1
|
Kobayashi Y, Suzuki Y, Seishima R, Chikaishi Y, Matsuoka H, Nakamura K, Shigeta K, Okabayashi K, Hiro J, Otsuka K, Uyama I, Saya H, Nishihara H, Suda K, Kitagawa Y. Utility of comprehensive genomic profiling combined with machine learning for prognostic stratification in stage II/III colorectal cancer after adjuvant chemotherapy. Int J Clin Oncol 2025; 30:926-934. [PMID: 40095334 DOI: 10.1007/s10147-025-02722-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND AND PURPOSE Accurate recurrence risk evaluation in patients with stage II and III colorectal cancer (CRC) remains difficult. Traditional histopathological methods frequently fall short in predicting outcomes after adjuvant chemotherapy. This study aims to evaluate the use of comprehensive genomic profiling combined with machine learning for prognostic risk stratification in patients with CRC. METHODS A machine learning model was developed using a training cohort of 52 patients with stage II/III CRC who underwent curative surgery at Fujita Health University Hospital. Genomic DNA was isolated from formalin-fixed, paraffin-embedded tissue sections and analyzed with a 160 cancer-related gene panel. The random forest algorithm was used to determine key genes affecting recurrence-free survival. The model was validated by developing a risk score with internal and external cohorts, including 44 patients from Keio University Hospital. RESULTS Six key genes (KRAS, KIT, SMAD4, ARID2, NF1, and FBXW7) were determined as significant prognostic risk predictors. A risk score system integrating these genes with clinicopathological factors effectively stratified patients in both internal (p < 0.001) and external cohorts (p = 0.017). CONCLUSIONS This study reveals that machine learning, combined with comprehensive genomic profiling, significantly improves prognostic risk stratification in patients with stage II/III CRC after adjuvant chemotherapy. This approach provides a promising tool for individualized treatment strategies, warranting further validation with larger cohorts.
Collapse
Affiliation(s)
- Yosuke Kobayashi
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | | | - Ryo Seishima
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan.
- Department of Surgery, Keio University School of Medicine, Tokyo, 160‑8582, Japan.
- Department of Surgery, Inagi Municipal Hospital, Tokyo, 206-0801, Japan.
| | - Yuko Chikaishi
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Hiroshi Matsuoka
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Kohei Nakamura
- Center for Cancer Genomics, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kohei Shigeta
- Department of Surgery, Keio University School of Medicine, Tokyo, 160‑8582, Japan
| | - Koji Okabayashi
- Department of Surgery, Keio University School of Medicine, Tokyo, 160‑8582, Japan
| | - Junichiro Hiro
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Koki Otsuka
- Department of Advanced Robotic and Endoscopic Surgery, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Ichiro Uyama
- Department of Advanced Robotic and Endoscopic Surgery, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Hiroshi Nishihara
- Center for Cancer Genomics, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Koichi Suda
- Department of Surgery, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, 160‑8582, Japan
| |
Collapse
|
2
|
Lișcu HD, Verga N, Atasiei DI, Badiu DC, Dumitru AV, Ultimescu F, Pavel C, Stefan RE, Manole DC, Ionescu AI. Biomarkers in Colorectal Cancer: Actual and Future Perspectives. Int J Mol Sci 2024; 25:11535. [PMID: 39519088 PMCID: PMC11546354 DOI: 10.3390/ijms252111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Biomarkers in colorectal cancer (CRC) are of great interest in the current literature due to improvements in techniques such as liquid biopsy and next-generation sequencing (NGS). However, screening methods vary globally, with multi-target stool DNA (mt-sDNA) predominantly used in the USA and, more recently, the Cologuard Plus; biomarkers such as the Galectins family and septins show promise in early detection. Gut microbiome assessments, such as Fusobacterium nucleatum, are under intense exploration. Diagnostic tests, such as circulating DNA analysis via NGS, exhibit effectiveness and are being increasingly adopted. Circulating tumor cells emerge as potential alternatives to traditional methods in terms of diagnosis and prognosis. Predictive biomarkers are well established in guidelines; nonetheless, with the aid of machine learning and artificial intelligence, these biomarkers may be improved. This review critically explores the actual dynamic landscape of CRC biomarkers and future, promising biomarkers involved in screening, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Horia-Dan Lișcu
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Nicolae Verga
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Dimitrie-Ionuț Atasiei
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
| | - Dumitru-Cristinel Badiu
- Department of Surgery, Bagdasar Arseni Clinical Emergency Hospital, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Adrian Vasile Dumitru
- Department of Pathology, University Emergency Hospital Bucharest, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Flavia Ultimescu
- Department of Pathology, Institute of Oncology Alexandru Trestioreanu, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Christopher Pavel
- Department of Gastroenterology, Floreasca Emergency Hospital Bucharest, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Roxana-Elena Stefan
- General Surgery Department, Clinic of General and Esophageal Surgery, Sf. Maria Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Diandra-Carmen Manole
- Department of Endocrinology, Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bd., 050474 Bucharest, Romania;
| | - Andreea-Iuliana Ionescu
- Discipline of Oncological Radiotherapy and Medical Imaging, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (H.-D.L.); (A.-I.I.)
- Radiotherapy Department, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| |
Collapse
|
3
|
van den Ende T, van der Pol Y, Creemers A, Moldovan N, Boers D, van Berge Henegouwen MI, Hulshof MC, Cillessen SA, van Grieken NC, Pegtel DM, Derks S, Bijlsma MF, Mouliere F, van Laarhoven HW. Genome-wide and panel-based cell-free DNA characterization of patients with resectable esophageal adenocarcinoma. J Pathol 2023; 261:286-297. [PMID: 37615198 DOI: 10.1002/path.6175] [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: 03/13/2023] [Revised: 06/08/2023] [Accepted: 07/06/2023] [Indexed: 08/25/2023]
Abstract
Circulating tumor DNA (ctDNA) holds promise in resectable esophageal adenocarcinoma (EAC) to predict patient outcome but is not yet sensitive enough to be clinically applicable. Our aim was to combine ctDNA mutation data with shallow whole-genome sequencing (sWGS)-derived copy number tumor fraction estimates (ichorCNA) to improve pathological response and survival prediction in EAC. In total, 111 stage II/III EAC patients with baseline (n = 111), post-neoadjuvant chemoradiotherapy (nCRT) (n = 68), and pre-surgery (n = 92) plasma samples were used for ctDNA characterization. sWGS (<5× coverage) was performed on all time-point samples, and copy number aberrations were estimated using ichorCNA. Baseline and pre-surgery samples were sequenced using a custom amplicon panel for mutation detection. Detection of baseline ctDNA was successful in 44.3% of patients by amplicon sequencing and 10.5% by ichorCNA. Combining both, ctDNA could be detected in 50.5% of patients. Baseline ctDNA positivity was related to higher T stage (cT3, 4) (p = 0.017). There was no relationship between pathological response and baseline ctDNA positivity. However, baseline ctDNA metrics (variant allele frequency > 1% or ichorCNA > 3%) were associated with a high risk of disease progression [HR = 2.23 (95% CI 1.22-4.07), p = 0.007]. The non-clearance of a baseline variant or ichorCNA > 3% in pre-surgery samples was related to early progression [HR = 4.58 (95% CI 2.22-9.46), p < 0.001]. Multi-signal analysis improves detection of ctDNA and can be used for prognostication of resectable EAC patients. Future studies should explore the potential of multi-modality sequencing for risk stratification and treatment adaptation based on ctDNA results. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Tom van den Ende
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Ymke van der Pol
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands
| | - Aafke Creemers
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Norbert Moldovan
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands
| | - Dries Boers
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands
| | - Mark I van Berge Henegouwen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, The Netherlands
| | - Maarten Ccm Hulshof
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, The Netherlands
- Department of Radiotherapy, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Saskia Agm Cillessen
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Nicole Ct van Grieken
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - D Michiel Pegtel
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands
| | - Sarah Derks
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Maarten F Bijlsma
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Florent Mouliere
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands
| | - Hanneke Wm van Laarhoven
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Jiang H, Zhou S, Li G. Novel biomarkers used for early diagnosis and tyrosine kinase inhibitors as targeted therapies in colorectal cancer. Front Pharmacol 2023; 14:1189799. [PMID: 37719843 PMCID: PMC10502318 DOI: 10.3389/fphar.2023.1189799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common and second most lethal type of cancer worldwide, presenting major health risks as well as economic costs to both people and society. CRC survival chances are significantly higher if the cancer is diagnosed and treated early. With the development of molecular biology, numerous initiatives have been undertaken to identify novel biomarkers for the early diagnosis of CRC. Pathological disorders can be diagnosed at a lower cost with the help of biomarkers, which can be detected in stool, blood, and tissue samples. Several lines of evidence suggest that the gut microbiota could be used as a biomarker for CRC screening and treatment. CRC treatment choices include surgical resection, chemotherapy, immunotherapy, gene therapy, and combination therapies. Targeted therapies are a relatively new and promising modality of treatment that has been shown to increase patients' overall survival (OS) rates and can inhibit cancer cell development. Several small-molecule tyrosine kinase inhibitors (TKIs) are being investigated as potential treatments due to our increasing awareness of CRC's molecular causes and oncogenic signaling. These compounds may inhibit critical enzymes in controlling signaling pathways, which are crucial for CRC cells' development, differentiation, proliferation, and survival. On the other hand, only one of the approximately 42 TKIs that demonstrated anti-tumor effects in pre-clinical studies has been licensed for clinical usage in CRC. A significant knowledge gap exists when bringing these tailored medicines into the clinic. As a result, the emphasis of this review is placed on recently discovered biomarkers for early diagnosis as well as tyrosine kinase inhibitors as possible therapy options for CRC.
Collapse
|
5
|
Muendlein A, Geiger K, Heinzle C, Gaenger S, Winder T, Severgnini L, Reimann P, Brandtner EM, Leiherer A, Drexel H, Decker T, Nonnenbroich C, Dechow T. Cell-free circulating RAS mutation concentrations significantly impact the survival of metastatic colorectal cancer patients. J Cancer Res Clin Oncol 2023; 149:6435-6444. [PMID: 36763171 DOI: 10.1007/s00432-023-04623-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/27/2023] [Indexed: 02/11/2023]
Abstract
PURPOSE RAS mutations are predictors of an adverse outcome in EGFR-targeted therapies and have been proposed as prognostic biomarkers of survival in metastatic colorectal cancer (mCRC). The analysis of circulating tumor DNA from plasma samples, known as liquid biopsies, has indicated that the RAS mutation status may change over time, potentially affecting patients' prognosis. To further evaluate the clinical validity of RAS mutation retesting using liquid biopsies, we prospectively investigated the impact of the circulating quantitative RAS mutation status on the course of mCRC. METHODS The present study included 81 consecutively recruited patients with mCRC. We used targeted next-generation sequencing of circulating cell-free DNA to determine and quantify plasma RAS mutation status. RESULTS Patients with a RAS mutation detected by liquid biopsy (37%; n = 30) were at increased risk of death during the follow-up period compared to RAS wild-type patients. Patients with evidence of a RAS mutation in the primary tumor but a putative RAS mutation loss in plasma (28%; n = 11) showed a prolonged survival compared to patients with a preserved RAS mutation status. Also, circulating RAS mutation concentrations significantly affected the outcome: The mortality risk of patients with a high RAS mutation concentration increased fivefold compared to subjects with a putative RAS mutation loss or low RAS mutation concentration. CONCLUSION Our results emphasize the clinical value of circulating RAS mutations in managing mCRC.
Collapse
Affiliation(s)
- Axel Muendlein
- Molecular Biology Laboratory, Vorarlberg Institute for Vascular Investigation and Treatment, Dornbirn, Austria.
| | - Kathrin Geiger
- Molecular Biology Laboratory, Vorarlberg Institute for Vascular Investigation and Treatment, Dornbirn, Austria
- Medical Central Laboratories, Feldkirch, Austria
| | - Christine Heinzle
- Molecular Biology Laboratory, Vorarlberg Institute for Vascular Investigation and Treatment, Dornbirn, Austria
- Medical Central Laboratories, Feldkirch, Austria
| | - Stella Gaenger
- Molecular Biology Laboratory, Vorarlberg Institute for Vascular Investigation and Treatment, Dornbirn, Austria
| | - Thomas Winder
- Department of Haematology and Oncology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Luciano Severgnini
- Department of Haematology and Oncology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
- Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Patrick Reimann
- Department of Haematology and Oncology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
- Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Eva Maria Brandtner
- Molecular Biology Laboratory, Vorarlberg Institute for Vascular Investigation and Treatment, Dornbirn, Austria
| | - Andreas Leiherer
- Molecular Biology Laboratory, Vorarlberg Institute for Vascular Investigation and Treatment, Dornbirn, Austria
- Medical Central Laboratories, Feldkirch, Austria
| | - Heinz Drexel
- Molecular Biology Laboratory, Vorarlberg Institute for Vascular Investigation and Treatment, Dornbirn, Austria
- Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
- Drexel University College of Medicine, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
6
|
Mirza S, Bhadresha K, Mughal MJ, McCabe M, Shahbazi R, Ruff P, Penny C. Liquid biopsy approaches and immunotherapy in colorectal cancer for precision medicine: Are we there yet? Front Oncol 2023; 12:1023565. [PMID: 36686736 PMCID: PMC9853908 DOI: 10.3389/fonc.2022.1023565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths globally, with nearly half of patients detected in the advanced stages. This is due to the fact that symptoms associated with CRC often do not appear until the cancer has reached an advanced stage. This suggests that CRC is a cancer with a slow progression, making it curable and preventive if detected in its early stage. Therefore, there is an urgent clinical need to improve CRC early detection and personalize therapy for patients with this cancer. Recently, liquid biopsy as a non-invasive or nominally invasive approach has attracted considerable interest for its real-time disease monitoring capability through repeated sample analysis. Several studies in CRC have revealed the potential for liquid biopsy application in a real clinical setting using circulating RNA/miRNA, circulating tumor cells (CTCs), exosomes, etc. However, Liquid biopsy still remains a challenge since there are currently no promising results with high specificity and specificity that might be employed as optimal circulatory biomarkers. Therefore, in this review, we conferred the plausible role of less explored liquid biopsy components like mitochondrial DNA (mtDNA), organoid model of CTCs, and circulating cancer-associated fibroblasts (cCAFs); which may allow researchers to develop improved strategies to unravel unfulfilled clinical requirements in CRC patients. Moreover, we have also discussed immunotherapy approaches to improve the prognosis of MSI (Microsatellite Instability) CRC patients using neoantigens and immune cells in the tumor microenvironment (TME) as a liquid biopsy approach in detail.
Collapse
Affiliation(s)
- Sheefa Mirza
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,Department of Internal Medicine, Common Epithelial Cancer Research Centre, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kinjal Bhadresha
- Hematology/Oncology Division, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Muhammed Jameel Mughal
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Science, The George Washington University, Washington, DC, United States
| | - Michelle McCabe
- Department of Anatomical Pathology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg, South Africa
| | - Reza Shahbazi
- Hematology/Oncology Division, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Paul Ruff
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,Department of Internal Medicine, Common Epithelial Cancer Research Centre, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,Department of Internal Medicine, Common Epithelial Cancer Research Centre, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,*Correspondence: Clement Penny,
| |
Collapse
|
7
|
Hayashi T, Yoshida Y, Yamada T, Tanaka K, Shimaoka H, Kajitani R, Munechika T, Nagano H, Matsumoto Y, Komono A, Sakamoto R, Aisu N, Yoshimatsu G, Yoshimura F, Hasegawa S. Relationship between perioperative oncological evaluation and recurrence using circulating tumor DNA with KRAS mutation in patients with colorectal cancer. Cancer Med 2022; 11:3126-3135. [PMID: 35312176 PMCID: PMC9385586 DOI: 10.1002/cam4.4677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 12/29/2022] Open
Abstract
Background The detection of circulating tumor DNA (ctDNA) in colorectal cancer (CRC) by liquid biopsy may have prognostic information. In this perioperative study, we evaluate if there is a relationship between mutant allele frequency (MAF) of Kirsten rat sarcoma viral oncogene homolog (KRAS) and tumor recurrence and how that could be useful in the early detection of recurrence. Methods Among 304 cases of colorectal cancer surgery, ctDNA was sampled from the perioperative blood of 84 patients with CRC with KRAS mutation (exon 4 p.A146T, exon 4 p.A146V, exon 2 p.G12A, exon 2 p.G12C, exon 2 p.G12D, exon 2 p.G12S, exon 2 p.G12V, exon 2 p.G13D, exon 3 p.Q61H) and analyzed using the digital polymerase chain reaction system. The median observation period was 26 months. Results Although the relationship between the perioperative MAF of KRAS and recurrence was not proved, tumor diameter, tumor depth, and stage were correlated with the preoperative MAF of KRAS (p = 0.034, p = 0.002, p = 0.008). However, tumor diameter, tumor depth, and stage did not correlate with MAF of KRAS at postoperative day 30. Conclusions In this study, pathological tumor size, tumor depth, and stage were correlated with preoperative MAF of KRAS, but it was unreliable to predict recurrence by detection of ctDNA with KRAS mutation in the perioperative period of colorectal surgery.
Collapse
Affiliation(s)
- Takaomi Hayashi
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoichiro Yoshida
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Teppei Yamada
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Keita Tanaka
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Hideki Shimaoka
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Ryuji Kajitani
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Taro Munechika
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Hideki Nagano
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshiko Matsumoto
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Akira Komono
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Ryohei Sakamoto
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Naoya Aisu
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Gumpei Yoshimatsu
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Fumihiro Yoshimura
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Suguru Hasegawa
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| |
Collapse
|
8
|
Parisi A, Porzio G, Pulcini F, Cannita K, Ficorella C, Mattei V, Delle Monache S. What Is Known about Theragnostic Strategies in Colorectal Cancer. Biomedicines 2021; 9:biomedicines9020140. [PMID: 33535557 PMCID: PMC7912746 DOI: 10.3390/biomedicines9020140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/08/2023] Open
Abstract
Despite the paradigmatic shift occurred in recent years for defined molecular subtypes in the metastatic setting treatment, colorectal cancer (CRC) still remains an incurable disease in most of the cases. Therefore, there is an urgent need for new tools and biomarkers for both early tumor diagnosis and to improve personalized treatment. Thus, liquid biopsy has emerged as a minimally invasive tool that is capable of detecting genomic alterations from primary or metastatic tumors, allowing the prognostic stratification of patients, the detection of the minimal residual disease after surgical or systemic treatments, the monitoring of therapeutic response, and the development of resistance, establishing an opportunity for early intervention before imaging detection or worsening of clinical symptoms. On the other hand, preclinical and clinical evidence demonstrated the role of gut microbiota dysbiosis in promoting inflammatory responses and cancer initiation. Altered gut microbiota is associated with resistance to chemo drugs and immune checkpoint inhibitors, whereas the use of microbe-targeted therapies including antibiotics, pre-probiotics, and fecal microbiota transplantation can restore response to anticancer drugs, promote immune response, and therefore support current treatment strategies in CRC. In this review, we aim to summarize preclinical and clinical evidence for the utilization of liquid biopsy and gut microbiota in CRC.
Collapse
Affiliation(s)
- Alessandro Parisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy; (G.P.); (K.C.); (C.F.)
| | - Giampiero Porzio
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy; (G.P.); (K.C.); (C.F.)
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Fanny Pulcini
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Katia Cannita
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy; (G.P.); (K.C.); (C.F.)
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Corrado Ficorella
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy; (G.P.); (K.C.); (C.F.)
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, via Angelo Maria Ricci 35A, 02100 Rieti, Italy;
| | - Simona Delle Monache
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Correspondence: ; Tel.: +39-086-243-3569
| |
Collapse
|
9
|
Lueong SS, Herbst A, Liffers ST, Bielefeld N, Horn PA, Tannapfel A, Reinacher-Schick A, Hinke A, Hegewisch-Becker S, Kolligs FT, Siveke JT. Serial Circulating Tumor DNA Mutational Status in Patients with KRAS-Mutant Metastatic Colorectal Cancer from the Phase 3 AIO KRK0207 Trial. Clin Chem 2020; 66:1510-1520. [PMID: 33257977 DOI: 10.1093/clinchem/hvaa223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 08/28/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND We assessed the usefulness of circulating tumor DNA (ctDNA) pre- or post-treatment initiation for outcome prediction and treatment monitoring in metastatic colorectal cancer (mCRC). METHODS Droplet digital PCR was used to measure absolute mutant V-Ki-ras2 Kirsten rat sarcoma viral oncogene ((mut)KRAS) ctDNA concentrations in 214 healthy controls (plasma and sera) and in 151 tissue-based mutKRAS positive patients with mCRC from the prospective multicenter phase 3 trial AIO KRK0207. Serial mutKRAS ctDNA was analyzed prior to and 2-3 weeks after first-line chemotherapy initiation with fluoropyrimidine, oxaliplatin, and bevacizumab in patients with mCRC and correlated with clinical parameters. RESULTS mut KRAS ctDNA was detected in 74.8% (113/151) of patients at baseline and in 59.6% (90/151) at follow-up. mutKRAS ctDNA at baseline and follow-up was associated with poor overall survival (OS) (hazard ratio [HR] =1.88, 95% confidence interval [CI] 1.20-2.95; HR = 2.15, 95% CI 1.47-3.15) and progression-free survival (PFS) (HR = 2.53, 95% CI 1.44-4.46; HR = 1.90, 95% CI 1.23-2.95), respectively. mutKRAS ctDNA clearance at follow-up conferred better disease control (P = 0.0075), better OS (log-rank P = 0.0018), and PFS (log-rank P = 0.0018). Measurable positive mutKRAS ctDNA at follow-up was the strongest and most significant independent prognostic factor on OS in multivariable analysis (HR = 2.31, 95% CI 1.40-3.25). CONCLUSIONS Serial analysis of circulating mutKRAS concentrations in mCRC has prognostic value. Post treatment mutKRAS concentrations 2 weeks after treatment initiation were associated with therapeutic response in multivariable analysis and may be an early response predictor in patients receiving first-line combination chemotherapy. CLINICALTRIALSGOV IDENTIFIER NCT00973609.
Collapse
Affiliation(s)
- Smiths S Lueong
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Medicine Essen, Essen, Germany.,Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Andreas Herbst
- Institute of Laboratory Medicine, University of Munich, Munich, Germany.,German Cancer Consortium (DKTK, Partner Site Munich) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Sven-Thorsten Liffers
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Medicine Essen, Essen, Germany.,Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Nicola Bielefeld
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Medicine Essen, Essen, Germany.,Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Peter A Horn
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany.,Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | | | - Anke Reinacher-Schick
- Department of Hematology, Oncology and Palliative Care, St. Josef-Hospital, Ruhr-University Bochum
| | - Axel Hinke
- CCRC: Cancer Clinical Research Consulting, Düsseldorf, Germany
| | | | - Frank T Kolligs
- German Cancer Consortium (DKTK, Partner Site Munich) and German Cancer Research Center, DKFZ, Heidelberg, Germany.,Department of Medicine, Division of- Gastroenterology, Hepatology & Infectiology, Helios Clinic Berlin-Buch, Berlin, Germany.,Department of Medicine II, University of Munich, Munich, Germany
| | - Jens T Siveke
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Medicine Essen, Essen, Germany.,Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| |
Collapse
|
10
|
Gandini S, Zanna I, De Angelis SP, Cocorocchio E, Queirolo P, Lee JH, Carlino MS, Mazzarella L, Achutti Duso B, Palli D, Raimondi S, Caini S. Circulating tumour DNA and melanoma survival: A systematic literature review and meta-analysis. Crit Rev Oncol Hematol 2020; 157:103187. [PMID: 33276181 DOI: 10.1016/j.critrevonc.2020.103187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
We reviewed and meta-analysed the available evidence (until December 2019) about circulating tumour DNA (ctDNA) levels and melanoma patients survival. We included twenty-six studies (>2000 patients overall), which included mostly stage III-IV cutaneous melanoma patients and differed widely in terms of systemic therapy received and somatic mutations that were searched. Patients with detectable ctDNA before treatment had worse progression-free survival (PFS) (summary hazard ratio (SHR) 2.47, 95 % confidence intervals (CI) 1.85-3.29) and overall survival (OS) (SHR 2.98, 95 % CI 2.26-3.92), with no difference by tumour stage. ctDNA detectability during follow-up was associated with poorer PFS (SHR 4.27, 95 %CI 2.75-6.63) and OS (SHR 3.91, 95 %CI 1.97-7.78); in the latter case, the association was stronger (p = 0.01) for stage IV vs. III melanomas. Between-estimates heterogeneity was low for all pooled estimates. ctDNA is a strong prognostic biomarker for advanced-stage melanoma patients, robust across tumour (e.g. genomic profile) and patients (e.g. systemic therapy) characteristics.
Collapse
Affiliation(s)
- Sara Gandini
- Molecular and Pharmaco-Epidemiology Unit Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Ines Zanna
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Simone Pietro De Angelis
- Molecular and Pharmaco-Epidemiology Unit Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Emilia Cocorocchio
- Division of Medical Oncology of Melanoma, Sarcoma and Rare Tumors, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Paola Queirolo
- Division of Medical Oncology of Melanoma, Sarcoma and Rare Tumors, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Jenny H Lee
- Department of Clinical Medicine, Macquarie University, Sydney, Australia
| | - Matteo S Carlino
- Department of Clinical Oncology, Westmead and Blacktown Hospitals, Melanoma Institute of Australia and the University of Sydney, Sydney, Australia
| | - Luca Mazzarella
- Molecular and Pharmaco-Epidemiology Unit Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Bruno Achutti Duso
- Molecular and Pharmaco-Epidemiology Unit Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Domenico Palli
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Sara Raimondi
- Molecular and Pharmaco-Epidemiology Unit Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Saverio Caini
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy.
| |
Collapse
|
11
|
Ljubic B, Pavlovski M, Alshehri J, Roychoudhury S, Bajic V, Van Neste C, Obradovic Z. Comorbidity network analysis and genetics of colorectal cancer. INFORMATICS IN MEDICINE UNLOCKED 2020. [DOI: 10.1016/j.imu.2020.100492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|