surveillance with surgery as needed is a safe treatment option and leads to non-inferior overall survival compared to standard esophagectomy after neoadjuvant chemoradiotherapy. This review will discuss the current status of active surveillance for esophageal and junctional cancer.
Collapse
Affiliation(s)
- Tamara J. Huizer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Sjoerd M. Lagarde
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Joost J.M.E. Nuyttens
- Department of Radiation Oncology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Lindsey Oudijk
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Manon C.W. Spaander
- Department of Gastroenterology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Roelf Valkema
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Bas P.L. Wijnhoven
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - SANO- study group
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Radiation Oncology, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Gastroenterology, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
7
|
Hosoya H, Carleton M, Tanaka K, Sworder B, Syal S, Sahaf B, Maltos AM, Silva O, Stehr H, Hovanky V, Duran G, Zhang T, Liedtke M, Arai S, Iberri D, Miklos D, Khodadoust MS, Sidana S, Kurtz DM. Deciphering response dynamics and treatment resistance from circulating tumor DNA after CAR T-cells in multiple myeloma. Nat Commun 2025; 16:1824. [PMID: 39979252 PMCID: PMC11842827 DOI: 10.1038/s41467-025-56486-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
Despite advances in treatments, multiple myeloma (MM) remains an incurable cancer where relapse is common. We developed a circulating tumor DNA (ctDNA) approach in order to characterize tumor genomics, monitor treatment response, and detect early relapse in MM. By sequencing 412 specimens from 64 patients with newly diagnosed or relapsed/refractory disease, we demonstrate the correlation between ctDNA and key clinical biomarkers, as well as patient outcomes. We further extend our approach to simultaneously track CAR-specific cell-free DNA (CAR-cfDNA) in patients undergoing anti-BCMA CAR T-cell (BCMA-CAR) therapy. We demonstrate that ctDNA levels following BCMA-CAR inversely correlate with relative time to progression (TTP), and that measurable residual disease (MRD) quantified by peripheral blood ctDNA (ctDNA-MRD) was concordant with clinical bone marrow MRD. Finally, we show that ctDNA-MRD can anticipate clinical relapse and identify the emergence of genomically-defined therapy-resistant clones. These findings suggest multiple clinical uses of ctDNA for MM in molecular characterization and disease surveillance.
Collapse
Affiliation(s)
- Hitomi Hosoya
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Mia Carleton
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Kailee Tanaka
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Brian Sworder
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine CA, USA
| | - Shriya Syal
- Center for Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Bita Sahaf
- Center for Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Alisha M Maltos
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Oscar Silva
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Henning Stehr
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Vanna Hovanky
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - George Duran
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Tian Zhang
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Michaela Liedtke
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Sally Arai
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - David Iberri
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - David Miklos
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Michael S Khodadoust
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Surbhi Sidana
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA.
| | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Liu Z, Shi Z, Jiang W, Shen Z, Chen W, Shen K, Sun Y, Tang Z, Wang X. Circulating tumor DNA analysis for prediction of prognosis and molecular insights in patients with resectable gastric cancer: results from a prospective study. MedComm (Beijing) 2025; 6:e70065. [PMID: 39830022 PMCID: PMC11742430 DOI: 10.1002/mco2.70065] [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: 06/02/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
This study aimed to evaluate the prognostic value of plasma circulating tumor DNA (ctDNA) level in patients with resectable gastric cancer (GC). A total of 59 patients were prospectively enrolled, with their ctDNA detected and paired tumor tissue collected at various peri-operative time points. Patients with higher 1-month post-operative ctDNA levels demonstrated shorter overall survival status (hazard ratio [HR] = 5.30, p = 0.0022) and a higher risk of recurrence (HR = 3.85, p = 0.011). The model combining ctDNA with conventional serum tumor markers for GC, including carcinoembryonic antigen, carbohydrate antigen 19-9, and CA72-4, shows high predictive effectiveness for GC prognosis with an area under the curve of 0.940 (p = 0.002), which is higher than net ctDNA and other models without ctDNA. Patients with lower ctDNA levels were more likely to have positive stromal programmed cell death ligand 1 expression (p = 0.046). Additionally, DCAF4L2 mutation was identified as the crucial gene mutation in ctDNA suggesting poor prognosis of patients with GC. Overall, this study highlights that post-operative ctDNA can serve as an effective biomarker for prognostic prediction and recurrence surveillance in resectable GC.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zhongyi Shi
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Wenchao Jiang
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhenbin Shen
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Weidong Chen
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Kuntang Shen
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Yihong Sun
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Department of General SurgeryZhongshan Hospital (Xiamen Branch)Fudan UniversityShanghaiChina
| | - Zhaoqing Tang
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Department of General SurgeryZhongshan Hospital (Xiamen Branch)Fudan UniversityShanghaiChina
| | - Xuefei Wang
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Department of General SurgeryZhongshan Hospital (Xiamen Branch)Fudan UniversityShanghaiChina
| |
Collapse
|
9
|
Li J, Wu C, Song Y, Fan Y, Li C, Li H, Zhang S. Exploring the Clinical Value of Perioperative ctDNA-Based Detection of Molecular Residual Disease in Patients With Esophageal Squamous Cell Carcinoma. Thorac Cancer 2025; 16:e70017. [PMID: 39966084 PMCID: PMC11835505 DOI: 10.1111/1759-7714.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE To explore the clinical value of molecular residual disease detection based on circulating tumor DNA (ctDNA-MRD) in the perioperative period of esophageal squamous cell carcinoma (ESCC) and to analyze the tumor escape mechanisms in MRD-positive cases. METHODS A total of 35 ESCC patients were prospectively enrolled. Preoperative and postoperative (1 month after surgery) blood and surgical tissue samples were analyzed. ctDNA variants were tracked in plasma to assess ctDNA-MRD, and whole-transcriptome sequencing was performed on MRD-positive and MRD-negative tissue samples. RESULTS Preoperative blood ctDNA was positive in 54.3% of patients, with a 31.6% positive predictive value for recurrence. One month postsurgery, the positive rate of ctDNA was 17.1%, with an 83.3% predictive value for recurrence. Both preoperative and postoperative ctDNA positivity were significant prognostic indicators (HR = 2.78, p < 0.05; HR = 4.42, p < 0.001). Multivariate analysis confirmed ctDNA as an independent prognostic factor (HR = 303.75, p < 0.001). Transcriptomic analysis revealed increased macrophage (W = 15 848; p < 0.01) and follicular helper T (Tfh) cell (W = 10 935; p < 0.01) levels in MRD-positive patients, suggesting a potential link to immune escape in tumors. CONCLUSIONS Plasma ctDNA measured 1 month postoperatively in ESCC patients can effectively detect MRD, and ctDNA-MRD serves as an independent risk factor for postoperative recurrence. The mechanism underlying MRD positivity may involve the polarization of Tfh cells and macrophages, aiding tumor cells in immune escape through the bloodstream.
Collapse
Affiliation(s)
- Jimin Li
- Department of Thoracic SurgeryCancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical SciencesTaiyuanChina
| | - Congcong Wu
- Department of Thoracic SurgeryCancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical SciencesTaiyuanChina
| | - Yongming Song
- Department of Thoracic SurgeryShanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Yuhui Fan
- Department of Thoracic SurgeryShanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Chao Li
- Department of Thoracic SurgeryShanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Haibo Li
- Department of Thoracic SurgeryJincheng Second People's HospitalJinchengChina
| | - Shuangping Zhang
- Department of Thoracic SurgeryCancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical SciencesTaiyuanChina
- Department of Thoracic SurgeryYuncheng Central HospitalYunchengChina
| |
Collapse
|
10
|
Hoshi Y, Matsuda S, Takeuchi M, Kawakubo H, Kitagawa Y. Liquid Biopsy and Multidisciplinary Treatment for Esophageal Cancer. Cancers (Basel) 2025; 17:196. [PMID: 39857978 PMCID: PMC11763614 DOI: 10.3390/cancers17020196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Esophageal cancer (EC) is one of the leading causes of cancer-related deaths globally. Surgery is the standard treatment for resectable EC after preoperative chemoradiotherapy or chemotherapy, followed by postoperative adjuvant chemotherapy in certain cases. Upper gastrointestinal endoscopy and computed tomography (CT) are predominantly performed to evaluate the efficacy of these treatments, but their sensitivity and accuracy for evaluating minimal residual disease remain unsatisfactory, thereby requiring the development of alternative methods. In recent years, interest has been increasing in using liquid biopsy to assess treatment responses. Liquid biopsy is a noninvasive technology for detecting cell components in the blood and other body fluids. It involves collecting a small sample of body fluid, which is then analyzed for the presence of components, including circulating tumor DNA (ctDNA), microRNA (miRNA), or circulating tumor cells (CTCs). Further, ctDNA and miRNA are analyzed with various techniques, including digital polymerase chain reaction (dPCR) and next-generation sequencing (NGS). CTCs are isolated by determining surface antigens using immunomagnetic techniques or by filtering the blood according to cell size and rigidity. Several studies indicate that investigating these materials helps predict EC prognosis and recurrence and possibly stratifies high-risk groups. Liquid biopsy may also apply to the selection of cases that have achieved a complete response through preoperative treatment to prevent surgery and preserve the esophagus, as well as identifying the suitability of postoperative chemotherapy and the timing of conversion surgery for unresectable EC. The potential of liquid biopsy to enhance treatment decisions will further advance EC treatment.
Collapse
Affiliation(s)
| | - Satoru Matsuda
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | | | | | | |
Collapse
|
11
|
Lin Z, Zhai M, Wang H, Li M, Liu L, Zhang P, Yan L, Liu H, Tao K, Zhang T. Longitudinal circulating tumor DNA monitoring in predicting response to short-course radiotherapy followed by neoadjuvant chemotherapy and camrelizumab in locally advanced rectal cancer: data from a Phase Ⅲ clinical trial (UNION). Cancer Lett 2025; 611:217442. [PMID: 39755361 DOI: 10.1016/j.canlet.2025.217442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 12/16/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
This study, conducted as part of a multicenter phase III clinical trial, aimed to assess the utility of circulating tumor DNA (ctDNA)-based minimal residual disease (MRD) in comparing the efficacy of short-course and long-course chemoradiotherapy (CRT) for locally advanced rectal cancer (LARC). A total of 244 plasma samples from 79 LARC patients undergoing neoadjuvant therapy (NAT) before surgery were collected at various time points. Targeted deep sequencing using a novel MRD panel was performed. During NAT, ctDNA levels declined significantly. Baseline ctDNA-MRD status did not correlate significantly with treatment response. Notably, compared to long-course radiotherapy, microsatellite instability increased significantly after short-course radiotherapy (shortRT). Additionally, ctDNA negativity or lower levels were significantly associated with pathological complete response (pCR). Clearance of ctDNA and MRD after shortRT correlated significantly with pCR. A predictive model based on ctDNA-MRD, combined with carcinoembryonic antigen (CEA), outperformed models using only MRD or only CEA in predicting pCR/non-pCR. These findings provide insights into NAT for LARC and highlight ctDNA-based MRD assessment's potential in tailoring treatment strategies, emphasizing the need for personalized approaches.
Collapse
Affiliation(s)
- Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Menglan Zhai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Mingjie Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Lichao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Linghua Yan
- Shanghai Tongshu Biotech Co Ltd, Shanghai, 201900, China
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
| |
Collapse
|
12
|
Marret G, Lamy C, Vacher S, Cabel L, Séné M, Ahmanache L, Courtois L, El Beaino Z, Klijanienko J, Martinat C, Servant N, Kamoun C, Halladjian M, Bronzini T, Balsat C, Laes JF, Prévot A, Sauvage S, Lienard M, Martin E, Genin B, Badois N, Lesnik M, Dubray-Vautrin A, Choussy O, Ghanem W, Taouachi R, Planchon JM, Bièche I, Le Tourneau C, Kamal M. Deciphering molecular relapse and intra-tumor heterogeneity in non-metastatic resectable head and neck squamous cell carcinoma using circulating tumor DNA. Oral Oncol 2025; 160:107111. [PMID: 39612700 DOI: 10.1016/j.oraloncology.2024.107111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/15/2024] [Accepted: 11/15/2024] [Indexed: 12/01/2024]
Abstract
OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) is characterized by significant genetic intra-tumor heterogeneity (ITH), which may hinder precision medicine strategies that depend on results from single tumor-biopsy specimens. Treatment response assessment relies on radiologic imaging, which cannot detect minimal residual disease (MRD). We assessed the relevance of circulating tumor DNA (ctDNA) as a biomarker for ITH and MRD in HNSCC. MATERIALS AND METHODS We recruited 41 non-metastatic resectable HNSCC patients treated with upfront curative-intent surgery in the prospective biobanking SCANDARE study (NCT03017573). Thirty-one patients (76 %) showed recurrent disease at a median follow-up of 41 months. Targeted next-generation sequencing was performed on resected tumor tissues, as well as on serial blood samples obtained at surgery, within 14 weeks after surgery, at six months and at recurrence. RESULTS ctDNA was detected in 21/41 patients at surgery (sensitivity: 51 %; 95 % CI, 35-67 %) and 15/22 patients at recurrence (sensitivity: 68 %; 95 % confidence interval [CI], 45-86 %). Among patients with mutations identified in longitudinal plasma samples, additional mutations missed in tumor tissues were reported in 3/21 patients (14 %), while emerging mutations were reported in 9/21 patients (43 %). In the postoperative surveillance setting, ctDNA-based MRD detection anticipated clinical recurrence with a median lead-time of 9.9 months (interquartile range, 8.0-14.5 months) in 17/27 patients (63 %). When detected within 14 weeks after surgery, MRD correlated with disease recurrence after adjusting for classical prognostic variables (HR = 3.0; 95 % CI, 1.1-7.9; p = 0.03). CONCLUSIONS ctDNA detection is a useful biomarker for ITH and MRD in resectable HNSCC patients.
Collapse
Affiliation(s)
- Grégoire Marret
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, Paris, France
| | - Constance Lamy
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, Paris, France
| | | | - Luc Cabel
- Department of Medical Oncology, Institut Curie, Paris, France
| | - Mathieu Séné
- Genetics Department, Institut Curie, Paris, France
| | | | | | | | | | | | - Nicolas Servant
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Choumouss Kamoun
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Maral Halladjian
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, Paris, France
| | - Thierry Bronzini
- Department of Pathology, Centre des Ressources Biologiques, Institut Curie, Paris, France
| | | | | | | | | | | | | | | | - Nathalie Badois
- Department of Oncologic Surgery, Institut Curie, PSL Research University, Paris & Saint-Cloud, France
| | - Maria Lesnik
- Department of Oncologic Surgery, Institut Curie, PSL Research University, Paris & Saint-Cloud, France
| | - Antoine Dubray-Vautrin
- Department of Oncologic Surgery, Institut Curie, PSL Research University, Paris & Saint-Cloud, France
| | - Olivier Choussy
- Department of Oncologic Surgery, Institut Curie, PSL Research University, Paris & Saint-Cloud, France
| | - Wahib Ghanem
- Department of Oncologic Surgery, Institut Curie, PSL Research University, Paris & Saint-Cloud, France
| | - Rabah Taouachi
- Department of Oncologic Surgery, Institut Curie, PSL Research University, Paris & Saint-Cloud, France
| | | | - Ivan Bièche
- Genetics Department, Institut Curie, Paris, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, Paris, France; INSERM U900 Research Unit, Institut Curie, Saint-Cloud, France.
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, Paris, France
| |
Collapse
|
13
|
Schoofs K, Ferro Dos Santos MR, De Wilde J, Roelandt S, Van de Velde S, Decruyenaere P, Meuris L, Thas O, Philippron A, Depypere L, Nafteux P, Vanommeslaeghe H, Van Daele E, Pattyn P, Vandesompele J, De Preter K. Therapy response monitoring in blood plasma from esophageal adenocarcinoma patients using cell-free DNA methylation profiling. Sci Rep 2024; 14:31112. [PMID: 39730941 DOI: 10.1038/s41598-024-82325-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/04/2024] [Indexed: 12/29/2024] Open
Abstract
Esophageal adenocarcinoma (EAC) is an aggressive cancer characterized by a high risk of relapse post-surgery. Current follow-up methods (serum carcinoembryonic antigen detection and PET-CT) lack sensitivity and reliability, necessitating a novel approach. Analyzing cell-free DNA (cfDNA) from blood plasma emerges as a promising avenue. This study aims to evaluate the cost-effective and genome-wide cell-free reduced representation bisulfite sequencing (cfRRBS) method combined with computational deconvolution for effective disease monitoring in EAC patients. cfDNA methylation profiling with cfRRBS was performed on 162 blood plasma samples from 33 EAC cancer patients and 28 blood plasma samples from 20 healthy donors. The estimated tumor fraction for EAC patients at the time of diagnosis was significantly different from the healthy donor plasma samples (one-sided Wilcoxon rank-sum test: p-value = 0.032). Tumor fractions above 15% and focal gains/amplifications in MYC (chr8), KRAS (chr12), EGFR (chr7) and NOTCH2 (chr1) were observed in four samples of distinct patients at the time metastatic disease was detected. This study showed feasibility to estimate tumor fractions in blood plasma of EAC patients based on cfDNA methylation using cfRRBS and computational deconvolution. Nevertheless, in this study only cancer patients with evidence of metastatic disease show high tumor fractions and copy number alterations.
Collapse
Affiliation(s)
- Kathleen Schoofs
- Translational Oncogenomics and Bioinformatics Lab, Center for Medical Biotechnology, VIB-UGent & CRIG, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Maísa R Ferro Dos Santos
- Translational Oncogenomics and Bioinformatics Lab, Center for Medical Biotechnology, VIB-UGent & CRIG, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Jilke De Wilde
- Translational Oncogenomics and Bioinformatics Lab, Center for Medical Biotechnology, VIB-UGent & CRIG, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Sofie Roelandt
- Translational Oncogenomics and Bioinformatics Lab, Center for Medical Biotechnology, VIB-UGent & CRIG, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
| | - Sofie Van de Velde
- Translational Oncogenomics and Bioinformatics Lab, Center for Medical Biotechnology, VIB-UGent & CRIG, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
| | - Philippe Decruyenaere
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Hematology, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Leander Meuris
- Department of Biochemistry and Microbiology, Center for Medical Biotechnology, VIB-UGent, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
| | - Olivier Thas
- I-BioStat, Data Science Institute, Hasselt University, Agoralaan Gebouw D, 3590, Diepenbeek, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
- National Institute for Applied Statistics Research Australia (NIASRA), University of Wollongong, Wollongong, Australia
| | - Annouck Philippron
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Department of Gastro-Intestinal Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Lieven Depypere
- Department of Thoracic Surgery, University Hospital Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Philippe Nafteux
- Department of Thoracic Surgery, University Hospital Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Hanne Vanommeslaeghe
- Department of Gastro-Intestinal Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Elke Van Daele
- Department of Gastro-Intestinal Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Piet Pattyn
- Department of Gastro-Intestinal Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Jo Vandesompele
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Katleen De Preter
- Translational Oncogenomics and Bioinformatics Lab, Center for Medical Biotechnology, VIB-UGent & CRIG, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium.
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium.
| |
Collapse
|
14
|
Zhao Q, Wang L, Yang X, Feng J, Chen Q. Preoperative inflammatory burden index for prognostication in esophageal squamous cell carcinoma undergoing radical resection. Sci Rep 2024; 14:30811. [PMID: 39730559 DOI: 10.1038/s41598-024-81237-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/25/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND The Inflammatory burden Index (IBI) is an effective predictor for a range of malignancies. However, the significance of IBI in esophageal squamous cell carcinoma (ESCC) needs to be further verified. The aim of this study was to verify the predictive power of IBI in ESCC undergoing radical resection. METHODS The current retrospective study, which comprised 408 ESCC patients randomized into either the primary or validation cohort, evaluated the relationships between IBI, clinical characteristics, and cancer-specific survival (CSS). Additionally, the nomogram model was also constructed and verified. RESULTS The IBI is significantly related to tumor length, vessel invasion, perineural invasion, and TNM stage. Compared to other hematological indices, the decision curve analyses (DCA) and receiver operating characteristic curve (ROC) confirmed the higher prognostic value of IBI, indicating the better clinical applicability. In patients with high IBI compared to the low IBI cohort, the 5-year CSS was considerably worse (total: 27.0% vs. 59.1%, P < 0.001; primary: 25.0% vs. 58.9%, P < 0.001; validation: 31.7% vs. 59.7%, P = 0.002). The IBI was shown to be an independent parameter by multivariate analyses (primary: HR = 2.352, P < 0.001; validation: HR = 1.683, P = 0.045). Finally, with the C-index of 0.675 (0.656-0.695) in the primary set and 0.662 (0.630-0.694) in the validation set for CSS in ESCC, an IBI-based nomogram was created and validated. CONCLUSION The predictive significance of IBI in ESCC patients undergoing radical resection was validated by this investigation. IBI may be utilized for preoperative evaluation of ESCC as it was found to be substantially correlated with prognosis.
Collapse
Affiliation(s)
- Qiang Zhao
- Department of Thoracic Surgery, Hangzhou Institute of Medicine (HIM), Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang province, China
| | - Liang Wang
- Department of Thoracic Surgery, Hangzhou Institute of Medicine (HIM), Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang province, China
| | - Xun Yang
- Department of Thoracic Surgery, Hangzhou Institute of Medicine (HIM), Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang province, China
| | - Jifeng Feng
- Department of Thoracic Surgery, Hangzhou Institute of Medicine (HIM), Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang province, China.
- Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang province, China.
| | - Qixun Chen
- Department of Thoracic Surgery, Hangzhou Institute of Medicine (HIM), Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang province, China.
- Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang province, China.
| |
Collapse
|
15
|
Nixon AB, Navarro FCP, Zhou KI, Abbott C, McDaniel L, Howard L, Brady JC, Liu Y, Jia J, Niedzwiecki D, Strickler J, Boyle SM, Chen RO, Uronis H. Ultra-sensitive, tumor-informed ctDNA profiling in pembrolizumab-treated esophagogastric cancer patients predicts clinical responses. RESEARCH SQUARE 2024:rs.3.rs-5349536. [PMID: 39764133 PMCID: PMC11702795 DOI: 10.21203/rs.3.rs-5349536/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
To explore whether ultra-sensitive circulating tumor DNA (ctDNA) profiling enables early prediction of treatment response and early detection of disease progression, we applied NeXT Personal, an ultra-sensitive bespoke tumor-informed liquid biopsy platform, to profile tumor samples from the KeyLargo study, a phase II trial in which metastatic esophagogastric cancer (mEGC) patients received capecitabine, oxaliplatin, and pembrolizumab. All 25 patients evaluated were ctDNA-positive at baseline. Minimal residual disease (MRD) events varied from 406,067 down to 1.5 parts per million (PPM) of ctDNA with a median limit of detection of 2.03 PPM. ctDNA dynamics were highly correlated with changes in tumor size (ρ = 0.59, p = 7.3×10-9). Lack of early molecular response (lack of ctDNA decrease) was associated with worse overall survival (OS) (HR 6.6, 95% CI 1.8-24.1, p = 0.005) and progression-free survival (PFS) (HR 15.4, 95% CI 2.7-87.0, p = 0.002). Lack of molecular clearance of ctDNA was associated with worse OS (HR 6.9, 95% CI 1.5-30.8, p = 0.012) and PFS (HR 19.2, 95% CI 2.4-152.8, p = 0.005). Molecular progression (ctDNA increase) preceded imaging-derived progression by a median lead time of 65 days. These results suggest that ultra-sensitive liquid biopsy approaches could improve treatment decision-making for mEGC patients receiving chemotherapy and immunotherapy.
Collapse
|
16
|
Rahadiani N, Stephanie M, Manatar AF, Krisnuhoni E. The Diagnostic Utility of cfDNA and ctDNA in Liquid Biopsies for Gastrointestinal Cancers over the Last Decade. Oncol Res Treat 2024; 48:125-141. [PMID: 39681095 DOI: 10.1159/000543030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND Cell-free DNA (cfDNA) is a fragmented DNA that is released into the blood through necrosis, apoptosis, phagocytosis, or active secretion. cfDNA includes a subclass called circulating tumor DNA (ctDNA) released from cancer cells and constitutes a varied proportion of the total cfDNA. Both cfDNA and ctDNA hold significant potential as diagnostic biomarkers in gastrointestinal cancers. SUMMARY cfDNA and ctDNA are promising diagnostic biomarkers for gastrointestinal cancers with varied diagnostic values in different types of cancers. cfDNA offers higher sensitivity that makes it more suitable for screening methods and constant monitoring, particularly in integration with conventional biomarkers or in a multimarker model. On the contrary, ctDNA gives a real-time picture of tumor genetics and is more suitable for definitive diagnosis due to its specificity for tumor-associated alterations. Different types of samples and methods of detection can influence sensitivity, and the amount of cfDNA is higher in serum but plasma is used for cfDNA analysis because it contains less cellular contamination. In summary, cfDNA is more sensitive than ctDNA, although they have comparable or slightly lower specificity. KEY MESSAGE Further studies are needed to create common guidelines, minimize the cost of analysis, and perform extensive clinical trials to demonstrate the utility of circulating cfDNA and ctDNA in the vast majority of gastrointestinal cancer stages. Therefore, with the advancement in these technologies, cfDNA and ctDNA will be highly beneficial and evolve cancer diagnostics and therapy.
Collapse
Affiliation(s)
- Nur Rahadiani
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia/Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Marini Stephanie
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia/Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Amelia Fossetta Manatar
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia/Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Ening Krisnuhoni
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia/Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| |
Collapse
|
17
|
Mahmud M, Munjal A, Savani M, Win H, Rozell U, Arshad J. Biomarker Testing and Role of Tyrosine Kinase Inhibitors and Immunotherapy for Esophageal Squamous Cell Carcinoma. FOREGUT: THE JOURNAL OF THE AMERICAN FOREGUT SOCIETY 2024; 4:467-474. [DOI: 10.1177/26345161241238748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) constitutes an aggressive subset of esophageal cancers that portends a poor prognosis. Management of ESCC has been historically challenging due to the limited effective therapeutic options. Broadening our understanding of the molecular landscape and identifying reliable biomarkers are essential in early detection, monitoring disease response and advancing treatment strategies. Recently, immunotherapy and tyrosine kinase inhibitors have changed the treatment algorithm of ESCC. In this review, we explore the molecular landscape and biomarkers that can aid in the management of ESCC and discuss the role of immunotherapy and tyrosine kinase inhibitors in the treatment of ESCC.
Collapse
Affiliation(s)
| | | | - Malvi Savani
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Hninyee Win
- University of Arizona Cancer Center, Tucson, AZ, USA
| | | | - Junaid Arshad
- University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
18
|
Tatalovic S, Doleschal B, Kupferthaler A, Grundner S, Burghofer J, Webersinke G, Schwendinger S, Jukic E, Zschocke J, Danhel L, Kirchweger A, Havranek L, Shalamberidze D, Rezaie D, Biebl M, Rumpold H, Kirchweger P. Circulating Tumor DNA (ctDNA) Dynamics Predict Early Response to Treatment in Metastasized Gastroesophageal Cancer (mGEC) After 2 Weeks of Systemic Treatment. Cancers (Basel) 2024; 16:3960. [PMID: 39682148 DOI: 10.3390/cancers16233960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/15/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
mGEC is associated with poor overall survival (OS) of approximately 4-10 months. CtDNA is emerging as a promising prognostic biomarker with high potential for early relapse detection. However, until now, there was little knowledge on serial ctDNA detection and its impact on early treatment evaluation and prognosis in mGEC. METHODS ctDNA detection (ddPCR) was carried out serially in 37 matched tissue (NGS) patients with mGEC prior to systemic treatment initiation and every two weeks thereafter until restaging (n = 173 samples). The results have been correlated with response to treatment (restaging CT), overall survival (OS), and progression-free survival (PFS). RESULTS The pretherapeutic detection rate was 77.8%. Response to treatment assessment was correct in 54.2% (pretherapeutically pos./neg.) and 85.7% (dynamics at week 4). Moreover, a decline in ctDNA (MAF in %) below 57.1% of the pretherapeutic value after 2 weeks of systemic treatment was accompanied by a sensitivity of 57.1% and a specificity of 90% (AUC = 0.73) for correct restaging assessment (response evaluation by CT after 3 months) evaluating 76.5% of patients correctly after only 2 weeks. In contrast to mere pretherapeutic ctDNA positivity (p = 0.445), a decline in ctDNA dynamics to under 57.1% of its initial value was significantly associated with OS (4.1 (95% Cl 2.1-6.1) vs. 13.6 (95% CI 10.4-16.6) months, p < 0.001) and PFS (3.2 (1.9-4.5) vs. 9.5 (95% CI 5.5-13.5) months, p = 0.001) after two weeks of treatment. Additionally, the change in detectability from positive pretherapeutic levels to negative during treatment was associated with similar survival as for patients who were always regarded as ctDNA-negative (9.5 (95%Cl 0.4-18.5) vs. 9.6 (95%Cl 1.3-17.9)). The absence of becoming undetectable was associated with worse survival (4.7 months). CONCLUSIONS ctDNA is a promising additional biomarker allowing for early evaluation of response to treatment and saving unevaluated treatment time for patients with mGEC, and could allow for an early change in treatment with anticipated prognostic benefit in the future.
Collapse
Affiliation(s)
- Stefan Tatalovic
- Department of Surgery, Ordensklinikum Linz, 4010 Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
- VYRAL, 4020 Linz, Austria
| | - Bernhard Doleschal
- Department of Internal Medicine I for Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz, 4010 Linz, Austria
| | - Alexander Kupferthaler
- Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
- Department of Diagnostic and Interventional Radiology, Ordensklinikum Linz, 4010 Linz, Austria
| | - Stephan Grundner
- Department of Diagnostic and Interventional Radiology, Ordensklinikum Linz, 4010 Linz, Austria
| | - Jonathan Burghofer
- Laboratory for Molecular Genetics Diagnostics, Ordensklinikum Linz, 4010 Linz, Austria
| | - Gerald Webersinke
- Laboratory for Molecular Genetics Diagnostics, Ordensklinikum Linz, 4010 Linz, Austria
| | - Simon Schwendinger
- Institute of Human Genetics, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Emina Jukic
- Institute of Human Genetics, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Johannes Zschocke
- Institute of Human Genetics, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Lorenz Danhel
- Department of Surgery, Ordensklinikum Linz, 4010 Linz, Austria
- VYRAL, 4020 Linz, Austria
| | - Antonia Kirchweger
- Department of Surgery, Ordensklinikum Linz, 4010 Linz, Austria
- VYRAL, 4020 Linz, Austria
| | - Lukas Havranek
- Department of Surgery, Ordensklinikum Linz, 4010 Linz, Austria
- VYRAL, 4020 Linz, Austria
| | - Demetre Shalamberidze
- Department of Surgery, Ordensklinikum Linz, 4010 Linz, Austria
- VYRAL, 4020 Linz, Austria
| | - Daniel Rezaie
- Department of Surgery, Ordensklinikum Linz, 4010 Linz, Austria
- VYRAL, 4020 Linz, Austria
| | - Matthias Biebl
- Department of Surgery, Ordensklinikum Linz, 4010 Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Holger Rumpold
- Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
- Department of Internal Medicine I for Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz, 4010 Linz, Austria
| | - Patrick Kirchweger
- Department of Surgery, Ordensklinikum Linz, 4010 Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
- VYRAL, 4020 Linz, Austria
| |
Collapse
|
19
|
Sewell M, Toumbacaris N, Tan KS, Bahadur N, Philip J, Shah NJ, Niederhausern A, Tavarez Martinez C, Zheng H, Boerner T, Janjigian YY, Maron SB, Bott MJ, Gray KD, Park BJ, Sihag S, Jones DR, Ku GY, Wu AJ, Molena D. Esophagectomy may have a role in stage IV esophageal adenocarcinoma. J Thorac Cardiovasc Surg 2024:S0022-5223(24)01087-0. [PMID: 39581309 DOI: 10.1016/j.jtcvs.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/29/2024] [Accepted: 11/10/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE We sought to determine whether aggressive local treatment provides a benefit in patients with stage IV esophageal adenocarcinoma and to determine factors associated with survival. METHODS Patients with clinical stage IV esophageal adenocarcinoma at diagnosis who underwent esophagectomy from 2010 to 2023 were identified from our prospectively maintained database. Clinicopathologic and demographic characteristics were compared among patients by stage. Overall survival was estimated using the Kaplan-Meier approach. RESULTS In total, 66 patients met the inclusion criteria. Of these, 30 (45%) had stage IVA disease, and 36 (55%) had stage IVB disease. Of the 36 patients with stage IVB disease, 26 had oligometastatic disease, and 10 had disseminated disease. All patients with stage IVA disease received standard neoadjuvant therapy followed by curative-intent surgery; 26 of these patients (87%) received chemoradiation. Patients with oligometastatic stage IVB disease underwent systemic therapy with the goal of surgical resection. Patients with disseminated stage IVB disease underwent palliative chemotherapy, which led to improvement in disease burden and performance of esophagectomy. Median time from the start of therapy to surgery was shorter for patients with stage IVA disease than patients with stage IVB disease (P < .001). Three-year progression-free survival was lower for patients with stage IVA disease (40% vs 56%), as was 3-year overall survival (57% vs 85%). Adjusted overall survival, from the start of therapy to most recent follow-up, was higher for patients with stage IVB disease. CONCLUSIONS Aggressive local treatment may provide a benefit for highly selected patients with advanced or metastatic esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Marisa Sewell
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nicolas Toumbacaris
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kay See Tan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nadia Bahadur
- Clinical & Translational Research Informatics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - John Philip
- Clinical & Translational Research Informatics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Neil J Shah
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Andrew Niederhausern
- Department of Translational Informatics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carlos Tavarez Martinez
- Clinical & Translational Research Informatics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Haiyu Zheng
- Clinical & Translational Research Informatics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Thomas Boerner
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yelena Y Janjigian
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Steve B Maron
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Matthew J Bott
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Katherine D Gray
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bernard J Park
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Smita Sihag
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Geoffrey Y Ku
- Gastroenterology, Hepatology, and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Abraham J Wu
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniela Molena
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
20
|
Yuan P, Huang ZH, Yang YH, Bao FC, Sun K, Chao FF, Liu TT, Zhang JJ, Xu JM, Li XN, Li F, Ma T, Li H, Li ZH, Zhang SF, Hu J, Qi Y. A 18F-FDG PET/CT-based deep learning-radiomics-clinical model for prediction of cervical lymph node metastasis in esophageal squamous cell carcinoma. Cancer Imaging 2024; 24:153. [PMID: 39533388 PMCID: PMC11556142 DOI: 10.1186/s40644-024-00799-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND To develop an artificial intelligence (AI)-based model using Radiomics, deep learning (DL) features extracted from 18F-fluorodeoxyglucose (18F-FDG) Positron emission tomography/Computed Tomography (PET/CT) images of tumor and cervical lymph node with clinical feature for predicting cervical lymph node metastasis (CLNM) in patients with esophageal squamous cell carcinoma (ESCC). METHODS The study included 300 ESCC patients from the First Affiliated Hospital of Zhengzhou University who were divided into a training cohort and an internal testing cohort with an 8:2 ratio. Another 111 patients from Shanghai Chest Hospital were included as the external cohort. For each sample, we extracted 428 PET/CT-based Radiomics features from the gross tumor volume (GTV) and cervical lymph node (CLN) delineated layer by layer and 256 PET/CT-based DL features from the maximum cross-section of GTV and CLN images We input these features into seven different machine learning algorithms and ultimately selected logistic regression (LR) as the model classifier. Subsequently, we evaluated seven models (Clinical, Radiomics, Radiomics-Clinical, DL-Clinical, DL-Radiomics, DL-Radiomics-Clinical) using Radiomics features, DL features and clinical feature. RESULTS The DL-Radiomics-Clinical (DRC) model demonstrated higher AUC of 0.955 and 0.916 compared to the other six models in both internal and external testing cohorts respectively. The DRC model achieved the highest accuracy among the seven models in both the internal and external test sets, with scores of 0.951 and 0.892, respectively. CONCLUSIONS Through the combination of Radiomics features and DL features from PET/CT imaging and clinical feature, we developed a predictive model exhibiting exceptional classification capabilities. This model can be considered as a non-invasive method for predication of CLNM in patients with ESCC. It might facilitate decision-making regarding to the extend of lymph node dissection, and to select candidates for postoperative adjuvant therapy.
Collapse
Affiliation(s)
- Ping Yuan
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China.
| | - Zhen-Hao Huang
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Yun-Hai Yang
- Surgical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fei-Chao Bao
- Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ke Sun
- Department of nuclear medicine and radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Fang-Fang Chao
- Department of nuclear medicine and radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Ting-Ting Liu
- Department of nuclear medicine and radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Jing-Jing Zhang
- Department of nuclear medicine and radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Jin-Ming Xu
- Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang province, China
| | - Xiang-Nan Li
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Feng Li
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Tao Ma
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Hao Li
- School of Artiffcial Intelligence, Sun Yat-sen University, Zhuhai, Guangdong province, China
| | - Zi-Hao Li
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Shan-Feng Zhang
- School of Basic Medical Science, Zhengzhou University, Zhengzhou, Henan province, China
| | - Jian Hu
- Thoracic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang province, China.
| | - Yu Qi
- Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan province, China.
| |
Collapse
|
21
|
Park SH, Lee HJ, Kim TI, Lee J, Han SY, Seo HI, Kim DU. Ultrashort Cell-Free DNA Fragments and Vimentin-Positive Circulating Tumor Cells for Predicting Early Recurrence in Patients with Biliary Tract Cancer. Diagnostics (Basel) 2024; 14:2462. [PMID: 39518429 PMCID: PMC11544859 DOI: 10.3390/diagnostics14212462] [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: 09/24/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Biliary tract cancer (BTC) is a rare but aggressive malignancy that requires surgical treatment. However, postoperative recurrence rates are high, and reliable predictors of recurrence are limited. This study aimed to investigate the effectiveness of cell-free DNA (cfDNA) and circulating tumor cells (CTCs) in predicting early recurrence after curative surgery and complete adjuvant therapy in patients with BTC. Methods: Twenty-four patients who underwent R0 and R1 resections and completed adjuvant therapy for BTC between September 2019 and March 2022 were followed up until March 2024. Patients were categorized into early recurrence (ER) and non-ER groups, using one year as the cutoff for recurrence. Results: The combination score derived from ultrashort fragments of cfDNA, vimentin-positive CTCs, and carbohydrate antigen (CA) 19-9 levels showed a statistically significant difference between the ER and non-ER groups (p-value < 0.001). The receiver operating characteristic curve from the combination score and CA 19-9 levels yielded areas under the curve of 0.891 and 0.750, respectively. Conclusions: Although further research is required, these findings suggest that cfDNA and CTCs may increase the accuracy of predicting postoperative recurrence in patients with BTC.
Collapse
Affiliation(s)
- Sung Hee Park
- Division of Gastroenterology, Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.H.P.); (H.J.L.); (J.L.)
| | - Hye Ji Lee
- Division of Gastroenterology, Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.H.P.); (H.J.L.); (J.L.)
| | - Tae In Kim
- Division of Gastroenterology, Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.H.P.); (H.J.L.); (J.L.)
| | - Jonghyun Lee
- Division of Gastroenterology, Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.H.P.); (H.J.L.); (J.L.)
| | - Sung Yong Han
- Division of Gastroenterology, Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; (S.H.P.); (H.J.L.); (J.L.)
- Department of Internal Medicine, Pusan National University College of Medicine, Yangsan 44955, Republic of Korea
| | - Hyung Il Seo
- Department of Surgery, Pusan National University College of Medicine, Yangsan 44955, Republic of Korea;
| | - Dong Uk Kim
- Department of Internal Medicine, Gumi Medical Center, CHA University, Gumi 39100, Republic of Korea;
| |
Collapse
|
22
|
Battaglin F, Lenz HJ. Clinical Applications of Circulating Tumor DNA Profiling in GI Cancers. JCO Oncol Pract 2024; 20:1481-1490. [PMID: 39531845 PMCID: PMC11567053 DOI: 10.1200/op.24.00167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/11/2024] [Accepted: 05/01/2024] [Indexed: 11/16/2024] Open
Abstract
Over the next few years, the analysis of circulating tumor DNA (ctDNA) through liquid biopsy is expected to enter clinical practice and revolutionize the approach to biomarker testing and treatment selection in GI cancers. In fact, growing evidence support the use of ctDNA testing as a noninvasive, effective, and highly specific tool for molecular profiling in GI cancers. Analysis of blood ctDNA has been investigated in multiple settings including early tumor detection, minimal residual disease evaluation, tumor diagnosis and evaluation of prognostic/predictive biomarkers for targeted treatment selection, longitudinal monitoring of treatment response, and identification of resistance mechanisms. Here, we review the clinical applications, advantages, and limitations of ctDNA profiling for precision oncology in GI cancers.
Collapse
Affiliation(s)
- Francesca Battaglin
- Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Heinz-Josef Lenz
- Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
23
|
Ge Q, Zhang ZY, Li SN, Ma JQ, Zhao Z. Liquid biopsy: Comprehensive overview of circulating tumor DNA (Review). Oncol Lett 2024; 28:548. [PMID: 39319213 PMCID: PMC11420644 DOI: 10.3892/ol.2024.14681] [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: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024] Open
Abstract
Traditional tumor diagnosis methods rely on tissue biopsy, which can be invasive and unsuitable for long-term monitoring of tumor dynamics. The advent of liquid biopsy has notably improved the overall management of patients with cancer. Liquid biopsy techniques primarily involve detection of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). The present review focuses on ctDNA because of its significance in tumor diagnosis, monitoring and treatment. The use of ctDNA-based liquid biopsy offers several advantages, including non-invasive or minimally invasive collection methods, the ability to conduct repeated assessment and comprehensive insights into tumor biology. It serves crucial roles in disease management by facilitating screening of high-risk patients, dynamically monitoring therapeutic responses and diagnosis. Furthermore, ctDNA can be used to demonstrate pseudo-progression, monitor postoperative tumor status and guide adaptive treatment plans. The present study provides a comprehensive review of ctDNA, exploring its origins, metabolism, detection methods, clinical role and the current challenges associated with its application.
Collapse
Affiliation(s)
- Qian Ge
- Graduate School, Xi'an Medical University, Xi'an, Shaanxi 710000, P.R. China
| | - Zhi-Yun Zhang
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, P.R. China
| | - Suo-Ni Li
- Department of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Jie-Qun Ma
- Department of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Zheng Zhao
- Department of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710000, P.R. China
| |
Collapse
|
24
|
Zhang WY, Chang YJ, Shi RH. Artificial intelligence enhances the management of esophageal squamous cell carcinoma in the precision oncology era. World J Gastroenterol 2024; 30:4267-4280. [PMID: 39492825 PMCID: PMC11525855 DOI: 10.3748/wjg.v30.i39.4267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common histological type of esophageal cancer with a poor prognosis. Early diagnosis and prognosis assessment are crucial for improving the survival rate of ESCC patients. With the advancement of artificial intelligence (AI) technology and the proliferation of medical digital information, AI has demonstrated promising sensitivity and accuracy in assisting precise detection, treatment decision-making, and prognosis assessment of ESCC. It has become a unique opportunity to enhance comprehensive clinical management of ESCC in the era of precision oncology. This review examines how AI is applied to the diagnosis, treatment, and prognosis assessment of ESCC in the era of precision oncology, and analyzes the challenges and potential opportunities that AI faces in clinical translation. Through insights into future prospects, it is hoped that this review will contribute to the real-world application of AI in future clinical settings, ultimately alleviating the disease burden caused by ESCC.
Collapse
Affiliation(s)
- Wan-Yue Zhang
- School of Medicine, Southeast University, Nanjing 221000, Jiangsu Province, China
| | - Yong-Jian Chang
- School of Cyber Science and Engineering, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Rui-Hua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
25
|
Kumar S, Poria R, Kala D, Nagraik R, Dhir Y, Dhir S, Singh B, Kaushik NK, Noorani MS, Kumar D, Gupta S, Kaushal A. Recent advances in ctDNA detection using electrochemical biosensor for cancer. Discov Oncol 2024; 15:517. [PMID: 39356360 PMCID: PMC11448507 DOI: 10.1007/s12672-024-01365-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
In the quest for early cancer diagnosis, early identification and treatment are paramount. Recently, ctDNA detection has emerged as a viable avenue for early screening of cancer. The examination of ctDNA in fluid biopsies has gained substantial attention in tumor diagnosis and therapy. Both the scientific community and industry are actively exploring this field. However, developing cost-effective, portable, and real-time ctDNA measurement methods using conventional gene detection equipment poses a significant challenge. This challenge has led to the exploration of alternative approaches. Electrochemical biosensors, distinguished by their heightened sensitivity, remarkable specificity, affordability, and excellent portability, have emerged as a promising avenue for ctDNA detection. This review is dedicated to the specific focus on ctDNA detection, highlighting recent advancements in this evolving detection technology. We aimed to reference previous studies related to ctDNA-targeted cancer detection using electrochemical biosensors to advocate the utilization of electrochemical biosensors in healthcare diagnostics. Further research is imperative for the effective integration of ctDNA analysis into point-of-care cancer testing. Innovative approaches utilizing multiple markers need to be explored to advance this technology and make substantial contributions to societal well-being.
Collapse
Affiliation(s)
- Sahil Kumar
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, 133203, Ambala, India
| | - Renu Poria
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, 133203, Ambala, India
| | - Deepak Kala
- NL-11 Centera Tetrahertz Laboratory, Institute of High Pressure Physics, Polish Academy of Sciences, 29/37 Sokolowska Street, Warsaw, 01142, Poland
| | - Rupak Nagraik
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
| | - Yashika Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, 133203, Ambala, India
| | - Sunny Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, 133203, Ambala, India
| | - Bharat Singh
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, 133203, Ambala, India
| | - Naveen Kumar Kaushik
- Department of Industrial Biotechnology, College of Biotechnology, Chaudhary Charan Singh Haryana Agricultural University, Hisar, Haryana, India
| | - Md Salik Noorani
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, 173229, Himachal Pradesh, India.
| | - Shagun Gupta
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, 133203, Ambala, India.
| | - Ankur Kaushal
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, 133203, Ambala, India.
| |
Collapse
|
26
|
Fu SW, Tang C, Tan X, Srivastava S. Liquid biopsy for early cancer detection: technological revolutions and clinical dilemma. Expert Rev Mol Diagn 2024; 24:937-955. [PMID: 39360748 DOI: 10.1080/14737159.2024.2408744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Liquid biopsy is an innovative advancement in oncology, offering a noninvasive method for early cancer detection and monitoring by analyzing circulating tumor cells, DNA, RNA, and other biomarkers in bodily fluids. This technique has the potential to revolutionize precision oncology by providing real-time analysis of tumor dynamics, enabling early detection, monitoring treatment responses, and tailoring personalized therapies based on the molecular profiles of individual patients. AREAS COVERED In this review, the authors discuss current methodologies, technological challenges, and clinical applications of liquid biopsy. This includes advancements in detecting minimal residual disease, tracking tumor evolution, and combining liquid biopsy with other diagnostic modalities for precision oncology. Key areas explored are the sensitivity, specificity, and integration of multi-omics, AI, ML, and LLM technologies. EXPERT OPINION Liquid biopsy holds great potential to revolutionize cancer care through early detection and personalized treatment strategies. However, its success depends on overcoming technological and clinical hurdles, such as ensuring high sensitivity and specificity, interpreting results amidst tumor heterogeneity, and making tests accessible and affordable. Continued innovation and collaboration are crucial to fully realize the potential of liquid biopsy in improving early cancer detection, treatment, and monitoring.
Collapse
Affiliation(s)
- Sidney W Fu
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Cong Tang
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Xiaohui Tan
- Division of LS Research, LSBioscience, LLC, Frederick, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
27
|
Qiu SF, Zhang QZ, Wu ZY, Liu MZ, Ding Q, Sun FM, Wang Y, Yang HX, Zheng L, Chen X, Wu L, Bai J, Liu JF, Chen CB. Establishment and validation of circulating cell-free DNA signatures for nasopharyngeal carcinoma detection. EBioMedicine 2024; 108:105321. [PMID: 39265506 PMCID: PMC11416236 DOI: 10.1016/j.ebiom.2024.105321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Early detection of nasopharyngeal carcinoma (NPC) poses a significant challenge. The absence of highly sensitive and specific diagnostic biomarkers for nasopharyngeal carcinoma contributes to the unfavourable prognosis of NPC patients. Here, we aimed to establish a non-invasive approach for detecting NPC using circulating cell-free DNA (cfDNA). METHODS We investigated the potential of next-generation sequencing (NGS) of peripheral blood cells as a diagnostic tool for NPC. We collected data on genome-wide nucleosome footprint (NF), 5'-end motifs, fragmentation patterns, CNV information, and EBV content from 553 Chinese subjects, including 234 NPC patients and 319 healthy individuals. Through case-control analysis, we developed a diagnostic model for NPC, and validated its detection capability. FINDINGS Our findings revealed that the frequencies of NF, fragmentation, and motifs were significantly higher in NPC patients compared to healthy controls. We developed an NPC score based on these parameters that accurately distinguished NPC from non-NPC cases according to the American Joint Committee on Cancer staging system from non-NPC (validation set: area under curve (AUC) = 99.9% (95% CI: 99.8%-100%), se: 98.15%, sp: 100%). This model showed superior performance over plasma EBV DNA. Additionally, the NPC score effectively differentiated between NPC patients and healthy controls, even after clinical treatment. Furthermore, the NPC score was found to be independent of potential confounders such as age, sex, or TNM stage. INTERPRETATION We have developed and verified a non-invasive approach with substantial potential for clinical application in detecting NPC. FUNDING A full list of funding bodies that contributed to this study can be found in Funding section.
Collapse
Affiliation(s)
- Su-Fang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China
| | | | - Zi-Yi Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Ming-Zhu Liu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Qin Ding
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China
| | - Fu-Ming Sun
- Berry Oncology Corporation, Beijing, 100102, China
| | - Yin Wang
- Berry Oncology Corporation, Beijing, 100102, China; Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, 350200, China
| | - Han-Xuan Yang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China
| | - Lu Zheng
- Berry Oncology Corporation, Beijing, 100102, China
| | - Xin Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Lin Wu
- Berry Oncology Corporation, Beijing, 100102, China
| | - Jian Bai
- Berry Oncology Corporation, Beijing, 100102, China; Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, 350200, China.
| | - Jing-Feng Liu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China.
| | - Chuan-Ben Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Provincial Key Laboratory of Tumor Biotherapy Fuzhou, 350200, China.
| |
Collapse
|
28
|
Murciano-Goroff YR, Hui ABY, Araujo Filho JA, Hamilton EG, Chabon JJ, Moding EJ, Bonilla RF, Lebow ES, Gomez D, Rimner A, Ginsberg MS, Offin M, Kundra R, Allaj V, Norton L, Reis-Filho JS, Razavi P, Drilon A, Jones DR, Isbell JM, Lai WV, Rudin CM, Alizadeh AA, Li BT, Diehn M. Early Circulating Tumor DNA Shedding Kinetics for Prediction of Platinum Sensitivity in Patients With Small Cell Lung Cancer. JCO Precis Oncol 2024; 8:e2400216. [PMID: 39231375 PMCID: PMC11376985 DOI: 10.1200/po.24.00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/06/2024] [Accepted: 07/18/2024] [Indexed: 09/06/2024] Open
Abstract
PURPOSE Small cell lung cancer (SCLC) is characterized by rapid progression after platinum resistance. Circulating tumor (ctDNA) dynamics early in treatment may help determine platinum sensitivity. MATERIALS AND METHODS Serial plasma samples were collected from patients receiving platinum-based chemotherapy for SCLC on the first 3 days of cycle one and on the first days of subsequent cycles with paired samples collected both before and again after infusions. Tumor-informed plasma analysis was carried out using CAncer Personalized Profiling by deep Sequencing (CAPP-Seq). The mean variant allele frequency (VAF) of all pretreatment mutations was tracked in subsequent blood draws and correlated with radiologic response. RESULTS ctDNA kinetics were assessed in 122 samples from 21 patients. Pretreatment VAF did not differ significantly between patients who did and did not respond to chemotherapy (mean 22.5% v 4.6%, P = .17). A slight increase in ctDNA on cycle 1, day 1 immediately post-treatment was seen in six of the seven patients with available draws (fold change from baseline: 1.01-1.44), half of whom achieved a response. All patients who responded had a >2-fold decrease in mean VAF on cycle 2 day 1 (C2D1). Progression-free survival (PFS) and overall survival (OS) were significantly longer in patients with a >2-fold decrease in mean VAF after one treatment cycle (6.8 v 2.6 months, log-rank P = .0004 and 21.7 v 6.4 months, log rank P = .04, respectively). CONCLUSION A >2-fold decrease in ctDNA concentration was observed by C2D1 in all patients who were sensitive to platinum-based therapy and was associated with longer PFS and OS.
Collapse
Affiliation(s)
- Yonina R Murciano-Goroff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Angela B-Y Hui
- Stanford Cancer Institute, Stanford University, Stanford, CA
| | - Jose A Araujo Filho
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Jacob J Chabon
- Stanford Cancer Institute, Stanford University, Stanford, CA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Rene F Bonilla
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Emily S Lebow
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michelle S Ginsberg
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Ritika Kundra
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Viola Allaj
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - David R Jones
- Department of Surgery, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - James M Isbell
- Department of Surgery, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - W Victoria Lai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA
| | - Bob T Li
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA
- Department of Radiation Oncology, Stanford University, Stanford, CA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA
| |
Collapse
|
29
|
Svrcek M, Voron T, André T, Smyth EC, de la Fouchardière C. Improving individualised therapies in localised gastro-oesophageal adenocarcinoma. Lancet Oncol 2024; 25:e452-e463. [PMID: 39214116 DOI: 10.1016/s1470-2045(24)00180-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/05/2024] [Accepted: 03/20/2024] [Indexed: 09/04/2024]
Abstract
Despite our increased understanding of the biological and molecular aspects of gastro-oesophageal tumourigenesis, the identification of prognostic or predictive factors remains challenging. Patients with resectable gastric and oesophageal adenocarcinoma are often treated similarly after surgical resection, regardless of their tumour biology, clinical characteristics, and histological treatment response. Substantial progress has been made in the past 5 years in managing patients with gastric or oesophageal adenocarcinoma, including the use of immune checkpoint inhibitors and new targeted therapies, leading to substantial improvements in clinical outcomes. These advancements have primarily been established in advanced and metastatic disease, while the management framework for local and locoregional disease is just beginning to shift. We provide an overview of existing data on biomarkers and tumour-related and host-related factors that are relevant to stratify patients into low-risk and high-risk recurrence groups, both before and after surgery, paving the way for more personalised treatment approaches.
Collapse
Affiliation(s)
- Magali Svrcek
- Department of Pathology, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | - Thibault Voron
- Digestive Surgery Department, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | - Thierry André
- Department of Medical Oncology, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | | | | |
Collapse
|
30
|
Deshpande D, Chhugani K, Ramesh T, Pellegrini M, Shifman S, Abedalthagafi MS, Alqahtani S, Ye J, Liu XS, Leek JT, Brazma A, Ophoff RA, Rao G, Butte AJ, Moore JH, Katritch V, Mangul S. The evolution of computational research in a data-centric world. Cell 2024; 187:4449-4457. [PMID: 39178828 PMCID: PMC11938813 DOI: 10.1016/j.cell.2024.07.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/26/2024]
Abstract
Computational data-centric research techniques play a prevalent and multi-disciplinary role in life science research. In the past, scientists in wet labs generated the data, and computational researchers focused on creating tools for the analysis of those data. Computational researchers are now becoming more independent and taking leadership roles within biomedical projects, leveraging the increased availability of public data. We are now able to generate vast amounts of data, and the challenge has shifted from data generation to data analysis. Here we discuss the pitfalls, challenges, and opportunities facing the field of data-centric research in biology. We discuss the evolving perception of computational data-driven research and its rise as an independent domain in biomedical research while also addressing the significant collaborative opportunities that arise from integrating computational research with experimental and translational biology. Additionally, we discuss the future of data-centric research and its applications across various areas of the biomedical field.
Collapse
Affiliation(s)
- Dhrithi Deshpande
- Titus Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| | - Karishma Chhugani
- Titus Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Tejasvene Ramesh
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sagiv Shifman
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Malak S Abedalthagafi
- Genomics Research Department, King Fahad Medical City, Riyadh, Saudi Arabia; Department of Pathology & Laboratory Medicine, Emory University Hospital, Atlanta, GA, USA
| | - Saleh Alqahtani
- The Liver Transplant Unit, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; The Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jimmie Ye
- Department of Epidemiology & Biostatistics, Institute for Human Genetics, University of California, San Francisco, 513 Parnassus Avenue S965F, San Francisco, CA 94143, USA
| | - Xiaole Shirley Liu
- GV20 Oncotherapy, One Broadway, 14th Floor, Kendall Square, Cambridge, MA 02142, USA
| | - Jeffrey T Leek
- Biostatistics and Oncology at the Johns Hopkins Bloomberg School of Public Health and Johns Hopkins Data Science Lab, John Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Alvis Brazma
- EMBL European Bioinformatics Institute, Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Roel A Ophoff
- Department of Psychiatry and Human Genetics, Center for Neurobehavioral Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gauri Rao
- Titus Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California, San Francisco, 490 Illinois Street, San Francisco, CA 94158, USA
| | - Jason H Moore
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, 700 N. San Vicente Boulevard, Pacific Design Center Suite G540, West Hollywood, CA 90068, USA
| | - Vsevolod Katritch
- Department of Quantitative and Computational Biology, USC Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90007, USA
| | - Serghei Mangul
- Department of Quantitative and Computational Biology, USC Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90007, USA.
| |
Collapse
|
31
|
Pan HF, Zheng ZF, Zhao ZY, Liu Z, Huang SH, Chi P. Prognostic Significance of Preoperative and Postoperative Evaluation of Combined Tumor Markers for Patients With Colon Cancer. Surg Laparosc Endosc Percutan Tech 2024; 34:335-344. [PMID: 38736427 DOI: 10.1097/sle.0000000000001126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 10/24/2022] [Indexed: 05/14/2024]
Abstract
BACKGROUND The combined value of the tumor markers carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) in patients with colon cancer (CC) is unclear. This study aimed to investigate the role of composite tumor markers in the prognosis of CC. METHODS Patients who underwent curative resection of colon adenocarcinoma were enrolled. The tumor marker status before and after the operation was used to divide the patients into groups according to the number of tumor markers with abnormal expression, and recurrence-free survival (RFS) and overall survival (OS) of different groups were compared. The impact of changes in composite tumor markers in the perioperative period on outcomes was further explored. RESULTS Ultimately, 531 patients were enrolled in the study. As the number of preoperative and postoperative elevated tumor markers increased, both RFS and OS rates became lower (both P <0.05). Further analysis revealed that the number of elevated tumor markers after resection can significantly affect the outcomes (both P <0.05). In patients with abnormal preoperative tumor markers, normalization of markers after surgery was a protective factor for prognosis (both P <0.05), and patients with postoperative elevated levels of both tumor markers had a 5.5-fold and 6-fold increase in the risk of recurrence and death. In addition, patients with elevated markers after surgery had a high risk of recurrence within 5 years after colectomy. CONCLUSIONS Postoperative tumor markers had a better ability to differentiate postoperative outcomes in patients with CC than preoperative tumor markers. Patients whose tumor markers normalized after surgery had a better prognosis.
Collapse
Affiliation(s)
- Hong-Feng Pan
- Departments of Colorectal Surgery
- General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Zhi-Fang Zheng
- Departments of Colorectal Surgery
- General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Ze-Yi Zhao
- Departments of Colorectal Surgery
- General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Zhun Liu
- Departments of Colorectal Surgery
- General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Sheng-Hui Huang
- Departments of Colorectal Surgery
- General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Pan Chi
- Departments of Colorectal Surgery
- General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| |
Collapse
|
32
|
Motegi A, Kageyama SI, Kashima Y, Hirata H, Hojo H, Nakamura M, Fujisawa T, Enokida T, Tahara M, Matsuura K, Zenda S. Detection of HPV DNA in Saliva of Patients with HPV-Associated Oropharyngeal Cancer Treated with Radiotherapy. Curr Oncol 2024; 31:4397-4405. [PMID: 39195311 PMCID: PMC11352770 DOI: 10.3390/curroncol31080328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND To investigate the technical feasibility of RT-PCR and direct sequencing to quantify HPV DNA in the saliva of patients with Human-Papilloma-Virus related oropharyngeal cancer (HPV-OPC), the level of which is known to predict prognosis after treatment. METHODS Nine patients with locally advanced HPV-OPC treated with definitive radiotherapy with chemotherapy or cetuximab, or radiotherapy alone between April 2016 and September 2017, were enrolled, two of whom also received induction chemotherapy. Saliva was collected before (baseline), during (mid-RT) and after (post-RT) radiotherapy. HPV-16 DNAs (E6 and E7) in saliva were quantified by RT-PCR and sequencing, the latter using a custom cancer panel. Correlations between HPV DNA levels and clinical outcomes were assessed. RESULTS Compared to the baseline, the relative cycle threshold (Ct) value of E6 and E7 reduced at the point of mid-RT in the majority of the patients (100% and 75% for E6 and E7, respectively). Similarly, the relative Ct value from the baseline to post-RT reduced in 86% and 100% of the patients for E6 and E7, respectively. During the follow-up period, three patients (33%) experienced disease progression. The relative baseline Ct values of these three patients were in the top 4 of all the patients. The sequences of HPV DNA were detected in five (83%) of six samples of the baseline saliva that underwent DNA sequencing, along with several gene mutations, such as TP53,CDKN2A and PIK3CA. CONCLUSIONS This study demonstrates that, in addition to detection and quantification of HPV DNA by RT-PCR, detection by sequencing of HPV-DNA using a customized cancer panel is technically possible.
Collapse
Affiliation(s)
- Atsushi Motegi
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Shun-ichiro Kageyama
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Yukie Kashima
- Department of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Hidenari Hirata
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
| | - Hidehiro Hojo
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
| | - Masaki Nakamura
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
| | - Takeshi Fujisawa
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
| | - Tomohiro Enokida
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
| | - Makoto Tahara
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
| | - Kazuto Matsuura
- Department of Head and Neck Surgery, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
| | - Sadamoto Zenda
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| |
Collapse
|
33
|
Bloom MD, Bashir B. Emerging role of circulating tumor DNA for early detection of recurrence in biliary tract cancers. J Gastrointest Oncol 2024; 15:1358-1362. [PMID: 38989432 PMCID: PMC11231843 DOI: 10.21037/jgo-24-224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/17/2024] [Indexed: 07/12/2024] Open
Affiliation(s)
- Matthew D. Bloom
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Babar Bashir
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology & Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
34
|
Nam H, Lee E, Yang H, Lee K, Kwak T, Kim D, Kim H, Yang M, Yang Y, Son S, Nam YH, Minn I. PROMER technology: A new real-time PCR tool enabling multiplex detection of point mutation with high specificity and sensitivity. Biol Methods Protoc 2024; 9:bpae041. [PMID: 38938409 PMCID: PMC11208725 DOI: 10.1093/biomethods/bpae041] [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: 01/15/2024] [Revised: 05/21/2024] [Accepted: 06/02/2024] [Indexed: 06/29/2024] Open
Abstract
Real-time polymerase chain reaction (real-time PCR) is a powerful tool for the precise quantification of nucleic acids in various applications. In cancer management, the monitoring of circulating tumor DNA (ctDNA) from liquid biopsies can provide valuable information for precision care, including treatment selection and monitoring, prognosis, and early detection. However, the rare and heterogeneous nature of ctDNA has made its precise detection and quantification challenging, particularly for ctDNA containing hotspot mutations. We have developed a new real-time PCR tool, PROMER technology, which enables the precise and sensitive detection of ctDNA containing cancer-driven single-point mutations. The PROMER functions as both a PRObe and priMER, providing enhanced detection specificity. We validated PROMER technology using synthetic templates with known KRAS point mutations and demonstrated its sensitivity and linearity of quantification. Using genomic DNA from human cancer cells with mutant and wild-type KRAS, we confirmed that PROMER PCR can detect mutant DNA. Furthermore, we demonstrated the ability of PROMER technology to efficiently detect mutation-carrying ctDNA from the plasma of mice with human cancers. Our results suggest that PROMER technology represents a promising new tool for the precise detection and quantification of DNA containing point mutations in the presence of a large excess of wild-type counterpart.
Collapse
Affiliation(s)
- Hwanhee Nam
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Esder Lee
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Hichang Yang
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Kyeyoon Lee
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Taeho Kwak
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Dain Kim
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Hyemin Kim
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Mihwa Yang
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Younjoo Yang
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Seungwan Son
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Young-Hyean Nam
- NuriBio Co., Ltd, Anyang-si, Gyeonggi-Do, 14058, Republic of Korea
| | - Il Minn
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, United States
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD 21287, United States
| |
Collapse
|
35
|
McClurg DP, Sanghera C, Mukherjee S, Fitzgerald RC, Jones CM. A systematic review of circulating predictive and prognostic biomarkers to aid the personalised use of radiotherapy in the radical treatment of patients with oesophageal cancer. Radiother Oncol 2024; 195:110224. [PMID: 38479442 DOI: 10.1016/j.radonc.2024.110224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The availability of circulating biomarkers that are predictive of treatment response or prognostic of overall outcome could enable the personalised and adaptive use of radiotherapy (RT) in patients with oesophageal adenocarcinoma (OAC) and squamous cell carcinoma (OSCC). METHODS A systematic review was carried out following Preferred Reporting Items for Systematic Reviews guidance. Medline, EMBASE, PubMed, Cochrane Library, CINAHL, Scopus and the Web of Science databases were searched for studies published between January 2005-February 2023 relating to circulating biomarkers evaluated in the context of neoadjuvant or definitive RT delivered for OAC/OSCC. Study quality was assessed using predefined criteria. RESULTS A total of 3012 studies were screened and 57 subsequently included, across which 61 biomarkers were reported. A majority (43/57,75.4%) of studies were of Asian origin and retrospective (40/57, 70.2%), with most (52/57, 91.2%) biomarkers reported in the context of patients with OSCC. There was marked inter-study heterogeneity in patient populations, treatment characteristics, biomarker measurement and the cut points used to define biomarker positivity. Nevertheless, there is evidence for the prognostic and predictive value of circulating tumour DNA and numerous miRNAs in OAC and OSCC, as well as for the prognostic and predictive value of circulating levels of CYFRA21.1 in OSCC. CONCLUSIONS There is consistent evidence for the potential predictive and prognostic value of a small number of biomarkers in OSCC and OAC, though these data are insufficient for translation to current clinical practice. Well-designed prospective studies are now required to validate their role in stratified and personalised RT treatment approaches.
Collapse
Affiliation(s)
- Dylan P McClurg
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Chandan Sanghera
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Somnath Mukherjee
- Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Christopher M Jones
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Oncology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
36
|
Boukovala M, Westphalen CB, Probst V. Liquid biopsy into the clinics: Current evidence and future perspectives. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100146. [PMID: 40027149 PMCID: PMC11863819 DOI: 10.1016/j.jlb.2024.100146] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 03/05/2025]
Abstract
As precision oncology has become a major part of the treatment landscape in oncology, liquid biopsies have developed as a particularly powerful tool as it surmounts several limitations of traditional tissue biopsies. These biopsies involve most commonly the isolation of circulating extracellular nucleic acids, including cell-free DNA (cfDNA) and circulating tumor DNA (ctDNA), as well as circulating tumor cells (CTCs), typically from blood. The clinical applications of liquid biopsies are diverse, encompassing the initial diagnosis and cancer detection, the application as a tool for prognostication in early and advanced tumor settings, the identification of potentially actionable alterations, the monitoring of response and resistance under systemic therapy and the detection of resistance mechanisms, the differentiation of distinct immune checkpoint blockade response patterns through serial samples, the prediction of immune checkpoint blockade responses based on initial liquid biopsy characteristics and the assessment of tumor heterogeneity. Moreover, molecular relapse monitoring in early-stage cancers and the personalization of adjuvant or additive therapy via MRD have become a major field of research in recent years. Compared to tissue biopsies, liquid biopsies are less invasive and can be collected serially, offering real-time molecular insights. Furthermore, liquid biopsies may allow for a more holistic evaluation of a patient's disease, as they assess material from all tumor sites and can theoretically reflect tumor heterogeneity. Furthermore, quicker turnaround-time also constitutes an advantage of liquid biopsies. Disadvantages or hurdles include the challenge of detecting low amounts of tumor deposits in peripheral blood or other fluids and the potential of different amounts tumor-shedding from different metastatic sites, as well as potentially false-positive from clonal hematopoietic mutations of indeterminate potential (CHIP) mutations. The clinical utility of liquid biopsies still must be validated in most settings and further research has to be done. Clinal trials including alternate bodily fluids and leveraging AI-technology are expected to revolutionize the field of liquid biopsies.
Collapse
|
37
|
Chen K, He Y, Wang W, Yuan X, Carbone DP, Yang F. Development of new techniques and clinical applications of liquid biopsy in lung cancer management. Sci Bull (Beijing) 2024; 69:1556-1568. [PMID: 38641511 DOI: 10.1016/j.scib.2024.03.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/12/2023] [Accepted: 01/17/2024] [Indexed: 04/21/2024]
Abstract
Lung cancer is an exceedingly malignant tumor reported as having the highest morbidity and mortality of any cancer worldwide, thus posing a great threat to global health. Despite the growing demand for precision medicine, current methods for early clinical detection, treatment and prognosis monitoring in lung cancer are hampered by certain bottlenecks. Studies have found that during the formation and development of a tumor, molecular substances carrying tumor-related genetic information can be released into body fluids. Liquid biopsy (LB), a method for detecting these tumor-related markers in body fluids, maybe a way to make progress in these bottlenecks. In recent years, LB technology has undergone rapid advancements. Therefore, this review will provide information on technical updates to LB and its potential clinical applications, evaluate its effectiveness for specific applications, discuss the existing limitations of LB, and present a look forward to possible future clinical applications. Specifically, this paper will introduce technical updates from the prospectives of engineering breakthroughs in the detection of membrane-based LB biomarkers and other improvements in sequencing technology. Additionally, it will summarize the latest applications of liquid biopsy for the early detection, diagnosis, treatment, and prognosis of lung cancer. We will present the interconnectedness of clinical and laboratory issues and the interplay of technology and application in LB today.
Collapse
Affiliation(s)
- Kezhong Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China; Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing 100044, China
| | - Yue He
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China; Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing 100044, China
| | - Wenxiang Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China; Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing 100044, China
| | - Xiaoqiu Yuan
- Peking University Health Science Center, Beijing 100191, China
| | - David P Carbone
- Thoracic Oncology Center, Ohio State University, Columbus 43026, USA.
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing 100044, China; Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing 100044, China.
| |
Collapse
|
38
|
Azad TD, Nanjo S, Jin MC, Chabon JJ, Kurtz DM, Chaudhuri AA, Connolly ID, Hui ABY, Liu CL, Merriott D, Ko R, Yoo C, Carter J, Chen E, Bonilla R, Hata A, Katakami N, Irie K, Yano S, Okimoto R, Bivona TG, Newman AM, Iv M, Nagpal S, Gephart MH, Alizadeh AA, Diehn M. Quantification of cerebrospinal fluid tumor DNA in lung cancer patients with suspected leptomeningeal carcinomatosis. NPJ Precis Oncol 2024; 8:121. [PMID: 38806586 PMCID: PMC11133465 DOI: 10.1038/s41698-024-00582-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 03/28/2024] [Indexed: 05/30/2024] Open
Abstract
Cerebrospinal fluid tumor-derived DNA (CSF-tDNA) analysis is a promising approach for monitoring the neoplastic processes of the central nervous system. We applied a lung cancer-specific sequencing panel (CAPP-Seq) to 81 CSF, blood, and tissue samples from 24 lung cancer patients who underwent lumbar puncture (LP) for suspected leptomeningeal disease (LMD). A subset of the cohort (N = 12) participated in a prospective trial of osimertinib for refractory LMD in which serial LPs were performed before and during treatment. CSF-tDNA variant allele fractions (VAFs) were significantly higher than plasma circulating tumor DNA (ctDNA) VAFs (median CSF-tDNA, 32.7%; median plasma ctDNA, 1.8%; P < 0.0001). Concentrations of tumor DNA in CSF and plasma were positively correlated (Spearman's ρ, 0.45; P = 0.03). For LMD diagnosis, cytology was 81.8% sensitive and CSF-tDNA was 91.7% sensitive. CSF-tDNA was also strongly prognostic for overall survival (HR = 7.1; P = 0.02). Among patients with progression on targeted therapy, resistance mutations, such as EGFR T790M and MET amplification, were common in peripheral blood but were rare in time-matched CSF, indicating differences in resistance mechanisms based on the anatomic compartment. In the osimertinib cohort, patients with CNS progression had increased CSF-tDNA VAFs at follow-up LP. Post-osimertinib CSF-tDNA VAF was strongly prognostic for CNS progression (HR = 6.2, P = 0.009). Detection of CSF-tDNA in lung cancer patients with suspected LMD is feasible and may have clinical utility. CSF-tDNA improves the sensitivity of LMD diagnosis, enables improved prognostication, and drives therapeutic strategies that account for spatial heterogeneity in resistance mechanisms.
Collapse
Affiliation(s)
- Tej D Azad
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Shigeki Nanjo
- Department of Medicine, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Respiratory Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | - Michael C Jin
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Jacob J Chabon
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - David M Kurtz
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Aadel A Chaudhuri
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Comprehensive Cancer Center, Rochester, MN, USA
| | - Ian D Connolly
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Angela Bik-Yu Hui
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Chih Long Liu
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - David Merriott
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Ryan Ko
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Christopher Yoo
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Justin Carter
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Emily Chen
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Rene Bonilla
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Akito Hata
- Department of Medical Oncology, Kobe Minimally Invasive Cancer Center, Kobe, Japan
| | - Nobuyuki Katakami
- Department of Medical Oncology, Takarazuka City Hospital, Hyogo, Japan
| | - Kei Irie
- Department of Pharmaceutics, Faculty of Pharmaceutical Science, Kobe Gakuin University, Kobe, Japan
| | - Seiji Yano
- Department of Respiratory Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | - Ross Okimoto
- Department of Medicine, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Aaron M Newman
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Michael Iv
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Seema Nagpal
- Department of Neurology, Stanford University, Stanford, CA, USA
| | | | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
39
|
Yue P, Bie F, Zhu J, Gao LR, Zhou Z, Bai G, Wang X, Zhao Z, Xiao ZF, Li Y, Zhou A, Liu WY, Jiao Y, Gao S. Minimal residual disease profiling predicts pathological complete response in esophageal squamous cell carcinoma. Mol Cancer 2024; 23:96. [PMID: 38730415 PMCID: PMC11084057 DOI: 10.1186/s12943-024-02006-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
Accurate presurgical prediction of pathological complete response (pCR) can guide treatment decisions, potentially avoiding unnecessary surgeries and improving the quality of life for cancer patients. We developed a minimal residual disease (MRD) profiling approach with enhanced sensitivity and specificity for detecting minimal tumor DNA from cell-free DNA (cfDNA). The approach was validated in two independent esophageal squamous cell carcinoma (ESCC) cohorts. In a cohort undergoing neoadjuvant, surgical, and adjuvant therapy (NAT cohort), presurgical MRD status precisely predicted pCR. All MRD-negative cases (10/10) were confirmed as pCR by pathological evaluation on the resected tissues. In contrast, MRD-positive cases included all the 27 non-pCR cases and only one pCR case (10/10 vs 1/28, P < 0.0001, Fisher's exact test). In a definitive radiotherapy cohort (dRT cohort), post-dRT MRD status was closely correlated with patient prognosis. All MRD-negative patients (25/25) remained progression-free during the follow-up period, while 23 of the 26 MRD-positive patients experienced disease progression (25/25 vs 3/26, P < 0.0001, Fisher's exact test; progression-free survival, P < 0.0001, log-rank test). The MRD profiling approach effectively predicted the ESCC patients who would achieve pCR with surgery and those likely to remain progression-free without surgery. This suggests that the cancer cells in these MRD-negative patients have been effectively eliminated and they could be suitable candidates for a watch-and-wait strategy, potentially avoiding unnecessary surgery.
Collapse
Affiliation(s)
- Pinli Yue
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China
| | - Fenglong Bie
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Jiarun Zhu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China
| | - Lin-Rui Gao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China
| | - Zhendiao Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China
| | - Guangyu Bai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China
| | - Xiaobing Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China
| | - Ziyi Zhao
- Harrow International School Shenzhen Qianhai, Shenzhen, China
| | - Ze-Fen Xiao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China
| | - Aiping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen-Yang Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China.
| | - Yuchen Jiao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China.
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Pan-jia-yuan South Ln, Chaoyang, District, Beijing, 100021, China.
| |
Collapse
|
40
|
Hashimoto T, Nakamura Y, Oki E, Kobayashi S, Yuda J, Shibuki T, Bando H, Yoshino T. Bridging horizons beyond CIRCULATE-Japan: a new paradigm in molecular residual disease detection via whole genome sequencing-based circulating tumor DNA assay. Int J Clin Oncol 2024; 29:495-511. [PMID: 38551727 PMCID: PMC11043144 DOI: 10.1007/s10147-024-02493-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/16/2024] [Indexed: 04/26/2024]
Abstract
Circulating tumor DNA (ctDNA) is the fraction of cell-free DNA in patient blood that originates from a tumor. Advances in DNA sequencing technologies and our understanding of the molecular biology of tumors have increased interest in exploiting ctDNA to facilitate detection of molecular residual disease (MRD). Analysis of ctDNA as a promising MRD biomarker of solid malignancies has a central role in precision medicine initiatives exemplified by our CIRCULATE-Japan project involving patients with resectable colorectal cancer. Notably, the project underscores the prognostic significance of the ctDNA status at 4 weeks post-surgery and its correlation to adjuvant therapy efficacy at interim analysis. This substantiates the hypothesis that MRD is a critical prognostic indicator of relapse in patients with colorectal cancer. Despite remarkable advancements, challenges endure, primarily attributable to the exceedingly low ctDNA concentration in peripheral blood, particularly in scenarios involving low tumor shedding and the intrinsic error rates of current sequencing technologies. These complications necessitate more sensitive and sophisticated assays to verify the clinical utility of MRD across all solid tumors. Whole genome sequencing (WGS)-based tumor-informed MRD assays have recently demonstrated the ability to detect ctDNA in the parts-per-million range. This review delineates the current landscape of MRD assays, highlighting WGS-based approaches as the forefront technique in ctDNA analysis. Additionally, it introduces our upcoming endeavor, WGS-based pan-cancer MRD detection via ctDNA, in our forthcoming project, SCRUM-Japan MONSTAR-SCREEN-3.
Collapse
Affiliation(s)
- Tadayoshi Hashimoto
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shin Kobayashi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Junichiro Yuda
- Department of Hematology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Taro Shibuki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
41
|
Lee JS, Cho EH, Kim B, Hong J, Kim YG, Kim Y, Jang JH, Lee ST, Kong SY, Lee W, Shin S, Song EY. Clinical Practice Guideline for Blood-based Circulating Tumor DNA Assays. Ann Lab Med 2024; 44:195-209. [PMID: 38221747 PMCID: PMC10813828 DOI: 10.3343/alm.2023.0389] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/06/2023] [Accepted: 01/06/2024] [Indexed: 01/16/2024] Open
Abstract
Circulating tumor DNA (ctDNA) has emerged as a promising tool for various clinical applications, including early diagnosis, therapeutic target identification, treatment response monitoring, prognosis evaluation, and minimal residual disease detection. Consequently, ctDNA assays have been incorporated into clinical practice. In this review, we offer an in-depth exploration of the clinical implementation of ctDNA assays. Notably, we examined existing evidence related to pre-analytical procedures, analytical components in current technologies, and result interpretation and reporting processes. The primary objective of this guidelines is to provide recommendations for the clinical utilization of ctDNA assays.
Collapse
Affiliation(s)
- Jee-Soo Lee
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Hye Cho
- Department of Laboratory Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Boram Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | - Young-gon Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoonjung Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Ja-Hyun Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
- Dxome Co. Ltd., Seongnam, Korea
| | - Sun-Young Kong
- Department of Laboratory Medicine, National Cancer Center, Goyang, Korea
| | - Woochang Lee
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Young Song
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
42
|
Zhang GL, Zhu QK, Ma TY, Weng CG, Zhang DD, Zeng H, Wang T, Gao F, Mi LL, Wang R. Clinical study of camrelizumab combined with docetaxel and carboplatin as a neoadjuvant treatment for locally advanced oesophageal squamous cell carcinoma. Dis Esophagus 2024; 37:doad073. [PMID: 38189470 DOI: 10.1093/dote/doad073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Herein, we aimed to evaluate the efficacy and safety of camrelizumab combined with docetaxel and carboplatin as a neoadjuvant treatment for locally advanced oesophageal squamous cell carcinoma (OSCC). Fifty-one patients with OSCC, treated from July 2020 to October 2022, were analyzed. Of them, 41 patients underwent surgery 4-8 weeks after undergoing two cycles of camrelizumab (200 mg IV Q3W) combined with docetaxel (75 mg/m2 IV Q3W) and carboplatin (area under the curve = 5-6 IV Q3W). The primary endpoint was the pathological complete response rate. All 51 patients (100%) experienced treatment-related grades 1-2 adverse events, and 2 patients (3.9%) experienced grade 4 events (including elevated alanine transaminase/aspartate transferase levels and Guillain-Barre syndrome). Fifty patients were evaluated for the treatment efficacy. Of them, 13 achieved complete response, and the objective response rate was 74%. Only 41 patients underwent surgical treatment. The pathological complete response rate was 17.1%, the major pathological response rate was 63.4%, and the R0 resection rate was 100%. Approximately 22% of the patients had tumor regression grades 0. Eight patients (19.5%) developed surgery-related complications. The median follow-up time was 18 months (range: 3-29 months). Four patients experienced disease progression, while four died. The median disease-free survival and overall survival were not reached. Camrelizumab combined with docetaxel and carboplatin is an effective and safe neoadjuvant treatment for locally advanced OSCC. This regimen may afford a potential strategy to treat patients with locally advanced OSCC.
Collapse
Affiliation(s)
- Guo-Liang Zhang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qi-Kun Zhu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tian-You Ma
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chen-Gang Weng
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dan-Dan Zhang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hui Zeng
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tao Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Feng Gao
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li-Li Mi
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Rui Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
43
|
Daban A, Beaussire-Trouvay L, Lévêque É, Alexandru C, Tennevet I, Langlois O, Veresezan O, Marguet F, Clatot F, Di Fiore F, Sarafan-Vasseur N, Fontanilles M. Prognostic value of circulating short-length DNA fragments in unresected glioblastoma patients. Transl Oncol 2024; 42:101897. [PMID: 38340682 PMCID: PMC10867437 DOI: 10.1016/j.tranon.2024.101897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/08/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Liquid biopsy application is still challenging in glioblastoma patients and the usefulness of short-length DNA (slDNA) fragments is not established. The aim was to investigate slDNA concentration as a prognostic marker in unresected glioblastoma patients. METHODS Patients with unresected glioblastoma and treated by radiochemotherapy (RT/TMZ) were included. Plasmas were prospectively collected at three times: before (pre-) RT, after (post-) RT and at the time of progression. Primary objective was to investigate the impact on survival of slDNA concentration [slDNA] variation during RT/TMZ. Secondary objectives were to explore the association between tumor volume, corticosteroid exposition and [slDNA]; and the impact of slDNA detection at pre-RT on survival. RESULTS Thirty-six patients were analyzed: 11 patients (30.6 %) experienced [slDNA] decrease during RT/TMZ, 22 patients (61.1 %) experienced increase and 3 patients (8.3 %) had stability. Decrease of [slDNA] during RT/TMZ was associated with better outcome compared to increase or stability: median OS, since end of RT, of 13.2 months [11.4 - NA] vs 10.1 months [7.8 - 12.6] and 6.8 months [4.5 - NA], p = 0.015, respectively. slDNA detection at pre-RT time was associated with improved OS: 11.7 months in the slDNA(+) group versus 8.8 months in the slDNA(-) group, p = 0.004. [slDNA] was not associated with corticosteroids exposition or tumor volume. No influence on survival was observed for both whole cfDNA concentration or slDNA peak size. CONCLUSION [slDNA] decrease during radiochemotherapy phase is a favorable prognostic marker on OS for unresected glioblastoma patients. Larger and independent cohorts are now required. TRIAL REGISTRATION ClinicalTrial, NCT02617745. Registered 1 December 2015, https://clinicaltrials.gov/ct2/show/NCT02617745?term=glioplak&draw=2&rank=1.
Collapse
Affiliation(s)
- Arthur Daban
- Department of Medical Oncology, Cancer Centre Henri Becquerel, Rue d'Amiens, 76038, Rouen, France
| | | | - Émilie Lévêque
- Clinical Research Unit, Cancer Centre Henri Becquerel, Rue d'Amiens, 76038, Rouen, France
| | - Cristina Alexandru
- Department of Medical Oncology, Cancer Centre Henri Becquerel, Rue d'Amiens, 76038, Rouen, France
| | - Isabelle Tennevet
- Department of Medical Oncology, Cancer Centre Henri Becquerel, Rue d'Amiens, 76038, Rouen, France
| | - Olivier Langlois
- Department of Neurosurgery, Rouen University Hospital, F-76031, 1 Rue de Germont, Rouen, CEDEX 76031, France
| | - Ovidiu Veresezan
- Department of Radiation Oncology, Henri Becquerel Cancer Center, 76038, Rouen, France
| | - Florent Marguet
- Univ Rouen Normandy, INSERM unit U1245 Brain and Cancer Genomics, Rouen, 76000 France; Department of Pathology, Rouen University Hospital, 1 Rue de Germont, Rouen, CEDEX 76031, France
| | - Florian Clatot
- Department of Medical Oncology, Cancer Centre Henri Becquerel, Rue d'Amiens, 76038, Rouen, France; Univ Rouen Normandy, INSERM unit U1245 Brain and Cancer Genomics, Rouen, 76000 France
| | - Frédéric Di Fiore
- Department of Medical Oncology, Cancer Centre Henri Becquerel, Rue d'Amiens, 76038, Rouen, France; Univ Rouen Normandy, INSERM unit U1245 Brain and Cancer Genomics, Rouen, 76000 France
| | | | - Maxime Fontanilles
- Department of Medical Oncology, Cancer Centre Henri Becquerel, Rue d'Amiens, 76038, Rouen, France; Univ Rouen Normandy, INSERM unit U1245 Brain and Cancer Genomics, Rouen, 76000 France.
| |
Collapse
|
44
|
Northcott J, Bartha G, Harris J, Li C, Navarro FC, Pyke RM, Hong M, Zhang Q, Ma S, Chen TX, Lai J, Udar N, Saldivar JS, Ayash E, Anderson J, Li J, Cui T, Le T, Chow R, Velasco RJ, Mallo C, Santiago R, Bruce RC, Goodman LJ, Chen Y, Norton D, Chen RO, Lyle JM. Analytical validation of NeXT Personal®, an ultra-sensitive personalized circulating tumor DNA assay. Oncotarget 2024; 15:200-218. [PMID: 38484152 PMCID: PMC10939476 DOI: 10.18632/oncotarget.28565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
We describe the analytical validation of NeXT Personal®, an ultra-sensitive, tumor-informed circulating tumor DNA (ctDNA) assay for detecting residual disease, monitoring therapy response, and detecting recurrence in patients diagnosed with solid tumor cancers. NeXT Personal uses whole genome sequencing of tumor and matched normal samples combined with advanced analytics to accurately identify up to ~1,800 somatic variants specific to the patient's tumor. A personalized panel is created, targeting these variants and then used to sequence cell-free DNA extracted from patient plasma samples for ultra-sensitive detection of ctDNA. The NeXT Personal analytical validation is based on panels designed from tumor and matched normal samples from two cell lines, and from 123 patients across nine cancer types. Analytical measurements demonstrated a detection threshold of 1.67 parts per million (PPM) with a limit of detection at 95% (LOD95) of 3.45 PPM. NeXT Personal showed linearity over a range of 0.8 to 300,000 PPM (Pearson correlation coefficient = 0.9998). Precision varied from a coefficient of variation of 12.8% to 3.6% over a range of 25 to 25,000 PPM. The assay targets 99.9% specificity, with this validation study measuring 100% specificity and in silico methods giving us a confidence interval of 99.92 to 100%. In summary, this study demonstrates NeXT Personal as an ultra-sensitive, highly quantitative and robust ctDNA assay that can be used to detect residual disease, monitor treatment response, and detect recurrence in patients.
Collapse
Affiliation(s)
| | | | | | - Conan Li
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | | | | - Qi Zhang
- Personalis, Inc., Fremont, CA 94555, USA
| | - Shuyuan Ma
- Personalis, Inc., Fremont, CA 94555, USA
| | | | - Janet Lai
- Personalis, Inc., Fremont, CA 94555, USA
| | - Nitin Udar
- Personalis, Inc., Fremont, CA 94555, USA
| | | | - Erin Ayash
- Personalis, Inc., Fremont, CA 94555, USA
| | | | - Jiang Li
- Personalis, Inc., Fremont, CA 94555, USA
| | - Tiange Cui
- Personalis, Inc., Fremont, CA 94555, USA
| | - Tu Le
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | | | | | | | | | | - Yi Chen
- Personalis, Inc., Fremont, CA 94555, USA
| | - Dan Norton
- Personalis, Inc., Fremont, CA 94555, USA
| | | | - John M. Lyle
- Personalis, Inc., Fremont, CA 94555, USA
- Co-last authors
| |
Collapse
|
45
|
Chen B, Liu S, Zhu Y, Wang R, Cheng X, Chen B, Dragomir MP, Zhang Y, Hu Y, Liu M, Li Q, Yang H, Xi M. Predictive role of ctDNA in esophageal squamous cell carcinoma receiving definitive chemoradiotherapy combined with toripalimab. Nat Commun 2024; 15:1919. [PMID: 38429311 PMCID: PMC10907344 DOI: 10.1038/s41467-024-46307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 02/15/2024] [Indexed: 03/03/2024] Open
Abstract
The combination of toripalimab (an anti-PD-1 antibody) with definitive chemoradiotherapy (CRT) demonstrated encouraging efficacy against locally advanced esophageal squamous cell carcinoma (ESCC) in the EC-CRT-001 phase II trial (NCT04005170). The primary endpoint of this trial was the clinical complete response rate (cCR), and the secondary endpoints included overall survival (OS), progression-free survival (PFS), duration of response, and quality of life. The exploratory analyses of EC-CRT-001 include exploring the role of circulating tumor DNA (ctDNA) and blood-based tumor mutational burden (bTMB) in predicting the response and survival. In total, 118 blood and 35 tissue samples from 42 enrolled patients were included in the analyses. We found that ctDNA-negative patients achieved a higher cCR compared to those with detectable ctDNA during CRT (83%, 19/23 vs. 39%, 7/18; p = 0.008) or post-CRT (78%, 21/27 vs. 30%, 3/10; p = 0.017). Patients with detectable ctDNA during CRT had shorter PFS (p = 0.014). Similarly, patients with post-CRT detectable ctDNA had a significantly shorter PFS (p = 0.012) and worse OS (p = 0.004). Moreover, patients with high bTMB levels during CRT had prolonged OS (p = 0.027). In conclusion, ctDNA and bTMB have the potential to predict treatment efficacy and survival in ESCC treated with CRT and immunotherapy.
Collapse
Affiliation(s)
- Baoqing Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Shiliang Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Yujia Zhu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Ruixi Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Xingyuan Cheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Biqi Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Mihnea P Dragomir
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yaru Zhang
- Nanjing Geneseeq Technology Inc, Nanjing, Jiangsu, PR China
| | - Yonghong Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Mengzhong Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Qiaoqiao Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China.
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China.
| | - Hong Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China.
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China.
| | - Mian Xi
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, PR China.
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China.
| |
Collapse
|
46
|
Yu J, Avriett TA, Ray CM, Kim RD. Circulating tumor DNA analysis guiding adjuvant treatment in resected stage III cholangiocarcinoma: a case report. J Gastrointest Oncol 2024; 15:485-490. [PMID: 38482231 PMCID: PMC10932665 DOI: 10.21037/jgo-23-815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/08/2023] [Indexed: 07/12/2024] Open
Abstract
Background Cholangiocarcinoma (CCA) is a rare and aggressive gastrointestinal cancer. Unfortunately, 60% to 70% of early-stage CCA patients experience disease recurrence after curative resection and standard adjuvant therapy. Currently, there is no reliable tool to identify CCA recurrence before radiographic detection. Longitudinal monitoring of circulating tumor DNA (ctDNA) has shown promising value in molecular identification of relapse prior to conventional surveillance in other solid tumors. However, there is a scarcity of data on ctDNA in CCA after curative surgery. Case Description An 81-year-old male with stage 3A intrahepatic CCA achieved radiographic remission after curative resection and was started on standard adjuvant capecitabine on post-operative day (POD) 50. Tumor-informed ctDNA tested positive on two consecutive occasions, with the titer increasing from 0.16 mean tumor molecule (MTM)/mL on POD 92 to 0.80 MTM/mL on POD 183, despite being on capecitabine. carbohydrate antigen 19-9 (CA19-9) also continued to increase from 175.6 U/mL on POD 92 to 7,594.9 U/mL on POD 217. Notably, surveillance computed tomography (CT) scans showed no evidence of disease (NED) on POD 126, 186, and 211. Molecular profiling and next-generation sequencing (NGS) panels from CCA tissue revealed microsatellite instability-high (MSI-H). After extensive discussions with the patient regarding the rising ctDNA titer despite being on capecitabine for nearly 6 months, we initiated pembrolizumab on POD 224 prior to radiographic recurrence. Given the tumor is MSI-H, and the preferred toxicity profile compared to the front-line chemotherapy option for CCA, we started pembrolizumab. ctDNA became undetectable, and CA19-9 returned to the reference range with pembrolizumab. As of the last follow-up on POD 876, the patient has continued pembrolizumab without noticeable side effects, and imaging continues to show NED, with persistent negative ctDNA and normal CA19-9 levels. Conclusions This case demonstrates the potential utility of tumor-informed ctDNA in CCA as (I) an early detection tool before radiographic recurrence; (II) a response monitoring tool as a surrogate biomarker that can guide therapy optimization; and (III) shows that early intervention with immunotherapy or potentially targeted agents based on ctDNA may lead to improved survival outcomes.
Collapse
Affiliation(s)
- James Yu
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Trenton A Avriett
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Christopher M Ray
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Richard D Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
47
|
Nielsen LR, Stensgaard S, Meldgaard P, Sorensen BS. ctDNA-based minimal residual disease detection in lung cancer patients treated with curative intended chemoradiotherapy using a clinically transferable approach. Cancer Treat Res Commun 2024; 39:100802. [PMID: 38428066 DOI: 10.1016/j.ctarc.2024.100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Reliable biomarkers are needed to identify tumor recurrence of non-small cell lung cancer (NSCLC) patients after chemoradiotherapy (CRT) with curative intent. This could improve consolidation therapy of progressing patients. However, the approach of existing studies has limited transferability to the clinic. MATERIALS AND METHODS A retrospective analysis of 135 plasma samples from 56 inoperable NSCLC patients who received CRT with curative intent was performed. Plasma samples collected at baseline, at the first check-up (average 1.6 months post-RT), and at the second check-up (average 4.5 months post-RT) were analyzed by deep sequencing with a commercially available cancer personalized profiling strategy (CAPP-Seq) using a tumor-agnostic approach. RESULTS Detection of circulating tumor DNA (ctDNA) at 4.5 months after therapy was significantly associated with higher odds of tumor recurrence (OR: 5.4 (CI: 1.1-31), Fisher's exact test: p-value = 0.022), and shorter recurrence-free survival (RFS) (HR: 4.1 (CI: 1.7-10); log-rank test: p-value = 9e-04). In contrast, detection of ctDNA at 1.6 months after therapy was not associated with higher odds of tumor recurrence (OR: 2.7 (CI: 0.67-12), Fisher's exact test: p-value = 0.13) or shorter RFS (HR: 1.5 (CI: 0.67-3.3); log-rank test: p-value = 0.32). CONCLUSION This study demonstrates that the detection of ctDNA can be used to identify minimal residual disease 4.5 months after CRT in NSCLC patients using a commercially available kit and a tumor-agnostic approach. Furthermore, the time point of collecting the plasma sample after CRT has decisive importance for the prognostic value of ctDNA. MICRO ABSTRACT This study analysed 135 plasma samples from 56 NSCLC patients treated with curative intent chemoradiotherapy using a tumor-agnostic approach. Detecting ctDNA at 4.5 months post-treatment was linked to higher recurrence odds, indicating ctDNA's potential as a biomarker for identifying residual disease after treatment with curative intent. Importantly, the study emphasizes the importance of timing for accurate ctDNA analysis results.
Collapse
Affiliation(s)
- Lærke Rosenlund Nielsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Simone Stensgaard
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Peter Meldgaard
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark; Department of Oncology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark.
| |
Collapse
|
48
|
Blewett T, Rhoades J, Liu R, Xiong K, Sridhar S, Crnjac A, Cheng J, Lawless AR, Frederick DT, Flaherty KT, Makrigiorgos GM, Adalsteinsson VA. MAESTRO-Pool Enables Highly Parallel and Specific Mutation-Enrichment Sequencing for Minimal Residual Disease Detection in Cohort Studies. Clin Chem 2024; 70:434-443. [PMID: 38069911 PMCID: PMC10847667 DOI: 10.1093/clinchem/hvad203] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/19/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND Tracing patient-specific tumor mutations in cell-free DNA (cfDNA) for minimal residual disease (MRD) detection is promising but challenging. Assaying more mutations and cfDNA stands to improve MRD detection but requires highly accurate, efficient sequencing methods and proper calibration to prevent false detection with bespoke tests. METHODS MAESTRO (Minor Allele Enriched Sequencing Through Recognition Oligonucleotides) uses mutation-specific oligonucleotide probes to enrich cfDNA libraries for tumor mutations and enable their accurate detection with minimal sequencing. A new approach, MAESTRO-Pool, which entails pooling MAESTRO probes for all patients and applying these to all samples from all patients, was used to screen for 22 333 tumor mutations from 9 melanoma patients in 98 plasma samples. This enabled quantification of MRD detection in patient-matched samples and false detection in unmatched samples from other patients. To detect MRD, a new dynamic MRD caller was used that computes a probability for MRD detection based on the number of mutations and cfDNA molecules sequenced, thereby calibrating for variations in each bespoke test. RESULTS MAESTRO-Pool enabled sensitive detection of MRD down to 0.78 parts per million (ppm), reflecting a 10- to 100-fold improvement over existing tests. Of the 8 MRD positive samples with ultra-low tumor fractions <10 ppm, 7 were either in upward-trend preceding recurrence or downward-trend aligning with response. Of 784 patient-unmatched tests, only one was found as MRD positive (tumor fraction = 2.7 ppm), suggesting high specificity. CONCLUSIONS MAESTRO-Pool enables massively parallel, tumor-informed MRD testing with concurrent benchmarking of bespoke MRD tests. Meanwhile, our new MRD caller enables more mutations and cfDNA molecules to be tested without compromising specificity. These improve the ability for detecting traces of MRD from blood.
Collapse
Affiliation(s)
- Timothy Blewett
- Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Justin Rhoades
- Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Ruolin Liu
- Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Kan Xiong
- Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Sainetra Sridhar
- Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Andjela Crnjac
- Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Ju Cheng
- Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Aleigha R Lawless
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Dennie T Frederick
- Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Gerassimos Mike Makrigiorgos
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Boston, MA, United States
| | - Viktor A Adalsteinsson
- Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
49
|
Mishra S, Srivastava P, Pandey A, Shukla S, Agarwal A, Husain N. Diagnostic Utility of Next-Generation Sequencing in Circulating Free DNA and a Comparison With Matched Tissue in Gallbladder Carcinoma. J Transl Med 2024; 104:100301. [PMID: 38092180 DOI: 10.1016/j.labinv.2023.100301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/15/2023] [Accepted: 12/05/2023] [Indexed: 01/14/2024] Open
Abstract
Mutation detection for therapy monitoring in cell-free DNA (cfDNA) is used clinically for some malignancies. Gallbladder carcinoma (GBC) presents a diagnostic challenge and has limited late-stage treatment options. To our knowledge, this novel study examines, for the first time, genomic alterations in cfDNA from GBC to assess diagnostic accuracy and therapeutic options. The concordance of somatic genomic changes in cfDNA and DNA from paired tumor tissue was analyzed. Paired serum and tissue samples from 40 histologically proven GBC, 20 cholecystitis, and 4 normal (noninflamed gallbladder) controls were included. Targeted next-generation sequencing with a 22-gene panel (Colon and Lung Cancer Research Panel v2, Thermo Scientific) in cfDNA and tumor tissue with high depth and uniform coverage on ION Personal Genome Machine (ION, PGM) was performed. A spectrum of 223 mutations in cfDNA and 225 mutations in formalin-fixed paraffin-embedded tissue DNA were identified in 22 genes. Mutations ranged from 1 to 17 per case. In cfDNA frequent alterations were in TP53 (85.0%), EGFR (52.5%), MET (35%) CTNNB1, SMAD4, BRAF (32.5%), PTEN (30%), FGFR3 and PIK3CA (27.5%), NOTCH1 (25.0%), and FBXW7 and ERBB4 (22.5%). At least one clinically actionable mutation was identified in all cfDNA samples. Paired samples shared 149 of 225 genetic abnormalities (66.2%). Individual gene mutation concordance ranged from 44.44% to 82.0% and was highest for EGFR (82.0%), BRAF and NOTCH1 (80.0%), TP53 (73.08%), MET (72.22%), and ERBB4 (71.42%) with a significant level of correlation (Spearman r = 0.91, P ≤ .0001). The sensitivity and specificity of the TP53 gene at the gene level was the highest (94.44% and 100.0%, respectively). Overall survival was higher for ERBB4 and ERBB2 mutant tumors. The adenocarcinoma subtype revealed specific genetic changes in ERBB4, SMAD4, ERBB2, PTEN, KRAS, and NRAS. NGS-based cfDNA mutation profiling can be used to diagnose GBC before surgery to guide treatment decisions. Targeted therapy identified in GBC included SMAD4, ERBB2, ERBB4, EGFR, KRAS, BRAF, PIK3CA, MET, and NRAS.
Collapse
Affiliation(s)
- Sridhar Mishra
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Pallavi Srivastava
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Anshuman Pandey
- Department of Gastrosurgery, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Saumya Shukla
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Akash Agarwal
- Department of Surgical Oncology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Nuzhat Husain
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
50
|
Wang R, Yang Y, Lu T, Cui Y, Li B, Liu X. Circulating cell-free DNA-based methylation pattern in plasma for early diagnosis of esophagus cancer. PeerJ 2024; 12:e16802. [PMID: 38313016 PMCID: PMC10838104 DOI: 10.7717/peerj.16802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/26/2023] [Indexed: 02/06/2024] Open
Abstract
With the increased awareness of early tumor detection, the importance of detecting and diagnosing esophageal cancer in its early stages has been underscored. Studies have consistently demonstrated the crucial role of methylation levels in circulating cell-free DNA (cfDNA) in identifying and diagnosing early-stage cancer. cfDNA methylation pertains to the methylation state within the genomic scope of cfDNA and is strongly associated with cancer development and progression. Several research teams have delved into the potential application of cfDNA methylation in identifying early-stage esophageal cancer and have achieved promising outcomes. Recent research supports the high sensitivity and specificity of cfDNA methylation in early esophageal cancer diagnosis, providing a more accurate and efficient approach for early detection and improved clinical management. Accordingly, this review aims to present an overview of methylation-based cfDNA research with a focus on the latest developments in the early detection of esophageal cancer. Additionally, this review summarizes advanced analytical technologies for cfDNA methylation that have significantly benefited from recent advancements in separation and detection techniques, such as methylated DNA immunoprecipitation sequencing (MeDIP-seq). Recent findings suggest that biomarkers based on cfDNA methylation may soon find successful applications in the early detection of esophageal cancer. However, large-scale prospective clinical trials are required to identify the potential of these biomarkers.
Collapse
Affiliation(s)
- Rui Wang
- School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yue Yang
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Tianyu Lu
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Youbin Cui
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Bo Li
- School of Public Health, Jilin University, Changchun, Jilin, China
| | - Xin Liu
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|