1
|
Budhbaware T, Rathored J, Shende S. Molecular methods in cancer diagnostics: a short review. Ann Med 2024; 56:2353893. [PMID: 38753424 PMCID: PMC11100444 DOI: 10.1080/07853890.2024.2353893] [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: 02/14/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND One of the ailments with the greatest fatality rates in the 21st century is cancer. Globally, molecular methods are widely employed to treat cancer-related disorders, and the body of research on this subject is growing yearly. A thorough and critical summary of the data supporting molecular methods for illnesses linked to cancer is required. OBJECTIVE In order to guide clinical practice and future research, it is important to examine and summarize the systematic reviews (SRs) that evaluate the efficacy and safety of molecular methods for disorders associated to cancer. METHODS We developed a comprehensive search strategy to find relevant articles from electronic databases like PubMed, Google Scholar, Web of Science (WoS), or Scopus. We looked through the literature and determined which diagnostic methods in cancer genetics were particularly reliable. We used phrases like 'cancer genetics', genetic susceptibility, Hereditary cancer, cancer risk assessment, 'cancer diagnostic tools', cancer screening', biomarkers, and molecular diagnostics, reviews and meta-analyses evaluating the efficacy and safety of molecular therapies for cancer-related disorders. Research that only consider treatment modalities that don't necessitate genetic or molecular diagnostics fall under the exclusion criteria. RESULTS The results of this comprehensive review clearly demonstrate the transformative impact of molecular methods in the realm of cancer genetics.This review underscores how these technologies have empowered researchers and clinicians to identify and understand key genetic alterations that drive malignancy, ranging from point mutations to structural variations. Such insights are instrumental in pinpointing critical oncogenic drivers and potential therapeutic targets, thus opening the door for methods in precision medicine that can significantly improve patient outcomes. LIMITATION The search does not specify a timeframe for publication inclusion, it may have missed recent advancements or changes in the field's landscape of molecular methods for cancer. As a result, it may not have included the most recent developments in the field. CONCLUSION After conducting an in-depth study on the molecular methods in cancer genetics, it is evident that these cutting-edge technologies have revolutionized the field of oncology, providing researchers and clinicians with powerful tools to unravel the complexities of cancer at the genetic level. The integration of molecular methods techniques has not only enhanced our understanding of cancer etiology, progression, and treatment response but has also opened new avenues for personalized medicine and targeted therapies, leading to improved patient outcomes.
Collapse
Affiliation(s)
- Tanushree Budhbaware
- Department of ‘School of Allied Health Sciences’, Central Research Laboratory (CRL) and Molecular Diagnostics, Datta Meghe Institute of Higher Education and Research, Sawangi (Meghe), Wardha, India
| | - Jaishriram Rathored
- Department of ‘School of Allied Health Sciences’, Central Research Laboratory (CRL) and Molecular Diagnostics, Datta Meghe Institute of Higher Education and Research, Sawangi (Meghe), Wardha, India
| | - Sandesh Shende
- Department of ‘School of Allied Health Sciences’, Central Research Laboratory (CRL) and Molecular Diagnostics, Datta Meghe Institute of Higher Education and Research, Sawangi (Meghe), Wardha, India
| |
Collapse
|
2
|
Cui S, Wang N, Liang Y, Meng Y, Shu X, Kong F. Advances in clinical trials on perioperative immune checkpoint inhibitors for resectable non-small cell lung cancer: A comprehensive review. Int Immunopharmacol 2024; 141:112903. [PMID: 39146783 DOI: 10.1016/j.intimp.2024.112903] [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: 05/13/2024] [Revised: 07/12/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
The reduction in lung cancer mortality rates over the past decade can be partially ascribed to advancements in immunotherapy. Immune checkpoint inhibitors (ICIs) have transformed the therapeutic landscape for advanced non-small cell lung cancer (NSCLC) and have recently been evaluated in multiple clinical trials to confirm their safety and efficacy in the neoadjuvant, adjuvant and perioperative settings for patients with resectable NSCLC. The Food and Drug Administration (FDA) has granted approval for adjuvant atezolizumab following platinum-doublet chemotherapy, neoadjuvant nivolumab and platinum-doublet chemotherapy, adjuvant pembrolizumab after platinum-doublet chemotherapy, and neoadjuvant/adjuvant pembrolizumab for resectable NSCLC, with potential forthcoming approvals for additional agents or indications. Novel data, approvals, and emerging research findings are dramatically shifting the accepted standards of care over just a few years. Despite these advances, the optimal application of these treatments is not entirely straightforward. This article summarizes the biological rationale for immunotherapy and the important clinical trials regarding perioperative ICIs. We also further outline the controversies and future directions to better guide the individualized treatment of NSCLC patients.
Collapse
Affiliation(s)
- Siyuan Cui
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road No.88, Xiqing District, Tianjin 300381, China; Tianjin Cancer Institute of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Na Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road No.88, Xiqing District, Tianjin 300381, China; Tianjin Cancer Institute of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Yangyueying Liang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road No.88, Xiqing District, Tianjin 300381, China; Tianjin Cancer Institute of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Yuan Meng
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road No.88, Xiqing District, Tianjin 300381, China; Tianjin Cancer Institute of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Xinyi Shu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road No.88, Xiqing District, Tianjin 300381, China; Tianjin Cancer Institute of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Fanming Kong
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Changling Road No.88, Xiqing District, Tianjin 300381, China; Tianjin Cancer Institute of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China.
| |
Collapse
|
3
|
Clasen K, Gani C, Schuetz L, Clasen S, Ballin N, Bonzheim I, Orth M, Ossowski S, Riess O, Niyazi M, Schroeder C, Kelemen O. Dynamics of cell-free tumor DNA correlate with early MRI response during chemoradiotherapy in rectal cancer. Radiat Oncol 2024; 19:153. [PMID: 39506775 PMCID: PMC11539469 DOI: 10.1186/s13014-024-02540-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND In locally advanced rectal cancer, the prediction of tumor response during and after neoadjuvant treatment remains challenging. In terms of organ preservation, adaptive radiotherapy, and intensified (total) neoadjuvant therapies, biomarkers are desirable for patient stratification. METHODS In 16 patients, weekly blood samples (n = 86) to detect cell-free tumor DNA (ctDNA) during long-course neoadjuvant chemoradiotherapy were analyzed. Data were correlated with initial tumor volumes, MRI response in week 2 and 5 of radiotherapy as well as with pathologic tumor response after resection and outcome parameters. RESULTS Most patients showed decreasing ctDNA during the course of radiochemotherapy. However, we found heterogenous dynamics of ctDNA and could identify three groups: (1) decline (2) no clear decline and/or late shedding (3) persistence of ctDNA. In seven patients we could detect significant amounts of ctDNA in week 5 or week 6 of treatment. In our pilot cohort, we did not find significant correlations of ctDNA dynamics with pathologic response or outcome parameters. However, patients with distinct decline of ctDNA had larger tumor volumes prior to treatment, and MRI imaging in week 2 and 5 revealed bigger absolute decrease of tumor volumes. If significant levels of ctDNA were found in week 5 and / or 6, patients showed less absolute tumor volume decrease in week 2 and 5. CONCLUSIONS Weekly measurement of ctDNA during radiochemotherapy is feasible and might represent a promising biomarker. Bigger initial primary tumors showed different ctDNA shedding profiles compared with smaller primary tumors and correlations of ctDNA dynamics with early imaging response were found.
Collapse
Affiliation(s)
- Kerstin Clasen
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany.
| | - Cihan Gani
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Leon Schuetz
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Stephan Clasen
- Department of Diagnostic and Interventional Radiology, District Hospital Reutlingen, Reutlingen, Germany
| | - Nadja Ballin
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Irina Bonzheim
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Michael Orth
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Stephan Ossowski
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Christopher Schroeder
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Olga Kelemen
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
4
|
Samaille T, Falcoz A, Cohen R, Laurent-Puig P, André T, Taieb J, Auclin E, Vernerey D. A novel risk classification model integrating CEA, ctDNA, and pTN stage for stage 3 colon cancer: a post hoc analysis of the IDEA-France trial. Oncologist 2024; 29:e1492-e1500. [PMID: 39011625 PMCID: PMC11546770 DOI: 10.1093/oncolo/oyae140] [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: 02/02/2024] [Accepted: 05/10/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND We assessed the added value of incorporating carcinoembryonic antigen (CEA) to circulating tumor DNA (ctDNA) and pathological TN (pTN) stage for risk classification in stage 3 colon cancer (CC). PATIENTS AND METHODS We retrospectively analyzed postoperative CEA values in patients with CC from the IDEA-France phase 3 trial. The relation between disease-free survival (DFS) and CEA was modeled through restricted cubic splines. Prognostic value of CEA, ctDNA, and pTN was assessed with the Kaplan-Meier method. Multivariate analysis was used to identify prognostic and predictive factors for DFS. RESULTS Among 696 patients (35%), CEA values were retrievable, and for 405 (20%) both CEA and ctDNA were available. An optimized CEA threshold of 2 ng/mL was identified, the 3-year DFS was 66.4% for patients above the threshold and 80.9% for those below (HR, 1.74; 95% CI, 1.33-2.28, P < .001). In multivariate analysis, CEA ≥ 2 ng/mL contributed significantly to model variability, becoming an independent prognostic factor for DFS (HR, 1.82; 95% CI,1.27-2.59), alongside ctDNA (HR, 1.88; 95% CI, 1.16-3.03) and pTN (HR, 1.78; 95% CI, 1.24-2.54). A novel integrated risk classification combining CEA, ctDNA, and pTN stage reclassified 19.8% of pT4/N2 patients as low risk and 2.5% of pT3/N1 patients as high risk. This new classification demonstrated the 3-year DFS of 80.8% for low-risk patients and 55.4% for high-risk patients (HR, 2.66, 95% CI, 1.84-3.86, P < .001). CONCLUSIONS Postoperative CEA value is a prognostic factor for DFS in stage 3 CC, independently of ctDNA and pTN. It advocates for systematic reporting in future adjuvant trials. Integrating both biomarkers with pTN could refine risk classification in stage 3 CC.
Collapse
Affiliation(s)
- Thomas Samaille
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne Université, Paris, France
| | - Antoine Falcoz
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France
| | - Romain Cohen
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne Université, Paris, France
| | - Pierre Laurent-Puig
- Institut du cancer Paris CARPEM, Georges Pompidou European Hospital, AP-HP, Université Paris Cité, Paris, France
| | - Thierry André
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne Université, Paris, France
| | - Julien Taieb
- Department of Gastroenterology and GI oncology, Georges Pompidou European Hospital, SIRIC CARPE, Université Paris-Cité, Paris, France
| | - Edouard Auclin
- Department of Medical Oncology, Georges Pompidou European Hospital, AP-HP, Université Paris Cité, Paris, France
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France
| |
Collapse
|
5
|
Kajiwara Y, Ueno H. Essential updates 2022-2023: Surgical and adjuvant therapies for locally advanced colorectal cancer. Ann Gastroenterol Surg 2024; 8:977-986. [PMID: 39502729 PMCID: PMC11533030 DOI: 10.1002/ags3.12853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 11/08/2024] Open
Abstract
Pivotal articles that had been published between 2022 and 2023 on surgical and perioperative adjuvant treatments for locally advanced colorectal cancer (CRC) were reviewed. This review focuses on new evidence in the following areas: optimization of surgical procedures for colon cancer, including the optimal length of bowel resection and use of the no-touch isolation technique; minimally invasive surgery for rectal cancer, such as laparoscopic transanal total mesorectal excision and robotic surgery; neoadjuvant treatments for rectal cancer, including total neoadjuvant therapy; neoadjuvant chemotherapy for colon cancer; and postoperative adjuvant chemotherapy for Stage II and III colon cancer. Although the current understanding may not enable perfect decision-making for patients and medical professionals, ongoing advancements are expected to result in more effective personalized treatment plans, ultimately improving the prognosis and quality of life of patients.
Collapse
Affiliation(s)
- Yoshiki Kajiwara
- Department of SurgeryNational Defense Medical CollegeTokorozawaJapan
| | - Hideki Ueno
- Department of SurgeryNational Defense Medical CollegeTokorozawaJapan
| |
Collapse
|
6
|
Ivatury SJ. ctDNA Testing for Minimal Residual Disease Assessment and Surveillance. Dis Colon Rectum 2024; 67:1364-1366. [PMID: 39432727 DOI: 10.1097/dcr.0000000000003490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Affiliation(s)
- Srinivas J Ivatury
- Department of Surgery and Perioperative Care, University of Texas at Austin Dell Medical School, Austin, Texas
| |
Collapse
|
7
|
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
|
8
|
Ben-David R, Mehrazin R, Attalla K, Wiklund P, Sfakianos JP. Tumor-informed circulating tumor DNA in urothelial carcinoma: a promising novel biomarker. Curr Opin Urol 2024; 34:464-470. [PMID: 39228216 DOI: 10.1097/mou.0000000000001221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
PURPOSE OF REVIEW Tumor-informed circulating tumor DNA (ctDNA) is an emerging biomarker in urothelial carcinoma. Recent clinical trials have investigated the integration of ctDNA into clinical decision-making in patients with muscle-invasive bladder cancer, their findings suggest that ctDNA may potentially revolutionize the way we stratify patients to different treatment modalities. RECENT FINDINGS ctDNA informed from TURBT specimens was found to be prognostic of disease outcomes among patients with localized nonmetastatic bladder cancer. Detectable precystectomy ctDNA status was associated with worse survival outcomes. Additionally, ctDNA status was predictive of adverse disease on radical cystectomy, including the likelihood of disease upstaging, lymph node involvement, and having a locally advanced disease (≥pT3a). In the postcystectomy minimal residual disease (MRD) period, ctDNA status may refine patient selection to adjuvant therapy, and if validated by ongoing clinical trials, patients with undetectable postcystectomy ctDNA status may forgo adjuvant treatment, regardless of pathological stage. On the contrary, patients with pre or postcystectomy detectable ctDNA status may benefit from treatment intensification. SUMMARY The integration of ctDNA in clinical decision-making has the potential to revolutionize the way we manage urothelial carcinoma by refining patient selection to different treatment modalities. This approach could ultimately lead to personalization of oncological care, with the potential to reduce both treatment-related and financial toxicity.
Collapse
Affiliation(s)
- Reuben Ben-David
- Department of Urology, Icahn School of Medicine at The Mount Sinai Hospital, New York, New York, USA
| | | | | | | | | |
Collapse
|
9
|
Trinidad-Gutiérrez I, Vázquez-Borrego MC, Aguilera-Fernández E, Velez-Castaño JE, Muriel-López CE, Rodríguez-Ortíz L, Gómez A, Berchez-Moreno F, Hervás C, Romero-Ruiz A, Arjona-Sánchez Á. Efficacy of systemic Chemotherapy on high-risk stage II and III Mucnious colon cancer. CHEMUCCA study part I. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108642. [PMID: 39213699 DOI: 10.1016/j.ejso.2024.108642] [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: 05/20/2024] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Locally advanced colon cancer is a high-risk condition for tumour recurrence with poor survival. The current treatment is surgery followed by adjuvant chemotherapy based on fluoropyrimidines and oxaliplatin. This approach has improved the oncological outcomes on this population, however the mucinous condition has not been studied in depth and although the evidence is weak, it is thought to have a worse response to systemic chemotherapy. The CHEMUCCA study aims to answer this question. OBJECTIVE To evaluate the effectiveness of adjuvant systemic chemotherapy using the disease-free survival for stage II and III mucinous colon cancer who underwent surgical resection plus systemic adjuvant chemotherapy vs. surgery alone. PATIENTS AND METHODS Retrospective analytical study including patients diagnosed with high-risk stage II and stage III colon cancer, treated between 2010 and 2021, with a minimum follow-up of 3 years. Demographic variables and tumour features were analysed. The primary endpoint was disease-free survival. Log rank test and Cox regression were used. RESULTS Of 1134 patients with high-risk stage II and III colon cancer disease, 206 (18,17 %) had mucinous histology and 928 (81,83 %) had non-mucinous histology. 708 patients who received adjuvant chemotherapy, 129 (62,62 %) in mucinous group and 579 (62,39 %) in the non-mucinous group. Adjuvant systemic chemotherapy in stage II and III mucinous colon cancer improved the DFS (HR = 0.58 [95 % CI 0.37-0.91]; p = 0,017). However, in a stratified analysis, patients with high-risk stage II mucinous colon cancer showed no benefit with this approach (HR = 0.4541 [95 % CI 0.19-1.03]; p = 0.06). CONCLUSION Adjuvant chemotherapy has demonstrated to be effective in locally advanced mucinous colorectal cancer improving the oncological outcomes. However, this benefit could be diminished in high-risk stage II mucinous colon cancer patients. The administration of adjuvant chemotherapy on this patient's sub-group must be balanced according to risk versus benefits.
Collapse
Affiliation(s)
| | - Mari C Vázquez-Borrego
- GE09 Peritoneal and Retroperitoneal Oncologic Surgery Research Group, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Department of Biochemistry and Molecular Biology, University of Cordoba, Córdoba, Spain
| | | | | | | | - Lidia Rodríguez-Ortíz
- Surgical Oncology Unit, Reina Sofia University Hospital, Córdoba, Spain; GE09 Peritoneal and Retroperitoneal Oncologic Surgery Research Group, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Antonio Gómez
- Department of Computer Science and Numerical Analysis, University of Cordoba, Spain
| | | | - Cesar Hervás
- Department of Computer Science and Numerical Analysis, University of Cordoba, Spain
| | - Antonio Romero-Ruiz
- GE09 Peritoneal and Retroperitoneal Oncologic Surgery Research Group, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Department of Biochemistry and Molecular Biology, University of Cordoba, Córdoba, Spain
| | - Álvaro Arjona-Sánchez
- Surgical Oncology Unit, Reina Sofia University Hospital, Córdoba, Spain; GE09 Peritoneal and Retroperitoneal Oncologic Surgery Research Group, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain.
| |
Collapse
|
10
|
Pericay C, Montagut C, Reina JJ, Melian M, Alcaide J, Tarazona N, Ruiz-Casado A, González-Flores E, Graña B, Grávalos C. SEOM-GEMCAD-TTD clinical guidelines for the adjuvant treatment of colon cancer (2023). Clin Transl Oncol 2024; 26:2812-2825. [PMID: 38914755 PMCID: PMC11467085 DOI: 10.1007/s12094-024-03559-5] [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: 06/03/2024] [Indexed: 06/26/2024]
Abstract
Colorectal cancer (CRC) has a 5-year overall survival rate of over 60%. The decrease in the rate of metastatic disease is due to screening programs and the population's awareness of healthy lifestyle. Similarly, advancements in surgical methods and the use of adjuvant chemotherapy have contributed to a decrease in the recurrence of resected disease. Before evaluating a patient's treatment, it is recommended to be discussed in a multidisciplinary tumor board. In stage II tumors, the pathologic characteristics of poor prognosis must be known (T4, number of lymph nodes analyzed less than 12, lymphovascular or perineural invasion, obstruction or perforation, poor histologic grade, presence of tumor budding) and it is mandatory to determine the MSI/MMR status for avoiding administering fluoropyridimidines in monotherapy to patients with MSI-H/dMMR tumors. In stage III tumors, the standard treatment consists of a combination of fluoropyrimidine (oral or intravenous) with oxaliplatin for 6 months although the administration of CAPOX can be considered for 3 months in low-risk tumors. Neoadjuvant treatment is not consolidated yet although immunotherapy is achieving very good preliminary results in MSI-H patients. The use of ctDNA to define the treatment and monitoring of resected tumors is only recommended within studies. These guidelines are intended to help decision-making to offer the best management of patients with non-metastatic colon cancer.
Collapse
Affiliation(s)
- Carles Pericay
- Medical Oncology Department, Hospital University, Mútua de Terrassa, Barcelona, Spain.
| | - Clara Montagut
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | - Juan José Reina
- Medical Oncology Department, Hospital University, Virgen Macarena, Seville, Spain
| | | | - Julia Alcaide
- Medical Oncology Department, Hospital University, Regional y Virgen de la Victoria, Málaga, Spain
| | - Noelia Tarazona
- Medical Oncology Department, Hospital Clínico University de Valencia, Valencia, Spain
| | - Ana Ruiz-Casado
- Medical Oncology Department, H.U. Puerta de Hierro, Madrid, Spain
| | | | - Begoña Graña
- Medical Oncology Department, Complexo Hospitalario Universitario, A Coruña, Spain
| | - Cristina Grávalos
- Medical Oncology Department, Instituto de Investigacion Sanitaria Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
11
|
Medford AJ, Carmeli AB, Ritchie A, Wagle N, Garraway L, Lander ES, Parikh A. A standing platform for cancer drug development using ctDNA-based evidence of recurrence. Nat Rev Cancer 2024; 24:810-821. [PMID: 39349822 DOI: 10.1038/s41568-024-00742-2] [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] [Accepted: 08/09/2024] [Indexed: 10/26/2024]
Abstract
The time required to conduct clinical trials limits the rate at which we can evaluate and deliver new treatment options to patients with cancer. New approaches to increase trial efficiency while maintaining rigor would benefit patients, especially in oncology, in which adjuvant trials hold promise for intercepting metastatic disease, but typically require large numbers of patients and many years to complete. We envision a standing platform - an infrastructure to support ongoing identification and trial enrolment of patients with cancer with early molecular evidence of disease (MED) after curative-intent therapy for early-stage cancer, based on the presence of circulating tumour DNA. MED strongly predicts subsequent recurrence, with the vast majority of patients showing radiographic evidence of disease within 18 months. Such a platform would allow efficient testing of many treatments, from small exploratory studies to larger pivotal trials. Trials enrolling patients with MED but without radiographic evidence of disease have the potential to advance drug evaluation because they can be smaller (given high probability of recurrence) and faster (given short time to recurrence) than conventional adjuvant trials. Circulating tumour DNA may also provide a valuable early biomarker of treatment effect, which would allow small signal-finding trials. In this Perspective, we discuss how such a platform could be established.
Collapse
Affiliation(s)
- Arielle J Medford
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | - Eric S Lander
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Science for America, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Aparna Parikh
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Kramer A, Rubio‐Alarcón C, van den Broek D, Vessies DCL, van't Erve I, Meijer GA, Vink GR, Schuuring E, Fijneman RJA, Coupé VMH, Retèl VP. A scenario-drafting study to explore potential future implementation pathways of circulating tumor DNA testing in oncology. Mol Oncol 2024; 18:2730-2742. [PMID: 38060377 PMCID: PMC11547223 DOI: 10.1002/1878-0261.13562] [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: 07/07/2023] [Revised: 11/17/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024] Open
Abstract
Circulating tumor DNA (ctDNA) detection has multiple promising applications in oncology, but the road toward implementation in clinical practice is unclear. We aimed to support the implementation process by exploring potential future pathways of ctDNA testing. To do so, we studied four ctDNA-testing applications in two cancer types and elicited opinions from 30 ctDNA experts in the Netherlands. Our results showed that the current available evidence differed per application and cancer type. Tumor profiling and monitoring treatment response were found most likely to be implemented in non-small cell lung cancer (NSCLC) within 5 years. For colorectal cancer, applications of ctDNA testing were found to be at an early stage in the implementation process. Demonstrating clinical utility was found a key aspect for successful implementation, but there was no consensus regarding the evidence requirements. The next step toward implementation is to define how clinical utility of biomarkers should be evaluated. Finally, these data indicate that specific challenges for each clinical application and tumor type should be appropriately addressed in a deliberative process involving all stakeholders to ensure implementation of ctDNA testing and timely access for patients.
Collapse
Affiliation(s)
- Astrid Kramer
- Department of Epidemiology and Data ScienceAmsterdam UMCThe Netherlands
| | | | - Daan van den Broek
- Department of Laboratory MedicineNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Daan C. L. Vessies
- Department of Laboratory MedicineNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Iris van't Erve
- Department of PathologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Gerrit A. Meijer
- Department of PathologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Geraldine R. Vink
- Department of Medical Oncology, University Medical Center UtrechtUniversity of UtrechtThe Netherlands
- Department of Research and DevelopmentIKNLUtrechtThe Netherlands
| | - Ed Schuuring
- Department of Pathology and Medical BiologyUniversity Medical Center GroningenThe Netherlands
| | | | | | - Valesca P. Retèl
- Department of Psychosocial Research and EpidemiologyNetherlands Cancer InstituteAmsterdamThe Netherlands
- Erasmus School of Health Policy and ManagementErasmus University RotterdamThe Netherlands
| |
Collapse
|
13
|
Sachdeva UM. Commentary: Circulating tumor DNA in resectable non-small cell lung cancer: Emerging canary in the coal mine. J Thorac Cardiovasc Surg 2024; 168:1362-1363. [PMID: 38522575 DOI: 10.1016/j.jtcvs.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Affiliation(s)
- Uma M Sachdeva
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, Mass.
| |
Collapse
|
14
|
Morasso C, Daveri E, Bonizzi A, Truffi M, Colombo F, Danelli P, Albasini S, Rivoltini L, Mazzucchelli S, Sorrentino L, Corsi F. Raman spectroscopy on dried blood plasma allows diagnosis and monitoring of colorectal cancer. MedComm (Beijing) 2024; 5:e774. [PMID: 39492836 PMCID: PMC11527808 DOI: 10.1002/mco2.774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/21/2024] [Accepted: 09/05/2024] [Indexed: 11/05/2024] Open
Abstract
Colorectal cancer (CRC) remains challenging to diagnose, necessitating the identification of a noninvasive biomarker that can differentiate it from other conditions such as inflammatory bowel diseases (IBD) and diverticular disease (DD). Raman spectroscopy (RS) stands out as a promising technique for monitoring blood biochemical profiles, with the potential to identify distinct signatures identifying CRC subjects. We performed RS analysis on dried plasma from 120 subjects: 32 CRC patients, 37 IBD patients, 20 DD patients, and 31 healthy controls. We also conducted longitudinal studies of CRC patient's postsurgery to monitor the spectral changes over time. We identified six spectral features that showed significant differences between CRC and non-CRC patients, corresponding to tryptophan, tyrosine, phenylalanine, lipids, carotenoids, and disulfide bridges. These features enabled the classification of CRC patients with an accuracy of 87.5%. Moreover, longitudinal analysis revealed that the spectral differences normalized over 6 months after surgery, indicating their association with the presence of the disease. Our study demonstrates the potential of RS to identify specific biomolecular signatures related to CRC. These results suggest that RS could be a novel screening and monitoring tool, providing valuable insights for the development of noninvasive and accurate diagnostic methods for CRC.
Collapse
Affiliation(s)
- Carlo Morasso
- Laboratory of NanomedicineIstituti Clinici Scientifici Maugeri IRCCSPaviaItaly
| | - Elena Daveri
- Translational Immunology UnitFondazione IRCCS Istituto Nazionale dei Tumori di MilanoMilanItaly
| | - Arianna Bonizzi
- Laboratory of NanomedicineIstituti Clinici Scientifici Maugeri IRCCSPaviaItaly
| | - Marta Truffi
- Laboratory of NanomedicineIstituti Clinici Scientifici Maugeri IRCCSPaviaItaly
| | - Francesco Colombo
- Division of General Surgery“Luigi Sacco” University HospitalASST Fatebenefratelli‐SaccoMilanItaly
| | - Piergiorgio Danelli
- Division of General Surgery“Luigi Sacco” University HospitalASST Fatebenefratelli‐SaccoMilanItaly
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Sara Albasini
- Breast UnitIstituti Clinici Scientifici Maugeri IRCCSPaviaItaly
| | - Licia Rivoltini
- Translational Immunology UnitFondazione IRCCS Istituto Nazionale dei Tumori di MilanoMilanItaly
| | | | - Luca Sorrentino
- Colorectal surgery unitFondazione IRCCS Istituto Nazionale dei Tumori di MilanoMilanItaly
| | - Fabio Corsi
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
- Breast UnitIstituti Clinici Scientifici Maugeri IRCCSPaviaItaly
| |
Collapse
|
15
|
Frydendahl A, Nors J, Rasmussen MH, Henriksen TV, Nesic M, Reinert T, Afterman D, Lauterman T, Kuzman M, Gonzalez S, Glavas D, Smadback J, Maloney D, Levativ J, Yahalom M, Ptashkin R, Tavassoly I, Donenhirsh Z, White E, Kandasamy R, Alon U, Nordentoft I, Lindskrog SV, Dyrskjøt L, Jaensch C, Løve US, Andersen PV, Thorlacius-Ussing O, Iversen LH, Gotschalck KA, Zviran A, Oklander B, Andersen CL. Detection of circulating tumor DNA by tumor-informed whole-genome sequencing enables prediction of recurrence in stage III colorectal cancer patients. Eur J Cancer 2024; 211:114314. [PMID: 39316995 DOI: 10.1016/j.ejca.2024.114314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Circulating tumor (ctDNA) can be used to detect residual disease after cancer treatment. Detecting low-level ctDNA is challenging, due to the limited number of recoverable ctDNA fragments at any target loci. In response, we applied tumor-informed whole-genome sequencing (WGS), leveraging thousands of mutations for ctDNA detection. METHODS Performance was evaluated in serial plasma samples (n = 1283) from 144 stage III colorectal cancer patients. Tumor/normal WGS was used to establish a patient-specific mutational fingerprint, which was searched for in 20x WGS plasma profiles. For reproducibility, paired aliquots of 172 plasma samples were analyzed in two independent laboratories. De novo variant calling was performed for serial plasma samples with a ctDNA level > 10 % (n = 17) to explore genomic evolution. RESULTS WGS-based ctDNA detection was prognostic of recurrence: post-operation (Hazard ratio [HR] 6.75, 95 %CI 3.18-14.3, p < 0.001), post-adjuvant chemotherapy (HR 28.9, 95 %CI 10.1-82.8; p < 0.001), and during surveillance (HR 22.8, 95 %CI 13.7-37.9, p < 0.0001). The 3-year cumulative incidence of ctDNA detection in recurrence patients was 95 %. ctDNA was detected a median of 8.7 months before radiological recurrence. The independently analyzed plasma aliquots showed excellent agreement (Cohens Kappa=0.9, r = 0.99). Genomic characterization of serial plasma revealed significant evolution in mutations and copy number alterations, and the timing of mutational processes, such as 5-fluorouracil-induced mutations. CONCLUSION Our study supports the use of WGS for sensitive ctDNA detection and demonstrates that post-treatment ctDNA detection is highly prognostic of recurrence. Furthermore, plasma WGS can identify genomic differences distinguishing the primary tumor and relapsing metastasis, and monitor treatment-induced genomic changes.
Collapse
Affiliation(s)
- Amanda Frydendahl
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Jesper Nors
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Mads H Rasmussen
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Tenna V Henriksen
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Marijana Nesic
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Thomas Reinert
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | - Eric White
- C2i Genomics Inc., New York, NY 10014, USA
| | | | - Ury Alon
- C2i Genomics, Ltd., Haifa, Israel
| | - Iver Nordentoft
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Sia V Lindskrog
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | | | - Uffe S Løve
- Department of Surgery, Viborg Regional Hospital, Denmark
| | - Per V Andersen
- Department of Surgery, Odense University Hospital, Denmark
| | | | - Lene H Iversen
- Department of Clinical Medicine, Aarhus University, Denmark; Department of Surgery, Aarhus University Hospital, Denmark
| | - Kåre A Gotschalck
- Department of Clinical Medicine, Aarhus University, Denmark; Department of Surgery, Randers Regional Hospital, Denmark
| | | | | | - Claus L Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Denmark.
| |
Collapse
|
16
|
Gandini A, Taieb J, Blons H, Netter J, Laurent-Puig P, Gallois C. Early-Onset colorectal Cancer: From the laboratory to the clinic. Cancer Treat Rev 2024; 130:102821. [PMID: 39236404 DOI: 10.1016/j.ctrv.2024.102821] [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: 07/02/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Colorectal cancer that occurs before age of 50 is defined as Early-Onset Colorectal Cancer (EOCRC). Its incidence has worryingly increased since the late 90 s and is expected to keep rising in the next future, despite Late-Onset CRC (LOCRC) is decreasing worldwide. Because of this, there is an urgent need to better understand this subset of patients in order to give them the best treatment possible. However, most of the literature is retrospective and often discordant. In this review, we aim to provide a general overview of the issue, endeavoring to highlight the current available knowledge. We decided to move from the beginning, investigating risk factors and inheritance, passing through diagnosis and clinical aspects, and to conclude with the translational part, focusing on the biology of the tumor. However, lot of questions remain open, including screening age and prognosis. Indeed, young patients tend to be treated more aggressively, even if a survival benefit has not been proven yet. Every clinician should be aware of the best practice for young people, and more translational studies are awaited in order to clarify is EOCRC represents a distinct biological entity.
Collapse
Affiliation(s)
- Annalice Gandini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris CARPEM, AP-HP Centre, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Julien Taieb
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris CARPEM, AP-HP Centre, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Hélène Blons
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Assistance Publique-Hôpitaux de Paris, Department of Biochemistry, Pharmacogenetics and Molecular Oncology, European Georges Pompidou Hospital, Paris Cancer Institute CARPEM, 20 Rue Leblanc, 75015, Paris, France; Department of Genetics and Molecular Medicine, Georges Pompidou European Hospital, APHP Centre, Paris, France
| | - Jeanne Netter
- Institut du Cancer Paris CARPEM, AP-HP Centre, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris CARPEM, APHP. Centre, Department of Biology, Hôpital Européen Georges Pompidou, Paris, France
| | - Claire Gallois
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris CARPEM, AP-HP Centre, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
17
|
Padillo-Ruiz J, Fresno C, Suarez G, Blanco G, Muñoz-Bellvis L, Justo I, García-Domingo MI, Ausania F, Muñoz-Forner E, Serrablo A, Martin E, Díez L, Cepeda C, Marin L, Alamo J, Bernal C, Pereira S, Calero F, Tinoco J, Paterna S, Cugat E, Fondevila C, Diego-Alonso E, López-Guerra D, Gomez M, Denninghoff V, Sabater L. Effects of the superior mesenteric artery approach versus the no-touch approach during pancreatoduodenectomy on the mobilization of circulating tumour cells and clusters in pancreatic cancer (CETUPANC): randomized clinical trial. BJS Open 2024; 8:zrae123. [PMID: 39485887 PMCID: PMC11529789 DOI: 10.1093/bjsopen/zrae123] [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: 02/09/2024] [Revised: 08/02/2024] [Accepted: 08/21/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Patients with pancreatic ductal adenocarcinoma present early postoperative systemic metastases, despite complete oncological resection. The aim of this study was to assess two pancreatoduodenectomy approaches with regard to intraoperative circulating tumour cells and cluster mobilization and their potential association with the development of distant metastasis. METHODS Patients with periampullary tumours who underwent open pancreatoduodenectomy were randomly allocated to either the no-touch approach or the superior mesenteric artery approach. A total of four intraoperative portal vein samples (at the beginning of the intervention, after portal vein disconnection from the tumour, after tumour resection, and before abdominal closure) were collected to measure circulating tumour cells and cluster numbers. Primary outcomes were the intraoperative number of circulating tumour cells and cluster mobilization. Further, their potential impact on 3-year distant metastasis disease-free survival and overall survival was assessed. RESULTS A total of 101 patients with periampullary tumours were randomized (51 in the superior mesenteric artery group and 50 in the no-touch group) and 63 patients with pancreatic ductal adenocarcinoma (34 in the superior mesenteric artery group and 29 in the no-touch group) were analysed. Circulating tumour cells and cluster mobilization were similar in both the no-touch group and the superior mesenteric artery group at all time points. There were no significant differences between surgical groups with regard to the median metastasis disease-free survival (12.4 (interquartile range 6.1-not reached) months in the superior mesenteric artery group and 18.1 (interquartile range 12.1-not reached) months in the no-touch group; P = 0.730). Patients with intraoperative cluster mobilization from the beginning to the end of surgery developed significantly more distant metastases within the first year after surgery (P = 0.023). Two intraoperative factors (the superior mesenteric artery approach (P = 0.025) and vein resection (P < 0.001)) were predictive factors for cluster mobilization. CONCLUSION Patients undergoing pancreatoduodenectomy using either the no-touch approach or the superior mesenteric artery approach had similar circulating tumour cells and cluster mobilization and similar overall survival and metastasis disease-free survival. A high intraoperative cluster dissemination during pancreatoduodenectomy was a predictive factor for early metastases in patients with pancreatic ductal adenocarcinoma. REGISTRATION NUMBER NCT03340844 (http://www.clinicaltrials.gov)-CETUPANC trial.
Collapse
Affiliation(s)
- Javier Padillo-Ruiz
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Cristóbal Fresno
- Health and Sciences Research Centre, Health and Sciences Faculty, Anahuac University, Huixquilucan, Mexico
| | - Gonzalo Suarez
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Gerardo Blanco
- Department of Surgery, Badajoz University Hospital, University of Extremadura, Badajoz, Spain
| | - Luis Muñoz-Bellvis
- Department of Surgery, University Hospital of Salamanca, Salamanca Biosanitary Institute, University of Salamanca, Salamanca, Spain
| | - Iago Justo
- Department of Surgery, University Hospital October 12 in Madrid, Madrid, Spain
| | | | - Fabio Ausania
- Hospital-Clinic, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona, Spain
| | - Elena Muñoz-Forner
- Department of Surgery, Valencia Clinical Hospital, University of Valencia, Biomedical Research Institute, INCLIVA, Valencia, Spain
| | - Alejandro Serrablo
- Department of Surgery, Miguel Servet University Hospital, Zaragoza, Spain
| | - Elena Martin
- Department of Surgery, Princess University Hospital, Madrid, Spain
| | - Luis Díez
- Department of Surgery, Clinical Hospital, Madrid, Spain
| | - Carmen Cepeda
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Luis Marin
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Jose Alamo
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Carmen Bernal
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Sheila Pereira
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Francisco Calero
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Jose Tinoco
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Sandra Paterna
- Department of Surgery, Miguel Servet University Hospital, Zaragoza, Spain
| | - Esteban Cugat
- Department of Surgery, Terrassa Mutual University Hospital, Terrassa, Spain
| | - Constantino Fondevila
- Hospital-Clinic, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona, Spain
| | - Elisa Diego-Alonso
- Department of Surgery, University Hospital of Salamanca, Salamanca Biosanitary Institute, University of Salamanca, Salamanca, Spain
| | - Diego López-Guerra
- Department of Surgery, Badajoz University Hospital, University of Extremadura, Badajoz, Spain
| | - Miguel Gomez
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Valeria Denninghoff
- Department of Surgery, Virgen del Rocío University Hospital, IBIS, University of Seville, Seville, Spain
| | - Luis Sabater
- Department of Surgery, Valencia Clinical Hospital, University of Valencia, Biomedical Research Institute, INCLIVA, Valencia, Spain
| |
Collapse
|
18
|
Hu Q, Chen L, Li K, Liu R, Sun L, Han T. Circulating tumor DNA: current implementation issues and future challenges for clinical utility. Clin Chem Lab Med 2024; 62:2094-2110. [PMID: 38109307 DOI: 10.1515/cclm-2023-1157] [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: 10/16/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
Over the past decades, liquid biopsy, especially circulating tumor DNA (ctDNA), has received tremendous attention as a noninvasive detection approach for clinical applications, including early diagnosis of cancer and relapse, real-time therapeutic efficacy monitoring, potential target selection and investigation of drug resistance mechanisms. In recent years, the application of next-generation sequencing technology combined with AI technology has significantly improved the accuracy and sensitivity of liquid biopsy, enhancing its potential in solid tumors. However, the increasing integration of such promising tests to improve therapy decision making by oncologists still has complexities and challenges. Here, we propose a conceptual framework of ctDNA technologies and clinical utilities based on bibliometrics and highlight current challenges and future directions, especially in clinical applications such as early detection, minimal residual disease detection, targeted therapy, and immunotherapy. We also discuss the necessities of developing a dynamic field of translational cancer research and rigorous clinical studies that may support therapeutic strategy decision making in the near future.
Collapse
Affiliation(s)
- Qilin Hu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Lujun Chen
- The General Hospital of Northern Theater Command Training Base for Graduate, China Medical University, Shenyang, P.R. China
| | - Kerui Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Ruotong Liu
- Clinical Medicine, Shenyang Medical College, Shenyang, P.R. China
| | - Lei Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, P.R. China
| |
Collapse
|
19
|
Li R, Chen J, Shen X, Lin Y, Tang J, Xiong G, Zhang K, Xiang M, Xie L, Hu F. A study of the clinical significance of mSEPT9 in monitoring recurrence and prognosis in patients with surgically treated colorectal cancer. PLoS One 2024; 19:e0312676. [PMID: 39466813 PMCID: PMC11515984 DOI: 10.1371/journal.pone.0312676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
OBJECTIVE To explore the medical significance of methylated septin9 (mSEPT9) in monitoring recurrence and prognostic assessment in individuals with surgically treated colorectal cancer (CRC). METHODS To investigate the role of Septin9 in colorectal cancer, we utilized the TIMER2.0 database to analyze its differential expression between tumor tissues and adjacent normal tissues. Colorectal cancer RNA-seq data from the TCGA database was downloaded and curated. The clinical relevance of mSEPT9 in colorectal cancer was explored by examining the correlation between Septin9 methylation levels and clinical characteristics using UALCAN and MethSurv software. Peripheral blood samples were obtained from 130 CRC subjects who underwent surgery for the detection of mSEPT9 and carcinoembryonic antigen (CEA) expression, along with collection of clinicopathological data such as age, gender, tumor site, TNM stage, and tumor differentiation. Patients were followed up for at least 3 years post-surgery until the death or final follow-up dates (31/12/2022). Additionally, peripheral blood samples were collected from 30 colorectal cancer surgery patients for mSEPT9 detection before and 7 days after surgery. RESULTS Through bioinformatic database analysis, we identified higher expression levels of SEPT9 mRNA in most tumor tissues compared to normal tissues. Similarly, both paired and unpaired CRC tissues exhibited elevated expression of Septin9 when compared to normal tissues. Following GO and KEGG analysis of Septin9 target genes, we discovered their significant associations with ncRNA metabolic processes, ribonucleoprotein complex biogenesis, spliceosomes, and viral carcinogenesis. Furthermore, the overexpression of mSeptin9 was observed in CRC tissues, and it demonstrated a correlation with colon cancer staging and histologic classification. In our clinical sample study, The positive rate of mSEPT9 in CRC patients 7 days after surgery was 43.44% lower than that of preoperative. The differences in TNM stage, tumor differentiation degree, and preoperative CEA expression level between the preoperative mSEPT9 positive and negative groups of CRC were statistically significant (P < 0.05). Recurrence free survival (RFS) and overall survival (OS) were shorter in the preoperative mSEPT9-positive group, meaning preoperative mSEPT9 status was a risk factor for CRC recurrence and prognosis (P < 0.05). The sensitivity, specificity, and AUC value of preoperative mSEPT9 and CEA levels for predicting postoperative recurrence in CRC patients were 88% vs. 72%, 56.19% vs. 55.24%, and 0.721 vs. 0.636 respectively, well the AUC value of the combined prediction of postoperative recurrence was 0.758. CONCLUSION The detection of mSEPT9 combined with CEA in preoperative plasma helps predict recurrence in colorectal cancer patients.
Collapse
Affiliation(s)
- Rong Li
- Department of Digestive Neoplasms, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan Province, China
| | - Jiaojiao Chen
- Radiotherapy Department, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Xin Shen
- Department of Digestive Neoplasms, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan Province, China
| | - Yanping Lin
- Department of Digestive Neoplasms, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan Province, China
| | - Jiadai Tang
- Department of Digestive Neoplasms, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan Province, China
| | - Guangrui Xiong
- Gastroenterology, Lincang People’s Hospital, Lincang, Yunnan Province, China
| | - Ke Zhang
- Department of Oncology, Baoshan People’s Hospital in Yunnan Province, Baoshan, Yunnan Province, China
| | - Mengying Xiang
- Department of Critical Care Medicine, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan Province, China
| | - Lin Xie
- Department of Digestive Neoplasms, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan Province, China
| | - Fengdi Hu
- Department of Digestive Neoplasms, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan Province, China
| |
Collapse
|
20
|
Melichar B. Circulating tumor DNA measurement: a new pillar of medical oncology? Clin Chem Lab Med 2024; 62:2091-2093. [PMID: 39239844 DOI: 10.1515/cclm-2024-0986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Affiliation(s)
- Bohuslav Melichar
- Department of Oncology, Palacký University Medical School and University Hospital Olomouc Olomouc, Czech Republic
- Department of Oncology and Radiotherapy, University Hospital Hradec Králové Hradec Králové, Czech Republic
| |
Collapse
|
21
|
Dubrovsky G, Ross A, Jalali P, Lotze M. Liquid Biopsy in Pancreatic Ductal Adenocarcinoma: A Review of Methods and Applications. Int J Mol Sci 2024; 25:11013. [PMID: 39456796 PMCID: PMC11507494 DOI: 10.3390/ijms252011013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a malignancy with one of the highest mortality rates. One limitation in the diagnosis and treatment of PDAC is the lack of an early and universal biomarker. Extensive research performed recently to develop new assays which could fit this role is available. In this review, we will discuss the current landscape of liquid biopsy in patients with PDAC. Specifically, we will review the various methods of liquid biopsy, focusing on circulating tumor DNA (ctDNA) and exosomes and future opportunities for improvement using artificial intelligence or machine learning to analyze results from a multi-omic approach. We will also consider applications which have been evaluated, including the utility of liquid biopsy for screening and staging patients at diagnosis as well as before and after surgery. We will also examine the potential for liquid biopsy to monitor patient treatment response in the setting of clinical trial development.
Collapse
Affiliation(s)
- Genia Dubrovsky
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA; (G.D.); (A.R.)
- Pittsburgh VA Medical Center, Pittsburgh, PA 15240, USA
| | - Alison Ross
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA; (G.D.); (A.R.)
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Michael Lotze
- Departments of Surgery, Immunology, and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
22
|
Williams CJ, Gray R, Hills RK, Shires M, Zhang L, Zhao Z, Gardner T, Sapanara N, Xu XM, Bai I, Yan D, Muranyi A, Dance S, Aghaei F, Hemmings G, Hale M, Kurkure U, Guetter C, Richman SD, Hutchins G, Seligmann JF, West NP, Singh S, Shanmugam K, Quirke P. Evaluation of CD3 and CD8 T-Cell Immunohistochemistry for Prognostication and Prediction of Benefit From Adjuvant Chemotherapy in Early-Stage Colorectal Cancer Within the QUASAR Trial. J Clin Oncol 2024; 42:3430-3442. [PMID: 39083705 PMCID: PMC11458110 DOI: 10.1200/jco.23.02030] [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: 09/18/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 08/02/2024] Open
Abstract
PURPOSE High densities of tumor infiltrating CD3 and CD8 T-cells are associated with superior prognosis in colorectal cancer (CRC). Their value as predictors of benefit from adjuvant chemotherapy is uncertain. PATIENTS AND METHODS Tumor tissue from 868 patients in the QUASAR trial (adjuvant fluorouracil/folinic acid v observation in stage II/III CRC) was analyzed by CD3 and CD8 immunohistochemistry. Pathologists, assisted by artificial intelligence, calculated CD3 and CD8 cell densities (cells/mm2) in the core tumor (CT) and invasive margin (IM). Participants were randomly partitioned into training and validation sets. The primary outcome was recurrence-free interval (RFI), 2-year RFI for assessment of biomarker-treatment interactions. Maximum-likelihood methods identified optimal high-risk/low-risk group cutpoints in the training set. Prognostic analyses were repeated in the validation set. RESULTS In the training set, the recurrence rate in the high-risk group was twice that in the low-risk group for all measures (CD3-CT: rate ratio [RR], 2.00, P = .0008; CD3-IM: 2.38, P < .00001; CD8-CT: 2.17, P = .0001; CD8-IM: 2.13, P = .0001). This was closely replicated in the validation set (RR, 1.96, 1.79, 1.72, 1.72, respectively). In multivariate analyses, prognostic effects were similar in colon and rectal cancers, and in stage II and III disease. Proportional reductions in recurrence with adjuvant chemotherapy were of similar magnitude in the high- and low-recurrence risk groups. Combining information from CD3-IM and CD3-CT (CD3 Score) generated high-, intermediate-, and low-risk groups with numbers needed to treat (NNTs) to prevent one disease recurrence being 11, 21, and 36, respectively. CONCLUSION Recurrence rates in the high-risk CD3/CD8 groups are twice those in the low-risk groups. Proportional reductions with chemotherapy are similar, allowing NNTs derived in QUASAR to be updated using contemporary, nonrandomized data sets.
Collapse
Affiliation(s)
- Christopher J.M. Williams
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Richard Gray
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Deceased
| | - Robert K. Hills
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Michael Shires
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Liping Zhang
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Zuo Zhao
- Roche Diagnostics Solutions, Imaging and Algorithms, Digital Pathology, Santa Clara, CA
| | - Tracie Gardner
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Nancy Sapanara
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Xiao-Meng Xu
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Isaac Bai
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Dongyao Yan
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Andrea Muranyi
- Roche Diagnostics Solutions, Research and Development, Oro Valley, AZ
| | - Sarah Dance
- Roche Diagnostics Limited, Medical Affairs, Burgess Hill, West Sussex, United Kingdom
| | - Faranak Aghaei
- Roche Diagnostics Solutions, Imaging and Algorithms, Digital Pathology, Santa Clara, CA
| | - Gemma Hemmings
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Michael Hale
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Uday Kurkure
- Roche Diagnostics Solutions, Imaging and Algorithms, Digital Pathology, Santa Clara, CA
| | - Christoph Guetter
- Roche Diagnostics Solutions, Imaging and Algorithms, Digital Pathology, Santa Clara, CA
| | - Susan D. Richman
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Gordon Hutchins
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Jenny F. Seligmann
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Nicholas P. West
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Shalini Singh
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Kandavel Shanmugam
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Philip Quirke
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
23
|
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
|
24
|
Tang H, Yu D, Zhang J, Wang M, Fu M, Qian Y, Zhang X, Ji R, Gu J, Zhang X. The new advance of exosome-based liquid biopsy for cancer diagnosis. J Nanobiotechnology 2024; 22:610. [PMID: 39380060 PMCID: PMC11463159 DOI: 10.1186/s12951-024-02863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Liquid biopsy is a minimally invasive method that uses biofluid samples instead of tissue samples for cancer diagnosis. Exosomes are small extracellular vesicles secreted by donor cells and act as mediators of intercellular communication in human health and disease. Due to their important roles, exosomes have been considered as promising biomarkers for liquid biopsy. However, traditional methods for exosome isolation and cargo detection methods are time-consuming and inefficient, limiting their practical application. In the past decades, many new strategies, such as microfluidic chips, nanowire arrays and electrochemical biosensors, have been proposed to achieve rapid, accurate and high-throughput detection and analysis of exosomes. In this review, we discussed about the new advance in exosome-based liquid biopsy technology, including isolation, enrichment, cargo detection and analysis approaches. The comparison of currently available methods is also included. Finally, we summarized the advantages and limitations of the present strategies and further gave a perspective to their future translational use.
Collapse
Affiliation(s)
- Haozhou Tang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
- Department of Orthopaedics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
| | - Dan Yu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jiahui Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Maoye Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Min Fu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yu Qian
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaoxin Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Runbi Ji
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jianmei Gu
- Departmemt of Clinical Laboratory Medicine, Nantong Tumor Hospital/Affiliated Tumor Hospital of Nantong University, Nantong, 226300, China.
- Affiliated Cancer Hospital of Nantong University, Nantong, 226300, China.
| | - Xu Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
25
|
Campani C, Imbeaud S, Couchy G, Ziol M, Hirsch TZ, Rebouissou S, Noblet B, Nahon P, Hormigos K, Sidali S, Seror O, Taly V, Ganne Carrie N, Laurent-Puig P, Zucman-Rossi J, Nault JC. Circulating tumour DNA in patients with hepatocellular carcinoma across tumour stages and treatments. Gut 2024; 73:1870-1882. [PMID: 39054058 DOI: 10.1136/gutjnl-2024-331956] [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: 01/12/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE Circulating tumour DNA (ctDNA) is a promising non-invasive biomarker in cancer. We aim to assess the dynamic of ctDNA in patients with hepatocellular carcinoma (HCC). DESIGN We analysed 772 plasmas from 173 patients with HCC collected at the time of diagnosis or treatment (n=502), 24 hours after locoregional treatment (n=154) and during follow-up (n=116). For controls, 56 plasmas from patients with chronic liver disease without HCC were analysed. All samples were analysed for cell free DNA (cfDNA) concentration, and for mutations in TERT promoter, CTNNB1, TP53, PIK3CA and NFE2L2 by sequencing and droplet-based digital PCR. Results were compared with 232 corresponding tumour samples. RESULTS In patients with active HCC, 40.2% of the ctDNA was mutated vs 14.6% in patients with inactive HCC and 1.8% in controls (p<0.001). In active HCC, we identified 27.5% of mutations in TERT promoter, 21.3% in TP53, 13.1% in CTNNB1, 0.4% in PIK3CA and 0.2% in NFE2L2, most of the times similar to those identified in the corresponding tumour. CtDNA mutation rate increased with advanced tumour stages (p<0.001). In 103 patients treated by percutaneous ablation, the presence and number of mutations in the ctDNA before treatment were associated with higher risk of death (p=0.001) and recurrence (p<0.001). Interestingly, cfDNA concentration and detectable mutations increased 24 hours after a locoregional treatment. Among 356 plasmas collected in 53 patients treated by systemic treatments, we detected mutations at baseline in 60.4% of the cases. In patients treated by atezolizumab-bevacizumab, persistence of mutation in ctDNA was associated with radiological progression (63.6% vs 36.4% for disappearance, p=0.019). In two patients progressing under systemic treatments, we detected the occurrence of mutations in CTNNB1 in the plasma that was subclonal in the tumour for one patient and not detectable in the tumour for the other one. CONCLUSION ctDNA offers dynamic information reflecting tumour biology. It represents a non-invasive tool useful to guide HCC clinical management.
Collapse
Affiliation(s)
- Claudia Campani
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Internal Medicine and Hepatology Unit, Department of Experimental and Clinical Medicine, University of Firenze, Florence, Italy
| | - Sandrine Imbeaud
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Gabrielle Couchy
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Marianne Ziol
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Pathology Department and Biological Resource Center Center (BB-0033-00027), Paris-Seine-Saint-Denis, University Hospital, Avicenne Hospital, APHP, Sorbonne Paris Nord University, Bobugny, France
| | - Theo Z Hirsch
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Sandra Rebouissou
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Bénédicte Noblet
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Pierre Nahon
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Liver Unit, Avicenne Hospital, APHP, University Sorbonne Paris Nord, Bobigny, France
| | - Katia Hormigos
- Cordeliers Research Center, INSERM, CNRS SNC 5096, Sorbonne University, Paris Cité University, Paris, France
| | - Sabrina Sidali
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Liver unit, Paris Cité University, Beaujon Hospital, APHP, DMU DIGEST, Clichy, France
| | - Olivier Seror
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Interventional Radiology Unit, Avicenne Hospital, APHP, Bobigny, Paris, France
| | - Valerie Taly
- Cordeliers Research Center, INSERM, CNRS SNC 5096, Sorbonne University, Paris Cité University, Paris, France
| | - Nathalie Ganne Carrie
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Liver Unit, Avicenne Hospital, APHP, University Sorbonne Paris Nord, Bobigny, France
| | - Pierre Laurent-Puig
- Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, Institut of Cancer Paris CARPEM, AP-HP-Hôpital Européen Georges Pompidou, Paris, France
| | - Jessica Zucman-Rossi
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, Institut of Cancer Paris CARPEM, AP-HP-Hôpital Européen Georges Pompidou, Paris, France
| | - Jean-Charles Nault
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Liver Unit, Avicenne Hospital, APHP, University Sorbonne Paris Nord, Bobigny, France
| |
Collapse
|
26
|
Bigotte Vieira M, Arai H, Nicolau C, Murakami N. Cancer Screening and Cancer Treatment in Kidney Transplant Recipients. KIDNEY360 2024; 5:1569-1583. [PMID: 39480669 DOI: 10.34067/kid.0000000000000545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
As the population ages and post-transplant survival improves, pretransplant and post-transplant malignancy are becoming increasingly common. In addition, rapid advances in cancer therapies and improving outcomes prompt us to rethink pretransplant cancer-free wait time and screening strategies. Although kidney transplant recipients (KTRs) are at higher risk of developing cancer, epidemiological data on how to best screen and treat cancers in KTRs are incomplete. Thus, current recommendations are still largely on the basis of studies in the general population, and their validity in KTRs is uncertain. Kidney transplant candidates without prior cancer should be evaluated for latent malignancies even in the absence of symptoms. Conversely, individuals with a history of malignancy require thorough monitoring to detect potential recurrences or de novo malignancies. When treating KTRs with cancer, reducing immunosuppression can enhance antitumor immunity, yet this also increases the risk of graft rejection. Optimal treatment and immunosuppression management remains undefined. As the emergence of novel cancer therapies adds complexity to this challenge, individualized risk-benefit assessment is crucial. In this review, we discuss up-to-date data on pretransplant screening and cancer-free wait time, as well as post-transplant cancer screening, prevention strategies, and treatment, including novel therapies such as immune checkpoint inhibitors and chimeric antigen receptor T-cell therapies.
Collapse
Affiliation(s)
- Miguel Bigotte Vieira
- Nephrology Department, Hospital Curry Cabral, Unidade Local de Saúde São José, Lisbon, Portugal
- NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Hiroyuki Arai
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Carla Nicolau
- Nephrology Department, Hospital Curry Cabral, Unidade Local de Saúde São José, Lisbon, Portugal
| | - Naoka Murakami
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
27
|
Lallas K, Kyrgidis A, Chrysostomidis A, Vakirlis E, Apalla Z, Lallas A. Clinical, dermatoscopic, histological and molecular predictive factors of distant melanoma metastasis: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2024; 202:104458. [PMID: 39074631 DOI: 10.1016/j.critrevonc.2024.104458] [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: 04/16/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Melanoma metastasis to distant sites is associated with diminished survival rates and poor prognosis. Except of Breslow thickness and ulceration that are currently used in melanoma staging, the investigation of additional clinicopathological, dermatoscopic and molecular factors that could predict tumors with aggressive biologic behavior is of paramount importance. METHODS A literature search was conducted in PubMed, Scopus, Cochrane databases and gray literature until November 2023. Observational studies (including cohorts and case-control studies) were included and clinical and histopathological factors of primary cutaneous melanomas, along with dermatoscopic and molecular predictors of distant metastasis (DM) and distant metastasis-free survival (DMFS) were assessed. Random - effect models were preferred, the results were presented as Hazard Ratios (HRs) with 95 %Confidence Intervals (CIs) and the I2 index quantified heterogeneity. Subgroup analysis according to AJCC stage and sensitivity analysis were also conducted. RESULTS One hundred forty-three and 101 studies were included in the qualitive and quantitative synthesis, respectively. Regarding clinical factors, males, compared to females, and head and neck location, compared to trunk, demonstrated higher risk for DM [n=36, HR 1.49, 95%CI 1.36 - 1.63, I2 33% and n=21, HR 1.24, 95 %CI 1.01 - 1.52, I2 62 %]. Both factors had similar effects on DMFS. Breslow thickness and ulceration were significant predictors or DM. Additional factors that posed an increased risk for DM were nodular (n=15, HR 2.51, 95 %CI 1.83 - 3.43, I2 56 %) and lentigo maligna subtypes (n=12, HR 1.87, 95 %CI 1.27 - 2.75, I2 0 %), compared to superficial spreading subtype, lymphovascular invasion (n=9, HR 2.05, 95 %CI 1.18 - 3.58, I2 78 %), SLN positivity and BRAF+ mutational status. In contrast, regression was a negative predictor of DM (n=15, HR 0.59, 95 %CI 0.44 - 0.79, I2 68 %). Two studies focused on dermatoscopic factors and found that low pigmentation and the presence of blue-white veil might predict DM development. The results of subgroup analysis for stage I-II patients were essentially similar and sensitivity analysis did not reveal significant alterations, despite the moderate or high heterogeneity in some categories. CONCLUSIONS Clinical and histological characteristics of the tumor along with dermatoscopic features and molecular parameters hold significant prognostic information and could be incorporated into models to predict melanomas with high metastatic potential.
Collapse
Affiliation(s)
- Konstantinos Lallas
- Department of Medical Oncology, School of Medicine, Faculty of Health Sciences, Aristotle University, Thessaloniki, Greece.
| | - Athanassios Kyrgidis
- Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Anestis Chrysostomidis
- Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; First Department of Dermatology, School of Medicine, Faculty of Health Sciences, Aristotle University, Thessaloniki, Greece
| | - Efstratios Vakirlis
- First Department of Dermatology, School of Medicine, Faculty of Health Sciences, Aristotle University, Thessaloniki, Greece
| | - Zoe Apalla
- Second Department of Dermatology, School of Medicine, Faculty of Health Sciences, Aristotle University, Thessaloniki, Greece
| | - Aimilios Lallas
- First Department of Dermatology, School of Medicine, Faculty of Health Sciences, Aristotle University, Thessaloniki, Greece
| |
Collapse
|
28
|
Conroy T, Castan F, Etienne PL, Rio E, Mesgouez-Nebout N, Evesque L, Vendrely V, Artignan X, Bouché O, Gargot D, Boige V, Bonichon-Lamichhane N, Louvet C, Morand C, de la Fouchardière C, Boilève A, Delaye M, Gourgou S, Pezzella V, Borg C. Total neoadjuvant therapy with mFOLFIRINOX versus preoperative chemoradiotherapy in patients with locally advanced rectal cancer: long-term results of the UNICANCER-PRODIGE 23 trial. Ann Oncol 2024; 35:873-881. [PMID: 38986769 DOI: 10.1016/j.annonc.2024.06.019] [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: 02/05/2024] [Revised: 05/27/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND The standard of care for the treatment of locally advanced rectal cancer (LARC) results in an excellent local disease control but the metastasis rates remain high. PRODIGE 23 demonstrated improved disease-free survival (DFS) and metastasis-free survival (MFS) with total neoadjuvant therapy versus standard of care in this population. Long-term analysis of overall survival (OS) is reported here. PATIENTS AND METHODS The study design, participants, and primary endpoint DFS have been reported for this multicenter, randomized, open-label, phase III trial investigating the neoadjuvant chemotherapy with mFOLFIRINOX (6 cycles) followed by chemoradiotherapy, surgery, and adjuvant chemotherapy (6 cycles), versus chemoradiotherapy, surgery, and adjuvant chemotherapy (12 cycles) in patients with locally advanced rectal adenocarcinoma under peritoneal reflection on magnetic resonance imaging, and staged cT3/T4. Key secondary endpoints included OS, MFS, and local and metastatic recurrence rate. RESULTS With a median follow-up of 82.2 months, the 7-year DFS was 67.6% [95% confidence interval (CI) 60.7% to 73.9%] and 62.5% (95% CI 55.6% to 68.6%) [restricted mean survival time (RMST) difference 5.73 months, 95% CI 0.05-11.41 months, P = 0.048] in the neoadjuvant chemotherapy and the standard-of-care groups, respectively. The 7-year MFS was 79.2% (95% CI 73.0% to 84.4%) in the neoadjuvant chemotherapy group and 72.3% (95% CI 65.8% to 77.8%) in the standard-of-care group (RMST difference 6.1 months, 95% CI 0.93-11.37 months, P = 0.021). The 7-year OS was 81.9% (95% CI 75.8% to 86.6%) in the neoadjuvant chemotherapy group and 76.1% (95% CI 69.7% to 81.2%) in the standard-of-care group (RMST difference 4.37 months, 95% CI 0.35-8.38 months, P = 0.033). The safety profile remained unchanged since the previous analysis. CONCLUSIONS Neoadjuvant chemotherapy with mFOLFIRINOX followed by chemoradiotherapy improved OS, confirmed long-term DFS and MFS benefits in LARC patients, and should be considered as one of the best options of care for these patients.
Collapse
Affiliation(s)
- T Conroy
- Institut de Cancérologie de Lorraine and INSERM, INSPIIRE, Université de Lorraine, Nancy.
| | - F Castan
- Institut Régional du Cancer de Montpellier, Université de Montpellier, Montpellier
| | - P-L Etienne
- CARIO, Hôpital Privé des côtes d'Armor, Plérin
| | - E Rio
- Institut de Cancérologie de l'Ouest-Site René Gauducheau, Saint-Herblain
| | | | | | - V Vendrely
- Centre Hospitalier et Universitaire de Bordeaux, Hôpital Haut-Lévêque, Pessac
| | - X Artignan
- Centre Hospitalier Privé Saint-Grégoire, Saint-Grégoire
| | - O Bouché
- CHU Reims, Université de Reims Champagne-Ardenne, Reims
| | - D Gargot
- Centre Hospitalier de Blois, Blois
| | | | | | - C Louvet
- Institut Mutualiste Montsouris, Paris
| | - C Morand
- Centre Hospitalier Départemental Vendée, site de la Roche-sur-Yon, La Roche-sur-Yon
| | | | | | | | - S Gourgou
- Institut Régional du Cancer de Montpellier, Université de Montpellier, Montpellier
| | | | - C Borg
- University Hospital of Besançon, CIC-BT1431, Besançon, France
| |
Collapse
|
29
|
Nakamura Y, Tsukada Y, Matsuhashi N, Murano T, Shiozawa M, Takahashi Y, Oki E, Goto M, Kagawa Y, Kanazawa A, Ohta T, Ouchi A, Bando H, Uchigata H, Notake C, Ikematsu H, Yoshino T. Colorectal Cancer Recurrence Prediction Using a Tissue-Free Epigenomic Minimal Residual Disease Assay. Clin Cancer Res 2024; 30:4377-4387. [PMID: 39110016 PMCID: PMC11443202 DOI: 10.1158/1078-0432.ccr-24-1651] [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: 05/29/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 10/02/2024]
Abstract
PURPOSE Posttreatment detection of ctDNA is strongly predictive of recurrence. Most minimal/molecular residual disease assays require prior tissue testing to guide ctDNA analysis, resulting in lengthy time to initial results and unevaluable patients. EXPERIMENTAL DESIGN We assessed a tissue-free assay (Guardant Reveal) that bioinformatically evaluates >20,000 epigenomic regions for ctDNA detection in 1,977 longitudinally collected postoperative plasma samples from 342 patients with resected colorectal cancer. RESULTS We observed sensitive and specific detection of minimal/molecular residual disease associated with clinically meaningful differences in recurrence-free intervals at each time point evaluated with a median lead time of 5.3 months. The longitudinal sensitivity in stage II or higher colon cancer was 81%. Sensitivity increased with serial measurement and varied by recurrence site: higher for liver (100%) versus lung (53%) and peritoneal (40%). Sensitivity among patients with rectal cancer was 60% owing to a high proportion of lung metastases. Specificity was 98.2% among 1,461 posttreatment samples (99.1% among those with follow-up longer than the upper IQR of the lead time observed in this study). CONCLUSIONS Our data demonstrate the potential clinical utility of ctDNA as a tool to improve the management of stage II and higher colorectal cancer with a methodology that is noninvasive, accessible, and allows for rapid evaluation to inform clinical decisions.
Collapse
Affiliation(s)
- Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Yuichiro Tsukada
- Department of Colorectal Surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery and Pediatric Surgery, Graduate School of Medicine, Gifu University, Gifu, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, Japan
| | - Tatsuro Murano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| | - Manabu Shiozawa
- Department of Gastroenterological Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Yusuke Takahashi
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University Hospital, Fukuoka, Japan
| | - Masahiro Goto
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Takatsuki, Japan
| | - Yoshinori Kagawa
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Japan
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Akiyoshi Kanazawa
- Department of Surgery, Shimane Prefectural Central Hospital, Izumo, Japan
| | - Takashi Ohta
- Department of Clinical Oncology, Kansai Rosai Hospital, Amagasaki, Japan
| | - Akira Ouchi
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Hiroshi Uchigata
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Chiemi Notake
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Hiroaki Ikematsu
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
30
|
Tivey A, Lee RJ, Clipson A, Hill SM, Lorigan P, Rothwell DG, Dive C, Mouliere F. Mining nucleic acid "omics" to boost liquid biopsy in cancer. Cell Rep Med 2024; 5:101736. [PMID: 39293399 PMCID: PMC11525024 DOI: 10.1016/j.xcrm.2024.101736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/22/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024]
Abstract
Treatments for cancer patients are becoming increasingly complex, and there is a growing desire from clinicians and patients for biomarkers that can account for this complexity to support informed decisions about clinical care. To achieve precision medicine, the new generation of biomarkers must reflect the spatial and temporal heterogeneity of cancer biology both between patients and within an individual patient. Mining the different layers of 'omics in a multi-modal way from a minimally invasive, easily repeatable, liquid biopsy has increasing potential in a range of clinical applications, and for improving our understanding of treatment response and resistance. Here, we detail the recent developments and methods allowing exploration of genomic, epigenomic, transcriptomic, and fragmentomic layers of 'omics from liquid biopsy, and their integration in a range of applications. We also consider the specific challenges that are posed by the clinical implementation of multi-omic liquid biopsies.
Collapse
Affiliation(s)
- Ann Tivey
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Rebecca J Lee
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Alexandra Clipson
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Steven M Hill
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Paul Lorigan
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Dominic G Rothwell
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Caroline Dive
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Florent Mouliere
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
31
|
Sweeney CJ, Petry R, Xu C, Childress M, He J, Fabrizio D, Gjoerup O, Morley S, Catlett T, Assaf ZJ, Yuen K, Wongchenko M, Shah K, Gupta P, Hegde P, Pasquina LW, Mariathasan S, Graf RP, Powles T. Circulating Tumor DNA Assessment for Treatment Monitoring Adds Value to PSA in Metastatic Castration-Resistant Prostate Cancer. Clin Cancer Res 2024; 30:4115-4122. [PMID: 38990098 PMCID: PMC11393539 DOI: 10.1158/1078-0432.ccr-24-1096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/01/2024] [Accepted: 07/09/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE Enzalutamide after abiraterone progression is commonly used in metastatic castration-resistant prostate cancer despite a low rate of clinical benefit. Analyzing IMbassador250, a phase III trial assessing enzalutamide with or without atezolizumab after abiraterone, we hypothesized that baseline and early changes in circulating tumor DNA (ctDNA) tumor fraction (TF) may identify patients more likely to exhibit survival benefit from enzalutamide. EXPERIMENTAL DESIGN ctDNA was quantified from plasma samples using a tissue-agnostic assay without buffy coat sequencing. Baseline ctDNA TF, changes in ctDNA TF from baseline to cycle 3 day 1 (C3D1), and detection at C3D1 alone were compared with overall response rate, radiographic progression-free survival (rPFS), median OS (mOS), and 50% reduction in PSA. RESULTS ctDNA TF detection at baseline and/or C3D1 was associated with shorter rPFS and OS in 494 evaluable patients. Detection of ctDNA TF at C3D1, with or without detection at cycle 1 day 1, was associated with worse rPFS and mOS than lack of detection. When ctDNA TF and PSA response at C3D1 were discordant, patients with (ctDNA TF undetected/PSA not reduced) had more favorable outcomes than (ctDNA TF detected/PSA reduced; mOS 22.1 vs. 16 months; P < 0.001). CONCLUSIONS In a large cohort of patients with metastatic castration-resistant prostate cancer receiving enzalutamide after abiraterone, we demonstrate the utility of a new tissue-agnostic assay for monitoring molecular response based on ctDNA TF detection and dynamics. ctDNA TF provides a minimally invasive, complementary biomarker to PSA testing and may refine personalized treatment approaches.
Collapse
Affiliation(s)
- Christopher J. Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide SA, Australia.
| | - Russell Petry
- Foundation Medicine, Inc., Cambridge, Massachusetts.
| | - Chang Xu
- Foundation Medicine, Inc., Cambridge, Massachusetts.
| | | | - Jie He
- Foundation Medicine, Inc., Cambridge, Massachusetts.
| | | | - Ole Gjoerup
- Foundation Medicine, Inc., Cambridge, Massachusetts.
| | | | | | - Zoe J. Assaf
- Genentech, Inc., South San Francisco, California.
| | - Kobe Yuen
- Genentech, Inc., South San Francisco, California.
| | | | - Kalpit Shah
- Genentech, Inc., South San Francisco, California.
| | | | - Priti Hegde
- Foundation Medicine, Inc., Cambridge, Massachusetts.
| | | | | | - Ryon P. Graf
- Foundation Medicine, Inc., Cambridge, Massachusetts.
| | - Thomas Powles
- Saint Bartholomew’s Hospital, London, United Kingdom.
| |
Collapse
|
32
|
Zhang R, Zou C, Zeng L, Zhang Y. Perioperative immunotherapy in nonsmall cell lung cancer. Curr Opin Oncol 2024:00001622-990000000-00210. [PMID: 39246174 DOI: 10.1097/cco.0000000000001098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
PURPOSE OF REVIEW To evaluate and summarize the current clinical efficacy, safety, treatment patterns, and potential biomarkers, to guide future treatment strategies for nonsmall cell lung cancer (NSCLC), improve patient prognosis, and provide a scientific basis for personalized therapy. RECENT FINDINGS In recent years, the class of immune checkpoint inhibitors (ICIs), with programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors at the helm, has catalyzed groundbreaking advancements within the perioperative treatment milieu for NSCLC. With the positive results of several phase III clinical trials, perioperative immunotherapy has been confirmed to significantly reduce the risk of postoperative recurrence in resectable NSCLC, becoming the new standard for perioperative treatment of stages II to III NSCLC. With the advent of the perioperative immunotherapy era, clinical issues such as the selection of the treatment population, the choice of regimen, the duration of treatment, whether patients with pCR need further adjuvant therapy, and the comprehensive management of patients throughout the perioperative period have attracted widespread attention. SUMMARY The perioperative treatment of NSCLC has fully entered the era of immunotherapy. Multiple clinical studies have confirmed that perioperative immunotherapy can significantly improve the survival benefit of resectable stages II to III NSCLC, establishing a new standard for the perioperative treatment of stages II to III NSCLC.
Collapse
Affiliation(s)
- Renzhi Zhang
- Department of Medical Oncology, Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang
| | - Chun Zou
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Liang Zeng
- Department of Medical Oncology, Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yongchang Zhang
- Department of Medical Oncology, Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
33
|
Chibaudel B, Raeisi M, Cohen R, Yothers G, Goldberg RM, Bachet JB, Wolmark N, Yoshino T, Schmoll HJ, Kerr R, Lonardi S, George TJ, Shacham-Shmueli E, Shi Q, André T, de Gramont A. Assessment of the Addition of Oxaliplatin to Fluoropyrimidine-Based Adjuvant Chemotherapy in Patients With High-Risk Stage II Colon Cancer: An ACCENT Pooled Analysis. J Clin Oncol 2024:JCO2400394. [PMID: 39231393 DOI: 10.1200/jco.24.00394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/30/2024] [Accepted: 06/17/2024] [Indexed: 09/06/2024] Open
Abstract
PURPOSE The adjuvant treatment for stage III colon cancer (CC) is chemotherapy combining fluoropyrimidine (FP) and oxaliplatin (OX). FP regimen plus OX (FPOX) may benefit in high-risk stage II CC. We performed a pooled analysis of pivotal MOSAIC and C-07 studies evaluating FPOX for the treatment of high-risk stage II CC according to prognostic factors, number of high-risk factors, and current clinicopathologic risk classification on the basis of T stage, tumor perforation, and number of lymph nodes examined. PATIENTS AND METHODS One thousand five hundred and ninety-five patients with stage II CC receiving FP or FPOX were pooled. The overall survival (OS) benefit of OX was analyzed using Kaplan-Meier curves and unadjusted Cox models stratified by study. Three thousand and fifty-nine patients with stage III CC were used only for interaction tests between the allocated chemotherapy and stage. RESULTS In the pooled analysis of stage II patients, independent prognostic factors in multivariable analysis were sex, age, perforation/obstruction, and tumor sidedness. There was a significant interaction in OS between stage and allocated chemotherapy with hazard ratios (HRs) of 1.03 for stage II (95% CI, 0.82 to 1.29; P = .813) and 0.82 for stage III (95% CI, 0.73 to 0.92; P = .001; Pint = .073). There was no benefit from the addition of OX to FP for any of the prognostic factors. The number of high-risk factors tested was not predictive of OX benefit. According to the currently agreed clinicopathologic risk classification, no OS benefit of OX was observed, as HR was 0.86 (95% CI, 0.63 to 1.18; P = .349). CONCLUSION No OS benefit of adjuvant OX was found in high-risk stage II CC, regardless of the definition used to characterize tumors as having a high risk for recurrence. Hence, our analysis suggests that OX should not be the standard of care for adjuvant chemotherapy for stage II CC, even in high-risk patients.
Collapse
Affiliation(s)
- Benoist Chibaudel
- Department of Medical Oncology, Franco-British Hospital, Fondation Cognacq-Jay, Cancérologie Paris Ouest, Levallois-Perret, France
| | | | - Romain Cohen
- Sorbonne Université and Department of Medical Oncology, Saint-Antoine Hospital, Paris, France
| | - Greg Yothers
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA
| | - Richard M Goldberg
- Department of Medicine, West Virginia University Cancer Institute, Morgantown, WV
| | - Jean-Baptiste Bachet
- Hepato-gastroenterology and Digestive Oncology Department, Sorbonne University, Pitié Salpêtrière Hospital, APHP, Paris, France
| | | | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hans-Joachim Schmoll
- Division of Clinical Research in Oncology, Martin-Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Rachel Kerr
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Sara Lonardi
- Medical Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Thomas J George
- Division of Hematology and Oncology, University of Florida, Gainesville, FL
| | | | - Qian Shi
- Department of Quantitative Health Science, Mayo Clinic, Rochester, MN
| | - Thierry André
- Sorbonne Université and Department of Medical Oncology, Saint-Antoine Hospital, Paris, France
- ARCAD Foundation, Paris, France
| | - Aimery de Gramont
- ARCAD Foundation, Paris, France
- Department of Medical Oncology, Franco-British Hospital, Levallois-Perret, France
| |
Collapse
|
34
|
Wehrle CJ, Hong H, Kamath S, Schlegel A, Fujiki M, Hashimoto K, Kwon DCH, Miller C, Walsh RM, Aucejo F. Tumor Mutational Burden From Circulating Tumor DNA Predicts Recurrence of Hepatocellular Carcinoma After Resection: An Emerging Biomarker for Surveillance. Ann Surg 2024; 280:504-513. [PMID: 38860385 DOI: 10.1097/sla.0000000000006386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
OBJECTIVE Describe the utility of circulating tumor DNA in the postoperative surveillance of hepatocellular carcinoma (HCC). BACKGROUND Current biomarkers for HCC like alpha-fetoprotein (AFP) are lacking. Circulating tumor DNA (ctDNA) has shown promise in colorectal and lung cancers, but its utility in HCC remains relatively unknown. METHODS Patients with HCC undergoing curative-intent resection from November 1, 2020, to July 1, 2023, received ctDNA testing using the Guardant360 platform. Tumor mutational burden (TMB) is calculated as the number of somatic mutations-per-megabase of genomic material identified. RESULTS Forty-seven patients had postoperative ctDNA testing. The mean follow-up was 27 months, and the maximum was 43.2 months. Twelve patients (26%) experienced recurrence. Most (n=41/47, 87.2%) had identifiable ctDNA postoperatively; 55.3% (n=26) were TMB-not detected versus 45.7% (n=21) TMB-detectable. Postoperative identifiable ctDNA was not associated with RFS ( P =0.518). Detectable TMB was associated with reduced RFS (6.9 vs 14.7 mo, P =0.049). There was a higher rate of recurrence in patients with TMB (n=9/21, 42.9%, vs n=3/26, 11.5%, P =0.02). Area under the curve for TMB-prediction of recurrence was 0.752 versus 0.550 for AFP. ROC analysis established a TMB cutoff of 4.8mut/mB for predicting post-operative recurrence ( P =0.002) and RFS ( P =0.025). AFP was not correlated with RFS using the lab-normal cutoff (<11 ng/mL, P =0.682) or the cutoff established by ROC analysis (≥4.6 ng/mL, P =0.494). TMB-high was associated with poorer RFS on cox-regression analysis (hazard ratio=5.386, 95% CI: 1.109-26.160, P =0.037), while microvascular invasion ( P =0.853) and AFP ( P =0.439) were not. CONCLUSIONS Identifiable TMB on postoperative ctDNA predicts HCC recurrence and outperformed AFP in this cohort. Perioperative ctDNA may be a useful surveillance tool following curative-intent hepatectomy. Larger-scale studies are needed to confirm this utility and investigate additional applications in HCC patients, including the potential for prophylactic treatment in patients with residual TMB after resection.
Collapse
Affiliation(s)
- Chase J Wehrle
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland
| | - Hanna Hong
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland
| | - Suneel Kamath
- Department of Hematology and Oncology, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH
| | - Andrea Schlegel
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland
| | - Masato Fujiki
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland
| | - Koji Hashimoto
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland
| | - David Choon Hyuck Kwon
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland
| | - Charles Miller
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland
| | - R Matthew Walsh
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland
| | - Federico Aucejo
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland
| |
Collapse
|
35
|
De Simoni E, Spagnolo F, Gandini S, Gaeta A, Rizzetto G, Molinelli E, Simonetti O, Offidani A, Queirolo P. Circulating tumor DNA-based assessment of molecular residual disease in non-metastatic melanoma. Cancer Treat Rev 2024; 129:102788. [PMID: 38908229 DOI: 10.1016/j.ctrv.2024.102788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
In patients with resected non-metastatic melanoma, the liquid biopsy for the assessment of molecular residual disease (MRD) by circulating tumour DNA (ctDNA) represents a promising tool to stratify the risk and to monitor tumour evolution. However, its validation requires the demonstration of analytical validity, clinical validity and utility. Indeed, the development of sensitive and specific assays can optimize prognostication and eventually help clinicians to modulate adjuvant treatments, in order to improve clinical outcomes. Data about ctDNA-guided prognosis stratification is emerging, but clinical trials assessing ctDNA-guided therapeutic decisions are still ongoing. This review aims to depict the role of ctDNA-based MRD assessment in patients with non-metastatic melanoma and to provide a roadmap to face challenges for its introduction into clinical practice.
Collapse
Affiliation(s)
- Edoardo De Simoni
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Francesco Spagnolo
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Surgical Sciences and Integrated Diagnostics (DISC), Plastic Surgery Division, University of Genova, Genova, Italy
| | - Sara Gandini
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Aurora Gaeta
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giulio Rizzetto
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Elisa Molinelli
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Paola Queirolo
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
36
|
Polasek TM, Peck RW. Beyond Population-Level Targets for Drug Concentrations: Precision Dosing Needs Individual-Level Targets that Include Superior Biomarkers of Drug Responses. Clin Pharmacol Ther 2024; 116:602-612. [PMID: 38328977 DOI: 10.1002/cpt.3197] [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: 11/05/2023] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
The purpose of precision dosing is to increase the chances of therapeutic success in individual patients. This is achieved in practice by adjusting doses to reach precision dosing targets determined previously in relevant populations, ideally with robust supportive evidence showing improved clinical outcomes compared with standard dosing. But is this implicit assumption of translatable population-level precision dosing targets correct and the best for all patients? In this review, the types of precision dosing targets and how they are determined are outlined, problems with the translatability of these targets to individual patients are identified, and ways forward to address these challengers are proposed. Achieving improved clinical outcomes to support precision dosing over standard dosing is currently hampered by applying population-level targets to all patients. Just as "one-dose-fits-all" may be an inappropriate philosophy for drug treatment overall, a "one-target-fits-all" philosophy may limit the broad clinical benefits of precision dosing. Defining individual-level precision dosing targets may be needed for greatest therapeutic success. Superior future precision dosing targets will integrate several biomarkers that together account for the multiple sources of drug response variability.
Collapse
Affiliation(s)
- Thomas M Polasek
- Centre for Medicine Use and Safety, Monash University, Melbourne, Victoria, Australia
- CMAX Clinical Research, Adelaide, South Australia, Australia
| | - Richard W Peck
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Pharma Research & Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
37
|
Hu H, Zhang J, Li Y, Wang X, Wang Z, Wang H, Kang L, Liu P, Lan P, Wu X, Zhen Y, Pei H, Huang Z, Zhang H, Chen W, Zeng Y, Lai J, Wei H, Huang X, Chen J, Chen J, Tao K, Xu Q, Peng X, Liang J, Cai G, Ding K, Ding Z, Hu M, Zhang W, Tang B, Hong C, Cao J, Huang Z, Cao W, Li F, Wang X, Wang C, Huang Y, Zhao Y, Cai Y, Ling J, Xie X, Wu Z, Shi L, Ling L, Liu H, Wang J, Huang M, Deng Y. Neoadjuvant Chemotherapy With Oxaliplatin and Fluoropyrimidine Versus Upfront Surgery for Locally Advanced Colon Cancer: The Randomized, Phase III OPTICAL Trial. J Clin Oncol 2024; 42:2978-2988. [PMID: 38564700 DOI: 10.1200/jco.23.01889] [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: 08/31/2023] [Revised: 11/29/2023] [Accepted: 02/02/2024] [Indexed: 04/04/2024] Open
Abstract
PURPOSE The role of neoadjuvant chemotherapy (NAC) in colon cancer remains unclear. This trial investigated whether 3 months of modified infusional fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) or capecitabine and oxaliplatin (CAPOX) as NAC could improve outcomes in patients with locally advanced colon cancer versus upfront surgery. PATIENTS AND METHODS OPTICAL was a randomized, phase III trial in patients with clinically staged locally advanced colon cancer (T3 with extramural spread into the mesocolic fat ≥5 mm or T4). Patients were randomly assigned 1:1 to receive six preoperative cycles of mFOLFOX6 or four cycles of CAPOX, followed by surgery and adjuvant chemotherapy (NAC group), or immediate surgery and the physician's choice of adjuvant chemotherapy (upfront surgery group). The primary end point was 3-year disease-free survival (DFS) assessed in the modified intention-to-treat (mITT) population. RESULTS Between January 2016 and April 2021, of the 752 patients enrolled, 744 patients were included in the mITT analysis (371 in the NAC group; 373 in the upfront surgery group). At a median follow-up of 48.0 months (IQR, 46.0-50.1), 3-year DFS rates were 82.1% in the NAC group and 77.5% in the upfront surgery group (stratified hazard ratio [HR], 0.74 [95% CI, 0.54 to 1.03]). The R0 resection was achieved in 98% of patients who underwent surgery in both groups. Compared with upfront surgery, NAC resulted in a 7% pathologic complete response rate (pCR), significantly lower rates of advanced tumor staging (pT3-4: 77% v 94%), lymph node metastasis (pN1-2: 31% v 46%), and potentially improved overall survival (stratified HR, 0.44 [95% CI, 0.25 to 0.77]). CONCLUSION NAC with mFOLFOX6 or CAPOX did not show a significant DFS benefit. However, this neoadjuvant approach was safe, resulted in substantial pathologic downstaging, and appears to be a viable therapeutic option for locally advanced colon cancer.
Collapse
Affiliation(s)
- Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianwei Zhang
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yunfeng Li
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Xiaozhong Wang
- Department of Gastrointestinal Surgery, Shantou Central Hospital, Shantou, People's Republic of China
| | - Ziqiang Wang
- Department of General Surgery, Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Liang Kang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ping Liu
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaojian Wu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yunhuan Zhen
- Department of Colorectal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Haiping Pei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhongcheng Huang
- Department of General Surgery, Hunan Provincial People's Hospital, Changsha, People's Republic of China
| | - Hao Zhang
- Department of General Surgery, Dongguan Kanghua Hospital, Dongguan, People's Republic of China
| | - Wenbin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yongming Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, People's Republic of China
| | - Jiajun Lai
- Department of Gastrointestinal Surgery, Yuebei People's Hospital, Shaoguan, People's Republic of China
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jiansi Chen
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Jigui Chen
- Department of Surgery, The Eighth Hospital of Wuhan, Wuhan, People's Republic of China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qingwen Xu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Xiang Peng
- Department of Gastrointestinal Surgery, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Junlin Liang
- Department of Coloproctological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Guanfu Cai
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangzhou, People's Republic of China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Cancer Center, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zhijie Ding
- Department of Gastrointestinal Surgery, Zhongshan Hospital, Xiamen University, Xiamen, People's Republic of China
| | - Ming Hu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Wei Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Bo Tang
- Department of General Surgery, The First Hospital Affiliated to Army Medical University, Chongqing, People's Republic of China
| | - Chuyuan Hong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jie Cao
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, People's Republic of China
| | - Zonghai Huang
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Wuteng Cao
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Fangqian Li
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xinhua Wang
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chao Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yandong Zhao
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yue Cai
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiayu Ling
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaoyu Xie
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zehua Wu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lishuo Shi
- Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Ling
- Department of Medical Statistics, School of Public Health, and Center for Migrant Health Policy, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Biostatistics Shared Resource, Rutgers Cancer Institute of New Jersey, Rutgers School of Public Health, Brunswick, NJ
| | - Jianping Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meijin Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
38
|
Marinello A, Tagliamento M, Pagliaro A, Conci N, Cella E, Vasseur D, Remon J, Levy A, Dall'Olio FG, Besse B. Circulating tumor DNA to guide diagnosis and treatment of localized and locally advanced non-small cell lung cancer. Cancer Treat Rev 2024; 129:102791. [PMID: 38963991 DOI: 10.1016/j.ctrv.2024.102791] [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: 04/23/2024] [Revised: 06/15/2024] [Accepted: 06/22/2024] [Indexed: 07/06/2024]
Abstract
Liquid biopsy is a minimally invasive method for biomarkers detection in body fluids, particularly in blood, which offers an elevated and growing number of clinical applications in oncology. As a result of the improvement in the techniques for DNA analysis, above all next-generation sequencing (NGS) assays, circulating tumor DNA (ctDNA) has become the most informing tumor-derived material for most types of cancer, including non-small cell lung cancer (NSCLC). Although ctDNA concentration is higher in patients with advanced tumors, it can be detected even in patients with early-stage disease. Therefore, numerous clinical applications of ctDNA in the management of early-stage lung cancer are emerging, such as lung cancer screening, the identification of minimal residual disease (MRD), and the prediction of relapse before radiologic progression. Moreover, a high number of clinical trials are ongoing to better define the impact of ctDNA evaluation in this setting. Aim of this review is to offer a comprehensive overview of the most relevant implementations in using ctDNA for the management of early-stage lung cancer, addressing available data, technical aspects, limitations, and future perspectives.
Collapse
Affiliation(s)
- Arianna Marinello
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; INSERM Unit 1030 - Molecular Radiotherapy and Therapeutic Innovation, Gustave Roussy, Villejuif, France
| | - Marco Tagliamento
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Department of Internal Medicine and Medical Specialties, University of Genova, Genova, Italy.
| | - Arianna Pagliaro
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Department of Medical Oncology, IRCCS Istituto Clinico Humanitas, Rozzano, Italy
| | - Nicole Conci
- Department of Medical Oncology, IRCCS Sant'Orsola-Malpighi, Bologna, Italy
| | - Eugenia Cella
- Department of Internal Medicine and Medical Specialties, University of Genova, Genova, Italy
| | - Damien Vasseur
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Jordi Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Antonin Levy
- Department of Radiotherapy, Gustave Roussy, Villejuif, France
| | | | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
39
|
Cañellas-Socias A, Sancho E, Batlle E. Mechanisms of metastatic colorectal cancer. Nat Rev Gastroenterol Hepatol 2024; 21:609-625. [PMID: 38806657 DOI: 10.1038/s41575-024-00934-z] [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] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
Despite extensive research and improvements in understanding colorectal cancer (CRC), its metastatic form continues to pose a substantial challenge, primarily owing to limited therapeutic options and a poor prognosis. This Review addresses the emerging focus on metastatic CRC (mCRC), which has historically been under-studied compared with primary CRC despite its lethality. We delve into two crucial aspects: the molecular and cellular determinants facilitating CRC metastasis and the principles guiding the evolution of metastatic disease. Initially, we examine the genetic alterations integral to CRC metastasis, connecting them to clinically marked characteristics of advanced CRC. Subsequently, we scrutinize the role of cellular heterogeneity and plasticity in metastatic spread and therapy resistance. Finally, we explore how the tumour microenvironment influences metastatic disease, emphasizing the effect of stromal gene programmes and the immune context. The ongoing research in these fields holds immense importance, as its future implications are projected to revolutionize the treatment of patients with mCRC, hopefully offering a promising outlook for their survival.
Collapse
Affiliation(s)
- Adrià Cañellas-Socias
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| | - Elena Sancho
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
40
|
Pu W, Chen F, Tang Y, Qu Y, Han Y, Zha J, Jin J, Kong F. Potential value of detection of minimal residual disease in colorectal cancer following radical resection. Chin J Cancer Res 2024; 36:442-454. [PMID: 39246709 PMCID: PMC11377885 DOI: 10.21147/j.issn.1000-9604.2024.04.07] [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: 05/29/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
Although there has been significant advancement in the identification and management of colorectal cancer (CRC) in recent years, there is still room for improvement in the current standard treatment regimen. One area of concern is the lack of reliable tumor markers to predict treatment efficacy and guide tailored care. Due to its dynamic, effective, and non-invasive benefits over tissue biopsy, the detection of minimal or molecular residual lesions (MRD) based on circulating tumor DNA (ctDNA) is beneficial to the clinical development of drugs for patients with CRC after radical treatment, as well as for continuous monitoring of tumor recurrence and malignancy molecular gene evolution. The detection of ctDNA can currently be used to guide individual postoperative auxiliary treatment decisions (upgrade or downgrade treatment) in CRC, stratify the risk of clinical recurrence more precisely, and predict the risk of recurrence in advance of imaging examination, according to a large number of observational or prospective clinical studies. With increasing clarity comes the possibility of selecting a regimen of treatment based on postoperative ctDNA, which also improves the accuracy of clinical recurrence risk assessment for CRC. Therefore, it is anticipated that the identification of ctDNA would alter the current framework for dealing with CRC and lead to individualized, stratified precision therapy; however, additional confirmation will require subsequent high-quality, prospective, large-scale randomized controlled studies. This article will provide an overview of the definition and clinical significance of MRD, the primary indications and technological challenges for MRD detection, along with the advancement in clinical research about ctDNA detection following radical resection of the CRC.
Collapse
Affiliation(s)
- Wenji Pu
- Medical Department of Shenzhen University/General Hospital of Shenzhen University/Academy of Clinical Medicine of Shenzhen University, Shenzhen 518055, China
- Department of Clinical Oncology, the University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Radiotherapy, National Cancer Center/National Cancer Clinical Medical Research Center/Shenzhen Hospital, Cancer Hospital of Peking Union Medical College, Chinese Academy of Medical Sciences, Shenzhen 518116, China
| | - Fang Chen
- Department of Clinical Oncology, the University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Yuan Tang
- Department of Radiotherapy, National Cancer Center/National Cancer Clinical Medical Research Center/Shenzhen Hospital, Cancer Hospital of Peking Union Medical College, Chinese Academy of Medical Sciences, Shenzhen 518116, China
| | - Yanling Qu
- Department of Clinical Oncology, the University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Yunzhu Han
- Department of Clinical Oncology, the University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Jiandong Zha
- Department of Clinical Oncology, the University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Jing Jin
- Department of Radiotherapy, National Cancer Center/National Cancer Clinical Medical Research Center/Shenzhen Hospital, Cancer Hospital of Peking Union Medical College, Chinese Academy of Medical Sciences, Shenzhen 518116, China
| | - Fengming Kong
- Department of Clinical Oncology, the University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
41
|
Crisafulli G. Mutational Signatures in Colorectal Cancer: Translational Insights, Clinical Applications, and Limitations. Cancers (Basel) 2024; 16:2956. [PMID: 39272814 PMCID: PMC11393898 DOI: 10.3390/cancers16172956] [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: 08/12/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
A multitude of exogenous and endogenous processes have the potential to result in DNA damage. While the repair mechanisms are typically capable of correcting this damage, errors in the repair process can result in mutations. The findings of research conducted in 2012 indicate that mutations do not occur randomly but rather follow specific patterns that can be attributed to known or inferred mutational processes. The process of mutational signature analysis allows for the inference of the predominant mutational process for a given cancer sample, with significant potential for clinical applications. A deeper comprehension of these mutational signatures in CRC could facilitate enhanced prevention strategies, facilitate the comprehension of genotoxic drug activity, predict responses to personalized treatments, and, in the future, inform the development of targeted therapies in the context of precision oncology. The efforts of numerous researchers have led to the identification of several mutational signatures, which can be categorized into different mutational signature references. In CRC, distinct mutational signatures are identified as correlating with mismatch repair deficiency, polymerase mutations, and chemotherapy treatment. In this context, a mutational signature analysis offers considerable potential for enhancing minimal residual disease (MRD) tests in stage II (high-risk) and stage III CRC post-surgery, stratifying CRC based on the impacts of genetic and epigenetic alterations for precision oncology, identifying potential therapeutic vulnerabilities, and evaluating drug efficacy and guiding therapy, as illustrated in a proof-of-concept clinical trial.
Collapse
|
42
|
Kramer A, Greuter MJE, Schraa SJ, Vink GR, Phallen J, Velculescu VE, Meijer GA, van den Broek D, Koopman M, Roodhart JML, Fijneman RJA, Retèl VP, Coupé VMH. Early evaluation of the effectiveness and cost-effectiveness of ctDNA-guided selection for adjuvant chemotherapy in stage II colon cancer. Ther Adv Med Oncol 2024; 16:17588359241266164. [PMID: 39175989 PMCID: PMC11339739 DOI: 10.1177/17588359241266164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/18/2024] [Indexed: 08/24/2024] Open
Abstract
Background Current patient selection for adjuvant chemotherapy (ACT) after curative surgery for stage II colon cancer (CC) is suboptimal, causing overtreatment of high-risk patients and undertreatment of low-risk patients. Postoperative circulating tumor DNA (ctDNA) could improve patient selection for ACT. Objectives We conducted an early model-based evaluation of the (cost-)effectiveness of ctDNA-guided selection for ACT in stage II CC in the Netherlands to assess the conditions for cost-effective implementation. Methods A validated Markov model, simulating 1000 stage II CC patients from diagnosis to death, was supplemented with ctDNA data. Five ACT selection strategies were evaluated: the current guideline (pT4, pMMR), ctDNA-only, and three strategies that combined ctDNA status with pT4 and pMMR status in different ways. For each strategy, the costs, life years, quality-adjusted life years (QALYs), recurrences, and CC deaths were estimated. Sensitivity analyses were performed to assess the impact of the costs of ctDNA testing, strategy adherence, ctDNA as a predictive biomarker, and ctDNA test performance. Results Model predictions showed that compared to current guidelines, the ctDNA-only strategy was less effective (+2.2% recurrences, -0.016 QALYs), while the combination strategies were more effective (-3.6% recurrences, +0.038 QALYs). The combination strategies were not cost-effective, since the incremental cost-effectiveness ratio was €67,413 per QALY, exceeding the willingness-to-pay threshold of €50,000 per QALY. Sensitivity analyses showed that the combination strategies would be cost-effective if the ctDNA test costs were lower than €1500, or if ctDNA status was predictive of treatment response, or if the ctDNA test performance improved substantially. Conclusion Adding ctDNA to current high-risk clinicopathological features (pT4 and pMMR) can improve patient selection for ACT and can also potentially be cost-effective. Future studies should investigate the predictive value of post-surgery ctDNA status to accurately evaluate the cost-effectiveness of ctDNA testing for ACT decisions in stage II CC.
Collapse
Affiliation(s)
- Astrid Kramer
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, De Boelelaan 1089a, Amsterdam, Noord-Holland 1081 HV, The Netherlands
| | - Marjolein J. E. Greuter
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Suzanna J. Schraa
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Geraldine R. Vink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Research and Development, IKNL, Utrecht, The Netherlands
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Victor E. Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerrit A. Meijer
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daan van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jeanine M. L. Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Valesca P. Retèl
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Veerle M. H. Coupé
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
43
|
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 2024. [PMID: 39155652 DOI: 10.1002/jso.27838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [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 Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Noah X Tocci
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Keyue Sun
- Cleveland Clinic Foundation, Lerner Research Institute, Inflammation & Immunity, Cleveland, Ohio, USA
| | - Chunbao Jiao
- Cleveland Clinic Foundation, Lerner Research Institute, Inflammation & Immunity, Cleveland, Ohio, USA
| | - Hanna Hong
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Abby Gross
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Erlind Allkushi
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Melis Uysal
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Maureen Whitsett Linganna
- Department of Gastroenterology, Hepatology, and Nutrition, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Katheryn Stackhouse
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Koji Hashimoto
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Andrea Schlegel
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
- Cleveland Clinic Foundation, Lerner Research Institute, Inflammation & Immunity, Cleveland, Ohio, USA
| | - R Matthew Walsh
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Charles Miller
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - David C H Kwon
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| | - Federico Aucejo
- Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, Ohio, USA
| |
Collapse
|
44
|
Tocci NX, Wehrle CJ, 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. Circulating tumor DNA in management of primary liver malignancy: A review of the literature and future directions. J Surg Oncol 2024. [PMID: 39155663 DOI: 10.1002/jso.27825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/14/2024] [Indexed: 08/20/2024]
Abstract
Primary liver malignancies are a serious and challenging global health concern. The most common primary tumors are hepatocellular carcinoma and cholangiocarcinoma. These diseases portend poor prognosis when presenting with progressive, extensive disease. There is a critical need for improved diagnosis, therapeutic intervention, and monitoring surveillance in liver-related malignancies. Liquid biopsy using ctDNA provides an opportunity for growth within these domains for liver-related malignancy. However, ctDNA is relatively understudied in this field compared with other solid tumor types, possibly due to the complex nature of the pathology. In this review, we aim to discuss ctDNA, the current literature, and future directions of this technology within primary liver malignancies.
Collapse
Affiliation(s)
- Noah X Tocci
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Chase J Wehrle
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Keyue Sun
- Lerner Research Institute, Inflammation & Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Chunbao Jiao
- Lerner Research Institute, Inflammation & Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Hanna Hong
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Abby Gross
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Erlind Allkushi
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Melis Uysal
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Maureen Whitsett Linganna
- Department of Gastroenterology, Hepatology, and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Katheryn Stackhouse
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Koji Hashimoto
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Andrea Schlegel
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Lerner Research Institute, Inflammation & Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - R Matthew Walsh
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Charles Miller
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - David C H Kwon
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Federico Aucejo
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
45
|
Slater S, Bryant A, Aresu M, Begum R, Chen HC, Peckitt C, Lazaro-Alcausi R, Carter P, Anandappa G, Khakoo S, Melcher L, Potter V, Marti FM, Huang J, Branagan G, George N, Abulafi M, Duff S, Raja A, Gupta A, West N, Bucheit L, Rich T, Chau I, Cunningham D, Starling N. Tissue-Free Liquid Biopsies Combining Genomic and Methylation Signals for Minimal Residual Disease Detection in Patients with Early Colorectal Cancer from the UK TRACC Part B Study. Clin Cancer Res 2024; 30:3459-3469. [PMID: 38864835 PMCID: PMC11325146 DOI: 10.1158/1078-0432.ccr-24-0226] [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: 01/24/2024] [Revised: 04/08/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
PURPOSE The absence of postoperative circulating tumor DNA (ctDNA) identifies patients with resected colorectal cancer (CRC) with low recurrence risk for adjuvant chemotherapy (ACT) de-escalation. Our study presents the largest resected CRC cohort to date with tissue-free minimal residual disease (MRD) detection. EXPERIMENTAL DESIGN TRACC (tracking mutations in cell-free tumor DNA to predict relapse in early colorectal cancer) included patients with stage I to III resectable CRC. Prospective longitudinal plasma collection for ctDNA occurred pre- and postsurgery, post-ACT, every 3 months for year 1 and every 6 months in years 2 and 3 with imaging annually. The Guardant Reveal assay evaluated genomic and methylation signals. The primary endpoint was 2-year recurrence-free survival (RFS) by postoperative ctDNA detection (NCT04050345). RESULTS Between December 2016 and August 2022, 1,203 were patients enrolled. Plasma samples (n = 997) from 214 patients were analyzed. One hundred forty-three patients were evaluable for the primary endpoint; 92 (64.3%) colon, 51 (35.7%) rectal; two (1.4%) stage I, 64 (44.8%) stage II, and 77 (53.8%) stage III. Median follow-up was 30.3 months (95% CI, 29.5-31.3). Two-year RFS was 91.1% in patients with ctDNA not detected postoperatively and 50.4% in those with ctDNA detected [HR, 6.5 (2.96-14.5); P < 0.0001]. Landmark negative predictive value (NPV) was 91.2% (95% CI, 83.9-95.9). Longitudinal sensitivity and specificity were 62.1% (95% CI, 42.2-79.3) and 85.9% (95% CI, 78.9-91.3), respectively. The median lead time from ctDNA detection to radiological recurrence was 7.3 months (IQR, 3.3-12.5; n = 9). CONCLUSIONS Tissue-free MRD detection with longitudinal sampling predicts recurrence in patients with stage I to III CRC without the need for tissue sequencing. The UK TRACC Part C study is currently investigating the potential for ACT de-escalation in patients with undetectable postoperative ctDNA, given the high NPV indicating a low likelihood of residual disease.
Collapse
Affiliation(s)
- Susanna Slater
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Annette Bryant
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Maria Aresu
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Ruwaida Begum
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Hsiang-Chi Chen
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Clare Peckitt
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | | | - Paul Carter
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | | | - Shelize Khakoo
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Lucinda Melcher
- North Middlesex University Hospital NHS Trust, London, United Kingdom
| | - Vanessa Potter
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Francisca M Marti
- Wrightington, Wigan and Leigh NHS Foundation Trust, Wigan, United Kingdom
| | - Joesph Huang
- Barking, Havering and Redbridge University Hospitals NHS Trust, Greater London, United Kingdom
| | | | - Nicol George
- Broomfield Hospital Mid and South Essex NHS Foundation Trust, Chelmsford, United Kingdom
| | - Muti Abulafi
- Croydon Health Services NHS Trust, Surrey, United Kingdom
| | - Sarah Duff
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Ashraf Raja
- Epsom and St Helier University Hospitals NHS Trust, Surrey, United Kingdom
| | - Ashish Gupta
- Epsom and St Helier University Hospitals NHS Trust, Surrey, United Kingdom
| | - Nicholas West
- Epsom and St Helier University Hospitals NHS Trust, Surrey, United Kingdom
| | | | | | - Ian Chau
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - David Cunningham
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Naureen Starling
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
46
|
Stetson D, Labrousse P, Russell H, Shera D, Abbosh C, Dougherty B, Barrett JC, Hodgson D, Hadfield J. Next-Generation Molecular Residual Disease Assays: Do We Have the Tools to Evaluate Them Properly? J Clin Oncol 2024; 42:2736-2740. [PMID: 38754043 DOI: 10.1200/jco.23.02301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/27/2024] [Accepted: 03/05/2024] [Indexed: 05/18/2024] Open
Affiliation(s)
- Dan Stetson
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, MA
| | - Paul Labrousse
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, MA
| | - Hugh Russell
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, MA
| | - David Shera
- Oncology Biometrics, AstraZeneca, Gaithersburg, MD
| | - Chris Abbosh
- Cancer Biomarker Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Brian Dougherty
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, MA
| | - J Carl Barrett
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, MA
| | - Darren Hodgson
- Cancer Biomarker Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - James Hadfield
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
47
|
Claus J, De Smet D, Breyne J, Wesolowski J, Himpe U, Demedts I, Martens GA. Patient-centric thresholding of Cobas® EGFR mutation Test v2 for surveillance of EGFR-mutated metastatic non-small cell lung cancer. Sci Rep 2024; 14:18191. [PMID: 39107402 PMCID: PMC11303541 DOI: 10.1038/s41598-024-68350-6] [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: 02/19/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
Cobas EGFR mutation Test v2 was FDA-approved as qualitative liquid biopsy for actionable EGFR variants in non-small cell lung cancer (NSCLC). It generates semiquantitative index (SQI) values that correlate with mutant allele levels, but decision thresholds for clinical use in NSCLC surveillance are lacking. We conducted long-term ctDNA monitoring in 20 subjects with EGFR-mutated NSCLC; resulting in a 155 on-treatment samples. We defined optimal SQI intervals to predict/rule-out progression within 12 weeks from sampling and performed orthogonal calibration versus deep-sequencing and digital PCR. SQI showed significant diagnostic power (AUC 0.848, 95% CI 0.782-0.901). SQI below 5 (63% of samples) had 93% (95% CI 87-96%) NPV, while SQI above 10 (25% of samples) had 69% (95% CI 56-80%) PPV. Cobas EGFR showed perfect agreement with sequencing (Kappa 0.860; 95% CI 0.674-1.00) and digital PCR. SQI values strongly (r: 0.910, 95% 0.821-0.956) correlated to mutant allele concentrations with SQI of 5 and 10 corresponding to 6-9 (0.2-0.3%) and 64-105 (1.1-1.6%) mutant allele copies/mL (VAF) respectively. Our dual-threshold classifier of SQI 0/5/10 yielded informative results in 88% of blood draws with high NPV and good overall clinical utility for patient-centric surveillance of metastatic NSCLC.
Collapse
Affiliation(s)
- Jonas Claus
- Department of Pulmonary Diseases, AZ Delta General Hospital, Roeselare, Belgium
| | - Dieter De Smet
- Department of Laboratory Medicine, AZ Delta General Hospital, Roeselare, Belgium
| | - Joke Breyne
- Department of Laboratory Medicine, AZ Delta General Hospital, Roeselare, Belgium
| | | | - Ulrike Himpe
- Department of Pulmonary Diseases, AZ Delta General Hospital, Roeselare, Belgium
| | - Ingel Demedts
- Department of Pulmonary Diseases, AZ Delta General Hospital, Roeselare, Belgium
| | - Geert A Martens
- Department of Laboratory Medicine, AZ Delta General Hospital, Roeselare, Belgium.
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium.
| |
Collapse
|
48
|
Guo G, Zhang Z, Zhang J, Wang D, Xu S, Liu G, Gao Y, Mei J, Yan Z, Zhao R, Wang M, Li T, Bu X. Predicting recurrent glioblastoma clinical outcome to immune checkpoint inhibition and low-dose bevacizumab with tumor in situ fluid circulating tumor DNA analysis. Cancer Immunol Immunother 2024; 73:193. [PMID: 39105794 PMCID: PMC11303371 DOI: 10.1007/s00262-024-03774-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/05/2024] [Indexed: 08/07/2024]
Abstract
OBJECTIVE Most recurrent glioblastoma (rGBM) patients do not benefit from immune checkpoint inhibition, emphasizing the necessity for response biomarkers. This study evaluates whether tumor in situ fluid (TISF) circulating tumor DNA (ctDNA) could serve as a biomarker for response to low-dose bevacizumab (Bev) plus anti-PD-1 therapy in rGBM patients, aiming to enhance systemic responses to immunotherapy. METHODS In this phase II trial, 32 GBM patients with first recurrence after standard therapy were enrolled and then received tislelizumab plus low-dose Bev each cycle. TISF samples were analyzed for ctDNA using a 551-gene panel before each treatment. RESULTS The median progression-free survival (mPFS) and overall survival (mOS) were 8.2 months (95% CI, 5.2-11.1) and 14.3 months (95% CI, 6.5-22.1), respectively. The 12-month OS was 43.8%, and the objective response rate was 56.3%. Patients with more than 20% reduction in the mutant allele fraction and tumor mutational burden after treatment were significantly associated with better prognosis compared to baseline TISF-ctDNA. Among detectable gene mutations, patients with MUC16 mutation, EGFR mutation & amplification, SRSF2 amplification, and H3F3B amplification were significantly associated with worse prognosis. CONCLUSIONS Low-dose Bev plus anti-PD-1 therapy significantly improves OS in rGBM patients, offering guiding significance for future individualized treatment strategies. TISF-ctDNA can monitor rGBM patients' response to combination therapy and guide treatment. CLINICAL TRIAL REGISTRATION This trial is registered with ClinicalTrials.gov, NCT05540275.
Collapse
Affiliation(s)
- Guangzhong Guo
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Ziyue Zhang
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Jiubing Zhang
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Dayang Wang
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Sensen Xu
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Guanzheng Liu
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Yushuai Gao
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Jie Mei
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Zhaoyue Yan
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Ruijiao Zhao
- Department of Pathology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Meiyun Wang
- Department of Radiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Tianxiao Li
- Henan Provincial Neurointerventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, China
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Xingyao Bu
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
49
|
Janssen FW, Lak NSM, Janda CY, Kester LA, Meister MT, Merks JHM, van den Heuvel-Eibrink MM, van Noesel MM, Zsiros J, Tytgat GAM, Looijenga LHJ. A comprehensive overview of liquid biopsy applications in pediatric solid tumors. NPJ Precis Oncol 2024; 8:172. [PMID: 39097671 PMCID: PMC11297996 DOI: 10.1038/s41698-024-00657-z] [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: 02/20/2024] [Accepted: 07/15/2024] [Indexed: 08/05/2024] Open
Abstract
Liquid biopsies are emerging as an alternative source for pediatric cancer biomarkers with potential applications during all stages of patient care, from diagnosis to long-term follow-up. While developments within this field are reported, these mainly focus on dedicated items such as a specific liquid biopsy matrix, analyte, and/or single tumor type. To the best of our knowledge, a comprehensive overview is lacking. Here, we review the current state of liquid biopsy research for the most common non-central nervous system pediatric solid tumors. These include neuroblastoma, renal tumors, germ cell tumors, osteosarcoma, Ewing sarcoma, rhabdomyosarcoma and other soft tissue sarcomas, and liver tumors. Within this selection, we discuss the most important or recent studies involving liquid biopsy-based biomarkers, anticipated clinical applications, and the current challenges for success. Furthermore, we provide an overview of liquid biopsy-based biomarker publication output for each tumor type based on a comprehensive literature search between 1989 and 2023. Per study identified, we list the relevant liquid biopsy-based biomarkers, matrices (e.g., peripheral blood, bone marrow, or cerebrospinal fluid), analytes (e.g., circulating cell-free and tumor DNA, microRNAs, and circulating tumor cells), methods (e.g., digital droplet PCR and next-generation sequencing), the involved pediatric patient cohort, and proposed applications. As such, we identified 344 unique publications. Taken together, while the liquid biopsy field in pediatric oncology is still behind adult oncology, potentially relevant publications have increased over the last decade. Importantly, steps towards clinical implementation are rapidly gaining ground, notably through validation of liquid biopsy-based biomarkers in pediatric clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Michael T Meister
- Princess Máxima Center, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Johannes H M Merks
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center, Utrecht, the Netherlands
- Wilhelmina Children's Hospital-Division of CHILDHEALTH, University Medical Center Utrech, University of Utrecht, Utrecht, the Netherlands
| | - Max M van Noesel
- Princess Máxima Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | | | - Godelieve A M Tytgat
- Princess Máxima Center, Utrecht, the Netherlands
- Department of Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Leendert H J Looijenga
- Princess Máxima Center, Utrecht, the Netherlands.
- Department of Pathology, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
50
|
Gottschalk Z, Cohen SA. Use of Circulating Tumor DNA to Guide Decision-making in Adjuvant Colon Cancer. Curr Oncol Rep 2024; 26:959-966. [PMID: 38842605 DOI: 10.1007/s11912-024-01565-y] [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: 05/28/2024] [Indexed: 06/07/2024]
Abstract
PURPOSE OF REVIEW The use of circulating tumor DNA (ctDNA) assays to guide clinical decision-making in early-stage colon cancer is an area of rapidly advancing active research. With assays clinically available, clinicians must be informed how to best use this novel tool to treat patients. RECENT FINDINGS Recent observational and prospective studies have suggested that ctDNA has potential to guide clinical decision-making in early-stage colon cancer by detecting minimal residual disease (MRD) and predicting recurrence risks. MRD-negative patients may be able to de-escalate or forgo adjuvant chemotherapy (ACT) without compromising disease-free survival or overall survival, while MRD-positive patients may benefit significantly from ACT. Recent and ongoing studies have given reason for optimism about the future of ctDNA as a useful biomarker for clinicians treating early-stage colon cancer. Data thus far are mostly limited to observational studies; inconsistent results highlight the need for caution. As more evidence emerges, ctDNA may become standard of care for colon cancer patients.
Collapse
Affiliation(s)
- Zachary Gottschalk
- Fred Hutchinson Cancer Center, 825 Eastlake Ave E, LG-465, Seattle, WA, 98177, USA
| | - Stacey A Cohen
- Fred Hutchinson Cancer Center, 825 Eastlake Ave E, LG-465, Seattle, WA, 98177, USA.
- University of Washington, Seattle, WA, USA.
| |
Collapse
|