1
|
Kramer A, van Schaik LF, van den Broek D, Meijer GA, Gutierrez Ibarluzea I, Galnares Cordero L, Fijneman RJA, Ligtenberg MJL, Schuuring E, van Harten WH, Coupé VMH, Retèl VP. Towards Recommendations for Cost-Effectiveness Analysis of Predictive, Prognostic, and Serial Biomarker Tests in Oncology. PHARMACOECONOMICS 2025; 43:483-497. [PMID: 39920559 PMCID: PMC12011951 DOI: 10.1007/s40273-025-01470-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/19/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Cost-effectiveness analysis (CEA) of biomarkers is challenging due to the indirect impact on health outcomes and the lack of sufficient fit-for-purpose data. Hands-on guidance is lacking. OBJECTIVE We aimed firstly to explore how CEAs in the context of three different types of biomarker applications have addressed these challenges, and secondly to develop recommendations for future CEAs. METHODS A scoping review was performed for three biomarker applications: predictive, prognostic, and serial testing, in advanced non-small cell lung cancer, early-stage colorectal cancer, and all-stage colorectal cancer, respectively. Information was extracted on the model assumptions and uncertainty, and the reported outcomes. An in-depth analysis of the literature was performed describing the impact of model assumptions in the included studies. RESULTS A total of 43 CEAs were included (31 predictive, 6 prognostic, and 6 serial testing). Of these, 40 utilized different sources for test and treatment parameters, and three studies utilized a single source. Test performance was included in 78% of these studies utilizing different sources, but this parameter was differently expressed across biomarker applications. Sensitivity analyses for test performance was only performed in half of these studies. For the linkage of test results to treatments outcomes, a minority of the studies explored the impact of suboptimal adherence to test results, and/or explored potential differences in treatment effects for different biomarker subgroups. Intermediate outcomes were reported by 67% of studies. CONCLUSIONS We identified various approaches for dealing with challenges in CEAs of biomarker tests for three different biomarker applications. Recommendations on assumptions, handling uncertainty, and reported outcomes were drafted to enhance modeling practices for future biomarker cost-effectiveness evaluations.
Collapse
Affiliation(s)
- Astrid Kramer
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Lucas F van Schaik
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Daan van den Broek
- Department for Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gerrit A Meijer
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Remond J A Fijneman
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marjolijn J L Ligtenberg
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wim H van Harten
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Health technology and Services Research, University of Twente, Enschede, The Netherlands
| | - Veerle M H Coupé
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Valesca P Retèl
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Taieb J, Souglakos J, Boukovinas I, Falcoz A, Pages F, Messaritakis I, Bennouna J, Artru P, Louvet C, Lepere C, Emile JF, Bouche O, Mazard T, Vernerey D, Vogiatzoglou K, Tzardi M, Sharma S, Liu MC, Sethi H, André T, Galon J, Laurent-Puig P. Combined Analyses of Circulating Tumor DNA and Immunoscore in Patients With Stage III Colon Cancer: A Post Hoc Analysis of the PRODIGE-GERCOR IDEA-France/HORG-IDEA-Greece Trials. J Clin Oncol 2025; 43:1564-1577. [PMID: 39903903 PMCID: PMC12054978 DOI: 10.1200/jco.24.00648] [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: 03/27/2024] [Revised: 10/08/2024] [Accepted: 12/02/2024] [Indexed: 02/06/2025] Open
Abstract
PURPOSE Immunoscore (IS) and circulating tumor DNA (ctDNA) are two emerging technologies in improving prognostication and tailoring adjuvant treatments in patients resected from a stage III colon cancer (CC). Here, we analyzed the prognostic value of the two biomarkers in patients who participated in the randomized phase III IDEA-France and HORG trials. METHODS Plasma samples were collected after surgery and before adjuvant chemotherapy. ctDNA analysis was performed using a clinically validated, personalized, tumor-informed 16-plex protein chain reaction assay. Multivariable analyses for time to recurrence (TTR; patients without recurrence or death due to CC) and overall survival (OS) were performed using ctDNA and IS results, along with other parameters including treatment duration and disease risk group. RESULTS Of the 554 patients with available ctDNA results, 445 were ctDNA-negative (80.3%) and 109 were ctDNA-positive (19.7%); baseline characteristics showed more T4/N2 and venous embolism/lymphatic invasion/perineural invasion+ in ctDNA-positive patients. With a median follow-up of 6.7 years, the 2-year TTR rate was 43.5% (95% CI, 34.1 to 52.6) for ctDNA-positive patients and 88.1% (95% CI, 84.7 to 90.8) for ctDNA-negative patients (P < .0001). ctDNA was confirmed as an independent prognostic marker for both TTR (adjusted hazard ratio [adjHR], 5.21 [95% CI, 3.59 to 7.58]; P < .001) and OS (adjHR, 4.84 [95% CI, 3.40 to 6.89]; P < .001). ctDNA remained the most significant prognostic factor irrespective of disease stage, treatment duration, and IS results. IS was not prognostic in ctDNA-positive patients but remained a significant prognostic tool for ctDNA-negative patients. CONCLUSION In this combined analysis of two adjuvant trials dedicated to patients with stage III CC after surgery, ctDNA was detectable in 19.7% of the patients and was confirmed as a major independent prognostic biomarker. IS seems to bring additional prognostic information in the 80.3% of patients who are ctDNA-negative.
Collapse
Affiliation(s)
- Julien Taieb
- Department of Gastroenterology and Gastrointestinal Oncology, CARPEM Comprehensive Cancer Center, Georges-Pompidou European Hospital, AP-HP, Paris, France
- SIRIC CARPEM, Université Paris-Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, CNRS, Université Paris-Cité, Sorbonne Université, USPC, Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - John Souglakos
- Department of Medical Oncology, University Hospital of Heraklion, Crete, Greece
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, Greece
| | | | - Antoine Falcoz
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France
- INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Bourgogne Franche-Comté University, Besançon, France
| | - Franck Pages
- Department of Immunology, Georges-Pompidou European Hospital, AP-HP, INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, SIRIC CARPEM, Université Paris-Cité, Paris, France
| | - Ippokratis Messaritakis
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, Greece
| | - Jaafar Bennouna
- Department of Medical Oncology, Hospital Foch, Suresnes, France
| | - Pascal Artru
- Department of Medical Oncology, Private Hospital Jean Mermoz—Ramsay Santé, Lyon, France
| | - Christophe Louvet
- Department of Medical Oncology, Institut Mutualiste Montsouris, Paris, France
| | - Celine Lepere
- Department of Gastroenterology and Gastrointestinal Oncology, CARPEM Comprehensive Cancer Center, Georges-Pompidou European Hospital, AP-HP, Paris, France
- SIRIC CARPEM, Université Paris-Cité, Paris, France
| | - Jean Francois Emile
- Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
| | - Olivier Bouche
- Department of Digestive Oncology, Hospital Robert Debré, Reims, France
| | - Thibault Mazard
- Department of Medical Oncology, Montpellier Cancer Institute (ICM), Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Montpellier, France
| | - Dewi Vernerey
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France
- INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Bourgogne Franche-Comté University, Besançon, France
| | | | - Maria Tzardi
- Laboratory of Pathology, University Hospital of Heraklion, Crete, Greece
| | | | | | | | - Thierry André
- Sorbonne Université, Paris, France
- Department of Medical Oncology, Hôpital Saint Antoine, Paris, France
| | - Jérome Galon
- Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, INSERM, SIRIC CARPEM, Sorbonne Université, Université Paris Cité, Paris, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, CNRS, Université Paris-Cité, Sorbonne Université, USPC, Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
- Department of Biology, Assistance Publique—Hôpitaux de Paris, European Georges Pompidou Hospital, Paris, France
| |
Collapse
|
3
|
Yuan T, Edelmann D, Moreno V, Georgii E, de Andrade E Sousa LB, Pelin H, Jiang X, Kather JN, Tagscherer KE, Roth W, Bewerunge-Hudler M, Brobeil A, Kloor M, Bläker H, Brenner H, Hoffmeister M. Identification and external validation of tumor DNA methylation panel for the recurrence risk stratification of stage II colon cancer. Transl Oncol 2025; 57:102405. [PMID: 40311420 DOI: 10.1016/j.tranon.2025.102405] [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: 12/13/2024] [Revised: 03/19/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Tailoring surveillance and treatment strategies for stage II colon cancer (CC) after curative surgery remains challenging, and personalized approaches are lacking. We aimed to identify a gene methylation panel capable of stratifying high-risk stage II CC patients for recurrence beyond traditional clinical variables. METHODS Genome-wide tumor tissue DNA methylation data were analyzed from 562 stage II CC patients who underwent surgery in Germany (DACHS study). The cohort was divided into a training set (N = 395) and an internal validation set (N = 131), with external validation performed on 97 stage II CC patients from Spain. DNA methylation markers were primarily selected using the Elastic Net Cox model. The resulting prognostic index (PI), a combination of clinical factors and selected methylation markers, was compared to baseline models using clinical variables or microsatellite instability (MSI), with discrimination and prediction accuracy assessed through time-dependent receiver operating characteristic curves (AUC) and Brier scores. RESULTS The final PI incorporated age, sex, tumor stage, location, and 27 DNA methylation markers. The PI consistently outperformed the baseline model including age, sex, and tumor stage in time-dependent AUC across validation cohorts (e.g., 1-year AUC and 95 % confidence interval: internal validation set, PI: 0.66, baseline model: 0.52; external validation set, PI: 0.72, baseline model: 0.64). In internal validation, the PI also showed a consistently improved time-dependent AUC compared with a combination of MSI and tumor stage only. Nevertheless, the PI did not improve the prediction accuracy of CC recurrence compared to the baseline model. CONCLUSIONS This study identified 27 tumor tissue DNA methylation biomarkers that improved the discriminative power in classifying recurrence risk among stage II colon cancer patients. While this methylation panel alone lacks sufficient prediction accuracy for clinical application, its discriminative improvement suggests potential value as part of a multimodal risk-stratification tool.
Collapse
Affiliation(s)
- Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Dominic Edelmann
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Víctor Moreno
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO). Hospitalet de Llobregat, Barcelona, Spain; Colorectal Cancer Group, ONCOBELL, Bellvitge Biomedical Research Institute (IDIBELL). Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), Madrid, Spain; Department of Clinical Sciences, Faculty of Medicine and health Sciences and Universitat de Barcelona Institute of Complex Systems (UBICS), University of Barcelona, Barcelona, Spain
| | | | | | | | - Xiaofeng Jiang
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | | | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany; Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Alexander Brobeil
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Matthias Kloor
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hendrik Bläker
- Institute of Pathology, University of Leipzig Medical Center, Leipzig, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
4
|
Hoang T, Choi MK, Oh JH, Kim J. Utility of circulating tumor DNA to detect minimal residual disease in colorectal cancer: A systematic review and network meta-analysis. Int J Cancer 2025. [PMID: 40293388 DOI: 10.1002/ijc.35442] [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: 12/13/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025]
Abstract
Circulating tumor DNA (ctDNA) is a promising biomarker for predicting minimal residual disease (MRD) and guiding treatment decisions in patients with colorectal cancer (CRC). This study aimed to examine the study designs and settings of ongoing clinical trials that use ctDNA to guide treatment decisions and to determine the best timing for detecting MRD in non-metastatic CRC. We searched PubMed, Embase, Web of Science, Cochrane Library, and clinicaltrials.gov for English language records. The ctDNA settings from the clinical trials were categorized by randomization to ctDNA testing, treatment options based on ctDNA results, and the timing of ctDNA testing relative to adjuvant therapy. For prospective studies, a network meta-analysis using a frequentist approach was conducted to examine the pairwise associations between different ctDNA timing strategies and MRD, defined as recurrence, relapse, and progression. The main approaches in ctDNA-based interventional trial designs were categorized as ctDNA-guided treatment, ctDNA-by-treatment, ctDNA-guided surveillance, and ctDNA-enriched adjuvant therapy for guiding treatment decisions, including both escalation and de-escalation strategies, and surveillance. Overall, both preoperative and postoperative ctDNA detection were linked to higher risks of progression, with pooled hazard ratios (95% confidence intervals) of 5.23 (2.10-13.00) and 7.95 (5.30-11.91), respectively. Among the timing strategies, ctDNA testing after adjuvant therapy was the most effective for identifying high-risk patients, strongly suggesting the presence of residual disease. This study comprehensively reviewed the clinical settings of ctDNA testing in ongoing trials and provided evidence supporting the selection of post-adjuvant therapy as the optimal timing for ctDNA testing.
Collapse
Affiliation(s)
- Tung Hoang
- Department of Cancer AI & Digital Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Republic of Korea
- Faculty of Pharmacy, University of Health Sciences, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Moon Ki Choi
- Center for Colorectal Cancer, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Jae Hwan Oh
- Center for Colorectal Cancer, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Jeongseon Kim
- Department of Cancer AI & Digital Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
5
|
Zhou Q, Chen X, Zeng B, Zhang M, Guo N, Wu S, Zeng H, Sun F. Circulating tumor DNA as a biomarker of prognosis prediction in colorectal cancer: a systematic review and meta-analysis. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:167-178. [PMID: 40265088 PMCID: PMC12010414 DOI: 10.1016/j.jncc.2024.05.007] [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: 10/21/2023] [Revised: 04/18/2024] [Accepted: 05/14/2024] [Indexed: 02/03/2025] Open
Abstract
OBJECTIVE Circulating tumor DNA (ctDNA) is increasingly being used as a potential biomarker in colorectal cancer (CRC) patients. However, the role of ctDNA in CRC prognosis prediction remains unclear. The objective is to systematically assess the clinical value of ctDNA in colorectal cancer prognosis prediction throughout the treatment cycle. METHODS PubMed, Web of Science, Embase, Cochrane Library, Scopus, and clinical trials.gov database was searched from January 2016 to April 2023. Observational studies and randomized clinical trials reporting on ctDNA and prognostic outcomes in CRC patients were included. Pooled hazard risk ratios (HRs) were calculated for the primary outcomes, relapse-free survival (RFS), and overall survival (OS). Random-effects models were preferred considering the potential heterogeneity. RESULTS Sixty-five cohort studies were included. Association between ctDNA and shorter RFS or OS was significant, especially after the full-course treatment recommended by the guidelines (HR = 8.92 [ 95 % CI: 6.02-13.22], P < 0.001, I2 = 73 %; HR = 3.05 [ 95 % CI: 1.72-5.41], P < 0.001, I2 = 48 %) for all types of CRC patients. Despite the presence of heterogeneity, subgroup analyses showed that the cancer type and ctDNA detection assays may be the underlying cause. Besides, ctDNA may detect recurrence earlier than radiographic progression, but no uniform sampling time point between studies might bring bias. However, ctDNA detection did not appear to correlate with pathological complete response achievement in patients with locally advanced rectal cancer. CONCLUSION ctDNA detection was significantly associated with poorer prognosis. The potential applications in prognostic prediction are promising and remain to be evaluated in other fields.
Collapse
Affiliation(s)
- Qingxin Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
- Tianjin Centers for Disease Control and Prevention, Tianjin, China
| | - Xiaowei Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
| | - Baoqi Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
- Central Laboratory, Tianjin Fifth Central Hospital (Peking University Binhai Hospital), Tianjin, China
| | - Meng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
| | - Nana Guo
- Hebei Centers for Disease Control and Prevention, Hebei, China
| | - Shanshan Wu
- Clinical Epidemiology and EBM Unit, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hongmei Zeng
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
- Key Laboratory of Major Disease Epidemiology, Ministry of Education (Peking University), Beijing, China
- Xinjiang Medical University, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
6
|
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
|
7
|
Bartolomucci A, Nobrega M, Ferrier T, Dickinson K, Kaorey N, Nadeau A, Castillo A, Burnier JV. Circulating tumor DNA to monitor treatment response in solid tumors and advance precision oncology. NPJ Precis Oncol 2025; 9:84. [PMID: 40122951 PMCID: PMC11930993 DOI: 10.1038/s41698-025-00876-y] [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: 10/02/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Circulating tumor DNA (ctDNA) has emerged as a dynamic biomarker in cancer, as evidenced by its increasing integration into clinical practice. Carrying tumor specific characteristics, ctDNA can be used to inform treatment selection, monitor response, and identify drug resistance. In this review, we provide a comprehensive, up-to-date summary of ctDNA in monitoring treatment response with a focus on lung, colorectal, and breast cancers, and discuss current challenges and future directions.
Collapse
Affiliation(s)
- Alexandra Bartolomucci
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Monyse Nobrega
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Tadhg Ferrier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nivedita Kaorey
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Alberto Castillo
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Department of Pathology, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
8
|
Li S, Li Y, Wei W, Gong C, Wang T, Li G, Yao F, Ou JH, Xu Y, Wu W, Jin L, Rao N, Nie Y, Yu F, Jia W, Li XR, Zhang J, Yang HW, Yang Y, Wu M, Li Q, Li F, Gong Y, Yi X, Liu Q. Dynamic ctDNA tracking stratifies relapse risk for triple negative breast cancer patients receiving neoadjuvant chemotherapy. Nat Commun 2025; 16:2786. [PMID: 40118933 PMCID: PMC11928511 DOI: 10.1038/s41467-025-57988-z] [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: 06/24/2024] [Accepted: 03/06/2025] [Indexed: 03/24/2025] Open
Abstract
Early Triple negative breast cancer (eTNBC) is the subtype with the worst outcome. Circulating tumor DNA (ctDNA) is shown to predict the prognosis of breast cancer, but its utility in eTNBC remains unclear. 130 stage II-III female eTNBC patients receiving neoadjuvant chemotherapy (NAC) have been enrolled prospectively and subjected to ctDNA analysis. ctDNA at post-NAC (pre-surgery) and post-surgery, but not at baseline, is associated with worse prognosis. A threshold of 1.1% maximum variant allele frequency at baseline stratifies patients with different relapse risk, which is validated internally and externally. A systemic tumor burden model integrating baseline and post-surgery ctDNA is independently prognostic (p = 0.022). Combining systemic tumor burden with pathologic response identifies a highly curable subgroup and a subgroup of high-risk eTNBC patients. ctDNA surveillance during follow-up identifies patients with high relapse risk. In conclusion, systemic ctDNA analysis demonstrates the utility of a systemic tumor burden model of ctDNA in risk stratification of eTNBC patients, which may guide future treatment escalation or de-escalation trials.
Collapse
Affiliation(s)
- Shunying Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yudong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Wei
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Chang Gong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ting Wang
- Department of thyroid, breast and vascular surgery, Xijing Hospital, The Air Force Medical University, Xi'an, China
| | - Guangxin Li
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Feng Yao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiang-Hua Ou
- Department of Breast Surgery, Xinjiang Cancer Hospital, Xinjiang Medical University, Urumqi, China
| | - Yan Xu
- Department of Breast and Thyroid Surgery, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Wei Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Jin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nanyan Rao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Nie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fengyan Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weijuan Jia
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xing-Rui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Hua-Wei Yang
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yaping Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mengzi Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qin Li
- Geneplus-Beijing Institute, Beijing, China
| | - Fang Li
- Geneplus-Beijing Institute, Beijing, China
| | - Yuhua Gong
- Geneplus-Beijing Institute, Beijing, China
| | - Xin Yi
- Geneplus-Beijing Institute, Beijing, China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
9
|
Faulkner LG, Howells L, Lehman S, Cowley C, Sidat Z, Shaw J, Thomas AL. Clinical Validation of Local Versus Commercial Genomic Testing in Cancer: A Comparison of Tissue and Plasma Concordance. Cancer Invest 2025; 43:119-140. [PMID: 39989311 DOI: 10.1080/07357907.2025.2464684] [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: 09/22/2024] [Accepted: 02/04/2025] [Indexed: 02/25/2025]
Abstract
Genomic sequencing of tumours improves patient outcomes through implementation of precision oncology. At present, genomic testing is mainly confined to research settings, with samples sent to biopharmaceutical companies for analysis. The ever-expanding catalogue approved of targeted therapies has created an urgent unmet need for local genomic testing facilities, to enable upscaling of testing. Here, we compare the outcomes of local (IonTorrent™) and commercial (Foundation Medicine) genomic testing collected from 30 cancer patients in from plasma and tissue samples. Overall concordance was high in both tissue (98%) and plasma (94.2%). Variants identified by both platforms had a strong correlation in variant allele frequencies (VAF%): plasma: r = 0.99 p < 0.0001, tissue: r = 0.91 p < 0.0001. However, numerous low VAF% variants resulted in low positive percentage agreement (tissue 78.8% plasma 16.1%) and positive predictive values (tissue 56.3% plasma 71.4%). Local sequencing demonstrated higher fidelity in detecting fusions but low fidelity in detecting indels. Overall, this study supports the use of local genomic testing for routine molecular diagnostics but highlights outstanding issues before widespread implementation. Processing of variants detected at low VAF% and the limit of detection of assays needs to be addressed. Construction of gene panels requires careful consideration, including incorporation of markers of genomic instability.
Collapse
Affiliation(s)
- Lucy G Faulkner
- Department of Oncology, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, UK
- Leicester Cancer Research Centre, Robert Kilpatrick Clinical Sciences Building, University of Leicester, Leicester, UK
| | - Lynne Howells
- Leicester Cancer Research Centre, Robert Kilpatrick Clinical Sciences Building, University of Leicester, Leicester, UK
| | - Susann Lehman
- Leicester Cancer Research Centre, Robert Kilpatrick Clinical Sciences Building, University of Leicester, Leicester, UK
| | - Caroline Cowley
- Leicester Cancer Research Centre, Robert Kilpatrick Clinical Sciences Building, University of Leicester, Leicester, UK
| | - Zahirah Sidat
- Hope Clinical Trials Facility, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Jacqui Shaw
- Leicester Cancer Research Centre, Robert Kilpatrick Clinical Sciences Building, University of Leicester, Leicester, UK
| | - Anne L Thomas
- Department of Oncology, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, UK
- Leicester Cancer Research Centre, Robert Kilpatrick Clinical Sciences Building, University of Leicester, Leicester, UK
| |
Collapse
|
10
|
Tsukamoto R, Sugimoto K, Ii Y, Irie T, Kawaguchi M, Kobari A, Tsuchiya Y, Honjo K, Kawai M, Ishiyama S, Takahashi M, Sakamoto K. Prognostic Impact of the Postoperative Carcinoembryonic Antigen Level after Curative Resection of Locally Advanced Rectal Cancer. J Anus Rectum Colon 2025; 9:69-78. [PMID: 39882227 PMCID: PMC11772802 DOI: 10.23922/jarc.2024-035] [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: 04/25/2024] [Accepted: 10/03/2024] [Indexed: 01/31/2025] Open
Abstract
Objectives This study was conducted to investigate whether preoperative or postoperative carcinoembryonic antigen (CEA) with a new cut-off value is more optimal for predicting long-term outcomes in patients with Stage II/III rectal cancer, and to investigate the effectiveness of postoperative adjuvant chemotherapy (POAC) based on the CEA values. Methods Serum CEA levels were measured preoperatively (pre-CEA) and postoperatively (post-CEA). The area under the receiver operating curve (AUROC) was used to determine a cut-off for CEA. The cut-off for CEA relative to recurrence-free survival (RFS) was established as that giving the highest AUROC. In comparison of superiority between pre- and post- CEA levels, Akaike's information criterion (AIC) was used in the Cox proportional-hazard regression model. Results The subjects were 323 patients who underwent curative surgical treatment for Stage II/III rectal cancer. AIC values indicated that RFS was better stratified by a post-CEA level with a cut-off of 2.3 ng/ml compared with other classifications of pre- or post- CEA. In Stage III or high-risk Stage II cases, there was no effect of POAC on RFS in those with post-CEA <2.3 ng/ml (p=0.39), but in those with post-CEA ≥2.3 ng/ml there was a trend for better RFS in patients who received POAC compared to those without POAC (p=0.06). Conclusions Patients with post-CEA ≥2.3 ng/ml had worse long-term outcomes compared with those with post-CEA <2.3 ng/ml. Post-CEA with a cut-off of 2.3 ng/ml may be useful in determining the indication for POAC for in Stage III or high-risk Stage II cases.
Collapse
Affiliation(s)
- Ryoichi Tsukamoto
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Kiichi Sugimoto
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuki Ii
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Takahiro Irie
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Megumi Kawaguchi
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Aya Kobari
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuki Tsuchiya
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Kumpei Honjo
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Masaya Kawai
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Shun Ishiyama
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Makoto Takahashi
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Kazuhiro Sakamoto
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Abidoye O, Ahn DH, Borad MJ, Wu C, Bekaii-Saab T, Chakrabarti S, Sonbol MB. Circulating Tumor DNA Testing for Minimal Residual Disease and Its Application in Colorectal Cancer. Cells 2025; 14:161. [PMID: 39936953 PMCID: PMC11817155 DOI: 10.3390/cells14030161] [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: 12/13/2024] [Revised: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Colorectal cancer (CRC) represents a heterogeneous group of diseases that imposes a considerable global and national health burden. Although most CRC patients are diagnosed at an early stage and undergo potentially curative treatment, a significant proportion experience recurrence. Currently, adjuvant chemotherapy decisions are primarily based on clinicopathological characteristics, which have well-recognized limitations in accurately identifying patients harboring minimal residual disease (MRD), often resulting in unnecessary chemotherapy exposure. Circulating tumor DNA (ctDNA) has emerged as a promising surrogate marker for MRD, offering a more precise approach to identifying patients at risk of recurrence after curative-intent surgery and refining adjuvant chemotherapy decisions. Growing evidence from multiple studies has demonstrated that ctDNA outperforms traditional clinicopathological factors as a marker for MRD. This review synthesizes key studies supporting the role of ctDNA in MRD detection for CRC patients and evaluates clinical trials investigating the application of ctDNA in guiding adjuvant therapy decisions. This emerging strategy holds the potential to transform the adjuvant treatment paradigm in colorectal cancer by optimizing therapeutic precision and minimizing unnecessary treatment.
Collapse
Affiliation(s)
- Oluseyi Abidoye
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA; (D.H.A.); (M.J.B.); (C.W.); (T.B.-S.)
| | - Daniel H. Ahn
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA; (D.H.A.); (M.J.B.); (C.W.); (T.B.-S.)
| | - Mitesh J. Borad
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA; (D.H.A.); (M.J.B.); (C.W.); (T.B.-S.)
| | - Christina Wu
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA; (D.H.A.); (M.J.B.); (C.W.); (T.B.-S.)
| | - Tanios Bekaii-Saab
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA; (D.H.A.); (M.J.B.); (C.W.); (T.B.-S.)
| | - Sakti Chakrabarti
- University Hospital Seidman Cancer Center, Cleveland, OH 44106, USA;
| | - Mohamad Bassam Sonbol
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA; (D.H.A.); (M.J.B.); (C.W.); (T.B.-S.)
| |
Collapse
|
12
|
Bleckman RF, Haag CMSC, Rifaela N, Beukema G, Mathijssen RHJ, Steeghs N, Gelderblom H, Desar IME, Cleven A, ter Elst A, Schuuring E, Reyners AKL. Levels of circulating tumor DNA correlate with tumor volume in gastro-intestinal stromal tumors: an exploratory long-term follow-up study. Mol Oncol 2024; 18:2658-2667. [PMID: 38790141 PMCID: PMC11547224 DOI: 10.1002/1878-0261.13644] [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: 10/10/2023] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 05/26/2024] Open
Abstract
Patients with gastro-intestinal stromal tumors (GISTs) undergoing tyrosine kinase inhibitor therapy are monitored with regular computed tomography (CT) scans, exposing patients to cumulative radiation. This exploratory study aimed to evaluate circulating tumor DNA (ctDNA) testing to monitor treatment response and compare changes in ctDNA levels with RECIST 1.1 and total tumor volume measurements. Between 2014 and 2021, six patients with KIT proto-oncogene, receptor tyrosine kinase (KIT) exon-11-mutated GIST from whom long-term plasma samples were collected prospectively were included in the study. ctDNA levels of relevant plasma samples were determined using the KIT exon 11 digital droplet PCR drop-off assay. Tumor volume measurements were performed using a semi-automated approach. In total, 94 of 130 clinically relevant ctDNA samples were analyzed. Upon successful treatment response, ctDNA became undetectable in all patients. At progressive disease, ctDNA was detectable in five out of six patients. Higher levels of ctDNA correlated with larger tumor volumes. Undetectable ctDNA at the time of progressive disease on imaging was consistent with lower tumor volumes compared to those with detectable ctDNA. In summary, ctDNA levels seem to correlate with total tumor volume at the time of progressive disease. Our exploratory study shows promise for including ctDNA testing in treatment follow-up.
Collapse
Affiliation(s)
- Roos F. Bleckman
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Charlotte M. S. C. Haag
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Naomi Rifaela
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Gerrieke Beukema
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Ron H. J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer InstituteErasmus University Medical CenterRotterdamThe Netherlands
| | - Neeltje Steeghs
- Department of Medical OncologyThe Netherlands Cancer Institute Antoni van LeeuwenhoekAmsterdamThe Netherlands
| | - Hans Gelderblom
- Department of Medical OncologyLeiden University Medical CenterThe Netherlands
| | - Ingrid M. E. Desar
- Department of Medical OncologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Arjen Cleven
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
- Department of Medical OncologyLeiden University Medical CenterThe Netherlands
| | - Arja ter Elst
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Ed Schuuring
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Anna K. L. Reyners
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| |
Collapse
|
13
|
Lișcu HD, Verga N, Atasiei DI, Badiu DC, Dumitru AV, Ultimescu F, Pavel C, Stefan RE, Manole DC, Ionescu AI. Biomarkers in Colorectal Cancer: Actual and Future Perspectives. Int J Mol Sci 2024; 25:11535. [PMID: 39519088 PMCID: PMC11546354 DOI: 10.3390/ijms252111535] [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: 10/04/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Biomarkers in colorectal cancer (CRC) are of great interest in the current literature due to improvements in techniques such as liquid biopsy and next-generation sequencing (NGS). However, screening methods vary globally, with multi-target stool DNA (mt-sDNA) predominantly used in the USA and, more recently, the Cologuard Plus; biomarkers such as the Galectins family and septins show promise in early detection. Gut microbiome assessments, such as Fusobacterium nucleatum, are under intense exploration. Diagnostic tests, such as circulating DNA analysis via NGS, exhibit effectiveness and are being increasingly adopted. Circulating tumor cells emerge as potential alternatives to traditional methods in terms of diagnosis and prognosis. Predictive biomarkers are well established in guidelines; nonetheless, with the aid of machine learning and artificial intelligence, these biomarkers may be improved. This review critically explores the actual dynamic landscape of CRC biomarkers and future, promising biomarkers involved in screening, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Horia-Dan Lișcu
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Nicolae Verga
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Dimitrie-Ionuț Atasiei
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
| | - Dumitru-Cristinel Badiu
- Department of Surgery, Bagdasar Arseni Clinical Emergency Hospital, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Adrian Vasile Dumitru
- Department of Pathology, University Emergency Hospital Bucharest, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Flavia Ultimescu
- Department of Pathology, Institute of Oncology Alexandru Trestioreanu, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Christopher Pavel
- Department of Gastroenterology, Floreasca Emergency Hospital Bucharest, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Roxana-Elena Stefan
- General Surgery Department, Clinic of General and Esophageal Surgery, Sf. Maria Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Diandra-Carmen Manole
- Department of Endocrinology, Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Andreea-Iuliana Ionescu
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| |
Collapse
|
14
|
Saldanha EF. The role of liquid biopsy as a catalyst for sustained progress in precision oncology - Perspective of the young committee of the international society of liquid biopsy. THE JOURNAL OF LIQUID BIOPSY 2024; 5:100156. [PMID: 40027940 PMCID: PMC11863974 DOI: 10.1016/j.jlb.2024.100156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 03/05/2025]
Affiliation(s)
- Erick F. Saldanha
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, ON, Canada
- Young Committee, International Society of Liquid Biopsy, Spain
| |
Collapse
|
15
|
Kramer A, Greuter MJE, Schraa SJ, Vink GR, Phallen J, Velculescu VE, Meijer GA, van den Broek D, Koopman M, Roodhart JML, Fijneman RJA, Retèl VP, Coupé VMH. Early evaluation of the effectiveness and cost-effectiveness of ctDNA-guided selection for adjuvant chemotherapy in stage II colon cancer. Ther Adv Med Oncol 2024; 16:17588359241266164. [PMID: 39175989 PMCID: PMC11339739 DOI: 10.1177/17588359241266164] [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: 01/04/2024] [Accepted: 06/18/2024] [Indexed: 08/24/2024] Open
Abstract
Background Current patient selection for adjuvant chemotherapy (ACT) after curative surgery for stage II colon cancer (CC) is suboptimal, causing overtreatment of high-risk patients and undertreatment of low-risk patients. Postoperative circulating tumor DNA (ctDNA) could improve patient selection for ACT. Objectives We conducted an early model-based evaluation of the (cost-)effectiveness of ctDNA-guided selection for ACT in stage II CC in the Netherlands to assess the conditions for cost-effective implementation. Methods A validated Markov model, simulating 1000 stage II CC patients from diagnosis to death, was supplemented with ctDNA data. Five ACT selection strategies were evaluated: the current guideline (pT4, pMMR), ctDNA-only, and three strategies that combined ctDNA status with pT4 and pMMR status in different ways. For each strategy, the costs, life years, quality-adjusted life years (QALYs), recurrences, and CC deaths were estimated. Sensitivity analyses were performed to assess the impact of the costs of ctDNA testing, strategy adherence, ctDNA as a predictive biomarker, and ctDNA test performance. Results Model predictions showed that compared to current guidelines, the ctDNA-only strategy was less effective (+2.2% recurrences, -0.016 QALYs), while the combination strategies were more effective (-3.6% recurrences, +0.038 QALYs). The combination strategies were not cost-effective, since the incremental cost-effectiveness ratio was €67,413 per QALY, exceeding the willingness-to-pay threshold of €50,000 per QALY. Sensitivity analyses showed that the combination strategies would be cost-effective if the ctDNA test costs were lower than €1500, or if ctDNA status was predictive of treatment response, or if the ctDNA test performance improved substantially. Conclusion Adding ctDNA to current high-risk clinicopathological features (pT4 and pMMR) can improve patient selection for ACT and can also potentially be cost-effective. Future studies should investigate the predictive value of post-surgery ctDNA status to accurately evaluate the cost-effectiveness of ctDNA testing for ACT decisions in stage II CC.
Collapse
Affiliation(s)
- Astrid Kramer
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, De Boelelaan 1089a, Amsterdam, Noord-Holland 1081 HV, The Netherlands
| | - Marjolein J. E. Greuter
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Suzanna J. Schraa
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Geraldine R. Vink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Research and Development, IKNL, Utrecht, The Netherlands
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Victor E. Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerrit A. Meijer
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daan van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jeanine M. L. Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Valesca P. Retèl
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Veerle M. H. Coupé
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Hanna GJ, Dennis MJ, Scarfo N, Mullin MS, Sethi RK, Sehgal K, Annino DJ, Goguen LA, Haddad RI, Tishler RB, Margalit DN, Uppaluri R, Schoenfeld JD, Rettig EM. Personalized ctDNA for Monitoring Disease Status in Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 2024; 30:3329-3336. [PMID: 38824449 PMCID: PMC11292193 DOI: 10.1158/1078-0432.ccr-24-0590] [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: 02/26/2024] [Revised: 04/08/2024] [Accepted: 05/17/2024] [Indexed: 06/03/2024]
Abstract
PURPOSE Many patients with locoregionally advanced human papillomavirus-negative head and neck squamous cell carcinoma (HNSCC) relapse. ctDNA has the potential to identify minimal residual disease, but its clinical utility for virus-negative HNSCC is not well understood. EXPERIMENTAL DESIGN We retrospectively evaluated a personalized, commercial ctDNA assay (Signatera, Natera) during clinical care of patients treated for predominantly newly diagnosed human papillomavirus-negative HNSCC. Signatera utilizes 16-plex PCR from matched tumor and blood. Objectives were to understand ctDNA detectability and correlate changes posttreatment with disease outcomes. RESULTS Testing was successful in 100/116 (86%) patients (median age: 65 years, 68% male, 65% smokers); testing failed in 16 (14%) because of insufficient tissue. Oral cavity (55, 47%) tumors were most common; most had stage III to IV disease (82, 71%), whereas 17 (15%) had distant metastases. Pretreatment, 75/100 patients with successful testing (75%) had detectable ctDNA (range: 0.03-4049.69 mean tumor molecules/mL). No clinical features predicted ctDNA detectability or levels (multivariate analysis). At a median follow-up of 5.1 months (range: 0.2-15.1), 55 (55%) had >1 test result (range: 1-7; 194 samples). Of 55 patients, 17 (31%) remained ctDNA positive after starting treatment. Progression-free survival was significantly worse for patients who were ctDNA positive versus ctDNA negative posttreatment (HR, 7.33; 95% confidence interval, 3.12-17.2; P < 0.001); 1-year overall survival was 89.1% versus 100%, respectively (HR, 7.46; 95% confidence interval, 0.46-119.5; P = 0.155). CONCLUSIONS Tumor-informed ctDNA testing is feasible in nonviral HNSCC. ctDNA positivity is an indicator of disease progression and associated with inferior survival. Further research is warranted to understand whether ctDNA may be leveraged to guide therapy in HNSCC.
Collapse
Affiliation(s)
- Glenn J. Hanna
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Michael J. Dennis
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Nicole Scarfo
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Michelle S. Mullin
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Rosh K.V. Sethi
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- Head and Neck Surgical Oncology, Brigham and Women’s Hospital, Boston, Massachusetts.
| | - Kartik Sehgal
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Donald J. Annino
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- Head and Neck Surgical Oncology, Brigham and Women’s Hospital, Boston, Massachusetts.
| | - Laura A. Goguen
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- Head and Neck Surgical Oncology, Brigham and Women’s Hospital, Boston, Massachusetts.
| | - Robert I. Haddad
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Roy B. Tishler
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Brigham Cancer Center, Boston, Massachusetts.
| | - Danielle N. Margalit
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Brigham Cancer Center, Boston, Massachusetts.
| | - Ravindra Uppaluri
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- Head and Neck Surgical Oncology, Brigham and Women’s Hospital, Boston, Massachusetts.
| | - Jonathan D. Schoenfeld
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Brigham Cancer Center, Boston, Massachusetts.
| | - Eleni M. Rettig
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- Head and Neck Surgical Oncology, Brigham and Women’s Hospital, Boston, Massachusetts.
| |
Collapse
|
17
|
Smith HG, Schlesinger NH, Krarup PM, Nordholm-Carstensen A. Current treatment of pT1 rectal cancers in Denmark: A retrospective national cohort study. Colorectal Dis 2024; 26:1175-1183. [PMID: 38807258 DOI: 10.1111/codi.17049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/28/2024] [Accepted: 05/05/2024] [Indexed: 05/30/2024]
Abstract
AIM Organ preservation strategies for patients with rectal cancer are increasingly common. In appropriately selected patients, local excision (LE) of pT1 cancers can reduce morbidity without compromising cancer-related outcomes. However, determining the need for completion surgery after LE can be challenging, and it is unknown if prior LE compromises subsequent total mesorectal excision (TME). The aim of this study is to describe the current management of patients with pT1 rectal cancers. METHOD This is a retrospective national cohort study of the Danish Colorectal Cancer Group database, including patients with newly diagnosed pT1 cancers between 2016 and 2020. Patients were stratified according to treatment into LE alone, completion TME after LE or upfront TME. The treatment and outcomes of these groups were compared. RESULTS A total of 1056 patients were included. Initial LE was performed in 715 patients (67.7%), of whom 194 underwent completion TME (27.1%). The remaining 341 patients underwent upfront TME (32.3%). Patients undergoing LE alone were more likely to be male with low rectal cancers and greater comorbidity. No differences in specimen quality or perioperative outcomes were noted between patients undergoing completion or upfront TME. Eighty-five patients (15.9%) had lymph node metastases (LNM). Pathological risk factors poorly discriminated between patients with and without LNM, with similar rates seen in patients with zero (14.1%), one (12.0%) or two (14.4%) risk factors. CONCLUSION LE is a key component of the treatment of pT1 rectal cancer and does not appear to affect the outcomes of completion TME. Patient selection for completion TME remains a major challenge, with current stratification methods appearing to be inadequate.
Collapse
Affiliation(s)
- Henry G Smith
- Abdominalcenter K, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Nis H Schlesinger
- Abdominalcenter K, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Peter-Martin Krarup
- Abdominalcenter K, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | | |
Collapse
|
18
|
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
|
19
|
Alba-Bernal A, Godoy-Ortiz A, Domínguez-Recio ME, López-López E, Quirós-Ortega ME, Sánchez-Martín V, Roldán-Díaz MD, Jiménez-Rodríguez B, Peralta-Linero J, Bellagarza-García E, Troyano-Ramos L, Garrido-Ruiz G, Hierro-Martín MI, Vicioso L, González-Ortiz Á, Linares-Valencia N, Velasco-Suelto J, Carbajosa G, Garrido-Aranda A, Lavado-Valenzuela R, Álvarez M, Pascual J, Comino-Méndez I, Alba E. Increased blood draws for ultrasensitive ctDNA and CTCs detection in early breast cancer patients. NPJ Breast Cancer 2024; 10:36. [PMID: 38750090 PMCID: PMC11096188 DOI: 10.1038/s41523-024-00642-6] [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: 11/04/2023] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Early breast cancer patients often experience relapse due to residual disease after treatment. Liquid biopsy is a methodology capable of detecting tumor components in blood, but low concentrations at early stages pose challenges. To detect them, next-generation sequencing has promise but entails complex processes. Exploring larger blood volumes could overcome detection limitations. Herein, a total of 282 high-volume plasma and blood-cell samples were collected for dual ctDNA/CTCs detection using a single droplet-digital PCR assay per patient. ctDNA and/or CTCs were detected in 100% of pre-treatment samples. On the other hand, post-treatment positive samples exhibited a minimum variant allele frequency of 0.003% for ctDNA and minimum cell number of 0.069 CTCs/mL of blood, surpassing previous investigations. Accurate prediction of residual disease before surgery was achieved in patients without a complete pathological response. A model utilizing ctDNA dynamics achieved an area under the ROC curve of 0.92 for predicting response. We detected disease recurrence in blood in the three patients who experienced a relapse, anticipating clinical relapse by 34.61, 9.10, and 7.59 months. This methodology provides an easily implemented alternative for ultrasensitive residual disease detection in early breast cancer patients.
Collapse
Affiliation(s)
- Alfonso Alba-Bernal
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Andalusia-Roche Network in Precision Medical Oncology, 41092, Sevilla, Spain
| | - Ana Godoy-Ortiz
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), 28029, Madrid, Spain
| | - María Emilia Domínguez-Recio
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
| | - Esperanza López-López
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
| | - María Elena Quirós-Ortega
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Andalusia-Roche Network in Precision Medical Oncology, 41092, Sevilla, Spain
| | - Victoria Sánchez-Martín
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), 28029, Madrid, Spain
| | - María Dunia Roldán-Díaz
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
| | - Begoña Jiménez-Rodríguez
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), 28029, Madrid, Spain
| | - Jesús Peralta-Linero
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
| | - Estefanía Bellagarza-García
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
| | - Laura Troyano-Ramos
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
| | - Guadalupe Garrido-Ruiz
- Radiology Department, Hospital Clinico Universitario Virgen de la Victoria de Malaga, 29010, Malaga, Spain
| | - M Isabel Hierro-Martín
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Unidad de Gestion Clinica Provincial de Anatomia Patologica de Malaga, Hospital Clinico Universitario Virgen de la Victoria de Malaga, 29010, Malaga, Spain
- University of Málaga, Faculty of Medicine, 29010, Malaga, Spain
| | - Luis Vicioso
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Unidad de Gestion Clinica Provincial de Anatomia Patologica de Malaga, Hospital Clinico Universitario Virgen de la Victoria de Malaga, 29010, Malaga, Spain
- University of Málaga, Faculty of Medicine, 29010, Malaga, Spain
| | - Álvaro González-Ortiz
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
| | - Noelia Linares-Valencia
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
| | - Jesús Velasco-Suelto
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
| | - Guillermo Carbajosa
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- University of Málaga, Faculty of Medicine, 29010, Malaga, Spain
| | - Alicia Garrido-Aranda
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Andalusia-Roche Network in Precision Medical Oncology, 41092, Sevilla, Spain
- Laboratorio de biologia molecular del cancer (LBMC), Centro de investigaciones medico-sanitarias (CIMES-UMA), 29010, Malaga, Spain
| | - Rocío Lavado-Valenzuela
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Andalusia-Roche Network in Precision Medical Oncology, 41092, Sevilla, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), 28029, Madrid, Spain
- Laboratorio de biologia molecular del cancer (LBMC), Centro de investigaciones medico-sanitarias (CIMES-UMA), 29010, Malaga, Spain
| | - Martina Álvarez
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Andalusia-Roche Network in Precision Medical Oncology, 41092, Sevilla, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), 28029, Madrid, Spain
- University of Málaga, Faculty of Medicine, 29010, Malaga, Spain
- Laboratorio de biologia molecular del cancer (LBMC), Centro de investigaciones medico-sanitarias (CIMES-UMA), 29010, Malaga, Spain
| | - Javier Pascual
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Andalusia-Roche Network in Precision Medical Oncology, 41092, Sevilla, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), 28029, Madrid, Spain
| | - Iñaki Comino-Méndez
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain.
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain.
- Andalusia-Roche Network in Precision Medical Oncology, 41092, Sevilla, Spain.
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), 28029, Madrid, Spain.
| | - Emilio Alba
- Unidad de Gestion Clinica Intercentros de Oncologia Medica, Hospitales Universitarios Regional y Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga (IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Andalusia-Roche Network in Precision Medical Oncology, 41092, Sevilla, Spain
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC - CB16/12/00481), 28029, Madrid, Spain
- University of Málaga, Faculty of Medicine, 29010, Malaga, Spain
| |
Collapse
|
20
|
Torresan S, de Scordilli M, Bortolot M, Di Nardo P, Foltran L, Fumagalli A, Guardascione M, Ongaro E, Puglisi F. Liquid biopsy in colorectal cancer: Onward and upward. Crit Rev Oncol Hematol 2024; 194:104242. [PMID: 38128627 DOI: 10.1016/j.critrevonc.2023.104242] [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: 10/05/2023] [Revised: 11/20/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths worldwide. In recent years, liquid biopsy has emerged as one of the most interesting areas of research in oncology, leading to innovative trials and practical changes in all aspects of CRC management. RNAs and cell free DNA (cfDNA) methylation are emerging as promising biomarkers for early diagnosis. Post-surgical circulating tumour DNA (ctDNA) can aid in evaluating minimal residual disease and personalising adjuvant treatment. In rectal cancer, ctDNA could improve response assessment to neoadjuvant therapy and risk stratification, especially in the era of organ-preservation trials. In the advanced setting, ctDNA analysis offers the opportunity to monitor treatment response and identify driver and resistance mutations more comprehensively than traditional tissue analysis, providing prognostic and predictive information. The aim of this review is to provide a detailed overview of the clinical applications and future perspectives of liquid biopsy in CRC.
Collapse
Affiliation(s)
- Sara Torresan
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Marco de Scordilli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; Department of Medicine, University of Udine, 33100 Udine, Italy.
| | - Martina Bortolot
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Paola Di Nardo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Luisa Foltran
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Arianna Fumagalli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Michela Guardascione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elena Ongaro
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Fabio Puglisi
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
21
|
Fan W, Xia Z, Chen R, Lin D, Li F, Zheng Y, Luo J, Xiong Y, Yu P, Gao W, Gong Y, Zhang F, Zhang S, Li L. Circulating tumor DNA analysis predicts recurrence and avoids unnecessary adjuvant chemotherapy in I-IV colorectal cancer. Ther Adv Med Oncol 2024; 16:17588359231220607. [PMID: 38282662 PMCID: PMC10822076 DOI: 10.1177/17588359231220607] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Background Circulating tumor DNA (ctDNA) has emerged as a biomarker that can define the risk of recurrence after curative-intent surgery for patients with colorectal cancer (CRC). However, beyond the predictive power of postoperative ctDNA detection, the efficacy and potential limitations of ctDNA detection urgently need to be fully elucidated in a large cohort of CRC. Objectives To define potentially cured CRC patients through ctDNA monitoring following surgery. Design A prospective, multicenter, observational study. Methods We enrolled 309 patients with stages I-IV CRC who underwent definitive surgery. Tumor tissues were sequenced by a custom-designed next-generation sequencing panel to identify somatic mutations. Plasma was analyzed using a ctDNA-based molecular residual disease (MRD) assay which integrated tumor-genotype-informed and tumor-genotype-naïve ctDNA analysis. The turnaround time of the assay was 10-14 days. Results Postoperative ctDNA was detected in 5.4%, 13.8%, 15%, and 30% of patients with stage I, II, III, and IV disease, respectively, and in 17.5% of all longitudinal samples. Patients with positive postsurgery MRD had a higher recurrence rate than those with negative postsurgery MRD [hazard ratio (HR), 13.17; p < 0.0001], producing a sensitivity of 64.6%, a specificity of 94.8%, a positive predictive value (PPV) of 75.6%, and a negative predictive value (NPV) of 91.5%. Furthermore, patients with positive longitudinal MRD also had a significantly higher recurrence rate (HR, 14.44; p < 0.0001), with increased sensitivity (75.0%), specificity (94.9%), PPV (79.6%), and NPV (93.4%). Subgroup analyses revealed that adjuvant therapy did not confer superior survival for patients with undetectable or detectable MRD. In addition, MRD detection was less effective in identifying lung-only and peritoneal metastases. Conclusion Postoperative ctDNA status is a strong predictor of recurrence independent of stage and microsatellite instability status. Longitudinal undetectable MRD could be used to define the potentially cured population in CRC patients undergoing curative-intent surgery.
Collapse
Affiliation(s)
- Wenhua Fan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhiyuan Xia
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | | | - Dagui Lin
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Fang Li
- Geneplus-Beijing, Beijing, China
| | - Yang Zheng
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jiongyong Luo
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | | | | | - Wei Gao
- Geneplus-Beijing, Beijing, China
| | | | - Feiran Zhang
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Jinping District, Shantou, Guangdong 515041, China
| | - Sen Zhang
- Department of Colorectal & Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning 530021, China
| | - Liren Li
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, China
| |
Collapse
|
22
|
Min L, Chen J, Yu M, Yang K, Liu D. Utilizing circulating tumour DNA as a prognostic predictor of gastric cancer: a meta-analysis. Biomarkers 2023; 28:427-436. [PMID: 37036017 DOI: 10.1080/1354750x.2023.2201664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/05/2023] [Indexed: 04/11/2023]
Abstract
Background: Circulating tumour DNA (ctDNA) has demonstrated robust diagnostic accuracy in several digestive cancers. However, the prognostic role of ctCDNA in gastric cancer (GC) is still controversial. This systematic review and meta-analysis aimed to evaluate the prognostic value of ctDNA in GC.Methods: PubMed, Web of Science and Cochrane databases were searched to identify studies reporting the use of ctDNA to predict GC outcome and all relevant studies published until November 2022 were enrolled for our analysis. Data were extracted by two authors independently and statistic analysis was conducted by R program with 'meta' and 'metafor' packages.Results: A total of 34 qualified articles with 5091 subjects were incorporated into our meta-analysis. The corresponding Hazard ratio (HR) of overall survival (OS), disease-free survival (DFS) and progression-free survival (PFS) were 2.74 (95% CI:2.24-3.35), 3.13 (95% CI:2.08-4.72) and 3.04 (95% CI:2.46-3.76), respectively, in GC patients.Conclusion: Blood-based ctDNA assay would be a potential novel biomarker for GC evaluation and prediction.Simple Summary: This is the integrated meta-analysis on the association of circulating tumour DNA (ctDNA) and prognosis of gastric cancer (GC) with an increasing number of studies exploring the prognostic value of GC in the last few years, which depicted that the detection of ctDNA could be a promising predictor in GC patients.
Collapse
Affiliation(s)
- Liang Min
- Department of Gastroenterology, the Second Xiangya Hospital, Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| | - Jinghua Chen
- Department of Oncology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Meihong Yu
- Department of Gastroenterology, the Second Xiangya Hospital, Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| | - Ke Yang
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Deliang Liu
- Department of Gastroenterology, the Second Xiangya Hospital, Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| |
Collapse
|
23
|
Yao S, Han Y, Yang M, Jin K, Lan H. Integration of liquid biopsy and immunotherapy: opening a new era in colorectal cancer treatment. Front Immunol 2023; 14:1292861. [PMID: 38077354 PMCID: PMC10702507 DOI: 10.3389/fimmu.2023.1292861] [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: 09/12/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Immunotherapy has revolutionized the conventional treatment approaches for colorectal cancer (CRC), offering new therapeutic prospects for patients. Liquid biopsy has shown significant potential in early screening, diagnosis, and postoperative monitoring by analyzing circulating tumor cells (CTC) and circulating tumor DNA (ctDNA). In the era of immunotherapy, liquid biopsy provides additional possibilities for guiding immune-based treatments. Emerging technologies such as mass spectrometry-based detection of neoantigens and flow cytometry-based T cell sorting offer new tools for liquid biopsy, aiming to optimize immune therapy strategies. The integration of liquid biopsy with immunotherapy holds promise for improving treatment outcomes in colorectal cancer patients, enabling breakthroughs in early diagnosis and treatment, and providing patients with more personalized, precise, and effective treatment strategies.
Collapse
Affiliation(s)
- Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yuejun Han
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Mengxiang Yang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Wullaert L, van Rees JM, Martens JWM, Verheul HMW, Grünhagen DJ, Wilting SM, Verhoef C. Circulating Tumour DNA as Biomarker for Colorectal Liver Metastases: A Systematic Review and Meta-Analysis. Cells 2023; 12:2520. [PMID: 37947598 PMCID: PMC10647834 DOI: 10.3390/cells12212520] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Circulating tumour DNA (ctDNA) is a potential biomarker that could contribute to more judicious patient selection for personalised treatment. This review and meta-analysis gives an overview of the current knowledge in the literature investigating the value of ctDNA in patients with colorectal liver metastases (CRLM). A systematic search was conducted in electronic databases for studies published prior to the 26th of May 2023. Studies investigating the association between ctDNA and oncological outcomes in patients undergoing curative-intent local therapy for CRLM were included. Meta-analyses were performed to pool hazard ratios (HR) for the recurrence-free survival (RFS) and overall survival (OS). A total of eleven studies were included and nine were eligible for meta-analyses. Patients with detectable ctDNA after surgery experienced a significantly higher chance of recurrence (HR 3.12, 95% CI 2.27-4.28, p < 0.000010) and shorter OS (HR 5.04, 95% CI 2.53-10.04, p < 0.00001) compared to patients without detectable ctDNA. A similar association for recurrence was found in patients with detectable ctDNA after the completion of adjuvant therapy (HR 6.39, 95% CI 2.13-19.17, p < 0.0009). The meta-analyses revealed no association between detectable ctDNA before surgery and the RFS and OS. These meta-analyses demonstrate the strong association between detectable ctDNA after treatment and oncological outcomes in CRLM patients.
Collapse
Affiliation(s)
- Lissa Wullaert
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (L.W.)
| | - Jan M. van Rees
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (L.W.)
| | - John W. M. Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Henk M. W. Verheul
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Dirk J. Grünhagen
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (L.W.)
| | - Saskia M. Wilting
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (L.W.)
| |
Collapse
|
25
|
Aalami AH, Shahriari A, Mazaheri M, Aalami F, Amirabadi A, Sahebkar A. Diagnostic accuracy of tumor M2-pyruvate kinase (tM2-PK) as a non-invasive biomarker in colorectal cancer: A systematic review and meta-analysis. Clin Biochem 2023; 120:110652. [PMID: 37757965 DOI: 10.1016/j.clinbiochem.2023.110652] [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: 06/16/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
INTRODUCTION The tumor pyruvate kinase M2 isoform (tM2-PK) is a glycolytic enzyme isoform that is present on the surface of rapidly proliferating cancer cells. The objective of this investigation was to assess the efficacy of the tM2-PK measurement assay in detecting colorectal cancer (CRC) through the analysis of serum/plasma and stool samples obtained from patients. METHODS The pooled diagnostic performance measures, including sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), the area under the curve (AUC), Q*index, and summary receiver-operating characteristic curve (SROC), were computed using the Meta-Disc V.1.4 and Comprehensive Meta-Analysis V.3.3 software. The statistical methods of I2 and chi-square were employed to assess the presence of heterogeneity. The estimation of publication bias was conducted through the implementation of Begg's rank correlation and Egger's regression asymmetry tests. RESULTS A total of 28 studies were found, involving 2900 participants (1560 cases and 1340 controls). The diagnostic accuracy of tM2-PK was calculated in CRC based on the pooled sensitivity of 83.70% (95% CI: 82.0% - 85.30%), specificity of 74.0% (95% CI: 72.0% - 76.0%), PLR of 4.432 (95% CI: 3.33 - 5.60), NLR of 0.187 (95% CI: 0.144 - 0.243), DOR of 30.182 (95% CI: 19.761 - 46.10) as well as AUC at 91.6%, and Q*-index at 85.0%. Publication bias was seen based on Begg's (p = 0.0006) and Egger's (p = 0.00015) tests. CONCLUSION The results demonstrate that tM2-PK exhibits promise as a fair marker for CTRC, with the potential to serve as a non-invasive biomarker.
Collapse
Affiliation(s)
- Amir Hossein Aalami
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Nephrology, University of Utah, Salt Lake City, UT, USA.
| | - Ali Shahriari
- Department of Internal Medicine, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Mohammad Mazaheri
- Department of Molecular, Cell and Systems Biology, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA, USA
| | - Farnoosh Aalami
- Student Research Committee, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Amir Amirabadi
- Department of Internal Medicine, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
26
|
Lim DH, Yoon H, Kim KP, Ryoo BY, Lee SS, Park DH, Song TJ, Hwang DW, Lee JH, Song KB, Kim SC, Hong SM, Hyung J, Yoo C. Analysis of Plasma Circulating Tumor DNA in Borderline Resectable Pancreatic Cancer Treated with Neoadjuvant Modified FOLFIRINOX: Clinical Relevance of DNA Damage Repair Gene Alteration Detection. Cancer Res Treat 2023; 55:1313-1320. [PMID: 37139665 PMCID: PMC10582539 DOI: 10.4143/crt.2023.452] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/04/2023] [Indexed: 05/05/2023] Open
Abstract
PURPOSE There are no reliable biomarkers to guide treatment for patients with borderline resectable pancreatic cancer (BRPC) in the neoadjuvant setting. We used plasma circulating tumor DNA (ctDNA) sequencing to search biomarkers for patients with BRPC receiving neoadjuvant mFOLFIRINOX in our phase 2 clinical trial (NCT02749136). MATERIALS AND METHODS Among the 44 patients enrolled in the trial, patients with plasma ctDNA sequencing at baseline or post-operation were included in this analysis. Plasma cell-free DNA isolation and sequencing were performed using the Guardant 360 assay. Detection of genomic alterations, including DNA damage repair (DDR) genes, were examined for correlations with survival. RESULTS Among the 44 patients, 28 patients had ctDNA sequencing data qualified for the analysis and were included in this study. Among the 25 patients with baseline plasma ctDNA data, 10 patients (40%) had alterations of DDR genes detected at baseline, inclu-ding ATM, BRCA1, BRCA2 and MLH1, and showed significantly better progression-free survival than those without such DDR gene alterations detected (median, 26.6 vs. 13.5 months; log-rank p=0.004). Patients with somatic KRAS mutations detected at baseline (n=6) had significantly worse overall survival (median, 8.5 months vs. not applicable; log-rank p=0.003) than those without. Among 13 patients with post-operative plasma ctDNA data, eight patients (61.5%) had detectable somatic alterations. CONCLUSION Detection of DDR gene mutations from plasma ctDNA at baseline was associated with better survival outcomes of pati-ents with borderline resectable pancreatic ductal adenocarcinoma treated with neoadjuvant mFOLFIRINOX and may be a prognostic biomarker.
Collapse
Affiliation(s)
- Dong-Hoon Lim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Hyunseok Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Kyu-pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Sang Soo Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Do Hyun Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Tae Jun Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Dae Wook Hwang
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Jae Hoon Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Ki Byung Song
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Song Cheol Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Jaewon Hyung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul,
Korea
| |
Collapse
|
27
|
Loft M, To YH, Gibbs P, Tie J. Clinical application of circulating tumour DNA in colorectal cancer. Lancet Gastroenterol Hepatol 2023; 8:837-852. [PMID: 37499673 DOI: 10.1016/s2468-1253(23)00146-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 07/29/2023]
Abstract
Liquid biopsies that detect circulating tumour DNA (ctDNA) have the potential to revolutionise the personalised management of colorectal cancer. For patients with early-stage disease, emerging clinical applications include the assessment of molecular residual disease after surgery, the monitoring of adjuvant chemotherapy efficacy, and early detection of recurrence during surveillance. In the advanced disease setting, data highlight the potential of ctDNA levels as a prognostic marker and as an early indicator of treatment response. ctDNA assessment can complement standard tissue-based testing for molecular characterisation, with the added ability to monitor emerging mutations under the selective pressure of targeted therapy. Here we provide an overview of the evidence supporting the use of ctDNA in colorectal cancer, the studies underway to address some of the outstanding questions, and the barriers to widespread clinical uptake.
Collapse
Affiliation(s)
- Matthew Loft
- Division of Personalised Oncology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Department of Medical Oncology, Western Health, Footscray, VIC, Australia
| | - Yat Hang To
- Division of Personalised Oncology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Peter Gibbs
- Division of Personalised Oncology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Department of Medical Oncology, Western Health, Footscray, VIC, Australia
| | - Jeanne Tie
- Division of Personalised Oncology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
28
|
Kan CM, Pei XM, Yeung MHY, Jin N, Ng SSM, Tsang HF, Cho WCS, Yim AKY, Yu ACS, Wong SCC. Exploring the Role of Circulating Cell-Free RNA in the Development of Colorectal Cancer. Int J Mol Sci 2023; 24:11026. [PMID: 37446204 PMCID: PMC10341751 DOI: 10.3390/ijms241311026] [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: 05/21/2023] [Revised: 06/25/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023] Open
Abstract
Circulating tumor RNA (ctRNA) has recently emerged as a novel and attractive liquid biomarker. CtRNA is capable of providing important information about the expression of a variety of target genes noninvasively, without the need for biopsies, through the use of circulating RNA sequencing. The overexpression of cancer-specific transcripts increases the tumor-derived RNA signal, which overcomes limitations due to low quantities of circulating tumor DNA (ctDNA). The purpose of this work is to present an up-to-date review of current knowledge regarding ctRNAs and their status as biomarkers to address the diagnosis, prognosis, prediction, and drug resistance of colorectal cancer. The final section of the article discusses the practical aspects involved in analyzing plasma ctRNA, including storage and isolation, detection technologies, and their limitations in clinical applications.
Collapse
Affiliation(s)
- Chau-Ming Kan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; (C.-M.K.); (H.F.T.)
| | - Xiao Meng Pei
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China; (X.M.P.); (M.H.Y.Y.)
| | - Martin Ho Yin Yeung
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China; (X.M.P.); (M.H.Y.Y.)
| | - Nana Jin
- Codex Genetics Limited, Shatin, Hong Kong SAR, China; (N.J.); (A.K.-Y.Y.); (A.C.-S.Y.)
| | - Simon Siu Man Ng
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Hin Fung Tsang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; (C.-M.K.); (H.F.T.)
| | - William Chi Shing Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China;
| | - Aldrin Kay-Yuen Yim
- Codex Genetics Limited, Shatin, Hong Kong SAR, China; (N.J.); (A.K.-Y.Y.); (A.C.-S.Y.)
| | - Allen Chi-Shing Yu
- Codex Genetics Limited, Shatin, Hong Kong SAR, China; (N.J.); (A.K.-Y.Y.); (A.C.-S.Y.)
| | - Sze Chuen Cesar Wong
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China; (X.M.P.); (M.H.Y.Y.)
| |
Collapse
|
29
|
Spindler KLG, Jakobsen A. Circulating tumor DNA: Response Evaluation Criteria in Solid Tumors - can we RECIST? Focus on colorectal cancer. Ther Adv Med Oncol 2023; 15:17588359231171580. [PMID: 37152423 PMCID: PMC10154995 DOI: 10.1177/17588359231171580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/05/2023] [Indexed: 05/09/2023] Open
Abstract
Interest in the measurement of circulating tumor DNA (ctDNA) in colorectal cancer (CRC) has increased during the past decade. The analysis of quantitative ctDNA changes as a general response evaluation criterion during systemic treatment is a scientific approach with high clinical potential, and results can be transferred to a pan-cancer concept if relevantly investigated. The purpose of this overview is to discuss the current evidence for ctDNA as a marker of response in metastatic CRC (mCRC) and to propose criteria for definitions of response to systemic therapies applicable in prospective clinical trials. We discuss the literature, which supports a new definition of ctDNA Response Evaluation Criteria in Solid Tumors. Finally, we discuss the challenges in preparations of the optimal trial design to establish the true clinical utility of ctDNA.
Collapse
Affiliation(s)
- Karen-Lise Garm Spindler
- Department of Oncology, Aarhus University
Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus
DK-8200, Denmark
| | - Anders Jakobsen
- Department of Oncology, Institute of Regional
Health Services, University of Southern Denmark, Vejle University Hospital,
Vejle, Denmark
| |
Collapse
|
30
|
Spindler KLG, Callesen LB, Arnold D. Time for a lead-time definition? Author response to 'Why the length of recurrence free survival or "lead-times" can be misleading. Comment on: Callesen LB, Takacova T, Hamfjord J, et al. Circulating DNA in patients undergoing loco-regional treatment of colorectal cancer metastases: a systematic review and meta-analysis'. Ther Adv Med Oncol 2023; 15:17588359231156393. [PMID: 36950273 PMCID: PMC10026177 DOI: 10.1177/17588359231156393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
Affiliation(s)
| | - Louise Bach Callesen
- Department of Oncology, Aarhus University
Hospital, Denmark and Department of Clinical Medicine, Aarhus University,
Denmark
| | - Dirk Arnold
- Department of Oncology and Hematology,
Asklepios Tumourzentrum Hamburg, AK Altona, Hamburg, Germany
| |
Collapse
|