1
|
Montalti R, Pepe F, Cassese G, Russo G, Malapelle U, Carlomagno C, Giglio MC, Falco G, Alagia M, De Simone G, Geboes K, Troncone G, Troisi RI, Rompianesi G. Prognostic role of postoperative persistence of ctDNA molecular signature after liver resection for colorectal liver metastases: Preliminary results from a prospective study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109706. [PMID: 40009912 DOI: 10.1016/j.ejso.2025.109706] [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: 06/06/2024] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
INTRODUCTION Liver resection represents the cornerstone treatment for Colo-Rectal Liver Metastases (CRLM), but it is still followed by a high recurrence rate. The identification of a valid and reproducible predictor of recurrence can play a key role in correct and timely management of the disease. In this scenario, liquid biopsy may represent an integrative, less invasive, and easily manageable tool to clinically stratify colorectal cancer patients.We aimed to investigate the prognostic role of persistent circulating tumor DNA (ctDNA) signature on CRLM recurrence after liver resection. METHODS A series of 51 consecutive patients undergoing liver resection for CRLM were prospectively enrolled between January 2020 and March 2023. Patients underwent liquid biopsy from peripheral blood samples before and after surgery. Other risk factors for disease recurrence were also investigated. RESULTS Twenty-nine patients were found to be positive for one of the clinically relevant molecular alterations detected by NGS, at preoperative (T0) ctDNA analysis. At univariate analysis, total tumor size bigger than 60 mm (p = 0.046, HR 2.486) and postoperative persistence of ctDNA molecular signature (p = 0.024, HR 2.831) were significantly associated with recurrence. Multivariable Cox Regression analysis showed that only postoperative persistence of ctDNA molecular signature was associated with recurrence (p = 0.027, HR 2.766). Similarly, 1-3 yr DFS was significantly lower in the subgroup with persistence of molecular signature at liquid biopsy (100-49.5 % vs 57.1-21.4 %, p = 0.018). CONCLUSION Postoperative persistence of ctDNA molecular signature could represent a useful tool to predict recurrence after liver resection for CRLM patients.
Collapse
Affiliation(s)
- Roberto Montalti
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy; Department of Public Health, Federico II University, 80131, Naples, Italy.
| | - Francesco Pepe
- Department of Public Health, Federico II University, 80131, Naples, Italy
| | - Gianluca Cassese
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, Federico II University, 80131, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, Federico II University, 80131, Naples, Italy
| | - Chiara Carlomagno
- Department of Clinical Medicine and Surgery, Division of Oncology, University Federico II, Naples, Italy
| | - Mariano Cesare Giglio
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Giacinto Falco
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Mariantonietta Alagia
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Giuseppe De Simone
- Department of Anesthesiology, Federico II University Hospital, Naples, Italy
| | - Karen Geboes
- Oncology Centrum, Ghent University Hospital, Belgium
| | - Giancarlo Troncone
- Department of Public Health, Federico II University, 80131, Naples, Italy
| | - Roberto Ivan Troisi
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Gianluca Rompianesi
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| |
Collapse
|
2
|
Raunkilde L, Andersen RF, Thomsen CB, Hansen TF, Jensen LH. A prospective study of methylated ctDNA in patients undergoing treatment for liver metastases from colorectal cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109586. [PMID: 39847896 DOI: 10.1016/j.ejso.2025.109586] [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: 06/21/2024] [Revised: 12/12/2024] [Accepted: 01/06/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Decision regarding local treatment of colorectal liver metastases (CRLM) is a multidisciplinary assessment, and liver intervention should be performed when the metastases are deemed resectable. There is no standard biomarker to aid neither this decision nor the postoperative treatment decisions. The present prospective, observational study aimed to investigate the potential clinical utility of a combined tumor-specific and organ-specific methylated circulating DNA assay in the perioperative setting of CRLM. MATERIAL AND METHODS The study included 56 cases with CRLM. Blood samples were drawn preoperatively and postoperatively. Multiplex methylation analysis of the markers NPY, KANK1, and GAL3ST3 (meth-ctDNA) was performed using droplet digital PCR. RESULTS The assay detected preoperative and postoperative meth-ctDNA in 37 % and 46 % of patients, respectively. Patients with negative preoperative meth-ctDNA had a longer median PFS compared to those with positive preoperative meth-ctDNA (HR = 2.2, 95 % CI 1.2-3.9, p < 0.01). In a multivariate analysis, preoperative negative meth-ctDNA was identified as a strong independent predictor of PFS (HR = 3.3, 95 % CI 1.5-7.2, p < 0.01). Similarly, patients with negative postoperative meth-ctDNA had longer median PFS (HR = 3.0, 95 % CI = 1.6-5.6, p < 0.001) and OS (HR = 4.1, 95 % CI 1.9-9.1, p < 0.001) compared to those with positive postoperative meth-ctDNA. CONCLUSION Preoperative meth-ctDNA may serve as an important biomarker to inform the multidisciplinary assessment and treatment planning of CRLM. Negative meth-ctDNA may indicate the optimal timing for liver intervention, whereas positive meth-ctDNA may indicate initiation or re-orientation of chemotherapy, or immediate local intervention. Our results confirm postoperative negative meth-ctDNA as a strong prognostic marker of survival.
Collapse
Affiliation(s)
- Louise Raunkilde
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark; Department of Regional Health Research, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.
| | - Rikke Fredslund Andersen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark; Department of Clinical Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark
| | - Caroline Brenner Thomsen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark; Department of Regional Health Research, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Lars Henrik Jensen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark; Department of Regional Health Research, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| |
Collapse
|
3
|
Kawashima M, Yamada T, Miyasaka T, Kanaka S, Kuriyama S, Uehara K, Matsuda A, Ohta R, Sonoda H, Taniai N, Yoshida H. Impact of Minimal Residual Disease on Early Recurrence of Liver Metastatic Colorectal Cancer. Cancer Sci 2025; 116:1366-1374. [PMID: 40059633 PMCID: PMC12044645 DOI: 10.1111/cas.16442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 05/02/2025] Open
Abstract
For patients with resectable colorectal liver metastases (CRLM), the efficacy of adjuvant chemotherapy remains a subject of debate. Several studies have concluded that postoperative circulating tumor DNA (ctDNA) is a marker of minimal residual disease (MRD) and is a useful prognostic factor in patients with nonmetastatic colorectal cancer. However, few studies have explored its application in cases involving metastases. This was an observational study that included CRLM patients who underwent primary and liver tumor resection. By examining targeted sequencing of 50 genes commonly mutated in CRC, we identified at least one somatic mutation in each patient's metastatic liver tumor. Blood samples were obtained before and 1-month after surgery. Fifty-three patients were included, and recurrence was diagnosed in 39 patients. Of those, 13 patients experienced early relapse. ctDNA was detected in 45 patients before surgery and 11 after. All MRD-positive patients experienced recurrence. Among them, nine had early recurrence. MRD-positive patients had poorer recurrence free survival (RFS, p < 0.0001) and overall survival (OS, p < 0.0005). Nine of 13 patients with early recurrence had MRD; however, two of 40 patients without early recurrence also had MRD (p < 0.0001). Among 42 MRD-negative patients, adjuvant chemotherapy had no impact of RFS (p = 0.84) or OS (p = 0.54). MRD proved valuable in predicting the risk of postoperative recurrence in patients with CRLM, particularly because MRD positivity emerged as a significant risk factor for early recurrence. Furthermore, it appears that adjuvant chemotherapy may not effectively improve the prognosis for MRD-negative patients.
Collapse
Affiliation(s)
- Mampei Kawashima
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| | - Takeshi Yamada
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| | | | - Shintaro Kanaka
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| | - Sho Kuriyama
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| | - Kay Uehara
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| | - Akihisa Matsuda
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| | - Ryo Ohta
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| | - Hiromichi Sonoda
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| | - Nobuhiko Taniai
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| | - Hiroshi Yoshida
- Department of Gastroenterological SurgeryNippon Medical SchoolTokyoJapan
| |
Collapse
|
4
|
Zhuang L, Park SH, Skates SJ, Prosper AE, Aberle DR, Hsu W. Advancing Precision Oncology Through Modeling of Longitudinal and Multimodal Data. ARXIV 2025:arXiv:2502.07836v2. [PMID: 39990791 PMCID: PMC11844620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cancer evolves continuously over time through a complex interplay of genetic, epigenetic, microenvironmental, and phenotypic changes. This dynamic behavior drives uncontrolled cell growth, metastasis, immune evasion, and therapy resistance, posing challenges for effective monitoring and treatment. However, today's data-driven research in oncology has primarily focused on cross-sectional analysis using data from a single modality, limiting the ability to fully characterize and interpret the disease's dynamic heterogeneity. Advances in multiscale data collection and computational methods now enable the discovery of longitudinal multimodal biomarkers for precision oncology. Longitudinal data reveal patterns of disease progression and treatment response that are not evident from single-timepoint data, enabling timely abnormality detection and dynamic treatment adaptation. Multimodal data integration offers complementary information from diverse sources for more precise risk assessment and targeting of cancer therapy. In this review, we survey methods of longitudinal and multimodal modeling, highlighting their synergy in providing multifaceted insights for personalized care tailored to the unique characteristics of a patient's cancer. We summarize the current challenges and future directions of longitudinal multimodal analysis in advancing precision oncology.
Collapse
Affiliation(s)
- Luoting Zhuang
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| | - Stephen H Park
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| | - Steven J Skates
- Harvard Medical School, Boston, MA 02115 USA, and also with Biostatistics Center, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Ashley E Prosper
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| | - Denise R Aberle
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| | - William Hsu
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| |
Collapse
|
5
|
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
|
6
|
Wehrle CJ, Tocci NX, Sun K, Jiao C, Hong H, Gross A, Allkushi E, Uysal M, Linganna MW, Stackhouse K, Hashimoto K, Schlegel A, Walsh RM, Miller C, Kwon DCH, Aucejo F. Utility of circulating tumor DNA in secondary liver malignancies: What we know and what is to come. J Surg Oncol 2025; 131:888-894. [PMID: 39155652 PMCID: PMC12120384 DOI: 10.1002/jso.27838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 07/27/2024] [Indexed: 08/20/2024]
Abstract
Secondary liver malignancies are a serious and challenging global health concern. Secondary metastasis to the liver is most commonly from colorectal cancer that has metastatically spread through splanchnic circulation. Metastatic diseases can portend poor prognosis due to the progressive nature typically found on detection. Improvements in detection of disease, monitoring therapy response, and monitoring for recurrence are crucial to the improvement in the management of secondary liver malignancies. Assessment of ctDNA in these patient populations poses an opportunity to impact the management of secondary liver malignancies. In this review, we aim to discuss ctDNA, the current literature, and future directions of this technology within secondary liver malignancies.
Collapse
Affiliation(s)
- Chase J. Wehrle
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Noah X. Tocci
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Keyue Sun
- Cleveland Clinic Foundation, Lerner Research Institute, Inflammation & ImmunityClevelandOhioUSA
| | - Chunbao Jiao
- Cleveland Clinic Foundation, Lerner Research Institute, Inflammation & ImmunityClevelandOhioUSA
| | - Hanna Hong
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Abby Gross
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Erlind Allkushi
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Melis Uysal
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Maureen Whitsett Linganna
- Department of Gastroenterology, Hepatology, and NutritionCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Katheryn Stackhouse
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Koji Hashimoto
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Andrea Schlegel
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
- Cleveland Clinic Foundation, Lerner Research Institute, Inflammation & ImmunityClevelandOhioUSA
| | - R. Matthew Walsh
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Charles Miller
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - David C. H. Kwon
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| | - Federico Aucejo
- Department of Hepato‐Pancreato‐Biliary & Liver Transplant SurgeryCleveland Clinic Foundation, Digestive Diseases and Surgery InstituteClevelandOhioUSA
| |
Collapse
|
7
|
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
|
8
|
Donica WRF, Shindorf ML, Philips P, Scoggins CR, Egger ME, Hayat TM, Martin RCG. Preoperative liquid biopsy for optimal patient selection in metastatic colorectal cancer. Surgery 2025; 179:108810. [PMID: 39307674 DOI: 10.1016/j.surg.2024.06.060] [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: 05/07/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 02/02/2025]
Abstract
OBJECTIVE In this pilot study, we sought to determine if preoperative circulating tumor DNA could be a useful predictor to avoid futile metastasectomy, predict early postoperative recurrence, and determine optimal chemotherapy duration during the management of patients with resectable metastatic colorectal cancer. METHODS Patients from 2021 to 2023 were enrolled prospectively and evaluated with circulating tumor DNA preoperatively and postoperatively for detection of recurrence. Clinicopathologic and treatment factors as well as disease-free survival were compared between those with undetectable versus detectable preoperative circulating tumor DNA. RESULTS Twenty-eight patients were evaluated, with a median follow-up time of 24 months. The median preoperative circulating tumor DNA level was 0.16 MTM/mL [0.00, 2.30]. Of the 10 patients (40%) with a preoperative circulating tumor DNA level of zero, 5 patients (50%) recurred between 4 and 18 months postoperatively. Among the 18 patients whose disease recurred, 10 patients (56%) had circulating tumor DNA detected postoperatively. Median change between preoperative and postoperative circulating tumor DNA levels was 0.00 [-0.02, 0.05] in those who did not recur and 0.00 [-7.04, 0.00] in those who recurred. When disease-free survival was evaluated by detectable versus undetectable preoperative circulating tumor DNA levels, there was no difference in disease-free survival estimates (P value = .11). On univariate Cox proportional hazards analysis, the preoperative circulating tumor DNA level, change between preoperative and postoperative circulating tumor DNA levels, and postoperative circulating tumor DNA levels did not influence disease-free survival. However, those with detectable postoperative circulating tumor DNA were 3.96 (95% confidence interval 1.30-12.06) times as likely to recur compared to those with undetectable postoperative circulating tumor DNA. CONCLUSION New technologies including use of circulating tumor DNA may help better predict which patients with colorectal liver metastases will undergo futile surgery. Our preliminary findings suggest that postoperative, and not preoperative, circulating tumor DNA is predictive of recurrence following metastasectomy. Use of circulating tumor DNA in guiding operative management should be done in conjunction with high-quality imaging and other serologic markers to determine which patients with colorectal liver metastases are likely to receive durable benefit from operative intervention.
Collapse
Affiliation(s)
- Walter R F Donica
- The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville, Louisville, KY
| | - Machenzie L Shindorf
- The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville, Louisville, KY
| | - Prejesh Philips
- The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville, Louisville, KY
| | - Charles R Scoggins
- The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville, Louisville, KY
| | - Michael E Egger
- The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville, Louisville, KY
| | - Traci M Hayat
- The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville, Louisville, KY
| | - Robert C G Martin
- The Hiram C. Polk Jr., MD Department of Surgery, University of Louisville, Louisville, KY.
| |
Collapse
|
9
|
Zhang ZN, Hao L, Han S, Li SS, Lin SX, Miao YD. Harnessing the prognostic power of preoperative systemic immune-inflammation index/albumin ratio in hepatocellular carcinoma resection. World J Gastrointest Surg 2025; 17:102261. [DOI: 10.4240/wjgs.v17.i2.102261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
The recent study by Chen et al, published in the World Journal of Gastroenterology, introduces a groundbreaking assessment tool-the preoperative systemic immune-inflammation index/albumin (SII/ALB) ratio-for patients with hepatocellular carcinoma (HCC) undergoing curative resection. This study not only establishes the independent prognostic significance of the SII/ALB ratio but also incorporates it into a predictive nomogram, enhancing its utility for clinical decision-making. The SII/ALB ratio, by integrating inflammatory and nutritional biomarkers, offers a novel lens through which the prognosis of HCC patients can be viewed, suggesting a more tailored approach to patient management. The development of the nomogram, validated for its accuracy in predicting patient outcomes, marks a pivotal advance, potentially guiding surgical decisions and postoperative care. However, the study's focus on a single-center cohort prompts the need for validation in a broader, more diverse patient population to ensure its applicability across various clinical settings. Moreover, longitudinal studies could elucidate the dynamic changes in SII/ALB post-surgery, offering insights into its potential as a continuous monitor for recurrence and long-term survival. This abstract aim to underscore the critical findings of Chen et al's study while calling for further research to explore the full potential of the SII/ALB ratio in the global management of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhao-Nan Zhang
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Liang Hao
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Shuang Han
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Shan-Shan Li
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Si-Xiang Lin
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Yan-Dong Miao
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| |
Collapse
|
10
|
Adam R. Transplantation for hepatic metastases of colorectal cancer: Toward a change of concept and a change of practice. J Visc Surg 2025:S1878-7886(25)00033-5. [PMID: 39984395 DOI: 10.1016/j.jviscsurg.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Affiliation(s)
- René Adam
- Centre Hépato-Biliaire, Hôpital Paul Brousse, Équipe de Recherche « Chronothérapie, Cancer et Transplantation », Université Paris-Saclay, 94800 Villejuif, France.
| |
Collapse
|
11
|
Li H, Shi M, Long X, Huang P, Peng C, He W, Li Y, Li B, Yuan Y, Qiu J, Zou R. Contrast-enhanced intraoperative ultrasound improved hepatic recurrence-free survival in initially unresectable colorectal cancer liver metastases. Dig Liver Dis 2025; 57:467-476. [PMID: 39343654 DOI: 10.1016/j.dld.2024.09.009] [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: 11/12/2023] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND We aimed to evaluate the role of Contrast-enhanced intraoperative ultrasound (CE-IOUS) with perfluorobutane microbubbles (Sonazoid) in improving the prognosis of patients with unresectable colorectal cancer liver metastases (CRLM). METHODS A total of 130 Patients with unresectable CRLM who underwent curative hepatic resection at our institute were retrospectively analyzed. Of these 130 enrolled patients, 67 underwent intraoperative ultrasound alone (IOUS group); 63 underwent additional CE-IOUS and IOUS (CE-IOUS group). Normalized inverse probability treatment weighting (IPTW) was employed to balance baseline characteristics between groups. Hepatic recurrence-free survival (HRFS) and overall survival (OS) were compared. RESULTS The treatment strategy was altered in 25 patients (25/63, 39.9%) due to the additional use of CE-IOUS. After applying IPTW, the CE-IOUS group exhibited a significantly lower rate of hepatic recurrence (hazard ratio [HR], 0.55; 95% confidence interval [CI] 0.32-0.95; P = 0.032). Subgroup analysis showed that CE-IOUS provided a significant benefit over IOUS in patients with bilobar liver metastases (P = 0.007), or with a number of live tumors < 3 (P = 0.021), or without DLM (P = 0.018), or with extrahepatic metastasis (P = 0.034), or with a minimum of 6 cycles of systemic therapy (P = 0.03). CONCLUSIONS CE-IOUS is necessary for unresectable CRLM after preoperative chemotherapy, as it enhances detection accuracy and improves the prognosis of unresectable CRLM patients.
Collapse
Affiliation(s)
- HuiFang Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Ming Shi
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Xingzhang Long
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Ultrasound, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Pinzhu Huang
- Department of Colorectal Surgery, the Sixth affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510060, PR China
| | - Chuan Peng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Ultrasound, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Wei He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Yuhong Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Binkui Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - Yunfei Yuan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | - JiLiang Qiu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China.
| | - Ruhai Zou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China; Department of Ultrasound, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China.
| |
Collapse
|
12
|
Diergaarde B, Young G, Hall DW, Mazloom A, Costa GL, Subramaniam S, Palomares MR, Garces J, Baehner FL, Schoen RE. Circulating Tumor DNA as a Marker of Recurrence Risk in Stage III Colorectal Cancer: The α-CORRECT Study. J Surg Oncol 2025. [PMID: 39865324 DOI: 10.1002/jso.27989] [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: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND AND OBJECTIVES Identification of colorectal cancer (CRC) patients at high risk of recurrence could be of substantial clinical use. We evaluated the association of ctDNA status, using a tumor-informed assay, with recurrence-free survival (RFS). METHODS Stage III CRC patients were enrolled between 2016 and 2020. Tumor tissue and serial (every 3 months for years 1-3, biannually for years 4-5) blood samples were collected. Utilizing whole-exome sequencing and selection of 50-200 variants for tumor informed assays, ctDNA status was determined using plasma cell-free DNA. RESULTS Of 137 patients enrolled, 124 with 1029 ctDNA results were included in the analyses. Median follow-up was 4.8 years. Plasma ctDNA status was strongly associated with risk of recurrence during the surveillance period (hazard ratio (HR) 49.6, 95% CI: 16.6-148.3; p < 0.0001), and at the postsurgical (HR 9.6, 95% CI: 3.2-29.5) and postdefinitive therapy timepoints (HR: 16.7, 95% CI: 6.9-40.3). The estimated 3-year RFS for ctDNA positive and ctDNA negative patients were, respectively, 54.5% and 96.1% after surgery, and 18.2% and 90.0% after definitive therapy. Multivariable analysis indicated ctDNA but not CEA was strongly prognostic for recurrence. CONCLUSIONS Our tumor-informed ctDNA assay was strongly prognostic for recurrence in patients with stage III colorectal cancer at all timepoints.
Collapse
Affiliation(s)
- Brenda Diergaarde
- Department of Human Genetics, School of Public Health, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Greg Young
- Exact Sciences Corporation, Madison, Wisconsin, USA
| | - David W Hall
- Exact Sciences Corporation, Madison, Wisconsin, USA
| | - Amin Mazloom
- Exact Sciences Corporation, Madison, Wisconsin, USA
| | - Gina L Costa
- Exact Sciences Corporation, Madison, Wisconsin, USA
| | | | | | - Jorge Garces
- Exact Sciences Corporation, Madison, Wisconsin, USA
| | | | - Robert E Schoen
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, and Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
13
|
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] [Download PDF] [Figures] [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
|
14
|
Wang DS, Pat Fong W, Wen L, Cai YY, Ren C, Wu XJ, Zhang TQ, Cao F, Zuo MX, Li BK, Zheng Y, Li LR, Chen G, Ding PR, Lu ZH, Zhang RX, Yuan YF, Pan ZZ, Li YH. Safety and efficacy of adjuvant FOLFOX/FOLFIRI with versus without hepatic arterial infusion of floxuridine in patients following colorectal cancer liver metastasectomy (HARVEST trial): A randomized controlled trial. Eur J Cancer 2025; 214:115154. [PMID: 39644535 DOI: 10.1016/j.ejca.2024.115154] [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: 10/01/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Hepatic artery infusion (HAI) chemotherapy, particularly with floxuridine (FUDR), has previously shown effectiveness in improving recurrence-free survival (RFS) in colorectal cancer (CRC) patients with colorectal liver metastases (CRLM). Nonetheless, its adjuvant use alongside modern systemic chemotherapy remains unevaluated. PATIENTS AND METHODS The HARVEST trial is an open-label, randomized, controlled study conducted from May 2018 to August 2021. CRC patients with resectable primary tumors and CRLM were recruited and randomized to receive standard systemic chemotherapy only (non-HAI group) or in combination with HAI-FUDR (HAI group). However, due to a FUDR manufacturing shortage, the study was terminated early after enrolling 92 patients. The primary endpoint was the 3-year RFS rate, with secondary endpoints including overall survival (OS), liver-specific RFS, and adverse events. RESULTS Of the 92 randomized patients, 77 were included in the modified intention-to-treat analysis. Three-year RFS rates were comparable between the HAI (N = 38) and non-HAI (N = 39) groups (31.4 % vs. 34.4 %; P = 0.28). However, improved 1-year RFS and a longer expected five-year OS were observed in the HAI group. While exploratory subgroup analysis suggested potential RFS benefits for patients with multiple liver metastases, RAS/BRAF mutations, and positive postoperative ctDNA methylation, multivariable analysis did not identify these as independent factors. Safety analysis showed comparable chemotherapy-related adverse events, except for a higher occurrence of ALT elevation in the HAI group. CONCLUSIONS While our study showed no significant difference in three-year RFS, adjuvant chemotherapy intensification with HAI-FUDR is feasible and may offer early benefits in RFS and long-term OS. Nonetheless, a larger sample size is needed for validation and identifying which patient subgroup might benefit from this regimen. TRIAL REGISTRATION ClinicalTrials.gov: NCT03500874.
Collapse
Affiliation(s)
- De-Shen Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - William Pat Fong
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lei Wen
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yan-Yu Cai
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chao Ren
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Jun Wu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tian-Qi Zhang
- Department of Minimally Invasive & Interventional Therapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fei Cao
- Department of Minimally Invasive & Interventional Therapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Meng-Xuan Zuo
- Department of Minimally Invasive & Interventional Therapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Bin-Kui Li
- Department of Hepatobiliary Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yun Zheng
- Department of Hepatobiliary Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li-Ren Li
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gong Chen
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Pei-Rong Ding
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhen-Hai Lu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rong-Xin Zhang
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yun-Fei Yuan
- Department of Hepatobiliary Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Zhi-Zhong Pan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Yu-Hong Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
15
|
Sutton TL, Patel RK, Watson KM, Gardner IH, Herzig DO, Tsikitis VL, Chen EY, Mayo SC. Liver-First Resection in Patients With Synchronous Colorectal Liver Metastases Is Associated With Inferior Recurrence-Free Survival: Reconsidering the Importance of the Primary Cancer. Dis Colon Rectum 2025; 68:32-40. [PMID: 39264063 DOI: 10.1097/dcr.0000000000003518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
BACKGROUND Synchronous colorectal liver metastases may be managed with primary-first, simultaneous, or liver-first resection. Relative oncologic outcomes based on treatment sequencing are understudied. OBJECTIVE This study aimed to assess oncologic survival outcomes in patients with synchronous colorectal liver metastases managed with each of the 3 treatment strategies, with respect to early or delayed removal of the primary tumor. DESIGN Retrospective analysis of the prospectively maintained database, with 1:1 propensity score matching of relevant clinicopathologic variables comparing liver-first to primary-first/simultaneous approaches. SETTINGS Single-institution, tertiary cancer center. PATIENTS Patients undergoing curative-intent hepatectomy for synchronous colorectal liver metastases from 2003 to 2019. MAIN OUTCOME MEASURES Overall and recurrence-free survival. RESULTS Of 151 patients, 23% (n = 35) had liver-first and 77% (n = 116; primary-first = 93 and simultaneous = 23) had primary-first/simultaneous approaches. The median follow-up was 45 months. Recurrence-free survival was worse for liver-first versus primary-first/simultaneous groups (median 12 versus 16 months, p = 0.02), driven by 3-year extrahepatic recurrence-free survival of 19%, 58%, and 50% for liver-first, primary-first, and simultaneous groups, respectively. Three-year overall survival was not significantly different at 86%, 79%, and 86%, respectively. Oncologic outcomes did not differ significantly between primary-first and simultaneous groups (all p > 0.4). Matching yielded 34 clinicopathologically similar patients per group (liver-first = 34, primary-first = 28, simultaneous = 6). The liver-first approach was associated with shorter recurrence-free survival (median 12 versus 23 months, p = 0.004), driven by extrahepatic recurrence-free survival (3 years: 20% versus 55%, p = 0.04). Overall survival was not significantly different at 3 years (79% versus 80%, p = 0.95) or 5 years (59% versus 59%, p > 0.99). LIMITATIONS This study has a retrospective design and a limited sample size. CONCLUSIONS A liver-first approach is associated with worse recurrence-free survival compared to primary-first or simultaneous resection, driven by extrahepatic recurrence. A prospective study of whether oncologic risk is associated with leaving the primary in situ is needed. Multidisciplinary treatment sequencing and enhanced postoperative surveillance for patients receiving liver-first resection are recommended. See Video Abstract . LA RESECCIN DEL HGADO PRIMERO EN PACIENTES CON METSTASIS HEPTICAS COLORRECTALES SINCRNICAS SE ASOCIA CON UNA SUPERVIVENCIA INFERIOR SIN RECURRENCIA RECONSIDERACIN DE LA IMPORTANCIA DEL CNCER PRIMARIO ANTECEDENTES:Las metástasis hepáticas colorrectales sincrónicas se pueden tratar con resección primaria, simultánea o hepática. Los resultados oncológicos relativos basados en la secuenciación del tratamiento están poco estudiados.OBJETIVO:Este estudio tuvo como objetivo evaluar los resultados de supervivencia oncológica en pacientes con metástasis hepáticas colorrectales sincrónicas tratadas con cada una de las tres estrategias de tratamiento, con respecto a la extirpación temprana o tardía del tumor primario.DISEÑO:Análisis retrospectivo de una base de datos mantenida prospectivamente, con coincidencia de propensión 1:1 de variables clínico-patológicas relevantes que comparan enfoques de hígado primero con enfoques primarios primero/simultáneos.AJUSTES:Centro oncológico terciario de una sola institución.PACIENTES:Pacientes sometidos a hepatectomía con intención curativa por metástasis hepáticas colorrectales sincrónicas entre 2003 y 2019.MEDIDAS PRINCIPALES DE RESULTADOS:Supervivencia general y libre de recurrencia.RESULTADOS:De 151 pacientes, el 23% (n=35) tuvo un abordaje hepático primero y el 77% (n=116; primario primero=93 y simultáneo=23) tuvo un abordaje primario primero/simultáneo. La mediana de seguimiento fue de 45 meses. La supervivencia libre de recurrencia fue peor para los grupos de hígado primero versus primario primero/simultáneo (mediana 12 versus 16 meses, p = 0,02), impulsada por la supervivencia libre de recurrencia extrahepática a tres años del 19%, 58% y 50% para grupos de hígado primero, primario primero y simultáneo, respectivamente. La supervivencia general a tres años no fue significativamente diferente: 86%, 79% y 86%, respectivamente. Los resultados oncológicos no difirieron significativamente entre los grupos primario-primero y simultáneo (todos p > 0,4). El emparejamiento produjo 34 pacientes clínico-patológicamente similares por grupo (hígado primero = 34, primario primero = 28/simultáneo = 6). El enfoque de hígado primero se asoció con una supervivencia libre de recurrencia más corta (mediana de 12 frente a 23 meses, p = 0,004), impulsada por la supervivencia libre de recurrencia extrahepática (3 años: 20 % frente a 55 %, p = 0,04). La supervivencia general no fue significativamente diferente a los 3 años (79% versus 80%, p = 0,95) o a los 5 años (59% versus 59%, p > 0,99).LIMITACIONES:Este estudio tiene un diseño retrospectivo y un tamaño de muestra limitado.CONCLUSIONES:Un enfoque de hígado primero se asocia con una peor supervivencia libre de recurrencia en comparación con la resección primaria o simultánea, impulsada por la recurrencia extrahepática. Es necesario un estudio prospectivo sobre si el riesgo oncológico se asocia con dejar el primario in situ . Se recomienda la secuenciación del tratamiento multidisciplinario y una vigilancia posoperatoria mejorada para los pacientes que reciben una primera resección del hígado. (Traducción-Yesenia Rojas-Khalil ).
Collapse
Affiliation(s)
- Thomas L Sutton
- Division of General Surgery, Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Ranish K Patel
- Division of General Surgery, Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Katherine M Watson
- Division of General Surgery, Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Ivy H Gardner
- Division of General Surgery, Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Daniel O Herzig
- Division of General Surgery, Department of Surgery, Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - V Liana Tsikitis
- Division of General Surgery, Department of Surgery, Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Emerson Y Chen
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
- Division of Hematology/Oncology, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Skye C Mayo
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
- Division of Surgical Oncology, Department of Surgery, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
16
|
Chen H, Yu F, Lu D, Huang S, Liu S, Zhang B, Shu K, Pu D. Enhanced Error Suppression for Accurate Detection of Low-Frequency Variants. Electrophoresis 2025; 46:65-75. [PMID: 39679747 DOI: 10.1002/elps.202400202] [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: 09/11/2024] [Revised: 10/21/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024]
Abstract
The identification of low-frequency variants remains challenging due to the inevitable high error rates of next-generation sequencing (NGS). Numerous promising strategies employ unique molecular identifiers (UMIs) for error suppression. However, their efficiency depends highly on redundant sequencing and quality control, leading to tremendous read waste and cost inefficiency. Here, we describe a novel approach, enhanced error suppression strategy (EES), that addresses these challenges by (1) optimizing data utilization and reducing read waste by utilizing single-read correction that reserves abundant single reads that complement other single reads or single-strand consensus sequences (SSCSs), and (2) effectively enhancing the accuracy of NGS by employing Bayes' theorem. EES significantly improves variant detection accuracy, achieving a background error rate of less than 4.4 × 10-5 per base pair. Additionally, the data utilization rate is dramatically increased, with a 22.9-fold enhancement in duplex consensus sequence (DCS) recovery compared to traditional methodologies. Furthermore, EES demonstrates superior error suppression performance across various base substitutions. In conclusion, EES represents a significant advancement in detecting low-frequency variants by improving data utilization and reducing sequencing errors. It potentially enhances the sensitivity and accuracy of NGS applications, proving highly valuable in clinical and research contexts where precise variant detection is critical.
Collapse
Affiliation(s)
- Huimin Chen
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing, China
| | - Fei Yu
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing, China
| | - Debin Lu
- Department of Neurology, The Second Affiliated Hospital of the Army Medical University of the People's Liberation Army, Chongqing, China
| | - Shiyue Huang
- Chongqing Yangjiaping Middle School, Chongqing, China
| | - Songrui Liu
- Chongqing Yangjiaping Middle School, Chongqing, China
| | - Boseng Zhang
- Chongqing Yangjiaping Middle School, Chongqing, China
| | - Kunxian Shu
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing, China
| | - Dan Pu
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing, China
| |
Collapse
|
17
|
Raval H, Bhattacharya S. Early Detection, Precision Treatment, Recurrence Monitoring: Liquid Biopsy Transforms Colorectal Cancer Therapy. Curr Cancer Drug Targets 2025; 25:586-619. [PMID: 38623975 DOI: 10.2174/0115680096295070240318075023] [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: 11/26/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 04/17/2024]
Abstract
Colorectal cancer (CRC) is a significant global health concern. We need ways to detect it early and determine the best treatments. One promising method is liquid biopsy, which uses cancer cells and other components in the blood to help diagnose and treat the disease. Liquid biopsies focus on three key elements: circulating tumor DNA (ctDNA), circulating microRNA (miRNA), and circulating tumor cells (CTC). By analyzing these elements, we can identify CRC in its early stages, predict how well a treatment will work, and even spot signs of cancer returning. This study investigates the world of liquid biopsy, a rapidly growing field. We want to understand how it can help us better recognize the molecular aspects of cancer, improve and diagnostics, tailor treatments to individual patients, and keep track of the disease over the long-term. We explored specific components of liquid biopsy, like extracellular vesicles and cell-free DNA, and how they are used to detect CRC. This review sheds light on the current state of knowledge and the many ways a liquid biopsy can be used in treating colorectal cancer. It can transform patient care, disease management, and clinical outcomes by offering non-invasive cancer-targeting solutions.
Collapse
Affiliation(s)
- Harshvardhan Raval
- Department of Pharmaceutics, School of Pharmacy and Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy and Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| |
Collapse
|
18
|
Mannucci A, Goel A. Stool and blood biomarkers for colorectal cancer management: an update on screening and disease monitoring. Mol Cancer 2024; 23:259. [PMID: 39558327 PMCID: PMC11575410 DOI: 10.1186/s12943-024-02174-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Biomarkers have revolutionized the management of colorectal cancer (CRC), facilitating early detection, prevention, personalized treatment, and minimal residual disease (MRD) monitoring. This review explores current CRC screening strategies and emerging biomarker applications. MAIN BODY We summarize the landscape of non-invasive CRC screening and MRD detection strategies, discuss the limitations of the current approaches, and highlight the promising potential of novel biomarker solutions. The fecal immunochemical test remained the cornerstone of CRC screening, but its sensitivity has been improved by assays that combined its performance with other stool analytes. However, their sensitivity for advanced adenomas and the patient compliance both remain suboptimal. Blood-based tests promise to increase compliance but require further refinement to compete with stool-based biomarker tests. The ideal scenario involves leveraging blood tests to increase screening participation, and simultaneously promote stool- and endoscopy-based screening among those who are compliant. Once solely reliant on upfront surgery followed by stage and pathology-driven adjuvant chemotherapy, the treatment of stage II and III colon cancer has undergone a revolutionary transformation with the advent of MRD testing after surgery. A decade ago, the concept of using a post-surgical test instead of stage and pathology to determine the need for adjuvant chemotherapy was disruptive. Today, a blood test may be more informative of the need for chemotherapy than the stage at diagnosis. CONCLUSION Biomarker research is not just improving, but bringing a transformative change to CRC clinical management. Early detection is not just getting better, but improving thanks to a multi-modality approach, and personalized treatment plans are not just becoming a reality, but a promising future with MRD testing.
Collapse
Affiliation(s)
- Alessandro Mannucci
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute at City of Hope, Monrovia, CA, USA
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Hospital, Milan, Italy
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute at City of Hope, Monrovia, CA, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
19
|
Jin KM, Bao Q, Zhao TT, Wang HW, Huang LF, Wang K, Xing BC. Comparing baseline VAF in circulating tumor DNA and tumor tissues predicting prognosis of patients with colorectal cancer liver metastases after curative resection. Clin Res Hepatol Gastroenterol 2024; 48:102464. [PMID: 39276854 DOI: 10.1016/j.clinre.2024.102464] [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: 01/10/2024] [Revised: 07/12/2024] [Accepted: 09/12/2024] [Indexed: 09/17/2024]
Abstract
INTRODUCTION The prognostic value of baseline variant allele frequency (VAF) in circulating tumor DNA (ctDNA) of colorectal cancer liver metastases (CRLM) patients after curative resection was rarely investigated. METHODS A single-center prospective study was performed to investigate the prognostic impact of baseline VAF in ctDNA and matched tumor tissues of CRLM patients after curative resection between May 2019 and May 2021 by the Illumina NovoSeq 6000 platform. The relationship of the tumor burden score (TBS) and the VAF in ctDNA and matched tumor tissues was evaluated by the Pearson correlation method. The survival curves of recurrence-free survival (RFS) and overall survival (OS) were plotted. Factors associated with RFS were calculated using Cox regression analysis, and an integrated prognostic model using significant baseline variables was proposed. RESULTS There were 121 patients with baseline ctDNA and matched tumor tissues enrolled in the study. A total of 417 mutations spanning 20 genes were identified in baseline tumor tissues of 119/121 (98.3 %) cases. The overall mutations in tumor tissues were completely covered by ctDNA in 52 of 121(43.0 %) patients. Baseline VAF in ctDNA but not in tumor tissues was significantly correlated to TBS of CRLM (R = 0.36, p < 0.001). Significantly longer RFS but not OS was observed in patients with lower VAF in ctDNA compared to those with higher one (p < 0.001 and p = 0.33 respectively). Multivariate Cox regression analysis showed higher VAF in baseline ctDNA was an independent risk factor for RFS. An integrated prognostic model including baseline metastasis location and VAF in ctDNA outperformed the traditional CRS model in predicting RFS. CONCLUSION Baseline VAF in ctDNA but not in tumor tissues influenced RFS of CRLM patients after curative resection.
Collapse
Affiliation(s)
- Ke-Min Jin
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China
| | - Quan Bao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China
| | - Ting-Ting Zhao
- Research Institute, GloriousMed Clinical Laboratory Co., Ltd., Shanghai, PR China
| | - Hong-Wei Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China
| | - Long-Fei Huang
- Research Institute, GloriousMed Clinical Laboratory Co., Ltd., Shanghai, PR China
| | - Kun Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China.
| | - Bao-Cai Xing
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China.
| |
Collapse
|
20
|
Kataoka K, Mori K, Nakamura Y, Watanabe J, Akazawa N, Hirata K, Yokota M, Kato K, Kotaka M, Yamazaki K, Kagawa Y, Mishima S, Ando K, Miyo M, Yukami H, Laliotis G, Sharma S, Palsuledesai CC, Rabinowitz M, Jurdi A, Liu MC, Aleshin A, Kotani D, Bando H, Taniguchi H, Takemasa I, Kato T, Yoshino T, Oki E. Survival benefit of adjuvant chemotherapy based on molecular residual disease detection in resected colorectal liver metastases: subgroup analysis from CIRCULATE-Japan GALAXY. Ann Oncol 2024; 35:1015-1025. [PMID: 39293512 DOI: 10.1016/j.annonc.2024.08.2240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND The prognostic role of circulating tumor DNA (ctDNA)-based molecular residual disease (MRD) detection and its utility for postsurgical risk stratification has been reported in colorectal cancer. In this study, we explored the use of ctDNA-based MRD detection in patients with colorectal liver metastases (CLM), for whom the survival benefit of adjuvant chemotherapy (ACT) after surgical resection remains unclear. METHODS Patients with CLM without extrahepatic disease from the GALAXY study (UMIN000039205) were included. The disease-free survival (DFS) benefit of ACT was evaluated in MRD-positive and -negative groups after adjusting for age, gender, number, and size of liver metastases, RAS status, and previous history of oxaliplatin for primary cancer. ctDNA was detected using a personalized, tumor-informed 16-plex polymerase chain reaction-next-generation sequencing (mPCR-NGS) assay. ctDNA-based MRD status was evaluated 2-10 weeks after curative surgery, before the start of ACT. RESULTS Among 6061 patients registered in GALAXY, 190 surgically resected CLM patients without any preoperative chemotherapy were included with a median follow-up of 24 months (1-48 months). ctDNA positivity in the MRD window was 32.1% (61/190). ACT was administered to 25.1% (48/190) of patients. In the MRD-positive group, 24-month DFS was higher for patients treated with ACT [33.3% versus not reached, adjusted hazard ratio (HR): 0.07, P < 0.0001]; whereas no benefit of ACT was seen in the MRD-negative group (24-month DFS: 72.3% versus 62.2%, adjusted HR: 0.68, P = 0.371). Multivariate analysis showed that the size of liver metastases (HR: 3.94, P = 0.031) was prognostic of DFS in the MRD-positive group. In the MRD-negative group, however, none of the clinicopathological factors were prognostic of DFS. CONCLUSIONS Our data suggest that ACT may offer notable clinical benefits in MRD-positive patients with CLM. MRD status-based risk stratification could be potentially incorporated in future clinical trials for CLM.
Collapse
Affiliation(s)
- K Kataoka
- Division of Lower GI Surgery, Department of Gastroenterological Surgery, Hyogo Medical University, Nishinomiya
| | - K Mori
- Department of Biostatistics, Clinical Research Center, Shizuoka Cancer Center, Sunto-gun
| | - Y Nakamura
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa; Translational Research Support Office, National Cancer Center Hospital East, Kashiwa; International Research Promotion Office, National Cancer Center Hospital East, Kashiwa
| | - J Watanabe
- Department of Colorectal Surgery, Kansai Medical University, Hirakata; Department of Surgery, Gastroenterological Center, Yokohama City University Medical Center, Yokohama
| | - N Akazawa
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai
| | - K Hirata
- Department of Surgery 1, School of Medicine, University of Occupational and Environmental Health, Kitakyushu
| | - M Yokota
- Department of General Surgery, Kurashiki Central Hospital, Kurashiki
| | - K Kato
- Department of Surgery, Teine-Keijinkai Hospital, Sapporo
| | - M Kotaka
- Gastrointestinal Cancer Center, Sano Hospital, Kobe
| | - K Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Sunto-gun
| | - Y Kagawa
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka; Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka
| | - S Mishima
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa
| | - K Ando
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | - M Miyo
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, Sapporo
| | - H Yukami
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | | | | | | | | | | | | | | | - D Kotani
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa
| | - H Bando
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa
| | - H Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya
| | - I Takemasa
- Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, Sapporo
| | - T Kato
- Department of Surgery, NHO Osaka National Hospital, Osaka
| | - T Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa; Department of Gastroenterological Surgery/Pediatric Surgery, Graduate School of Medicine, Gifu University, Gifu; Kindai University Faculty of Medicine, Higashiosaka City, Japan
| | - E Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka.
| |
Collapse
|
21
|
Battaglin F, Lenz HJ. Clinical Applications of Circulating Tumor DNA Profiling in GI Cancers. JCO Oncol Pract 2024; 20:1481-1490. [PMID: 39531845 PMCID: PMC11567053 DOI: 10.1200/op.24.00167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/11/2024] [Accepted: 05/01/2024] [Indexed: 11/16/2024] Open
Abstract
Over the next few years, the analysis of circulating tumor DNA (ctDNA) through liquid biopsy is expected to enter clinical practice and revolutionize the approach to biomarker testing and treatment selection in GI cancers. In fact, growing evidence support the use of ctDNA testing as a noninvasive, effective, and highly specific tool for molecular profiling in GI cancers. Analysis of blood ctDNA has been investigated in multiple settings including early tumor detection, minimal residual disease evaluation, tumor diagnosis and evaluation of prognostic/predictive biomarkers for targeted treatment selection, longitudinal monitoring of treatment response, and identification of resistance mechanisms. Here, we review the clinical applications, advantages, and limitations of ctDNA profiling for precision oncology in GI cancers.
Collapse
Affiliation(s)
- Francesca Battaglin
- Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Heinz-Josef Lenz
- Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Kalil JA, Krzywon L, Petrillo SK, Tsamchoe M, Zlotnik O, Lazaris A, Metrakos P. Feasibility of ctDNA in detecting minimal residual disease and predicting recurrence for colorectal cancer liver metastases. Front Oncol 2024; 14:1418696. [PMID: 39439963 PMCID: PMC11493539 DOI: 10.3389/fonc.2024.1418696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Approximately 50% of patients diagnosed with colorectal cancer develop colorectal cancer liver metastases (CRLM). Although curative intent liver resection provides 5-year survival of 40-50%, up to 70% of patients develop recurrence of CRLM. Detection of minimal residual disease (MRD) is essential for timely, optimized treatment. This study evaluated the feasibility and utility of using circulating tumor DNA (ctDNA) to identify MRD and predict disease recurrence. Methods Patients with CRLM that underwent liver resection and had known KRAS or PIK3CA mutations were retrospectively identified. Serial blood samples were collected every 3 months following surgery for disease surveillance. ctDNA was isolated from the samples and analyzed with digital PCR (dPCR). Results KRAS and PIK3CA mutations were identified by dPCR in 29 patients over 115 timepoints. In patients with detectable ctDNA at time of liver resection, 81% (13/16) developed disease recurrence, while 46% (6/13) of the patients with undetectable ctDNA recurred (p=0.064). Presence of ctDNA was detected in 27.6% (8/29) of the initial postoperative samples. Radiologic recurrence was later diagnosed in 100% (8/8) of these patients, while 52% (11/21) who had undetectable ctDNA postoperatively recurred (p=0.026). Detectable ctDNA postoperatively was associated with a shorter disease-free survival (DFS) of 9 months vs 13 months in patients who had undetectable ctDNA (HR 2.95, 95% CI 1.16-7.49; p=0.02). Conclusion Liquid biopsy using dPCR can identify low levels of ctDNA, enabling early detection of disease recurrence. Additionally, the presence of ctDNA postoperatively was predictive of recurrence. This study corroborates current literature and provides rational for moving toward a clinical trial using ctDNA and dPCR to detect MRD after CRLM resection.
Collapse
Affiliation(s)
- Jennifer A. Kalil
- Department of Surgery, Royal Victoria Hospital - McGill University Health Center, Montréal, QC, Canada
- Research Institute, McGill University Health Center, Montréal, QC, Canada
| | - Lucyna Krzywon
- Research Institute, McGill University Health Center, Montréal, QC, Canada
| | | | - Migmar Tsamchoe
- Department of Anatomy and Cell Biology, McGill University Health Center, Montréal, QC, Canada
| | - Oran Zlotnik
- Department of Surgery, Royal Victoria Hospital - McGill University Health Center, Montréal, QC, Canada
- Research Institute, McGill University Health Center, Montréal, QC, Canada
| | - Anthoula Lazaris
- Research Institute, McGill University Health Center, Montréal, QC, Canada
| | - Peter Metrakos
- Department of Surgery, Royal Victoria Hospital - McGill University Health Center, Montréal, QC, Canada
- Research Institute, McGill University Health Center, Montréal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University Health Center, Montréal, QC, Canada
| |
Collapse
|
23
|
Yang J, Yu C, Li H, Peng D, Zhou Q, Yao J, Lv J, Fang S, Shi J, Wei Y, Wang G, Cai S, Zhang Z, Zhang Z, Zhou J. Evaluation of Molecular Residual Disease by a Fixed Panel in Resectable Colorectal Cancer. Cancer Res Treat 2024; 56:1183-1196. [PMID: 38726508 PMCID: PMC11491239 DOI: 10.4143/crt.2023.1371] [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: 12/28/2023] [Accepted: 04/29/2024] [Indexed: 10/16/2024] Open
Abstract
PURPOSE Molecular residual disease (MRD) is a promising biomarker in colorectal cancer (CRC) for prognosis and guiding treatment, while the whole-exome sequencing (WES) based tumor-informed assay is standard for evaluating MRD based on circulating tumor DNA (ctDNA). In this study, we assessed the feasibility of a fixed-panel for evaluating MRD in CRC. MATERIALS AND METHODS Seventy-five patients with resectable stage I-III CRC were enrolled. Tumor tissues obtained by surgery, and preoperative and postoperative day 7 blood samples were collected. The ctDNA was evaluated using the tumor-agnostic and tumor-informed fixed assays, as well as the WES-based and panel-based personalized assays in randomly selected patients. RESULTS The tumor-informed fixed assay had a higher preoperative positive rate than the tumor-agnostic assay (73.3% vs. 57.3%). The preoperative ctDNA status failed to predict disease-free survival (DFS) in either of the fixed assays, while the tumor-informed fixed assay-determined postoperative ctDNA positivity was significantly associated with worse DFS (hazard ratio [HR], 20.74; 95% confidence interval [CI], 7.19 to 59.83; p < 0.001), which was an independent predictor by multivariable analysis (HR, 28.57; 95% CI, 7.10 to 114.9; p < 0.001). Sub-cohort analysis indicated the WES-based personalized assay had the highest preoperative positive rate (95.1%). The two personalized assays and the tumor-informed fixed assay demonstrated same results in postoperative landmark (HR, 26.34; 95% CI, 6.01 to 115.57; p < 0.001), outperforming the tumor-agnostic fixed panel (HR, 3.04; 95% CI, 0.94 to 9.89; p=0.052). CONCLUSION Our study confirmed the prognostic value of the ctDNA positivity at postoperative day 7 by the tumor-informed fixed panel. The tumor-informed fixed panel may be a cost-effective method to evaluate MRD, which warrants further studies in future.
Collapse
Affiliation(s)
- Jian Yang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Chengqing Yu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Haoran Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Di Peng
- Burning Rock Biotech, Guangdong, China
| | | | - Jun Yao
- Department of General Surgery, The Dushu Lake Hospital Affiliated to Soochow University, Jiangsu, China
| | - Juan Lv
- Burning Rock Biotech, Guangdong, China
| | | | | | - Yijun Wei
- Department of General Surgery, The Dushu Lake Hospital Affiliated to Soochow University, Jiangsu, China
| | | | | | | | - Zixiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Jian Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Jiangsu, China
| |
Collapse
|
24
|
Boeken T, Pellerin O, Bourreau C, Palle J, Gallois C, Zaanan A, Taieb J, Lahlou W, Di Gaeta A, Al Ahmar M, Guerra X, Dean C, Laurent Puig P, Sapoval M, Pereira H, Blons H. Clinical value of sequential circulating tumor DNA analysis using next-generation sequencing and epigenetic modifications for guiding thermal ablation for colorectal cancer metastases: a prospective study. LA RADIOLOGIA MEDICA 2024; 129:1530-1542. [PMID: 39183242 DOI: 10.1007/s11547-024-01865-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION While thermal ablation is now a standard treatment option for oligometastatic colorectal cancer patients, selecting those who will benefit most from locoregional therapies remains challenging. This proof-of-concept study is the first to assess the feasibility of routine testing of ctDNA before and after thermal ablation with curative intent, analyzed by next-generation sequencing (NGS) and methylation specific digital droplet PCR (ddPCR). Our prospective study primary objective was to assess the prognostic value of ctDNA before thermal ablation. METHODS This single-center prospective study from November 2021 to June 2022 included colorectal cancer patients referred for curative-intent thermal ablation. Cell-free DNA was tested at different time points by next-generation sequencing and detection of WIF1 and NPY genes hypermethylation using ddPCR. The ctDNA was considered positive if either a tumor mutation or hypermethylation was detected; recurrence-free survival was used as the primary endpoint. RESULTS The study enrolled 15 patients, and a total of 60 samples were analyzed. The median follow-up after ablation was 316 days, and median recurrence-free survival was 250 days. CtDNA was positive for 33% of the samples collected during the first 24 h. The hazard ratio for progression according to the presence of baseline circulating tumor DNA was estimated at 0.14 (CI 95%: 0.03-0.65, p = 0.019). The dynamics are provided, and patients with no recurrence were all negative at H24 for ctDNA. DISCUSSION This study shows the feasibility of routine testing of ctDNA before and after thermal ablation with curative intent. We report that circulating tumor DNA is detectable in patients with low tumor burden using 2 techniques. This study emphasizes the potential of ctDNA for discerning patients who are likely to benefit from thermal ablation from those who may not, which could shape future referrals. The dynamics of ctDNA before and after ablation shed light on the need for further research and larger studies.
Collapse
Affiliation(s)
- Tom Boeken
- Department of Vascular and Oncological Interventional Radiology, AP-HP, INSERM PARCC U 970, Hôpital Européen Georges Pompidou, HEKA INRIA, Université de Paris Cité, Paris, France.
| | - Olivier Pellerin
- Department of Vascular and Oncological Interventional Radiology, AP-HP, INSERM PARCC U 970, Hôpital Européen Georges Pompidou, HEKA INRIA, Université de Paris Cité, Paris, France
| | | | - Juliette Palle
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Européen Georges Pompidou, SIRIC CARPEM, Université Paris Cité, Paris, France
| | - Claire Gallois
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Européen Georges Pompidou, SIRIC CARPEM, Université Paris Cité, Paris, France
| | - Aziz Zaanan
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Européen Georges Pompidou, SIRIC CARPEM, Université Paris Cité, Paris, France
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Européen Georges Pompidou, SIRIC CARPEM, Université Paris Cité, Paris, France
| | - Widad Lahlou
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Européen Georges Pompidou, SIRIC CARPEM, Université Paris Cité, Paris, France
| | - Alessandro Di Gaeta
- Department of Vascular and Oncological Interventional Radiology, AP-HP, INSERM PARCC U 970, Hôpital Européen Georges Pompidou, HEKA INRIA, Université de Paris Cité, Paris, France
| | - Marc Al Ahmar
- Department of Vascular and Oncological Interventional Radiology, AP-HP, INSERM PARCC U 970, Hôpital Européen Georges Pompidou, HEKA INRIA, Université de Paris Cité, Paris, France
| | - Xavier Guerra
- Department of Vascular and Oncological Interventional Radiology, AP-HP, INSERM PARCC U 970, Hôpital Européen Georges Pompidou, HEKA INRIA, Université de Paris Cité, Paris, France
| | | | - Pierre Laurent Puig
- Department of Biochemistry, Pharmacogenetics and Molecular Oncology (ONSTeP), AP-HP, Hôpital Européen Georges Pompidou, Paris Cancer Institute CARPEM, Université de Paris Cité, Paris, France
| | - Marc Sapoval
- Department of Vascular and Oncological Interventional Radiology, AP-HP, INSERM PARCC U 970, Hôpital Européen Georges Pompidou, HEKA INRIA, Université de Paris Cité, Paris, France
| | - Helena Pereira
- Centre d'investigation Clinique 1418 Épidémiologie Clinique, AP-HP, INSERM, Hôpital Européen Georges Pompidou, Clinical Research Unit, Paris, France
| | - Hélène Blons
- Department of Biochemistry, Pharmacogenetics and Molecular Oncology (ONSTeP), AP-HP, Hôpital Européen Georges Pompidou, Paris Cancer Institute CARPEM, Université de Paris Cité, Paris, France
| |
Collapse
|
25
|
Bürtin F, Elias L, Hinz S, Forster M, Hildebrandt G, Frerker B, Bock F. ctDNA responds to neoadjuvant treatment in locally advanced rectal cancer. J Cancer Res Clin Oncol 2024; 150:428. [PMID: 39307893 PMCID: PMC11417078 DOI: 10.1007/s00432-024-05944-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Liquid biopsy is a minimally invasive procedure investigating tumor mutations. METHODS In our retrospective study, we investigated whether molecular therapy monitoring of patients receiving neoadjuvant radio(chemo)therapy on a daily routine is possible in 17 patients with locally advanced rectal cancer. Six patients received short-course radiotherapy (5 × 5 Gy) with subsequent surgery, six patients were treated according RAPIDO protocol with short-course radiotherapy followed by chemotherapy (FOLFOX4) and subsequent surgery and five patients received conventional neoadjuvant radiochemotherapy with 5-FU followed by surgery. Response was assessed by Dworak. Liquid biopsies were taken before and immediately after neoadjuvant radiotherapy to isolate and ultradeeply sequence cell free DNA with a panel of 127 genes. Somatic mutations were determined bioinformatically by comparison with normal DNA from leukocytes to distinguish them from germline variants or aging mutations. RESULTS In 12 patients (71%) at least one somatic mutation was detected. In 8/12 patients a decrease and in 4/12 an increase or mixed response in ctDNA was seen. Statistical correlation between ctDNA analysis and clinical response could not be seen. CONCLUSION ctDNA is responding to neoadjuvant therapy and liquid biopsy is easily integrated into a daily routine. As part of translational research this protocol leaves room for further investigations.
Collapse
Affiliation(s)
- Florian Bürtin
- Department of General Surgery, Rostock University Medical Center, Rostock, Germany
| | - Liema Elias
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Medical Center Schleswig- Holstein, Kiel, Germany
| | - Sebastian Hinz
- Department of General Surgery, Rostock University Medical Center, Rostock, Germany
| | - Michael Forster
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Medical Center Schleswig- Holstein, Kiel, Germany
| | - Guido Hildebrandt
- Department of Radiotherapy and Radiation Oncology, Rostock University Medical Center, Südring 75, 18059, Rostock, Germany
| | - Bernd Frerker
- Department of Radiotherapy and Radiation Oncology, Rostock University Medical Center, Südring 75, 18059, Rostock, Germany
| | - Felix Bock
- Department of Radiotherapy and Radiation Oncology, Rostock University Medical Center, Südring 75, 18059, Rostock, Germany.
| |
Collapse
|
26
|
Cutts R, Ulrich L, Beaney M, Robert M, Coakley M, Bunce C, Crestani GW, Hrebien S, Kalashnikova E, Wu HT, Dashner S, Sethi H, Aleshin A, Liu M, Ring A, Okines A, Smith IE, Barry P, Turner NC, Garcia-Murillas I. Association of post-operative ctDNA detection with outcomes of patients with early breast cancers. ESMO Open 2024; 9:103687. [PMID: 39216186 PMCID: PMC11402396 DOI: 10.1016/j.esmoop.2024.103687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/08/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND In early breast cancer (EBC) patients, we aimed to determine whether circulating tumor DNA (ctDNA) analysis following primary surgery, before systemic therapy, identified molecular residual disease and was associated with risk of relapse and relapse-free survival (RFS). METHODS Plasma was collected, retrospectively, before surgery, 1-14 weeks post-operatively, and before adjuvant therapy, and in a subset of patients after adjuvant therapy. A personalized, tumor-informed, multiplex PCR next generation sequencing assay (Signatera™) was used for ctDNA detection and quantification. The primary objective was to compare RFS and distant recurrence-free survival (DRFS) in patients with detected versus non-detected ctDNA. RESULTS A total of 48 patients with EBC (median age 50.5 years) [34 hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-), 5 HER2+, 9 triple-negative breast cancer) were included. ctDNA was detected in 64.5% (20/31) of patients before surgery, and 35.4% (17/48) after surgery. ctDNA detection before surgery was associated with tumor grade (P = 0.019), ctDNA detection after surgery was associated with receptor subtype (P = 0.01). Patients with ctDNA detected after surgery had worse DRFS [hazard ratio = 5.5, 95% confidence interval (CI) 1.1-28.5, P = 0.04]. RFS in patients with ctDNA detected after surgery was worse than in those with lack of ctDNA detection, although not statistically significant (hazard ratio = 3.7, 95% CI 0.9-15.7, P = 0.073). Patients with ctDNA detected preoperatively or post-operatively had a trend towards worse RFS (hazard ratio = 7.8, 95% CI 0.9-63.7, P = 0.05) and DRFS (hazard ratio = 6.8, 95% CI 0.8-57, P = 0.07) compared with those with ctDNA undetected at both timepoints. ctDNA detection anticipated clinical relapse with a median lead time of 16 months. CONCLUSIONS In patients with treatment-naive EBC, ctDNA is detectable after surgery. The absence of ctDNA at a single post-surgical timepoint is associated with improved DRFS, supporting the development of future trials studying de-escalation of systemic therapy.
Collapse
Affiliation(s)
- R Cutts
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London
| | - L Ulrich
- Breast Unit, Royal Marsden Hospital, London, UK
| | - M Beaney
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London
| | - M Robert
- Department of Breast Medical Oncology and Neuro-Oncology, Early Therapeutic Unit, Institute of Oncology de l'Ouest, St Herblain, France
| | - M Coakley
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London
| | - C Bunce
- Clinical Trials Unit, Royal Marsden Hospital, London, UK
| | - G W Crestani
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London
| | - S Hrebien
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London
| | | | - H-T Wu
- Natera, Inc., Austin, USA
| | | | | | | | - M Liu
- Natera, Inc., Austin, USA
| | - A Ring
- Breast Unit, Royal Marsden Hospital, London, UK
| | - A Okines
- Breast Unit, Royal Marsden Hospital, London, UK
| | - I E Smith
- Breast Unit, Royal Marsden Hospital, London, UK
| | - P Barry
- Breast Unit, Royal Marsden Hospital, London, UK
| | - N C Turner
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London; Breast Unit, Royal Marsden Hospital, London, UK; The Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - I Garcia-Murillas
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London.
| |
Collapse
|
27
|
Negri F, Gnetti L, Aschele C. Editorial: Advances in molecular biology knowledge of rectal cancer and forthcoming role of liquid biopsy. Front Oncol 2024; 14:1476174. [PMID: 39267833 PMCID: PMC11390347 DOI: 10.3389/fonc.2024.1476174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Affiliation(s)
- Francesca Negri
- Gastroenterology and Endoscopy Unit, University Hospital of Parma, Parma, Italy
| | - Letizia Gnetti
- Department of Pathology, University Hospital of Parma, Parma, Italy
| | - Carlo Aschele
- Medical Oncology Unit, Department of Oncology, Ospedale Sant'Andrea, La Spezia, Italy
| |
Collapse
|
28
|
da Silva TF, de Azevedo JC, Teixeira EB, Casseb SMM, Moreira FC, de Assumpção PP, dos Santos SEB, Calcagno DQ. From haystack to high precision: advanced sequencing methods to unraveling circulating tumor DNA mutations. Front Mol Biosci 2024; 11:1423470. [PMID: 39165643 PMCID: PMC11333322 DOI: 10.3389/fmolb.2024.1423470] [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: 04/25/2024] [Accepted: 07/11/2024] [Indexed: 08/22/2024] Open
Abstract
Identifying mutations in cancer-associated genes to guide patient treatments is essential for precision medicine. Circulating tumor DNA (ctDNA) offers valuable insights for early cancer detection, treatment assessment, and surveillance. However, a key issue in ctDNA analysis from the bloodstream is the choice of a technique with adequate sensitivity to identify low frequent molecular changes. Next-generation sequencing (NGS) technology, evolving from parallel to long-read capabilities, enhances ctDNA mutation analysis. In the present review, we describe different NGS approaches for identifying ctDNA mutation, discussing challenges to standardized methodologies, cost, specificity, clinical context, and bioinformatics expertise for optimal NGS application.
Collapse
Affiliation(s)
- Tamires Ferreira da Silva
- Programa de Residência Multiprofissional em Saúde (Oncologia), Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Brazil
| | - Juscelino Carvalho de Azevedo
- Programa de Residência Multiprofissional em Saúde (Oncologia), Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Brazil
| | | | | | | | | | | | - Danielle Queiroz Calcagno
- Programa de Residência Multiprofissional em Saúde (Oncologia), Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Brazil
| |
Collapse
|
29
|
Alsaab HO, Alzahrani MS, Bahauddin AA, Almutairy B. Circulating tumor DNA (ctDNA) application in investigation of cancer: Bench to bedside. Arch Biochem Biophys 2024; 758:110066. [PMID: 38906310 DOI: 10.1016/j.abb.2024.110066] [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: 04/10/2024] [Revised: 06/02/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Now, genomics forms the core of the precision medicine concept. Comprehensive investigations of tumor genomes have made it possible to characterize tumors at the molecular level and, specifically, to identify the fundamental processes that cause condition. A variety of kinds of tumors have seen better outcomes for patients as a result of the development of novel medicines to tackle these genetic-driving processes. Since therapy may exert selective pressure on cancers, non-invasive methods such as liquid biopsies can provide the opportunity for rich reservoirs of crucial and real-time genetic data. Liquid biopsies depend on the identification of circulating cells from tumors, circulating tumor DNA (ctDNA), RNA, proteins, lipids, and metabolites found in patient biofluids, as well as cell-free DNA (cfDNA), which exists in those with cancer. Although it is theoretically possible to examine biological fluids other than plasma, such as pleural fluid, urine, saliva, stool, cerebrospinal fluid, and ascites, we will limit our discussion to blood and solely cfDNA here for the sake of conciseness. Yet, the pace of wider clinical acceptance has been gradual, partly due to the increased difficulty of choosing the best analysis for the given clinical issue, interpreting the findings, and delaying proof of value from clinical trials. Our goal in this review is to discuss the current clinical value of ctDNA in cancers and how clinical oncology systems might incorporate procedures for ctDNA testing.
Collapse
Affiliation(s)
- Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, 21944, Saudi Arabia.
| | - Mohammad S Alzahrani
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
| | - Ammar A Bahauddin
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina Al-Munawarah, Saudi Arabia.
| | - Bandar Almutairy
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia.
| |
Collapse
|
30
|
Kataoka K, Yamada T, Yamazaki K, Mori K, Matsuhashi N, Shiozawa M, Iwai T, Goto M, Yasui M, Takii Y, Suto T, Takamizawa Y, Takase N, Sharma S, Ensor J, Jurdi A, Liu MC, Ikeda M, Kanemitsu Y. Trial Protocol of a Phase II Study of mFOLFOXIRI after Metastasectomy in Patients with Oligometastatic Colorectal Cancer (FANTASTIC Study). J Anus Rectum Colon 2024; 8:246-252. [PMID: 39086881 PMCID: PMC11286365 DOI: 10.23922/jarc.2024-025] [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: 03/21/2024] [Accepted: 04/23/2024] [Indexed: 08/02/2024] Open
Abstract
Background The survival benefit of adjuvant chemotherapy after surgical resection of oligometastases from colorectal cancer (CRC) remains unclear. The prognostic role of circulating-tumor DNA (ctDNA) was reported recently and a risk stratification strategy based on monitoring minimal/molecular residual disease (MRD) has been proposed, however, which drug regimen is most effective for ctDNA-positive patients is unknown. Methods/Design Oligometastatic CRC patients planning to undergo surgery were registered in this study. After metastasectomy, the registered patients were enrolled in the treatment arm, in which 8 courses of modified-FOLFOXIRI (mFOLFOXIRI; irinotecan 150 mg/m2, oxaliplatin 85 mg/m2, l-leucovorin (l-LV) 200 mg/m2, and 46-h continuous infusion of 5-fluorouracil (5-FU) 2400 mg/m2 every 2 weeks) followed by 4 courses of 5-FU/l-LV are administered. The patients who did not meet the eligibility criteria for the treatment arm or did not consent to mFOLFOXIRI enrolled in the observation arm in which standard of care treatment is provided. Prospective blood collections for retrospective ctDNA analysis are scheduled pre-surgery, and at 28 days, 4 and 7 months after surgery. The primary endpoint is treatment compliance at 8 courses of mFOLFOXIRI and the key secondary endpoints are the ctDNA-positivity rate and survival outcomes in ctDNA-positive and -negative groups. A total of 85 patients will be enrolled from 11 institutions. First patient-in was on July 2020. Accrual completed in February 2024. Discussion This study will potentially identify a better treatment strategy for patients with resectable oligometastatic CRC having postsurgical ctDNA positivity, compared to the current standard of care approaches.
Collapse
Affiliation(s)
- Kozo Kataoka
- Division of Lower GI, Department of Gastroenterological Surgery, Hyogo Medical University, Nishinomiya, Japan
| | - Takeshi Yamada
- Department of Gastrointestinal Surgery, Nippon Medical School, Tokyo, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shunto, Japan
| | - Keita Mori
- Department of Biostatistics, Clinical Research Center, Shizuoka Cancer Center, Shunto, Japan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery・Pediatric Surgery, Gifu University School of Medicine, Gifu, Japan
| | - Manabu Shiozawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Kanagawa, Japan
| | - Takuma Iwai
- Department of Gastrointestinal Surgery, Nippon Medical School, Tokyo, Japan
| | - Masahiro Goto
- The Second Department of Internal Medicine, Osaka Medical College Hospital, Takatsuki, Japan
| | - Masayoshi Yasui
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Yasumasa Takii
- Department of Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Takeshi Suto
- Department of Gastrointestinal Surgery, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Yasuyuki Takamizawa
- Department of Colorectal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Naoto Takase
- Department of Medical Oncology, Takarazuka City Hospital, Takarazuka, Japan
| | | | | | | | | | - Masataka Ikeda
- Division of Lower GI, Department of Gastroenterological Surgery, Hyogo Medical University, Nishinomiya, Japan
| | - Yukihide Kanemitsu
- Department of Colorectal Surgery, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
31
|
Parikh AR, Chee BH, Tsai J, Rich TA, Price KS, Patel SA, Zhang L, Ibrahim F, Esquivel M, Van Seventer EE, Jarnagin JX, Raymond VM, Corvera CU, Hirose K, Nakakura EK, Corcoran RB, Van Loon K, Atreya CE. Minimal Residual Disease using a Plasma-Only Circulating Tumor DNA Assay to Predict Recurrence of Metastatic Colorectal Cancer Following Curative Intent Treatment. Clin Cancer Res 2024; 30:2964-2973. [PMID: 38695832 PMCID: PMC11247320 DOI: 10.1158/1078-0432.ccr-23-3660] [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: 11/28/2023] [Revised: 02/28/2024] [Accepted: 04/30/2024] [Indexed: 07/16/2024]
Abstract
PURPOSE Minimal residual disease (MRD) detection can identify the recurrence in patients with colorectal cancer (CRC) following definitive treatment. We evaluated a plasma-only MRD assay to predict recurrence and survival in patients with metastatic CRC who underwent curative intent procedures (surgery and/or radiotherapy), with or without (neo)adjuvant chemotherapy. The primary objective of this study was to assess the correlation of postprocedure tumor cell-free DNA detection status with radiographic disease recurrence. EXPERIMENTAL DESIGN Preprocedure and postprocedure longitudinal samples were collected from 53 patients and analyzed with a multiomic MRD assay detecting circulating tumor DNA (ctDNA) from genomic and epigenomic signals. Preprocedure and postprocedure ctDNA detection correlated with recurrence-free and overall survival (OS). RESULTS From 52 patients, 230/233 samples were successfully analyzed. At the time of data cutoff, 36 (69.2%) patients recurred with median follow-up of 31 months. Detectable ctDNA was observed in 19/42 patients (45.2%) with ctDNA analyzed 3 weeks postprocedure. ctDNA detection 3 weeks postprocedure was associated with shorter median recurrence-free survival (RFS; HR, 5.27; 95% CI, 2.31-12.0; P < 0.0001) and OS (HR, 12.83; 95% CI, 3.6-45.9; P < 0.0001). Preprocedure ctDNA detection status was not associated with RFS but was associated with improved OS (HR, 4.65; 95% CI, 1.4-15.2; P = 0.0111). Undetectable ctDNA preprocedure had notable long-term OS, >90% 3 years postprocedure. CONCLUSIONS In this cohort of oligometastatic CRC, detection of ctDNA preprocedure or postprocedure was associated with inferior outcomes even after accounting for known prognostic clinicopathologic variables. This suggests ctDNA may enhance current risk stratification methods helping the evaluation of novel treatments and surveillance strategies toward improving patient outcomes.
Collapse
Affiliation(s)
- Aparna R Parikh
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts
| | - Bryant H Chee
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center (HDFCCC), San Francisco, California
| | - Jill Tsai
- Guardant Health, Palo Alto, California
| | | | | | | | - Li Zhang
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center (HDFCCC), San Francisco, California
- Department of Medicine, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Faaiz Ibrahim
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center (HDFCCC), San Francisco, California
| | - Mikaela Esquivel
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center (HDFCCC), San Francisco, California
| | - Emily E Van Seventer
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Joy X Jarnagin
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts
| | - Victoria M Raymond
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts
| | - Carlos U Corvera
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center (HDFCCC), San Francisco, California
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Kenzo Hirose
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center (HDFCCC), San Francisco, California
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Eric K Nakakura
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center (HDFCCC), San Francisco, California
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Ryan B Corcoran
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts
| | - Katherine Van Loon
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center (HDFCCC), San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Chloe E Atreya
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center (HDFCCC), San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| |
Collapse
|
32
|
O’Donnell CDJ, Naleid N, Siripoon T, Zablonski KG, Storandt MH, Selfridge JE, Hallemeier CL, Conces ML, Jethwa KR, Bajor DL, Thiels CA, Warner SG, Starlinger PP, Atwell TD, Mitchell JL, Mahipal A, Jin Z. Circulating Tumor DNA Predicts Early Recurrence Following Locoregional Therapy for Oligometastatic Colorectal Cancer. Cancers (Basel) 2024; 16:2407. [PMID: 39001469 PMCID: PMC11240520 DOI: 10.3390/cancers16132407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
(1) Background: Local therapies offer a potentially curative approach for patients with oligometastatic colorectal cancer (CRC). An evidence-based consensus recommendation for systemic therapy following definitive locoregional therapy is lacking. Tumor-informed circulating tumor DNA (ctDNA) might provide information to help guide management in this setting. (2) Methods: A multi-institutional retrospective study was conducted, including patients with CRC that underwent curative-intent locoregional therapy to an isolated site of metastatic disease, followed by tumor-informed ctDNA assessment. The Kaplan-Meier method and log-rank tests were used to compare disease-free survival based on ctDNA results. ctDNA test performance was compared to carcinoembryonic antigen (CEA) test results using McNemar's test. (3) Results: Our study cohort consisted of 87 patients treated with locoregional interventions who underwent ctDNA testing. The initial ctDNA test post-intervention was positive in 28 patients and negative in 59 patients. The median follow-up time was 14.0 months. Detectable ctDNA post-intervention was significantly associated with early disease recurrence, with a median disease-free survival (DFS) of 6.63 months compared to 21.30 months in ctDNA-negative patients (p < 0.001). ctDNA detected a numerically higher proportion of recurrences than CEA (p < 0.097). Post-intervention systemic therapy was not associated with improved DFS (p = 0.745). (4) Conclusions: ctDNA results are prognostically important in oligometastatic CRC, and further prospective studies are urgently needed to define its role in guiding clinical decisions.
Collapse
Affiliation(s)
- Conor D. J. O’Donnell
- Mayo Clinic School of Graduate Education, Mayo Clinic College of Medicine, Mayo Building, Rochester, MN 55905, USA; (C.D.J.O.)
| | - Nikolas Naleid
- Department of Medicine, University Hospitals of Cleveland, Lakeside Building, 11100 Euclid Avenue, Cleveland, OH 44016, USA
| | - Teerada Siripoon
- Mayo Clinic School of Graduate Education, Mayo Clinic College of Medicine, Mayo Building, Rochester, MN 55905, USA; (C.D.J.O.)
- Division of Medical Oncology, Department of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Kevin G. Zablonski
- Department of Medicine, University Hospitals of Cleveland, Lakeside Building, 11100 Euclid Avenue, Cleveland, OH 44016, USA
| | - Michael H. Storandt
- Mayo Clinic School of Graduate Education, Mayo Clinic College of Medicine, Mayo Building, Rochester, MN 55905, USA; (C.D.J.O.)
| | - Jennifer E. Selfridge
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Madison L. Conces
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Krishan R. Jethwa
- Department of Radiation Oncology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - David L. Bajor
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Cornelius A. Thiels
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Susanne G. Warner
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Patrick P. Starlinger
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Thomas D. Atwell
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Jessica L. Mitchell
- Division of Medical Oncology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Amit Mahipal
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zhaohui Jin
- Division of Medical Oncology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
33
|
Liu Y, Lu XN, Guo HM, Bao C, Zhang J, Jin YN. Development and validation of a circulating tumor DNA-based optimization-prediction model for short-term postoperative recurrence of endometrial cancer. World J Clin Cases 2024; 12:3385-3394. [PMID: 38983398 PMCID: PMC11229938 DOI: 10.12998/wjcc.v12.i18.3385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Endometrial cancer (EC) is a common gynecological malignancy that typically requires prompt surgical intervention; however, the advantage of surgical management is limited by the high postoperative recurrence rates and adverse outcomes. Previous studies have highlighted the prognostic potential of circulating tumor DNA (ctDNA) monitoring for minimal residual disease in patients with EC. AIM To develop and validate an optimized ctDNA-based model for predicting short-term postoperative EC recurrence. METHODS We retrospectively analyzed 294 EC patients treated surgically from 2015-2019 to devise a short-term recurrence prediction model, which was validated on 143 EC patients operated between 2020 and 2021. Prognostic factors were identified using univariate Cox, Lasso, and multivariate Cox regressions. A nomogram was created to predict the 1, 1.5, and 2-year recurrence-free survival (RFS). Model performance was assessed via receiver operating characteristic (ROC), calibration, and decision curve analyses (DCA), leading to a recurrence risk stratification system. RESULTS Based on the regression analysis and the nomogram created, patients with postoperative ctDNA-negativity, postoperative carcinoembryonic antigen 125 (CA125) levels of < 19 U/mL, and grade G1 tumors had improved RFS after surgery. The nomogram's efficacy for recurrence prediction was confirmed through ROC analysis, calibration curves, and DCA methods, highlighting its high accuracy and clinical utility. Furthermore, using the nomogram, the patients were successfully classified into three risk subgroups. CONCLUSION The nomogram accurately predicted RFS after EC surgery at 1, 1.5, and 2 years. This model will help clinicians personalize treatments, stratify risks, and enhance clinical outcomes for patients with EC.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Xiao-Ning Lu
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Hui-Ming Guo
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Chan Bao
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Juan Zhang
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Yu-Ni Jin
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| |
Collapse
|
34
|
Liu M, Bao Q, Zhao T, Huang L, Zhang D, Wang Y, Yan X, Wang H, Jin K, Liu W, Wang K, Xing B. Pre-hepatectomy dynamic circulating tumor DNA to predict pathologic response to preoperative chemotherapy and post-hepatectomy recurrence in patients with colorectal liver metastases. Hepatol Int 2024; 18:1029-1039. [PMID: 38427145 DOI: 10.1007/s12072-023-10628-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/15/2023] [Indexed: 03/02/2024]
Abstract
OBJECTIVE To evaluate the predictive value of pre-hepatectomy dynamic circulating tumor DNA (ctDNA) on pathologic response to preoperative chemotherapy and recurrence after liver resection for colorectal liver metastases (CRLM). BACKGROUND Pathologic response is a predictor of clinical outcomes for patients undergoing hepatectomy for CRLM. Postoperative ctDNA has been proven to be sensitive for recurrence detection. However, few studies investigate the impact of pre-hepatectomy ctDNA on pathologic response and recurrence. METHODS Patients with potential resectable CRLM underwent preoperative chemotherapy and hepatectomy between 2018 and 2021 was considered for inclusion. Plasma ctDNA was collected before and after preoperative chemotherapy. Pathologic response was analyzed for all patients after liver resection. Recurrence free survival was compared between patients with different ctDNA status and different pathologic response. The relation between ctDNA and pathologic response was also analyzed. RESULTS A total of 114 patients were included. ctDNA was detectable in 108 of 114 patients (94.7%) before chemotherapy, in 56 of 114 patients (49.1%) after chemotherapy. Patients with ctDNA positive at baseline and negative after chemotherapy had significantly longer RFS (median RFS 17 vs 7 months, p = 0.001) and HRFS (median HRFS unreached vs 8 months, p < 0.001) than those with ctDNA persistently positive after chemotherapy. Two patients (1.6%) had a pathologic complete response and 56 patients (45.2%) had a pathologic major response. Post-chemotherapy ctDNA- was associated with improved major pathologic response (53.4% vs 32.1%, p = 0.011). In the multivariable analysis, ctDNA- after chemotherapy (HR 0.51, 95% CI 0.28-0.93), major pathologic response (HR 0.34, 95% CI 0.19-0.62) and surgery combined with radiofrequency ablation (HR 2.62, 95% CI 1.38-5.00) were independently associated with RFS (all p < 0.05). CONCLUSIONS Pre-hepatectomy dynamic monitoring of ctDNA could predict pathologic response to preoperative chemotherapy and post-hepatectomy recurrence in CRLM patients. Negative ctDNA after preoperative chemotherapy was associated with better tumor regression grade and recurrence-free survival, which might be used to guide pre-hepatectomy chemotherapy and predict prognosis.
Collapse
Affiliation(s)
- Ming Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Quan Bao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Tingting Zhao
- GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Shanghai, People's Republic of China
| | - Longfei Huang
- GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Shanghai, People's Republic of China
| | - Danhua Zhang
- GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Shanghai, People's Republic of China
| | - Yanyan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Xiaoluan Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Hongwei Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Kemin Jin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Wei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China
| | - Kun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China.
| | - Baocai Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Biliary-Pancreatic Surgery I, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing, People's Republic of China.
| |
Collapse
|
35
|
Li Y, Xu J, Hu X, Chen Y, Liu F, Chen Y, Ma X, Dong Q, Sun L, Mo S, Zhang L, He X, Tong S, Wu H, Li W, Cai S, Zhu S, Pan Q, Peng J. Personalized circulating tumor DNA monitoring improves recurrence surveillance and management after curative resection of colorectal liver metastases: a prospective cohort study. Int J Surg 2024; 110:2776-2787. [PMID: 38445460 DOI: 10.1097/js9.0000000000001236] [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: 10/21/2023] [Accepted: 02/15/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Approximately 60% of patients with colorectal liver metastases (CRLM) experience relapse within 2 years after radical resection, previous studies have proven that repeat local treatment (LT) could prolong survival, however, it is difficult to seize the window for LT due to the lack of a high-sensitive surveillance method. In this study, the authors aim to examine the value of longitudinal circulating tumor DNA (ctDNA) in guiding adjuvant chemotherapy, optimizing clinical surveillance strategy, and thereby improving CRLM outcomes. MATERIALS AND METHODS The authors conducted a prospective clinical trial using a personalized, tumor-informed ctDNA assay to monitor 60 CRLM patients undergoing resection with curative intent. Formalin-fixed paraffin-embedded tumor samples were collected after surgery. Blood samples were collected before surgery, 30 days after surgery (post-OP), and every third month until relapse or up to 2 years. RESULTS A total of 394 plasma samples from 60 eligible patients were analyzed, with a median follow-up time of 31.3 months. Landmark analyses revealed that detectable ctDNA at post-OP (HR, 4.8), postadjuvant chemotherapy (HR, 6.0), and end-of-treatment (HR, 5.6) were associated with higher recurrence risk ( P <0.001). Post-OP ctDNA positivity served as the only independent prognostic marker in the multivariant analysis (HR, 5.1; P <0.001). Longitudinal ctDNA analysis identified relapsed patients at both sensitivity and specificity of 100%. Most (75%) patients were found with radiological relapse within 6 months after the first detectable ctDNA with a median lead time of 3.5 months. In relapsed patients, 73.2% had oligometastatic disease and 61% were liver-restricted, of which 72.0% received repeat LTs, and 60.0% achieved a secondary no evidence of disease status. CONCLUSIONS Longitudinal ctDNA monitoring assists in early prediction of relapse, and thereby improves survival of CRLM patients by increased secondary resection rate and secondary no evidence of disease rate.
Collapse
Affiliation(s)
- Yaqi Li
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Jing Xu
- BGI Genomics, BGI-Shenzhen, Shenzhen, People's Republic of China
| | - Xiang Hu
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Yikuan Chen
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Fangqi Liu
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Yun Chen
- BGI Genomics, BGI-Shenzhen, Shenzhen, People's Republic of China
| | - Xiaoji Ma
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Qiduo Dong
- BGI Genomics, BGI-Shenzhen, Shenzhen, People's Republic of China
| | - Lei Sun
- Tianjin Medical Laboratory BGI, BGI-Tianjin, Tianjin
| | - Shaobo Mo
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Long Zhang
- Department of Colorectal Surgery
- Cancer Institute, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Xingfeng He
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Shanyou Tong
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Huizi Wu
- BGI Genomics, BGI-Shenzhen, Shenzhen, People's Republic of China
| | - Wenhua Li
- Department of Medical Oncology
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Sanjun Cai
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Shida Zhu
- Shenzhen Engineering Laboratory for Innovative Molecular Diagnostics
- BGI Genomics, BGI-Shenzhen, Shenzhen, People's Republic of China
| | - Qi Pan
- Department of Hepatic Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| | - Junjie Peng
- Department of Colorectal Surgery
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai
| |
Collapse
|
36
|
Belmont E, Bansal VV, Yousef MMG, Zeineddine MA, Su D, Dhiman A, Liao CY, Polite B, Eng OS, Fournier KF, White MG, Turaga KK, Shen JP, Shergill A. Multi-Institutional Study Evaluating the Role of Circulating Tumor DNA in the Management of Appendiceal Cancers. JCO Precis Oncol 2024; 8:e2300531. [PMID: 38723230 DOI: 10.1200/po.23.00531] [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: 09/22/2023] [Revised: 02/06/2024] [Accepted: 04/01/2024] [Indexed: 06/09/2024] Open
Abstract
PURPOSE Conventional surveillance methods are poorly sensitive for monitoring appendiceal cancers (AC). This study investigated the utility of circulating tumor DNA (ctDNA) in evaluating systemic therapy response and recurrence after surgery for AC. METHODS Patients from two specialized centers who underwent tumor-informed ctDNA testing (Signatera) were evaluated to determine the association between systemic therapy and ctDNA detection. In addition, the accuracy of ctDNA detection during surveillance for the diagnosis of recurrence after complete cytoreductive surgery (CRS) for grade 2-3 ACs with peritoneal metastases (PM) was investigated. RESULTS In this cohort of 94 patients with AC, most had grade 2-3 tumors (84.0%) and PM (84.0%). Fifty patients completed the assay in the presence of identifiable disease, among which ctDNA was detected in 4 of 7 (57.1%), 10 of 16 (62.5%), and 19 of 27 (70.4%) patients with grade 1, 2, and 3 diseases, respectively. Patients who had recently received systemic chemotherapy had ctDNA detected less frequently (7 of 16 [43.8%] v 26 of 34 [76.5%]; odds ratio, 0.22 [95% CI, 0.06 to 0.82]; P = .02). Among 36 patients with complete CRS for grade 2-3 AC-PM, 16 (44.4%) developed recurrence (median follow-up, 19.6 months). ctDNA detection was associated with shorter recurrence-free survival (median 11.3 months v not reached; hazard ratio, 14.1 [95% CI, 1.7 to 113.8]; P = .01) and showed high accuracy for the detection of recurrence (sensitivity 93.8%, specificity 85.0%). ctDNA was more sensitive than carcinoembryonic antigen (62.5%), CA19-9 (25.0%), and CA125 (18.8%) and was the only elevated biomarker in four (25%) patients with recurrence. CONCLUSION This study revealed a reduced ctDNA detection frequency after systemic therapy and accurate recurrence assessment after CRS. These findings underscore the role of ctDNA as a predictive and prognostic biomarker for grade 2-3 AC-PM management.
Collapse
Affiliation(s)
- Erika Belmont
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL
| | - Varun V Bansal
- Division of Surgical Oncology, Yale School of Medicine, New Haven, CT
| | - Mahmoud M G Yousef
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mohammad A Zeineddine
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - David Su
- Division of Surgical Oncology, Yale School of Medicine, New Haven, CT
| | - Ankit Dhiman
- Department of Surgery, Medical College of Georgia, Augusta, GA
| | - Chih-Yi Liao
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL
| | - Blasé Polite
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL
| | - Oliver S Eng
- Department of Surgery, University of California, Irvine, Orange, CA
| | - Keith F Fournier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael G White
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kiran K Turaga
- Division of Surgical Oncology, Yale School of Medicine, New Haven, CT
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ardaman Shergill
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL
| |
Collapse
|
37
|
Hashimoto T, Nakamura Y, Oki E, Kobayashi S, Yuda J, Shibuki T, Bando H, Yoshino T. Bridging horizons beyond CIRCULATE-Japan: a new paradigm in molecular residual disease detection via whole genome sequencing-based circulating tumor DNA assay. Int J Clin Oncol 2024; 29:495-511. [PMID: 38551727 PMCID: PMC11043144 DOI: 10.1007/s10147-024-02493-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/16/2024] [Indexed: 04/26/2024]
Abstract
Circulating tumor DNA (ctDNA) is the fraction of cell-free DNA in patient blood that originates from a tumor. Advances in DNA sequencing technologies and our understanding of the molecular biology of tumors have increased interest in exploiting ctDNA to facilitate detection of molecular residual disease (MRD). Analysis of ctDNA as a promising MRD biomarker of solid malignancies has a central role in precision medicine initiatives exemplified by our CIRCULATE-Japan project involving patients with resectable colorectal cancer. Notably, the project underscores the prognostic significance of the ctDNA status at 4 weeks post-surgery and its correlation to adjuvant therapy efficacy at interim analysis. This substantiates the hypothesis that MRD is a critical prognostic indicator of relapse in patients with colorectal cancer. Despite remarkable advancements, challenges endure, primarily attributable to the exceedingly low ctDNA concentration in peripheral blood, particularly in scenarios involving low tumor shedding and the intrinsic error rates of current sequencing technologies. These complications necessitate more sensitive and sophisticated assays to verify the clinical utility of MRD across all solid tumors. Whole genome sequencing (WGS)-based tumor-informed MRD assays have recently demonstrated the ability to detect ctDNA in the parts-per-million range. This review delineates the current landscape of MRD assays, highlighting WGS-based approaches as the forefront technique in ctDNA analysis. Additionally, it introduces our upcoming endeavor, WGS-based pan-cancer MRD detection via ctDNA, in our forthcoming project, SCRUM-Japan MONSTAR-SCREEN-3.
Collapse
Affiliation(s)
- Tadayoshi Hashimoto
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shin Kobayashi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Junichiro Yuda
- Department of Hematology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Taro Shibuki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
38
|
Popescu I, Dudău AM, Dima S, Herlea V, Croitoru VM, Dinu IM, Miron M, Lupescu I, Croitoru-Cazacu IM, Dumitru R, Croitoru AE. Multimodal Treatment of Metastatic Rectal Cancer in a Young Patient: Case Report and Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:696. [PMID: 38792879 PMCID: PMC11123219 DOI: 10.3390/medicina60050696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/14/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024]
Abstract
Metastatic colorectal cancer requires a multidisciplinary and individualized approach. Herein, we reported the case of a young woman diagnosed with metastatic rectal cancer who received an individualized multimodal treatment strategy that resulted in a remarkable survival. There were several particular aspects of this case, such as the early onset of the disease, the successful use of conversion therapy, the application of liquid biopsy to guide treatment, and the specific nature of the bone metastasis. To offer more insights for navigating such challenges in patients with metastatic colorectal cancer, we have conducted a literature review to find more data related to the particularities of this case. The incidence of early onset colorectal cancer is on the rise. Data suggests that it differs from older-onset colorectal cancer in terms of its pathological, epidemiological, anatomical, metabolic, and biological characteristics. Conversion therapy and surgical intervention provide an opportunity for cure and improve outcomes in metastatic colorectal cancer. It is important to approach each case individually, as every patient with limited liver disease should be considered as a candidate for secondary resection. Moreover, liquid biopsy has an important role in the individualized management of metastatic colorectal cancer patients, as it offers additional information for treatment decisions.
Collapse
Affiliation(s)
- Ionuț Popescu
- Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania; (I.P.); (V.M.C.)
| | - Ana-Maria Dudău
- Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania; (I.P.); (V.M.C.)
- Medical Oncology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (I.M.D.); (M.M.); (I.M.C.-C.); (A.E.C.)
| | - Simona Dima
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 020021 Bucharest, Romania; (S.D.); (V.H.); (I.L.); (R.D.)
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Vlad Herlea
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 020021 Bucharest, Romania; (S.D.); (V.H.); (I.L.); (R.D.)
- Pathology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Vlad M. Croitoru
- Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania; (I.P.); (V.M.C.)
- Medical Oncology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (I.M.D.); (M.M.); (I.M.C.-C.); (A.E.C.)
| | - Ioana Mihaela Dinu
- Medical Oncology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (I.M.D.); (M.M.); (I.M.C.-C.); (A.E.C.)
| | - Monica Miron
- Medical Oncology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (I.M.D.); (M.M.); (I.M.C.-C.); (A.E.C.)
| | - Ioana Lupescu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 020021 Bucharest, Romania; (S.D.); (V.H.); (I.L.); (R.D.)
- Radiology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Irina M. Croitoru-Cazacu
- Medical Oncology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (I.M.D.); (M.M.); (I.M.C.-C.); (A.E.C.)
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 020021 Bucharest, Romania; (S.D.); (V.H.); (I.L.); (R.D.)
| | - Radu Dumitru
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 020021 Bucharest, Romania; (S.D.); (V.H.); (I.L.); (R.D.)
- Radiology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Adina Emilia Croitoru
- Medical Oncology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (I.M.D.); (M.M.); (I.M.C.-C.); (A.E.C.)
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 020021 Bucharest, Romania; (S.D.); (V.H.); (I.L.); (R.D.)
| |
Collapse
|
39
|
Radomski SN, Ali S, Lafaro KJ, Shubert C, Hidalgo M, Chung H, Christenson ES. The utilization of circulating tumor DNA to predict the risk and location of relapse after curative-intent local therapy in oligometastatic colorectal cancer. J Gastrointest Surg 2024; 28:534-537. [PMID: 38583907 DOI: 10.1016/j.gassur.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 04/09/2024]
Affiliation(s)
- Shannon N Radomski
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Sundas Ali
- Department of Medicine, Ascension Saint Agnes Hospital, Baltimore, Maryland, United States
| | - Kelly J Lafaro
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Christopher Shubert
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Madison Hidalgo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Haniee Chung
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Eric S Christenson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.
| |
Collapse
|
40
|
Northcott J, Bartha G, Harris J, Li C, Navarro FC, Pyke RM, Hong M, Zhang Q, Ma S, Chen TX, Lai J, Udar N, Saldivar JS, Ayash E, Anderson J, Li J, Cui T, Le T, Chow R, Velasco RJ, Mallo C, Santiago R, Bruce RC, Goodman LJ, Chen Y, Norton D, Chen RO, Lyle JM. Analytical validation of NeXT Personal®, an ultra-sensitive personalized circulating tumor DNA assay. Oncotarget 2024; 15:200-218. [PMID: 38484152 PMCID: PMC10939476 DOI: 10.18632/oncotarget.28565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
We describe the analytical validation of NeXT Personal®, an ultra-sensitive, tumor-informed circulating tumor DNA (ctDNA) assay for detecting residual disease, monitoring therapy response, and detecting recurrence in patients diagnosed with solid tumor cancers. NeXT Personal uses whole genome sequencing of tumor and matched normal samples combined with advanced analytics to accurately identify up to ~1,800 somatic variants specific to the patient's tumor. A personalized panel is created, targeting these variants and then used to sequence cell-free DNA extracted from patient plasma samples for ultra-sensitive detection of ctDNA. The NeXT Personal analytical validation is based on panels designed from tumor and matched normal samples from two cell lines, and from 123 patients across nine cancer types. Analytical measurements demonstrated a detection threshold of 1.67 parts per million (PPM) with a limit of detection at 95% (LOD95) of 3.45 PPM. NeXT Personal showed linearity over a range of 0.8 to 300,000 PPM (Pearson correlation coefficient = 0.9998). Precision varied from a coefficient of variation of 12.8% to 3.6% over a range of 25 to 25,000 PPM. The assay targets 99.9% specificity, with this validation study measuring 100% specificity and in silico methods giving us a confidence interval of 99.92 to 100%. In summary, this study demonstrates NeXT Personal as an ultra-sensitive, highly quantitative and robust ctDNA assay that can be used to detect residual disease, monitor treatment response, and detect recurrence in patients.
Collapse
Affiliation(s)
| | | | | | - Conan Li
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | | | | - Qi Zhang
- Personalis, Inc., Fremont, CA 94555, USA
| | - Shuyuan Ma
- Personalis, Inc., Fremont, CA 94555, USA
| | | | - Janet Lai
- Personalis, Inc., Fremont, CA 94555, USA
| | - Nitin Udar
- Personalis, Inc., Fremont, CA 94555, USA
| | | | - Erin Ayash
- Personalis, Inc., Fremont, CA 94555, USA
| | | | - Jiang Li
- Personalis, Inc., Fremont, CA 94555, USA
| | - Tiange Cui
- Personalis, Inc., Fremont, CA 94555, USA
| | - Tu Le
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | | | | | | | | | | - Yi Chen
- Personalis, Inc., Fremont, CA 94555, USA
| | - Dan Norton
- Personalis, Inc., Fremont, CA 94555, USA
| | | | - John M. Lyle
- Personalis, Inc., Fremont, CA 94555, USA
- Co-last authors
| |
Collapse
|
41
|
Baumgartner JM, Botta GP. Role of Circulating Tumor DNA Among Patients with Colorectal Peritoneal Metastases. J Gastrointest Cancer 2024; 55:41-46. [PMID: 37436640 PMCID: PMC11096195 DOI: 10.1007/s12029-023-00959-8] [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] [Accepted: 07/01/2023] [Indexed: 07/13/2023]
Abstract
PURPOSE This was a review of circulating tumor DNA (ctDNA) in patients with peritoneal metastases from colorectal cancer. METHODS We searched the PubMed database for studies reporting detection of ctDNA in patients with colorectal cancer (CRC) and with peritoneal metastases (PM) from colorectal cancer (CRPM). We extracted data on the population included, number of subjects, study design, type of ctDNA assay used and schedule, and the major findings from these publications. RESULTS We identified 13 studies for review investigating ctDNA, using a variety of ctDNA assays, among 1787 patients with CRC without PM, as well as four eligible published and one unpublished (in press) studies, which included 255 patients with PM from any primary site and 61 patients with CRPM. Among the 13 studies investigating ctDNA among CRC without PM, posttreatment surveillance ctDNA was associated with recurrence and was generally more sensitive than imaging or tumor markers. Among the five studies including patients with PM, ctDNA was not universally able to detect the presence of PM, but when present, ctDNA predicted worse outcomes. CONCLUSION Circulating-tumor DNA is a potentially useful surveillance tool for patients with CRC. However, the sensitivity of ctDNA to detect CRPM is variable and warrants further inquiry.
Collapse
Affiliation(s)
- Joel M Baumgartner
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, USA.
| | - Gregory P Botta
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
42
|
Chen ZG, Ji XM, Xu YX, Fong WP, Liu XY, Liang JY, Tan Q, Wen L, Cai YY, Wang DS, Li YH. Methylated ctDNA predicts early recurrence risk in patients undergoing resection of initially unresectable colorectal cancer liver metastases. Ther Adv Med Oncol 2024; 16:17588359241230752. [PMID: 38425989 PMCID: PMC10903215 DOI: 10.1177/17588359241230752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
Background Patients with initially unresectable colorectal cancer liver metastases (IU-CRLM) might benefit from using an effective systemic treatment followed by resection of liver metastases but the curative success rate is quite low. Indeed, nearly one-third of patients exhibit early recurrence within the first 6 months after surgery, and these individuals often have poor overall survival. Objectives This study aims to clarify the application value of serial circulating tumor DNA (ctDNA) analysis in predicting the clinical outcome of IU-CRLM patients following liver metastasectomy. Design A retrospective study was conducted on a cohort of patients with IU-CRLM between February 2018 and April 2021. Methods Plasma samples at different time points during CRLM treatment [baseline (BL), preoperation (PRE), postoperation (POST), end-of-treatment (EOT), and progressive disease (PD)] were retrospectively collected from patients with initially unresectable CRLM enrolled at the Sun Yat-sen University Cancer Center. Dynamic changes of SEPTIN 9 (SEPT9) and Neuropeptide Y (NPY) methylated circulating tumor DNA (MetctDNA) levels in serial plasma samples were detected using droplet-digital PCR (ddPCR). Results SEPT9 and NPY genes were hypermethylated in colon cancer cell lines and tissues while no difference was observed between primary and metastatic tumors. Patients with MetctDNA positive at POST or EOT had significantly lower recurrence-free survival (RFS) compared to patients with MetctDNA negative at these time points [POST: Hazard ratio (HR) 9.44, 95% confidence interval (CI) 5.15-17.30, p < 0.001; EOT: HR 11.48, 95% CI 3.27-40.31, p < 0.001]. Multivariate analysis demonstrated that POST (OR 33.96, 95% CI 4.03-286.10, p = 0.001) and EOT (OR 18.36, 95% CI 1.14-295.71, p = 0.04) MetctDNA was an independent risk factor for early recurrence. Time-dependent receiver operating characteristic curve (T-ROC) analysis revealed that area under the curve (AUC) value was greatest at the relapse time point of 6 months post-intervention, with POST-AUC and EOT-AUC values of 0.74 (95% CI 0.66-0.81) and 0.73 (95% CI 0.53-0.94), respectively. Serial MetctDNA analysis showed that RFS was significantly lower in patients with no MetctDNA clearance compared with those with MetctDNA clearance (HR 26.05, 95% CI 4.92-137.81, p < 0.001). Conclusion Our study confirmed that serial ctDNA analysis of NPY and SEPT9 gene methylation could effectively predict early recurrence in IU-CRLM patients, especially at POST and EOT.
Collapse
Affiliation(s)
- Zhi-Gang Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - Xiao-Meng Ji
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, P.R. China
| | - Yu-Xia Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Molecular Diagnostics Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - William Pat Fong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - Xiao-Yun Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Molecular Diagnostics Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - Jie-Ying Liang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - Qiong Tan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - Lei Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - Yan-Yu Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - De-Shen Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, P.R. China
| | - Yu-Hong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, P.R. China
| |
Collapse
|
43
|
Sandberg E, Nunes L, Edqvist PH, Mathot L, Chen L, Edgren T, Al Nassralla S, Glimelius B, Landegren U, Sjöblom T. Sensitive and Specific Analyses of Colorectal Cancer Recurrence through Multiplex superRCA Mutation Detection in Blood Plasma. Cancers (Basel) 2024; 16:549. [PMID: 38339300 PMCID: PMC10854605 DOI: 10.3390/cancers16030549] [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: 12/22/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Mutation analysis of circulating tumor DNA (ctDNA) has applications in monitoring of colorectal cancer (CRC) patients for recurrence. Considering the low tumor fraction of ctDNA in cell-free DNA (cfDNA) isolated from blood plasma, the sensitivity of the detection method is important. Here, plasma DNA collected at diagnosis and follow-up from 25 CRC patients was analyzed using a multiplex superRCA mutation detection assay. The assay was also performed on genomic DNA (gDNA) from tumor and normal tissue from 20 of these patients. The lower limit of detection for most sequence variants was in the range of 10-5, while when analyzing cfDNA from plasma with a typical input of 33 ng, the practical detection limit was ~10-4 or 0.01% mutant allele frequency (MAF). In 17 of 19 patients with identified hotspot mutations in tumor gDNA, at least one hotspot mutation could be detected in plasma DNA at the time of diagnosis. The MAF increased at subsequent time points in four of the patients who experienced a clinical relapse. Multiplex superRCA analysis of the remaining six patients did not reveal any hotspot mutations. In conclusion, multiplex superRCA assays proved suitable for monitoring CRC patients by analyzing hotspot mutations in cfDNA, and dynamic changes in MAF were observed in patients with clinical relapse.
Collapse
Affiliation(s)
- Emma Sandberg
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (E.S.); (L.N.); (P.-H.E.); (L.M.); (L.C.); (S.A.N.); (B.G.)
| | - Luís Nunes
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (E.S.); (L.N.); (P.-H.E.); (L.M.); (L.C.); (S.A.N.); (B.G.)
| | - Per-Henrik Edqvist
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (E.S.); (L.N.); (P.-H.E.); (L.M.); (L.C.); (S.A.N.); (B.G.)
| | - Lucy Mathot
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (E.S.); (L.N.); (P.-H.E.); (L.M.); (L.C.); (S.A.N.); (B.G.)
| | - Lei Chen
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (E.S.); (L.N.); (P.-H.E.); (L.M.); (L.C.); (S.A.N.); (B.G.)
- Rarity Bioscience AB, SE-752 37 Uppsala, Sweden;
| | - Tomas Edgren
- Rarity Bioscience AB, SE-752 37 Uppsala, Sweden;
| | - Shahed Al Nassralla
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (E.S.); (L.N.); (P.-H.E.); (L.M.); (L.C.); (S.A.N.); (B.G.)
| | - Bengt Glimelius
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (E.S.); (L.N.); (P.-H.E.); (L.M.); (L.C.); (S.A.N.); (B.G.)
| | - Ulf Landegren
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (E.S.); (L.N.); (P.-H.E.); (L.M.); (L.C.); (S.A.N.); (B.G.)
| | - Tobias Sjöblom
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (E.S.); (L.N.); (P.-H.E.); (L.M.); (L.C.); (S.A.N.); (B.G.)
| |
Collapse
|
44
|
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
|
45
|
Yuan H, Xu F, Wang S, Liu D, Zhang H, Zhang J, Shi M, Yan C, Zhu Z. Analysis of circulating tumor DNA identifies distinct therapeutic response to intraperitoneal and intravenous paclitaxel plus S-1 in gastric cancer patients with peritoneal metastasis. Ther Adv Med Oncol 2024; 16:17588359231225038. [PMID: 38249327 PMCID: PMC10799602 DOI: 10.1177/17588359231225038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) is a promising technique for predicting curative effects and monitoring tumor recurrence. The utility of ctDNA for gastric cancer with peritoneal dissemination remains elusive. OBJECTIVES To assess the feasibility of ctDNA in predicting tumor response to chemotherapy in gastric cancer with peritoneal dissemination. DESIGN This was a prospective study. METHODS We enrolled 30 patients with gastric cancer peritoneal metastasis, treated with intraperitoneal and intravenous paclitaxel plus S-1. Peripheral blood samples of patients were prospectively collected at baseline, after treatment initiation accompanied by computed tomography scan and disease progression. Mutational profiles from ctDNA were analyzed to evaluate its association with chemotherapeutic response. RESULTS Tumor protein 53 (TP53) was the most frequently altered gene at baseline blood samples. Although baseline TP53 mutation was not related to therapeutic response, patients with TP53 mutation had worse progression-free survival (PFS) and overall survival (OS). Additionally, baseline ctDNA content fraction (CCF) was found to be significantly lower in responders than non-responders. Meanwhile, patients with high CCF had a trend of worse PFS and OS. Combining TP53 alteration and CCF, the prognosis of TP53-wt patients could be further stratified. Patients with CCF-low_TP53-wt had markedly longer survival than those with CCF-high_TP53-wt. CONCLUSION Our study highlighted the significance of ctDNA in predicting potential clinical outcomes in gastric cancer patients during chemotherapy. TRIAL REGISTRATION ChiCTR-IIR-16009802 (Chinese Clinical Trial Registry).
Collapse
Affiliation(s)
- Hong Yuan
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Xu
- Genecast Biotechnology Co. Ltd, Wuxi, China
| | | | - Di Liu
- Genecast Biotechnology Co. Ltd, Wuxi, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Wuxi, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai 200025, China
- Shanghai Hospital of Civil Aviation Administration of China, Shanghai, China
- Department of Oncology, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Wuxi, China
| | - Chao Yan
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai 200025, China
| | - Zhenggang Zhu
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Ajithkumar P, Vasantharajan SS, Pattison S, McCall JL, Rodger EJ, Chatterjee A. Exploring Potential Epigenetic Biomarkers for Colorectal Cancer Metastasis. Int J Mol Sci 2024; 25:874. [PMID: 38255946 PMCID: PMC10815915 DOI: 10.3390/ijms25020874] [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: 11/30/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Metastatic progression is a complex, multistep process and the leading cause of cancer mortality. There is growing evidence that emphasises the significance of epigenetic modification, specifically DNA methylation and histone modifications, in influencing colorectal (CRC) metastasis. Epigenetic modifications influence the expression of genes involved in various cellular processes, including the pathways associated with metastasis. These modifications could contribute to metastatic progression by enhancing oncogenes and silencing tumour suppressor genes. Moreover, specific epigenetic alterations enable cancer cells to acquire invasive and metastatic characteristics by altering cell adhesion, migration, and invasion-related pathways. Exploring the involvement of DNA methylation and histone modification is crucial for identifying biomarkers that impact cancer prediction for metastasis in CRC. This review provides a summary of the potential epigenetic biomarkers associated with metastasis in CRC, particularly DNA methylation and histone modifications, and examines the pathways associated with these biomarkers.
Collapse
Affiliation(s)
- Priyadarshana Ajithkumar
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (P.A.)
| | - Sai Shyam Vasantharajan
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (P.A.)
| | - Sharon Pattison
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - John L. McCall
- Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Euan J. Rodger
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (P.A.)
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (P.A.)
- School of Health Sciences and Technology, UPES University, Dehradun 248007, India
| |
Collapse
|
47
|
Yang L, Yang J, Kleppe A, Danielsen HE, Kerr DJ. Personalizing adjuvant therapy for patients with colorectal cancer. Nat Rev Clin Oncol 2024; 21:67-79. [PMID: 38001356 DOI: 10.1038/s41571-023-00834-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2023] [Indexed: 11/26/2023]
Abstract
The current standard-of-care adjuvant treatment for patients with colorectal cancer (CRC) comprises a fluoropyrimidine (5-fluorouracil or capecitabine) as a single agent or in combination with oxaliplatin, for either 3 or 6 months. Selection of therapy depends on conventional histopathological staging procedures, which constitute a blunt tool for patient stratification. Given the relatively marginal survival benefits that patients can derive from adjuvant treatment, improving the safety of chemotherapy regimens and identifying patients most likely to benefit from them is an area of unmet need. Patient stratification should enable distinguishing those at low risk of recurrence and a high chance of cure by surgery from those at higher risk of recurrence who would derive greater absolute benefits from chemotherapy. To this end, genetic analyses have led to the discovery of germline determinants of toxicity from fluoropyrimidines, the identification of patients at high risk of life-threatening toxicity, and enabling dose modulation to improve safety. Thus far, results from analyses of resected tissue to identify mutational or transcriptomic signatures with value as prognostic biomarkers have been rather disappointing. In the past few years, the application of artificial intelligence-driven models to digital images of resected tissue has identified potentially useful algorithms that stratify patients into distinct prognostic groups. Similarly, liquid biopsy approaches involving measurements of circulating tumour DNA after surgery are additionally useful tools to identify patients at high and low risk of tumour recurrence. In this Perspective, we provide an overview of the current landscape of adjuvant therapy for patients with CRC and discuss how new technologies will enable better personalization of therapy in this setting.
Collapse
Affiliation(s)
- Li Yang
- Department of Gastroenterology, Sichuan University, Chengdu, China
| | - Jinlin Yang
- Department of Gastroenterology, Sichuan University, Chengdu, China
| | - Andreas Kleppe
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
- Centre for Research-based Innovation Visual Intelligence, UiT The Arctic University of Norway, Tromsø, Norway
| | - Håvard E Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Radcliffe Department of Medicine, Oxford University, Oxford, UK
| | - David J Kerr
- Radcliffe Department of Medicine, Oxford University, Oxford, UK.
| |
Collapse
|
48
|
Zhu M, Taylor WR, Mahoney DW, Then SS, Berger CK, Burger KN, Gonser AM, Doering KA, Xie H, Foote PH, Kaiser MW, Allawi HT, Hubbard JM, Kisiel JB. Plasma Assay of Cell-Free Methylated DNA Markers of Colorectal Cancer: A Tumor-Agnostic Approach to Monitor Recurrence and Response to Anticancer Therapies. Cancers (Basel) 2023; 15:5778. [PMID: 38136324 PMCID: PMC10742045 DOI: 10.3390/cancers15245778] [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: 11/11/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Radiographic surveillance of colorectal cancer (CRC) after curative-intent therapy is costly and unreliable. Methylated DNA markers (MDMs) detected primary CRC and metastatic recurrence with high sensitivity and specificity in cross-sectional studies. This study evaluated using serial MDMs to detect recurrence and monitor the treatment response to anti-cancer therapies. METHODS A nested case-control study was drawn from a prospective cohort of patients with CRC who completed curative-intent therapy for CRC of all stages. Plasma MDMs were assayed vis target enrichment long-probe quantitative-amplified signal assays, normalized to B3GALT6, and analyzed in combination with serum carcinoembryonic antigen to yield an MDM score. Clinical information, including treatment and radiographic measurements of the tumor burden, were longitudinally collected. RESULTS Of the 35 patients, 18 had recurrence and 17 had no evidence of disease during the study period. The MDM score was positive in 16 out of 18 patients who recurred and only 2 of the 17 patients without recurrence. The MDM score detected recurrence in 12 patients preceding clinical or radiographic detection of recurrent CRC by a median of 106 days (range 90-232 days). CONCLUSIONS Plasma MDMs can detect recurrent CRC prior to radiographic detection; this tumor-agnostic liquid biopsy approach may assist cancer surveillance and monitoring.
Collapse
Affiliation(s)
- Mojun Zhu
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA (H.X.)
| | - William R. Taylor
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA (A.M.G.); (K.A.D.); (P.H.F.)
| | - Douglas W. Mahoney
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN 55905, USA
| | - Sara S. Then
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA (A.M.G.); (K.A.D.); (P.H.F.)
| | - Calise K. Berger
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA (A.M.G.); (K.A.D.); (P.H.F.)
| | - Kelli N. Burger
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN 55905, USA
| | - Anna M. Gonser
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA (A.M.G.); (K.A.D.); (P.H.F.)
| | - Karen A. Doering
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA (A.M.G.); (K.A.D.); (P.H.F.)
| | - Hao Xie
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA (H.X.)
| | - Patrick H. Foote
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA (A.M.G.); (K.A.D.); (P.H.F.)
| | | | - Hatim T. Allawi
- Exact Sciences Corporation, Madison, WI 53719, USA; (M.W.K.)
| | | | - John B. Kisiel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA (A.M.G.); (K.A.D.); (P.H.F.)
| |
Collapse
|
49
|
Zavarykina TM, Lomskova PK, Pronina IV, Khokhlova SV, Stenina MB, Sukhikh GT. Circulating Tumor DNA Is a Variant of Liquid Biopsy with Predictive and Prognostic Clinical Value in Breast Cancer Patients. Int J Mol Sci 2023; 24:17073. [PMID: 38069396 PMCID: PMC10706922 DOI: 10.3390/ijms242317073] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
This paper introduces the reader to the field of liquid biopsies and cell-free nucleic acids, focusing on circulating tumor DNA (ctDNA) in breast cancer (BC). BC is the most common type of cancer in women, and progress with regard to treatment has been made in recent years. Despite this, there remain a number of unresolved issues in the treatment of BC; in particular, early detection and diagnosis, reliable markers of response to treatment and for the prediction of recurrence and metastasis, especially for unfavorable subtypes, are needed. It is also important to identify biomarkers for the assessment of drug resistance and for disease monitoring. Our work is devoted to ctDNA, which may be such a marker. Here, we describe its main characteristics and potential applications in clinical oncology. This review considers the results of studies devoted to the analysis of the prognostic and predictive roles of various methods for the determination of ctDNA in BC patients. Currently known epigenetic changes in ctDNA with clinical significance are reviewed. The possibility of using ctDNA as a predictive and prognostic marker for monitoring BC and predicting the recurrence and metastasis of cancer is also discussed, which may become an important part of a precision approach to the treatment of BC.
Collapse
Affiliation(s)
- Tatiana M. Zavarykina
- N.M. Emanuel Institute of Biochemical Physics of the Russian Academy of Sciences, Moscow 119334, Russia;
- “B.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology of Ministry of Health of the Russian Federation, Moscow 117997, Russia; (S.V.K.); (G.T.S.)
| | - Polina K. Lomskova
- N.M. Emanuel Institute of Biochemical Physics of the Russian Academy of Sciences, Moscow 119334, Russia;
| | - Irina V. Pronina
- Institute of General Pathology and Pathophysiology, Moscow 125315, Russia;
| | - Svetlana V. Khokhlova
- “B.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology of Ministry of Health of the Russian Federation, Moscow 117997, Russia; (S.V.K.); (G.T.S.)
| | - Marina B. Stenina
- “N.N. Blokhin National Medical Research Center of Oncology of Ministry of Health of the Russian Federation, Moscow 115522, Russia;
| | - Gennady T. Sukhikh
- “B.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology of Ministry of Health of the Russian Federation, Moscow 117997, Russia; (S.V.K.); (G.T.S.)
| |
Collapse
|
50
|
Callesen LB, Hansen TF, Andersen RF, Pallisgaard N, Kramer S, Schlander S, Rafaelsen SR, Boysen AK, Jensen LH, Jakobsen A, Spindler KLG. ctDNA-guided adjuvant treatment after radical-intent treatment of metastatic spread from colorectal cancer-the first interim results from the OPTIMISE study. Acta Oncol 2023; 62:1742-1748. [PMID: 37738268 DOI: 10.1080/0284186x.2023.2259083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/10/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND Patients with detectable ctDNA after radical-intent treatment of metastatic spread from colorectal cancer (mCRC) have a very high risk of recurrence, which may be prevented with intensified adjuvant chemotherapy (aCTh). In the OPTIMISE study, we investigate ctDNA-guided aCTh after radical-intent treatment of mCRC. Here we present results from the preplanned interim analysis. MATERIAL AND METHODS The study is an open-label 1:1 randomized clinical trial comparing ctDNA-guided aCTh against standard of care (SOC), with a run-in phase investigating feasibility measures. Key inclusion criteria; radical-intent treatment for mCRC and clinically eligible for triple-agent chemotherapy. Patients underwent a PET-CT scan before randomization. ctDNA analyses of plasma samples were done by ddPCR, detecting CRC-specific mutations and methylation of the NPY gene. In the ctDNA-guided arm, ctDNA positivity led to an escalation strategy with triple-agent chemotherapy, and conversely ctDNA negativity led to a de-escalation strategy by shared-decision making. Patients randomized to the standard arm were treated according to SOC. Feasibility measures for the run-in phase were; the inclusion of 30 patients over 12 months in two Danish hospitals, compliance with randomization >80%, rate of PET-CT-positive findings <20%, and eligibility for triple-agent chemotherapy >80%. RESULTS Thirty-two patients were included. The rate of PET-CT-positive cases was 22% (n = 7/32). Ninety-seven percent of the patients were randomized. Fourteen patients were randomly assigned to SOC and sixteen to ctDNA-guided adjuvant treatment and follow-up. All analyses of baseline plasma samples in the ctDNA-guided arm passed the quality control, and 19% were ctDNA positive. The median time to result was three working days. All ctDNA-positive patients were eligible for triple-agent chemotherapy. CONCLUSION The study was proven to be feasible and continues in the planned large-scale phase II trial. Results from the OPTIMISE study will potentially optimize the adjuvant treatment of patients undergoing radical-intent treatment of mCRC, thereby improving survival and reducing chemotherapy-related toxicity.
Collapse
Affiliation(s)
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Rikke Fredslund Andersen
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Niels Pallisgaard
- Department of Pathology, Zealand University Hospital, Næstved, Denmark
| | - Stine Kramer
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Sven Schlander
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Rafael Rafaelsen
- Department of Radiology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | | | - Lars Henrik Jensen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Anders Jakobsen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | | |
Collapse
|