1
|
Mouhanna P, Ståhlberg A, Andersson D, Albu‐Kareem A, Elinder E, Eriksson O, Kavanagh A, Kovács A, Larsson KF, Linderholm B, Uminska M, Österlund T, Howell SJ, Ekholm M. Integration of personalised ultrasensitive ctDNA monitoring of patients with metastatic breast cancer to reduce imaging requirements. Int J Cancer 2025; 156:1509-1517. [PMID: 39692755 PMCID: PMC11826139 DOI: 10.1002/ijc.35292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/19/2024]
Abstract
Circulating tumour DNA (ctDNA) is an emerging biomarker for monitoring cancers. The personalised disease monitoring in metastatic breast cancer (PDM-MBC) study is an ongoing study instigated to evaluate ctDNA as a biomarker to individualise imaging requirements in patients with MBC. Patients receiving first-line endocrine therapy (aromatase inhibitor + cyclin-dependent kinase 4/6 inhibitor) had plasma samples collected pre-treatment, weeks 2 and 4, and concurrently with imaging until progressive disease (PD). Here, we apply an experimental analytical workflow for ultrasensitive ctDNA analysis, utilising personalised ctDNA panels designed from mutations identified in tumour tissue, and present results for 24 patients. Twenty patients (83%) had detectable ctDNA pre-treatment. The median progression-free survival was 25.6 months, and 13 patients experienced PD, with rising ctDNA detected at or prior to PD in 12 patients (92%). If imaging had been omitted until the detection of rising ctDNA for at least one mutation, 68% (n = 71) of the scans performed amongst ctDNA-positive patients would have been avoided. Our results demonstrate that integration of personalised ctDNA monitoring of patients with MBC has potential to substantially reduce the imaging needs in patients showing ctDNA response to treatment.
Collapse
Affiliation(s)
- Pia Mouhanna
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of OncologyRyhov County HospitalJönköpingSweden
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical Genetics and GenomicsSahlgrenska University HospitalGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
| | - Daniel Andersson
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | | | | | - Olle Eriksson
- Futurum – The Academy for Health and CareJönköping CountySweden
| | - Amy Kavanagh
- Department of Medical OncologyThe Christie NHS Foundation TrustManchesterUK
| | - Anikó Kovács
- Department of Clinical PathologySahlgrenska University HospitalGothenburgSweden
| | - Karolina F. Larsson
- Sahlgrenska Center for Cancer Research, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of OncologySahlgrenska University HospitalGothenburgSweden
| | - Barbro Linderholm
- Sahlgrenska Center for Cancer Research, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of OncologySahlgrenska University HospitalGothenburgSweden
| | - Monika Uminska
- Department of OncologyKalmar County HospitalKalmarSweden
| | - Tobias Österlund
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical Genetics and GenomicsSahlgrenska University HospitalGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
| | - Sacha J. Howell
- Department of Medical OncologyThe Christie NHS Foundation TrustManchesterUK
- Division of Cancer Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Maria Ekholm
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of OncologyRyhov County HospitalJönköpingSweden
- Department of Biomedical and Clinical Sciences, Division of OncologyLinköping UniversityLinköpingSweden
| |
Collapse
|
2
|
Fornerod MJJ, Alvarez-Fernandez A, Füredi M, Rajendran AA, Prieto-Simón B, Voelcker NH, Guldin S. Block copolymer-assembled nanopore arrays enable ultrasensitive label-free DNA detection. NANOSCALE HORIZONS 2025; 10:760-769. [PMID: 39905896 PMCID: PMC11795167 DOI: 10.1039/d4nh00466c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025]
Abstract
DNA detection via nanoporous-based electrochemical biosensors is a promising method for rapid pathogen identification and disease diagnosis. These sensors detect electrical current variations caused by DNA hybridization in a nanoporous layer on an electrode. Current fabrication techniques for the typically micrometers-thick nanoporous layer often suffer from insufficient control over nanopore dimensions and involve complex fabrication steps, including handling and stacking of a brittle porous membrane. Here, we introduce a bottom-up fabrication process based on the self-assembly of high molecular weight block copolymers with sol-gel precursors to create an inorganic nanoporous thin film directly on electrode surfaces. This approach eliminates the need for elaborate manipulation of the nanoporous membrane, provides fine control over the structural features, and enables surface modification with DNA capture probes. Using this nanoarchitecture with a thickness of 150 nm, we detected DNA sequences derived from 16S rRNA gene fragments of the E. coli genome electrochemically in less than 20 minutes, achieving a limit of detection of 30 femtomolar (fM) and a limit of quantification of 500 fM. This development marks a significant step towards a portable, rapid, and accurate DNA detection system.
Collapse
Affiliation(s)
| | - Alberto Alvarez-Fernandez
- Department of Chemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK.
- Materials Physics Center (MPC) - CSICUPV/EHU, Paseo Manuel de Lardizabal 5, Donostia-San Sebastián (Gipuzkoa), 20018, Spain
| | - Máté Füredi
- Department of Chemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK.
- Semilab Co. Ltd., Prielle Kornélia u. 2, Budapest, 1117, Hungary
| | | | - Beatriz Prieto-Simón
- Institute of Chemical Research of Catalonia, The Barcelona Institute of Science and Technology, Av. Països Catalans, 16, Tarragona, 43007, Spain
- ICREA, Pg. Lluís Companys 23, Barcelona, 08010, Spain
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, 3168, Australia
| | - Stefan Guldin
- Department of Chemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK.
- Technical University of Munich, Department of Life Science Engineering, Gregor-Mendel-Straße 4, Freising, 85354, Germany
- TUMCREATE, 1 CREATE Way, Singapore 138602, Singapore
| |
Collapse
|
3
|
Kotsifa E, Saffioti F, Mavroeidis VK. Cholangiocarcinoma: The era of liquid biopsy. World J Gastroenterol 2025; 31:104170. [PMID: 40124277 PMCID: PMC11924015 DOI: 10.3748/wjg.v31.i11.104170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 03/13/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive and heterogeneous malignancy arising from the epithelial cells of the biliary tract. The limitations of the current methods in the diagnosis of CCA highlight the urgent need for new, accurate tools for early cancer detection, better prognostication and patient monitoring. Liquid biopsy (LB) is a modern and non-invasive technique comprising a diverse group of methodologies aiming to detect tumour biomarkers from body fluids. These biomarkers include circulating tumour cells, cell-free DNA, circulating tumour DNA, RNA and extracellular vesicles. The aim of this review is to explore the current and potential future applications of LB in CCA management, with a focus on diagnosis, prognostication and monitoring. We examine both its significant potential and the inevitable limitations associated with this technology. We conclude that LB holds considerable promise, but further research is necessary to fully integrate it into precision oncology for CCA.
Collapse
Affiliation(s)
- Evgenia Kotsifa
- The Second Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, Athens 11527, Greece
| | - Francesca Saffioti
- Department of Gastroenterology and Hepatology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, United Kingdom
- University College London Institute for Liver and Digestive Health and Sheila Sherlock Liver Unit, Royal Free Hospital and University College London, London NW3 2QG, United Kingdom
- Division of Clinical and Molecular Hepatology, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina 98124, Italy
| | - Vasileios K Mavroeidis
- Department of Transplant Surgery, North Bristol NHS Trust, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Department of Gastrointestinal Surgery, North Bristol NHS Trust, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Department of HPB Surgery, Bristol Royal Infirmary, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS2 8HW, United Kingdom
| |
Collapse
|
4
|
Yin H, Zhang M, Zhang Y, Zhang X, Zhang X, Zhang B. Liquid biopsies in cancer. MOLECULAR BIOMEDICINE 2025; 6:18. [PMID: 40108089 PMCID: PMC11923355 DOI: 10.1186/s43556-025-00257-8] [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: 07/22/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/22/2025] Open
Abstract
Cancer ranks among the most lethal diseases worldwide. Tissue biopsy is currently the primary method for the diagnosis and biological analysis of various solid tumors. However, this method has some disadvantages related to insufficient tissue specimen collection and intratumoral heterogeneity. Liquid biopsy is a noninvasive approach for identifying cancer-related biomarkers in peripheral blood, which allows for repetitive sampling across multiple time points. In the field of liquid biopsy, representative biomarkers include circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and exosomes. Many studies have evaluated the prognostic and predictive roles of CTCs and ctDNA in various solid tumors. Although these studies have limitations, the results of most studies appear to consistently demonstrate the correlations of high CTC counts and ctDNA mutations with lower survival rates in cancer patients. Similarly, a reduction in CTC counts throughout therapy may be a potential prognostic indicator related to treatment response in advanced cancer patients. Moreover, the biochemical characteristics of CTCs and ctDNA can provide information about tumor biology as well as resistance mechanisms against targeted therapy. This review discusses the current clinical applications of liquid biopsy in cancer patients, emphasizing its possible utility in outcome prediction and treatment decision-making.
Collapse
Affiliation(s)
- Hang Yin
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Manjie Zhang
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Yu Zhang
- Dalian Medical University, Dalian, 116000, China
| | - Xuebing Zhang
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Xia Zhang
- Dalian Fifth People's Hospital, Dalian, 116000, China.
| | - Bin Zhang
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China.
| |
Collapse
|
5
|
Sakai T, Omura G, Eguchi K, Sakai A, Matsumoto Y, Fushimi C, Yoshimoto S. Prognostic prediction using recursive partitioning analysis of patients undergoing salvage surgery for locally recurrent oral squamous cell carcinoma. Int J Clin Oncol 2025:10.1007/s10147-025-02739-9. [PMID: 40095336 DOI: 10.1007/s10147-025-02739-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/02/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND The prognosis of patients with locally recurrent oral cavity squamous cell carcinoma (LR-OCSCC) remains poor even when salvage surgery can be performed. Therefore, the current study aimed to identify adverse prognostic factors in these patients and use recursive partitioning analysis (RPA), a machine learning statistical method, to develop a prognostic classification based on these factors. METHODS The clinical data of 75 patients who underwent salvage surgery for LR-OCSCC at the National Cancer Center between 2010 and 2022 were retrospectively reviewed. Prognostic factors were analyzed using Cox proportional hazards regression models. Patients were classified by survival outcomes using RPA. Survival rates were determined using the Kaplan-Meier method. RESULTS The 3-year overall survival (OS) and locoregional recurrence rates for all patients who underwent salvage surgery were 53.4% and 32.7%, respectively. The univariate Cox proportional hazards regression analysis identified concurrent regional recurrence, previous history of radiotherapy to the neck, and recurrence within 12 months from initial treatment as adverse prognostic factors. RPA was performed using these variables and patients were classified into low-, intermediate-, and high-risk groups based on a combination of concurrent regional recurrence and time to recurrence. The 3-year OS rates for the low-, intermediate-, and high-risk groups were 73.3%, 53.3%, and 24.4%, respectively. CONCLUSION A novel prognostic classification for LR-OCSCC salvage surgery was developed to facilitate development of treatment strategies and identification of patients that would not benefit from this procedure.
Collapse
Affiliation(s)
- Toshihiko Sakai
- Department of Head and Neck Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan.
| | - Go Omura
- Department of Head and Neck Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Kohtaro Eguchi
- Department of Head and Neck Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Azusa Sakai
- Department of Head and Neck Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Yoshifumi Matsumoto
- Department of Head and Neck Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Chihiro Fushimi
- Department of Head and Neck Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Seiichi Yoshimoto
- Department of Head and Neck Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| |
Collapse
|
6
|
Zhang Q, Deng M, Gao Q, Zhou X, Guo Y, Wang Y, Fu Y, Zhang JL, Chen S, Hou G. Performance of Raman Spectroscopy in biopsy tissue for rapid diagnosis of Tracheobronchial Tuberculosis: A prospective study. Photodiagnosis Photodyn Ther 2025; 53:104556. [PMID: 40089169 DOI: 10.1016/j.pdpdt.2025.104556] [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: 02/06/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
Tracheobronchial tuberculosis (TBTB) is a specific form of pulmonary tuberculosis characterized by Mycobacterium tuberculosis involvement in the tracheobronchial tree. Rapid and accurate diagnostic tools for TBTB are crucial. Raman spectroscopy (RS) is a noninvasive tool that accesses molecular vibrations and sample characteristics, enabling the creation of a molecular fingerprint of biological samples, which has enormous potential on clinical diagnosis for TBTB. This study aimed to develop and validate a diagnostic model based on RS in bronchial biopsies for identifying TBTB. The training set included patients with TBTB (n = 18), airway malignant diseases (n = 20), and normal mucosal tissue biopsies as the healthy controls (n = 20). The spectral analysis results indicated that differential changes in tissue biomolecules, particularly certain amino acids, among the three groups. K-Nearest Neighbors (KNN), principal component analysis-linear discriminant analysis (PCA-LDA), principal component analysis-support vector machine (PCA-SVM) and decision tree methods were implemented to classify this same spectral data set. The PCA-SVM method exhibited highest classification accuracy, with a sensitivity of 88.89 % and an area under the receiver operating characteristic curve (AUROC) of 0.919. Subsequently, an independent validation set comprising 121 patients with suspected TBTB was enrolled to evaluate the performance of RS model using the PCA-SVM method. The sensitivity of RS model was 87.69 % (57/65) for diagnosing TBTB, higher than that of sputum smear, bronchial brush smear, the Bactec MGIT 960 Culture of bronchoalveolar lavage fluid, and comparable to GeneXpert sensitivity. In conclusion, the RS model using bronchial tissue provides a rapid and accurate method of identifying TBTB, showing potential as a powerful noninvasive tool for TBTB diagnosis under bronchoscopy.
Collapse
Affiliation(s)
- Qin Zhang
- National Center for Respiratory Medicine, Beijing 100029, PR China; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing 100029, PR China; National Clinical Research Center for Respiratory Diseases, Beijing 100029, PR China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, PR China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Mingming Deng
- National Center for Respiratory Medicine, Beijing 100029, PR China; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing 100029, PR China; National Clinical Research Center for Respiratory Diseases, Beijing 100029, PR China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, PR China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Qian Gao
- National Center for Respiratory Medicine, Beijing 100029, PR China; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing 100029, PR China; National Clinical Research Center for Respiratory Diseases, Beijing 100029, PR China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, PR China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, PR China
| | - Xiaoming Zhou
- Respiratory Department, Center for Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100037, PR China
| | - Yu Guo
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110167, PR China
| | - Yuexiang Wang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110167, PR China
| | - Yinghui Fu
- The Ninth Department of Tuberculosis, Shenyang Thoracic Hospital, Shenyang 110096, PR China
| | | | - Shuo Chen
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110167, PR China
| | - Gang Hou
- National Center for Respiratory Medicine, Beijing 100029, PR China; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing 100029, PR China; National Clinical Research Center for Respiratory Diseases, Beijing 100029, PR China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, PR China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, PR China.
| |
Collapse
|
7
|
Chen PH, Tsai TM, Lu TP, Lu HH, Pamart D, Kotronoulas A, Herzog M, Micallef JV, Hsu HH, Chen JS. Accurate Diagnosis of High-Risk Pulmonary Nodules Using a Non-Invasive Epigenetic Biomarker Test. Cancers (Basel) 2025; 17:916. [PMID: 40149253 DOI: 10.3390/cancers17060916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Accurate non-invasive tests to improve early detection and diagnosis of lung cancer are urgently needed. However, no regulatory-approved blood tests are available for this purpose. We aimed to improve pulmonary nodule classification to identify malignant nodules in a high-prevalence patient group. METHODS This study involved 806 participants with undiagnosed nodules larger than 5 mm, focusing on assessing nucleosome levels and histone modifications (H3.1 and H3K27Me3) in circulating blood. Nodules were classified as malignant or benign. For model development, the data were randomly divided into training (n = 483) and validation (n = 121) datasets. The model's performance was then evaluated using a separate testing dataset (n = 202). RESULTS Among the patients, 755 (93.7%) had a tissue diagnosis. The overall malignancy rate was 80.4%. For all datasets, the areas under curves were as follows: training, 0.74; validation, 0.86; and test, 0.79 (accuracy range: 0.80-0.88). Sensitivity showed consistent results across all datasets (0.91, 0.95, and 0.93, respectively), whereas specificity ranged from 0.37 to 0.64. For smaller nodules (5-10 mm), the model recorded accuracy values of 0.76, 0.88, and 0.85. The sensitivity values of 0.91, 1.00, and 0.94 further highlight the robust diagnostic capability of the model. The performance of the model across the reporting and data system (RADS) categories demonstrated consistent accuracy. CONCLUSIONS Our epigenetic biomarker panel detected non-small-cell lung cancer early in a high-risk patient group with high sensitivity and accuracy. The epigenetic biomarker model was particularly effective in identifying high-risk lung nodules, including small, part-solid, and non-solid nodules, and provided further evidence for validation.
Collapse
Affiliation(s)
- Pei-Hsing Chen
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei City 106, Taiwan
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei City 100, Taiwan
| | - Tung-Ming Tsai
- Department of Surgical Oncology, National Taiwan University Cancer Center, College of Medicine, National Taiwan University, Taipei City 106, Taiwan
| | - Tzu-Pin Lu
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei City 100, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei City 100, Taiwan
| | - Hsiao-Hung Lu
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei City 100, Taiwan
| | - Dorian Pamart
- Belgian Volition SRL, 22 Rue Phocas Lejeune, Parc Scientifique Crealys, 5032 Isnes, Belgium
| | | | - Marielle Herzog
- Belgian Volition SRL, 22 Rue Phocas Lejeune, Parc Scientifique Crealys, 5032 Isnes, Belgium
| | | | - Hsao-Hsun Hsu
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei City 100, Taiwan
- Department of Surgical Oncology, National Taiwan University Cancer Center, College of Medicine, National Taiwan University, Taipei City 106, Taiwan
| | - Jin-Shing Chen
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei City 100, Taiwan
| |
Collapse
|
8
|
Sabit H, Attia MG, Mohamed N, Taha PS, Ahmed N, Osama S, Abdel-Ghany S. Beyond traditional biopsies: the emerging role of ctDNA and MRD on breast cancer diagnosis and treatment. Discov Oncol 2025; 16:271. [PMID: 40050490 PMCID: PMC11885725 DOI: 10.1007/s12672-025-01940-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Breast cancer management has traditionally relied on tissue biopsies and imaging, which offer limited insights into the disease. However, the discovery of circulating tumor DNA (ctDNA) and minimal residual disease (MRD) detection has revolutionized our approach to breast cancer. ctDNA, which is fragmented tumor DNA found in the bloodstream, provides a minimally invasive way to understand the tumor's genomic landscape, revealing heterogeneity and critical mutations that biopsies may miss. MRD, which indicates cancer cells that remain after treatment, can now be detected using ctDNA and other advanced methods, improving our ability to predict disease recurrence. This allows for personalized adjuvant therapies based on individual MRD levels, avoiding unnecessary treatments for patients with low MRD. This review discusses how ctDNA and MRD represent a paradigm shift towards personalized, genomically guided cancer care, which has the potential to significantly improve patient outcomes in breast cancer.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt.
| | - Manar G Attia
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Nouran Mohamed
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Pancé S Taha
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Nehal Ahmed
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Salma Osama
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| |
Collapse
|
9
|
Sasaki T, Iwaya T, Yaegashi M, Idogawa M, Hiraki H, Abe M, Koizumi Y, Sasaki N, Yashima-Abo A, Fujisawa R, Endo F, Tange S, Otsuka K, Sasaki A, Masuda M, Fujita M, Nakagawa H, Takahashi F, Sasaki Y, Tokino T, Nishizuka SS. Impact of Sensitive Circulating Tumor DNA Monitoring on CT Scan Intervals During Postoperative Colorectal Cancer Surveillance. ANNALS OF SURGERY OPEN 2025; 6:e549. [PMID: 40134478 PMCID: PMC11932597 DOI: 10.1097/as9.0000000000000549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/07/2025] [Indexed: 03/27/2025] Open
Abstract
Objective This study investigated whether digital polymerase chain reaction (dPCR)-based circulating tumor DNA (ctDNA) monitoring can allow longer intervals between computed tomography (CT) scans during postoperative surveillance of colorectal cancer (CRC). Background Practical guidelines still recommend intensive postoperative surveillance of CRC using periodical CT scans and serum carcinoembryonic antigen testing. Methods The longitudinal dynamics of ctDNA for 52 patients with CRC as measured by dPCR using probes targeting 87 individual tumor-specific mutations (1-5 per patient) were compared with results from conventional (ie, clinical) surveillance using serum tumor markers and CT. Results A total of 382 CT procedures were carried out for the patient cohort (3.3/year per patient) and the median lead time from ctDNA relapse to clinical relapse was 182 days (range, 0-376 days). If the CT interval was annual, potential delays in the detection of clinical relapse would have occurred for 7 of the 10 patients who experienced clinical relapse (9 of 13 events), with a median delay of 164 days (range, 0-267 days). If annual CT surveillance was performed together with ctDNA monitoring, 218 (57.1%) CTs would not have been needed to detect the first clinical relapse. In addition, the ctDNA monitoring would have provided a lead time of 339 days for detection of clinical relapse (range, 42-533 days). Conclusions Our findings suggest that the ctDNA monitoring as part of postoperative surveillance and clinical relapse detection for patients with CRC could allow the CT interval to be lengthened. Trial Registration This trial was registered with University Hospital Medical Information Network Clinical Trial Registry (UMIN000045114).
Collapse
Affiliation(s)
- Tomoko Sasaki
- From the Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Takeshi Iwaya
- Department of Clinical Oncology, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Mizunori Yaegashi
- From the Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Masashi Idogawa
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Hayato Hiraki
- Division of Biomedical Research and Development, Iwate Medical University Institute for Biomedical Sciences, Yahaba, Japan
| | - Masakazu Abe
- Division of Biomedical Research and Development, Iwate Medical University Institute for Biomedical Sciences, Yahaba, Japan
- Department of Urology, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Yuka Koizumi
- From the Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Japan
- Division of Biomedical Research and Development, Iwate Medical University Institute for Biomedical Sciences, Yahaba, Japan
| | - Noriyuki Sasaki
- From the Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Japan
- Division of Biomedical Research and Development, Iwate Medical University Institute for Biomedical Sciences, Yahaba, Japan
| | - Akiko Yashima-Abo
- Division of Biomedical Research and Development, Iwate Medical University Institute for Biomedical Sciences, Yahaba, Japan
| | - Ryosuke Fujisawa
- From the Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Fumitaka Endo
- Department of Clinical Oncology, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Shoichiro Tange
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Koki Otsuka
- Department of Advanced Robotic and Endoscopic Surgery, Fujita health University School of Medicine, Toyoake, Japan
| | - Akira Sasaki
- From the Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Japan
| | - Mari Masuda
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Masashi Fujita
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Fumiaki Takahashi
- Division of Medical Engineering, Department of Information Science, Iwate Medical University Center for Liberal Arts and Sciences, Yahaba, Japan
| | - Yasushi Sasaki
- Biology Division, Department of Liberal Arts and Sciences, Center for Medical Education, Sapporo Medical University, Sapporo, Japan
| | - Takashi Tokino
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Satoshi S. Nishizuka
- Division of Biomedical Research and Development, Iwate Medical University Institute for Biomedical Sciences, Yahaba, Japan
| |
Collapse
|
10
|
Szostakowska-Rodzos M, Grzybowska EA, Mysliwy I, Zub R, Jagiello-Gruszfeld A, Rubach M, Konieczna A, Fabisiewicz A. The Combined Assessment of CTC and ESR1 Status in Liquid Biopsy Samples Enhances the Clinical Value of Prediction in Metastatic Breast Cancer. Int J Mol Sci 2025; 26:2038. [PMID: 40076662 PMCID: PMC11900918 DOI: 10.3390/ijms26052038] [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: 01/28/2025] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Monitoring of metastatic breast cancer (mBC) is an important issue in the clinical management of patients. Liquid biopsy has become a non-invasive method for detecting and monitoring cancer in body fluids. The presence of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) in peripheral blood indicates poor prognosis and may contribute to early detection of progression, but assessment of these levels is still not routine clinical management. The main objective of this study was to estimate the frequency and clinical value of the ESR1 and PIK3CA mutations identified in circulating free DNA (cfDNA.) The second goal was to evaluate whether simultaneous evaluation of CTCs and mutation status in cfDNA increases the prognostic value of liquid biopsy. The results of the analysis of the CTC number and ESR1 and PIK3CA mutations in blood collected from 179 patients with metastatic breast cancer show that ESR1 mutations are more frequent in patients with advanced luminal breast cancer regardless of the type of the treatment. ESR1 mutations appear primarily during progression, as no mutations were found in primary tumor samples. The main conclusion of the study is that combined assessment of CTCs and ESR1 status in liquid biopsy may improve the prognostic value of liquid biopsy.
Collapse
Affiliation(s)
- Malgorzata Szostakowska-Rodzos
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.S.-R.); (I.M.)
| | - Ewa A. Grzybowska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.S.-R.); (I.M.)
| | - Izabella Mysliwy
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.S.-R.); (I.M.)
| | - Renata Zub
- Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland;
| | | | - Maryna Rubach
- Cancer Chemotherapy Day Unit, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland;
| | - Aleksandra Konieczna
- Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland;
| | - Anna Fabisiewicz
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.S.-R.); (I.M.)
| |
Collapse
|
11
|
Glueck V, Grimm C, Postl M, Brueffer C, Segui N, Alcaide M, Oton L, Chen Y, Saal LH, Hofstetter G, Polterauer S, Muellauer L. ctDNA as an Objective Marker for Postoperative Residual Disease in Primary Advanced High-Grade Serous Ovarian Cancer. Cancers (Basel) 2025; 17:786. [PMID: 40075633 PMCID: PMC11899276 DOI: 10.3390/cancers17050786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES The surgeon's subjective intraoperative evaluation is the standard of care to assess postoperative residual disease (RD) in advanced epithelial ovarian cancer (EOC). We investigated the feasibility of ctDNA as an objective marker for postoperative RD. METHODS This prospective study included 27 patients with advanced ovarian cancer (FIGO IIIA1-IVB) who underwent primary surgery between July 2021 and July 2022. Blood samples were analyzed preoperatively and on days 2 (d2) and 10 (d10) postoperatively. Low-coverage whole genome sequencing (WGS) was used to identify structural variants (SVs) at single-base pair resolution, single nucleotide variants (SNVs), and indels in tumor tissue to develop personalized, tumor-informed digital polymerase chain reaction (dPCR) fingerprint assays for each patient. RESULTS dPCR fingerprint assays were successfully developed for all patients by identifying one to eight SVs/SNVs per patient. ctDNA was detected in 96% (n = 26/27) of patients preoperatively and in 81% (n = 22/27) of patients at d10. Median ctDNA levels at d10 were significantly higher in patients with postoperative RD (median 367.38 copies (cps)/mL, 2.84% variant allele frequency; VAF) than in patients without postoperative RD (median 0.92 cps/mL, 0.017% VAF, p < 0.001). In patients with postoperative RD, ctDNA levels increased from the preoperative stage to d10 in seven out of eight patients (p = 0.016). In patients with complete tumor resection, ctDNA levels decreased from the preoperative stage to d10 in 17/19 patients (p < 0.001). CONCLUSIONS A tumor-informed personalized ctDNA approach demonstrated feasibility, providing extremely high detection rates pre- and postoperatively. These results indicate that this approach could potentially be used for postoperative RD assessment in patients with primary advanced EOC.
Collapse
Affiliation(s)
- Valentina Glueck
- Gynecologic Cancer Unit, Division of General Gynecology and Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.); (S.P.)
- Department of Obstetrics and Gynecology, Klinikum Starnberg, 82319 Starnberg, Germany
| | - Christoph Grimm
- Gynecologic Cancer Unit, Division of General Gynecology and Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.); (S.P.)
| | - Magdalena Postl
- Gynecologic Cancer Unit, Division of General Gynecology and Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.); (S.P.)
| | - Christian Brueffer
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
- Division of Oncology, Lund University Cancer Center, Skåne University Hospital Comprehensive Cancer Center, Lund University, 221 00 Lund, Sweden
| | - Nuria Segui
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
| | - Miguel Alcaide
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
| | - Lucia Oton
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
| | - Yilun Chen
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
| | - Lao H. Saal
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
- Division of Oncology, Lund University Cancer Center, Skåne University Hospital Comprehensive Cancer Center, Lund University, 221 00 Lund, Sweden
| | - Gerda Hofstetter
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.H.); (L.M.)
| | - Stephan Polterauer
- Gynecologic Cancer Unit, Division of General Gynecology and Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.); (S.P.)
| | - Leonhard Muellauer
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.H.); (L.M.)
| |
Collapse
|
12
|
Lv D, Lan B, Guo Q, Yi Z, Qian H, Guan Y, Peng X, Chen T, Ma F. Exploration of the clonal evolution and construction of the tumor clonal evolution rate as a prognostic indicator in metastatic breast cancer. BMC Med 2025; 23:122. [PMID: 40001125 PMCID: PMC11863457 DOI: 10.1186/s12916-025-03959-6] [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: 07/11/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Tumor heterogeneity and clonal evolution are related to the treatment resistance and disease progression in metastatic breast cancer (MBC). However, the process of clonal evolution and their relationship to prognosis remain unclear. This study aimed to elucidate the evolution of MBC through circulating tumor DNA (ctDNA) analysis and to develop a novel indicator for predicting treatment efficacy and prognosis. METHODS This multicenter retrospective study enrolled MBC patients who underwent next-generation sequencing between April 2016 and October 2022. The clonal evolution of tumors was inferred using PyClone and CITUP software. RESULTS The study included 406 MBC patients. A cohort of 139 patients from the National Cancer Center served as the training cohort, while 267 patients from other centers comprised the validation cohort. In the training cohort, clonal analysis revealed that most MBCs exhibited branched clonal evolution, while a minority showed linear evolution. The branched evolution pattern was associated with slower disease progression (HR, 0.53; 95% CI, 0.32-0.87; P = 0.012). We introduced tumor clonal evolution rate (TER) as a novel concept to reflect the speed of clonal evolution. Survival analysis demonstrated that compared to the TER-high group, patients in the TER-low group had better progression-free survival (PFS) (HR, 0.62; 95% CI, 0.40-0.96; P = 0.033) and overall survival (OS) (HR, 0.45; 95% CI, 0.24-0.85; P = 0.013). Similarly, in the validation cohort, although the median OS was not reached, patients in the TER-low group had better prognosis compared to those in the TER-high group (HR, 0.41; 95% CI, 0.21-0.83; P < 0.001). CONCLUSIONS Patients with branched evolution have better treatment efficacy than those with linear evolution. The TER shows potential as a biomarker for treatment efficacy and prognosis, providing new evidence that ctDNA is a valuable molecular indicator for predicting treatment outcomes in metastatic breast cancer.
Collapse
Affiliation(s)
- Dan Lv
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bo Lan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qihan Guo
- Department of Computer Science and Technology & Institute of Artificial Intelligence & BNRist, Tsinghua University, Beijing, 100084, China
| | - Zongbi Yi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yanfang Guan
- Geneplus-Beijing Institute, Beijing, 102206, China
| | - Xuenan Peng
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ting Chen
- Department of Computer Science and Technology & Institute of Artificial Intelligence & BNRist, Tsinghua University, Beijing, 100084, China.
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
13
|
Luan X, Gao Y, Pan Y, Huang Z, Zeng F, He G, He B, Ye D, Song Y. Bifunctional Nanoassembly Enables Metabolism-Driven Microfluidic Blood Screening Guided by MRI Localization for Cancer Monitoring. Anal Chem 2025; 97:3395-3403. [PMID: 39900559 DOI: 10.1021/acs.analchem.4c05427] [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: 02/05/2025]
Abstract
Early detection and precise tumor localization are critical for improving treatment outcomes and enabling more targeted and minimally invasive therapies as biotechnology evolves. However, endogenous biomarkers from early lesions face significant challenges, such as short circulation times and blood dilution, which hinder early diagnostic efforts. In this study, we present a multimodal nanosensor specifically engineered to target cancer by responding to CD44 and tumor-associated enzymes within the microenvironment. Following systemic administration, the nanosensor selectively accumulates at the disease site, delivering hexaminolevulinate (HAL) to produce protoporphyrin IX (PpIX) as a synthetic biomarker, thus amplifying disease signals for analysis via a microfluidics-based device. Concurrently, embedded Gd2O3 nanoclusters facilitate tumor visualization through magnetic resonance imaging (MRI). Beyond tumor diagnosis, this innovative methodology supports the multimodal monitoring of drug response through the assessment of blood reporter signals and MRI imaging. This multifunctional system addresses critical limitations in traditional cancer diagnostics, which typically rely on sequential blood biomarker tests, followed by imaging. Our approach enhances diagnostic efficiency, minimizes the need for invasive procedures, and promotes more accurate and personalized cancer care.
Collapse
Affiliation(s)
- Xiaowei Luan
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Yanfeng Gao
- School of Medical Imaging, Wannan Medical College, Wuhu 241002, China
| | - Yongchun Pan
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Zheng Huang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Fei Zeng
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Guanzhong He
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yujun Song
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| |
Collapse
|
14
|
Obinah MPB, Al-Halafi SA, Dreisig K, Poulsen TS, Johansen C, Litman T, Bojesen SE, Høgdall E, Chakera AH, Hölmich LR. Circulating tumor DNA for surveillance in high-risk melanoma patients: a study protocol. Acta Oncol 2025; 64:229-233. [PMID: 39930781 PMCID: PMC11833324 DOI: 10.2340/1651-226x.2025.42515] [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: 11/17/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND AND PURPOSE Melanoma is one of the deadliest skin cancers and challenges clinicians worldwide due to rising incidence, potential aggressiveness, and propensity for metastasis, necessitating comprehensive follow-up programs after primary treatment. Circulating tumor DNA (ctDNA) is a promising biomarker that may indicate disease progression earlier than traditional surveillance methods, including 18F-FDG PET-CT, ultrasound, and clinical examination. This study examines ctDNA detection in blood as a minimally invasive method for early identification of progression following primary treatment of melanoma. The aim is to overcome the limitations of current methods, potentially improving prognosis and survival. PATIENTS/MATERIAL AND METHODS Patients with high risk of recurrence following primary treatment of melanoma are offered inclusion. Blood sampling is performed at each follow-up visit. In case of recurrence, patient-specific mutations are identified through next-generation sequencing (NGS) of formalin and paraffin embedded tissue from diagnostic routine. Detection of mutation-specific ctDNA is performed on blood using digital droplet polymerase chain reaction (ddPCR) or NGS. This allows determination of the value and sensitivity of ctDNA for early detection of recurrence. RESULTS AND INTERPRETATION For validation purposes, we conducted a small pilot study using blood samples from 10 patients who had experienced recurrence and had a clinically confirmed BRAF V600E mutation. Detection of BRAF V600E ctDNA using ddPCR varied from 0/5 (0%) in DNA harvested from 4 mL plasma, to 3/5 (60%) in DNA from 8 mL of plasma. These results show promise and highlight the importance of high sensitivity and sampling volumes to ensure accurate detection of low levels of ctDNA.
Collapse
Affiliation(s)
- Magnús P B Obinah
- Dept. of Plastic Surgery, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen Denmark.
| | - Sarah A Al-Halafi
- Dept. of Plastic Surgery, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen Denmark
| | - Karin Dreisig
- Dept. of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen Denmark
| | - Tim S Poulsen
- Molecular Unit, Dept. of Pathology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen Denmark
| | - Christoffer Johansen
- Center for Cancer Late Effect Research CASTLE, Dept. of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Thomas Litman
- Dept. of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stig E Bojesen
- Dept. of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen Denmark; Dept. of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Estrid Høgdall
- Molecular Unit, Dept. of Pathology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen Denmark; Dept. of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Lisbet R Hölmich
- Dept. of Plastic Surgery, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen Denmark; Dept. of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Kim KH, Park C, Beom SH, Kim MH, Kim CG, Kim HR, Jung M, Shin SJ, Rha SY, Kim HS. An open-label, phase IB/II study of abemaciclib with paclitaxel for tumors with CDK4/6 pathway genomic alterations. ESMO Open 2025; 10:104106. [PMID: 39874900 PMCID: PMC11799963 DOI: 10.1016/j.esmoop.2024.104106] [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: 05/28/2024] [Revised: 10/16/2024] [Accepted: 12/04/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Disruption of cyclin D-dependent kinases (CDKs), particularly CDK4/6, drives cancer cell proliferation via abnormal protein phosphorylation. This open-label, single-arm, phase Ib/II trial evaluated the efficacy of the CDK4/6 inhibitor, abemaciclib, combined with paclitaxel against CDK4/6-activated tumors. PATIENTS AND METHODS Patients with locally advanced or metastatic solid tumors with CDK4/6 pathway aberrations were included. Based on phase Ib, the recommended phase II doses were determined as abemaciclib 100 mg twice daily and paclitaxel 70 mg/m2 on days 1, 8, and 15, over 4-week-long cycles. The primary endpoint for phase II was the overall response rate (ORR). The secondary endpoints included the clinical benefit rate (CBR), progression-free survival (PFS), overall survival (OS), and safety. Tissue-based next-generation sequencing and exploratory circulating tumor DNA analyses were carried out. RESULTS Between February 2021 and April 2022, 30 patients received abemaciclib/paclitaxel (median follow-up: 15.7 months), and 27 were included in the efficacy analysis. CDK4/6 amplification (50%) and CCND1/3 amplification (20%) were common activating mutations. The ORR was 7.4%, with two partial responses, and the CBR was 66.7% (18/27 patients). The median OS and PFS were 9.9 months [95% confidence interval (CI) 5.7-14.0 months] and 3.5 months (95% CI 2.6-4.3 months), respectively. Grade 3 adverse events (50%, 21 events) were mainly hematologic. Genetic analysis revealed a 'poor genetic status' subgroup characterized by mutations in key signaling pathways (RAS, Wnt, PI3K, and NOTCH) and/or CCNE amplification, correlating with poorer PFS. CONCLUSION Abemaciclib and paclitaxel showed moderate clinical benefits for CDK4/6-activated tumors. We identified a poor genetic group characterized by bypass signaling pathway activation and/or CCNE amplification, which negatively affected treatment response and survival. Future studies with homogeneous patient groups are required to validate these findings.
Collapse
Affiliation(s)
- K H Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - C Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University Cancer Research Institute, Seoul, Republic of Korea
| | - S-H Beom
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - M H Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - C G Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - H R Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - M Jung
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - S J Shin
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - S Y Rha
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - H S Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Niu S, Sun T, Wang M, Yao L, He T, Wang Y, Zhang H, Li X, Xu Y. Multiple time points for detecting circulating tumor DNA to monitor the response to neoadjuvant therapy in breast cancer: a meta-analysis. BMC Cancer 2025; 25:115. [PMID: 39844103 PMCID: PMC11752932 DOI: 10.1186/s12885-025-13526-0] [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: 03/06/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Not all breast cancer (BC) patients can benefit from neoadjuvant therapy (NAT). A poor response may result in patients missing the best opportunity for treatment, ultimately leading to a poor prognosis. Thus, to identify an effective predictor that can assess and predict patient response at early time points, we focused on circulating tumor DNA (ctDNA), which is a vital noninvasive liquid biopsy biomarker. We performed a meta-analysis to explore the predictive value of response by monitoring ctDNA at four time points of NAT using pathologic complete response (pCR) and residual cancer burden (RCB). METHODS By searching Embase, PubMed, the Cochrane Library, and the Web of Science until December 24, 2023, we selected studies concerning the relationship between ctDNA and response or prognosis. We analysed the results at the following various time points: baseline (T0), first cycle of NAT (T1), mid-treatment (MT), and end of NAT (EOT). pCR and RCB were used to evaluate the response as the primary endpoint. The secondary endpoint was to investigate the relationship between ctDNA and prognosis. Odds ratios (ORs) and hazard ratios (HRs) were used as effect indicators. RESULTS Thirteen reports from twelve studies were eligible for inclusion in this meta-analysis. The results demonstrated that ctDNA negativity was associated with pCR at T1 (OR = 0.34; 95% CI: 0.21-0.57), MT (OR = 0.35; 95% CI: 0.20-0.60), and EOT (OR = 0.38; 95% CI: 0.22-0.66). When RCB was used to evaluate responses, ctDNA negativity was associated with RCB-0/I at the MT (OR = 0.34; 95% CI: 0.21-0.55) and EOT (OR = 0.26; 95% CI: 0.15-0.46). Furthermore, ctDNA positivity at T1 predicted a worse prognosis for patients (HR = 2.73; 95% CI: 1.29-5.75). We also performed a subgroup analysis to more accurately assess the predictive value of ctDNA for triple-negative breast cancer. CONCLUSIONS Our meta-analysis suggested that the ctDNA status at the early stage of NAT can predict patient response, which provides evidence for adjusting personalized treatment strategies and improving patient survival. PROSPERO REGISTRATION NUMBER CRD42024496465.
Collapse
Affiliation(s)
- Shuyi Niu
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Tie Sun
- The Third Department of General Surgery, People's Hospital of China Medical University (Liaoning Provincial People's Hospital), Shenyang, Liaoning, 110001, China
| | - Mozhi Wang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Litong Yao
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Tianyi He
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Yusong Wang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Hengjun Zhang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Xiang Li
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Yingying Xu
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
17
|
Abidoye O, Ahn DH, Borad MJ, Wu C, Bekaii-Saab T, Chakrabarti S, Sonbol MB. Circulating Tumor DNA Testing for Minimal Residual Disease and Its Application in Colorectal Cancer. Cells 2025; 14:161. [PMID: 39936953 DOI: 10.3390/cells14030161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Colorectal cancer (CRC) represents a heterogeneous group of diseases that imposes a considerable global and national health burden. Although most CRC patients are diagnosed at an early stage and undergo potentially curative treatment, a significant proportion experience recurrence. Currently, adjuvant chemotherapy decisions are primarily based on clinicopathological characteristics, which have well-recognized limitations in accurately identifying patients harboring minimal residual disease (MRD), often resulting in unnecessary chemotherapy exposure. Circulating tumor DNA (ctDNA) has emerged as a promising surrogate marker for MRD, offering a more precise approach to identifying patients at risk of recurrence after curative-intent surgery and refining adjuvant chemotherapy decisions. Growing evidence from multiple studies has demonstrated that ctDNA outperforms traditional clinicopathological factors as a marker for MRD. This review synthesizes key studies supporting the role of ctDNA in MRD detection for CRC patients and evaluates clinical trials investigating the application of ctDNA in guiding adjuvant therapy decisions. This emerging strategy holds the potential to transform the adjuvant treatment paradigm in colorectal cancer by optimizing therapeutic precision and minimizing unnecessary treatment.
Collapse
Affiliation(s)
| | - Daniel H Ahn
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA
| | | | - Christina Wu
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA
| | | | | | | |
Collapse
|
18
|
Yang X, Yang D, Qi X, Luo X, Zhang G. Endocrine treatment mechanisms in triple-positive breast cancer: from targeted therapies to advances in precision medicine. Front Oncol 2025; 14:1467033. [PMID: 39845328 PMCID: PMC11753220 DOI: 10.3389/fonc.2024.1467033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Triple-positive breast cancer (TPBC), defined by the co-expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), poses unique therapeutic challenges due to complex signaling interactions and resulting treatment resistance. This review summarizes key findings on the molecular mechanisms and cross-talk among ER, PR, and HER2 pathways, which drive tumor proliferation and resistance to conventional therapies. Current strategies in TPBC treatment, including endocrine and HER2-targeted therapies, are explored alongside emerging approaches such as immunotherapy and CRISPR/Cas9 gene editing. Additionally, we discuss the tumor microenvironment (TME) and its role in treatment resistance, highlighting promising avenues for intervention through combination therapies and predictive biomarkers. By addressing these interdependent pathways and optimizing therapeutic strategies, precision medicine holds significant potential for improving TPBC patient outcomes and advancing individualized cancer care.
Collapse
Affiliation(s)
| | | | | | | | - Guangmei Zhang
- Department of Medical Oncology, Third Division, Jilin City Second People’s Hospital, Jilin, China
| |
Collapse
|
19
|
Smilkou S, Ntzifa A, Tserpeli V, Balgkouranidou I, Papatheodoridi A, Razis E, Linardou H, Papadimitriou C, Psyrri A, Zagouri F, Kakolyris S, Lianidou E. Detection rate for ESR1 mutations is higher in circulating-tumor-cell-derived genomic DNA than in paired plasma cell-free DNA samples as revealed by ddPCR. Mol Oncol 2025. [PMID: 39754401 DOI: 10.1002/1878-0261.13787] [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: 08/12/2024] [Revised: 09/21/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025] Open
Abstract
Plasma cell-free DNA (cfDNA) analysis to track estrogen receptor 1 (ESR1) mutations is highly beneficial for the identification of tumor molecular dynamics and the improvement of personalized treatments for patients with metastatic breast cancer (MBC). Plasma-cfDNA is, up to now, the most frequent liquid biopsy analyte used to evaluate ESR1 mutational status. Circulating tumor cell (CTC) enumeration and molecular characterization analysis provides important clinical information in patients with MBC. In this study, we investigated whether analysis of CTCs and circulating tumor DNA (ctDNA) provide similar or complementary information for the analysis of ESR1 mutations. We analyzed both plasma-cfDNA (n = 90) and paired CTC-derived genomic DNA (gDNA; n = 42) from 90 MBC patients for seven ESR1 mutations. Eight out of 90 (8.9%) plasma-cfDNA samples tested using the ddPLEX Mutation Detection Assay (Bio-Rad, Hercules, CA, USA), were found positive for one ESR1 mutation, whereas 11/42 (26.2%) CTC-derived gDNA samples were found positive for at least one ESR1 mutation. Direct comparison of paired samples (n = 42) revealed that the ESR1 mutation rate was higher in CTC-derived gDNA (11/42, 26.2%) than in plasma-cfDNA (6/42, 14.3%) samples. Our results, using this highly sensitive ddPLEX assay, reveal a higher percentage of mutations in CTC-derived gDNAs than in paired ctDNA in patients with MBC. CTC-derived gDNA analysis should be further evaluated as an important and complementary tool to ctDNA for identifying patients with ESR1 mutations and for guiding individualized therapy.
Collapse
Affiliation(s)
- Stavroula Smilkou
- Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Greece
| | - Aliki Ntzifa
- Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Greece
| | - Victoria Tserpeli
- Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Greece
| | - Ioanna Balgkouranidou
- Department of Medical Oncology, University General Hospital of Alexandroupolis, Greece
| | - Alkistis Papatheodoridi
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Christos Papadimitriou
- Oncology Unit, Aretaieion University Hospital, National and Kapodistrian University of Athens, Greece
| | - Amanda Psyrri
- Department of Medical Oncology, Second Department of Internal Medicine, "Attikon" University General Hospital, Athens Medical School, National and Kapodistrian University of Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stylianos Kakolyris
- Department of Medical Oncology, University General Hospital of Alexandroupolis, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Greece
| |
Collapse
|
20
|
Jamieson A, McConechy MK, Lum A, Senz J, Dowhy T, Huntsman DG, McAlpine JN. Selective utilization of circulating tumor DNA testing enables disease monitoring in endometrial and ovarian carcinomas. J Gynecol Oncol 2025; 36:e5. [PMID: 38909641 PMCID: PMC11791001 DOI: 10.3802/jgo.2025.36.e5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/06/2024] [Accepted: 05/13/2024] [Indexed: 06/25/2024] Open
Abstract
OBJECTIVE Biomarkers reflecting real-time response to therapy and recurrence are lacking. We assessed the clinical value of detecting cell-free circulating tumor DNA (ctDNA) mutations in endometrial cancer (EC) and ovarian cancer (OC) patients. METHODS EC/OC patients undergoing primary surgery were consented for tissue banking and 2-year serial blood draws. Tumor tissue DNA and plasma ctDNA underwent next generation sequencing using a targeted gene panel for somatic mutations. RESULTS Of 44 patients (24 EC, 17 OC, 2 synchronous endometrial and ovarian carcinomas [SEOC] and 1 endocervical adenocarcinoma [EA]) at least one somatic mutation was identified in tumor tissue in 40 (91%, 20/24 EC, all OC/SEOC/EA), and in preoperative plasma ctDNA in 12 (27%) patients (6/24 [25%] EC and 6/17 [35%] OC). Detection of preoperative ctDNA mutations was associated with advanced stage, higher preoperative CA125, and subsequent disease recurrence. In 5/12 (42%) patients with preoperative ctDNA mutations, examination/imaging suggested clinical stage I however final pathology revealed stage II/III. In 11 patients where serial timepoints were assessed during treatment for ctDNA and CA125, ctDNA clearance preceded normalization of CA125. Thirteen patients developed recurrent disease (4 EC, 8 OC, 1 EA); 8 in whom ctDNA mutations were detected postoperatively, and 4 followed through time of recurrence with ctDNA mutations identified 2-5 months prior to clinical/radiologic/biomarker progression in 3. CONCLUSION ctDNA can reflect larger tumor volume/metastases, treatment response and subsequent disease recurrence in EC and OC. Careful patient selection is critical to direct resources to patients most likely to benefit, considering disease burden and risk group.
Collapse
Affiliation(s)
- Amy Jamieson
- Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, Canada
| | | | - Amy Lum
- Department of Molecular Oncology, University of British Columbia, Vancouver, Canada
| | - Janine Senz
- Department of Molecular Oncology, University of British Columbia, Vancouver, Canada
| | | | - David G Huntsman
- Imagia Canexia Health, Inc., Vancouver, Canada
- Department of Molecular Oncology, University of British Columbia, Vancouver, Canada
| | - Jessica N McAlpine
- Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
21
|
Martens GA, Demol J, Dedeurwaerdere F, De Smet K, Wesolowski J, De Smet D. Surveillance of Disease Progression in Metastatic Breast Cancer by Molecular Counting of Circulating Tumor DNA Using Plasma-SeqSensei Breast Cancer in Vitro Diagnostics Assay. J Mol Diagn 2025; 27:25-35. [PMID: 39521246 DOI: 10.1016/j.jmoldx.2024.08.011] [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: 01/18/2024] [Revised: 07/22/2024] [Accepted: 08/28/2024] [Indexed: 11/16/2024] Open
Abstract
Circulating tumor DNA (ctDNA) quantification surpasses cancer antigen 15 to 3 for metastatic breast cancer surveillance. Clinical translation, however, is limited because of uncertainties about the optimal method and clinically valid ctDNA decision thresholds. Plasma-SeqSensei Breast Cancer IVD kit (PSS) is a novel assay for ctDNA molecular counting, detecting ≥0.06% variant allele fractions in AKT1, ERBB2, ESR1, KRAS, PIK3CA, and TP53. PSS was validated against droplet digital PCR (ddPCR) in 201 samples from 16 subjects with PIK3CA/TP53-mutated cancers, longitudinally sampled for a median of 93 (range, 18 to 113) weeks, three to five weekly. PSS and ddPCR ctDNA levels correlate significantly (Spearman ρ, 0.923; 95% CI, 0.898-0.941) across 0% to 43% variant allele frequency (VAF) range. PSS predicts 12-week progression with high clinical accuracy (area under the curve, 0.848; 95% CI, 0.790-0.894). PSS validates a previously developed ddPCR classifier: <10 copies/mL (0.25% VAF); excludes >100 copies/mL (2.5% VAF); and confirms progression, with negative predictive value (95% CI) of 83% (76%-88%) and positive predictive value (95% CI) of 91% (81%-96%) (weighted κ, 0.856; 95% CI, 0.797-0.915). PSS thus confirms robust clinical thresholds (10 to 100 copies/mL, 0.25% to 2.5% VAF) for metastatic breast cancer surveillance, using absolute molecular counting.
Collapse
Affiliation(s)
- Geert A Martens
- Department of Laboratory Medicine, AZ Delta General Hospital, Roeselare, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.
| | - Jan Demol
- Department of Oncology, AZ Delta General Hospital, Roeselare, Belgium
| | | | - Kristof De Smet
- Department of Radiology, AZ Delta General Hospital, Roeselare, Belgium
| | | | - Dieter De Smet
- Department of Laboratory Medicine, AZ Delta General Hospital, Roeselare, Belgium
| |
Collapse
|
22
|
Du T, Yuan Y, Sun S, Gao Z, Li X. Integrating traditional biomarkers and emerging predictors to assess neoadjuvant chemotherapy efficacy in breast cancer: a multifactorial analysis of Ki-67, CDK4, EGFR, TILs and ctDNA. BMC Womens Health 2024; 24:674. [PMID: 39736579 DOI: 10.1186/s12905-024-03486-1] [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: 08/07/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
OBJECTIVE This study aimed to analyse the correlation between the expression of cell proliferation-associated antigen (Ki-67), cell cycle protein-dependent kinase 4 (CDK4), epidermal growth factor receptor (EGFR), tumour-infiltrating lymphocytes (TILs) and circulating tumour DNA (ctDNA) with the outcome and prognosis of patients with breast cancer (BC) undergoing neoadjuvant chemotherapy (NACT). METHODS We retrospectively analysed the clinicopathological data of 231 patients with BC who underwent preoperative NACT at XX Hospital between 1 January 2018 and 31 December 2021. Logistic regression models were used to analyse factors influencing NACT efficacy. The Cox risk regression model was used to analyse prognostic factors. The TILs were assessed on pre-treatment biopsies, and ctDNA levels were monitored during NACT. Propensity score matching and subgroup analyses were performed. RESULTS After 4-6 cycles of chemotherapy, the response rate was 77.92% (180/231), with 58.87% (136/231) achieving pathological complete response (pCR). Multifactorial analysis showed that tumour, node and metastasis (TNM) stage II, EGFR positivity, low Ki-67 expression, CDK4 negativity, non-triple-negative subtypes and effective NACT results were associated with higher pCR rates. Higher TIL levels correlated with increased pCR rates (72.4% for high TILs vs 39.1% for low TILs, p < 0.001). The ctDNA levels decreased significantly in patients with pCR compared with patients without pCR during NACT (p < 0.001). After propensity score matching, the 3-year disease-free survival rate was significantly higher in the pCR group (88.9% vs 71.1%, p = 0.003). Subgroup analysis revealed varying pCR rates and predictive biomarkers across BC subtypes. CONCLUSION The TNM classification, EGFR, Ki-67, CDK4 expression, BC subtype and NACT results have predictive value for pCR in patients with BC. Lower TNM classification, lower Ki-67 expression and EGFR positivity are associated with better outcomes. High TIL levels and significant decreases in ctDNA during NACT correlate with improved response and prognosis. These findings highlight the potential for integrating traditional clinicopathological factors with novel biomarkers for personalised treatment strategies in BC.
Collapse
Affiliation(s)
- Tianzhao Du
- Central Laboratory, Cancer Hospital of China Medical University, Dalian Medical University Clinical Oncology College, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Ye Yuan
- Central Laboratory, Cancer Hospital of China Medical University, Dalian Medical University Clinical Oncology College, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Shulan Sun
- Central Laboratory, Cancer Hospital of China Medical University, Dalian Medical University Clinical Oncology College, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China.
| | - Zhichao Gao
- Central Laboratory, Cancer Hospital of China Medical University, Dalian Medical University Clinical Oncology College, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Xiaoshuai Li
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| |
Collapse
|
23
|
Nagano S, Kurokawa Y, Hagi T, Yoshioka R, Takahashi T, Saito T, Yamamoto K, Momose K, Yamashita K, Tanaka K, Makino T, Nakajima K, Eguchi H, Doki Y. Extensive methylation analysis of circulating tumor DNA in plasma of patients with gastric cancer. Sci Rep 2024; 14:30739. [PMID: 39730450 DOI: 10.1038/s41598-024-79252-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/07/2024] [Indexed: 12/29/2024] Open
Abstract
DNA methylation is known to be involved in tumor progression. This is the first study to perform an extensive methylation analysis of plasma circulating tumor DNA (ctDNA) using targeted bisulfite sequencing in gastric cancer (GC) patients to evaluate the usefulness of ctDNA methylation as a new biomarker. Sixteen patients who received chemotherapy for recurrent GC were included. After confirmation of the methylation status of 63 genes using the Cancer Genome Atlas (TCGA) dataset, the methylation status in paired tumor and non-tumor tissues and plasma were investigated using targeted bisulfite sequencing in these genes. Forty-four of the 63 genes were significantly hypermethylated in GC patients in the TCGA cohort. Of these 44 genes, hierarchical clustering showed that five (SPG20, FBN1, SDC2, TFPI2, SEPT9) were particularly hypermethylated in tumor compared to non-tumor tissues in our GC cohort. In plasma methylation analysis, patients with high methylation of these genes had significantly worse overall survival than those with low methylation (log-rank P = 0.009). In a patient who underwent blood sampling at multiple points, the methylation levels of these five genes varied closely with clinical tumor status. The plasma ctDNA methylation levels of these five genes could be useful as a noninvasive prognostic biomarker for GC.
Collapse
Affiliation(s)
- Shinnosuke Nagano
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan.
| | - Takaomi Hagi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Ryo Yoshioka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Kazuyoshi Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Kota Momose
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita City, Osaka, 565-0871, Japan
| |
Collapse
|
24
|
Uchibori M, Hosomichi K, Hoshimoto Y, Sasaki M, Aoki T, Tajima A, Ota Y, Kimura M. The Efficacy of Liquid Biopsy of Total cfDNA for Predicting Systemic Metastasis in Japanese Patients With Oral Squamous Cell Carcinoma. Head Neck 2024. [PMID: 39731308 DOI: 10.1002/hed.28054] [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: 02/22/2024] [Revised: 11/28/2024] [Accepted: 12/17/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND The use of liquid biopsy of total cell-free DNA (cfDNA) to identify otherwise undetectable cancers has attracted interest; however, its efficacy remains unknown. We explored whether analysis using total cfDNA is efficacious for Japanese patients with oral squamous cell carcinoma (OSCC). METHODS We collected total cfDNA from nine patients with OSCC preoperatively, 1 month postoperatively, and every 3 months thereafter to analyze this association. We used a target DNA sequence for genetic mutation analysis of tumor tissues collected from 33 patients, including the aforementioned nine patients. RESULTS Patients with good disease control showed negligible changes in preoperative and postoperative total cfDNA concentrations. A rapid increase in total cfDNA concentration was observed in patients who developed systemic metastases. Patients whose tumor tissue DNA showed genetic mutations had the same mutations in preoperative circulating tumor DNA. CONCLUSIONS Our data suggested that analyzing total cfDNA is efficacious for patients with OSCC.
Collapse
Affiliation(s)
- Masahiro Uchibori
- Department of Oral and Maxillofacial Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Kazuyoshi Hosomichi
- Laboratory of Computational Genomics, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yasutaka Hoshimoto
- Department of Oral and Maxillofacial Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Masashi Sasaki
- Department of Oral and Maxillofacial Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Takayuki Aoki
- Department of Oral and Maxillofacial Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshihide Ota
- Department of Oral and Maxillofacial Surgery, Tokai University School of Medicine, Isehara, Japan
| | | |
Collapse
|
25
|
Eboshida N, Hamada A, Higaki M, Obayashi F, Ito N, Yamasaki S, Tani R, Shintani T, Koizumi K, Yanamoto S. Potential role of circulating tumor cells and cell-free DNA as biomarkers in oral squamous cell carcinoma: A prospective single-center study. PLoS One 2024; 19:e0309178. [PMID: 39729421 DOI: 10.1371/journal.pone.0309178] [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: 05/30/2024] [Accepted: 08/06/2024] [Indexed: 12/29/2024] Open
Abstract
Metastasis in patients with oral squamous cell carcinoma has been associated with a poor prognosis. However, sensitive and reliable tests for monitoring their occurrence are unavailable, with the exception of PET-CT. Circulating tumor cells and cell-free DNA have emerged as promising biomarkers for determining treatment efficacy and as prognostic predictors in solid tumors such as breast cancer and colorectal cancer. Hence, this study aimed to determine the potential role of liquid biopsy, circulating tumor cells, and cell-free DNA as biomarkers of oral squamous cell carcinoma. Thirteen patients with primary oral squamous cell carcinoma who visited our hospital between 2022 and 2023 were recruited, and plasma samples were collected from each patient preoperatively and postoperatively. We examined the relationship between the prognosis, the number of circulating tumor cells per four milliliters of peripheral blood, and the amount of cell-free DNA per milliliter of serum or the gene mutation in cell-free DNA. We observed no correlation between the number of preoperative circulating tumor cells and metastatic events. However, the number of circulating tumor cell clusters or the amount of preoperative cell-free DNA in metastatic cases was higher than that in non-metastatic cases. In oral squamous cell carcinoma, circulating tumor cell clusters or cell-free DNA levels may help inform management decisions regarding metastasis. However, further studies are required to provide a possible window for therapeutic interventions.
Collapse
Affiliation(s)
- Natsuki Eboshida
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Atsuko Hamada
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mirai Higaki
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Fumitaka Obayashi
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nanako Ito
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Sachiko Yamasaki
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ryouji Tani
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoaki Shintani
- Center of Oral Clinical Examination, Hiroshima University Hospital, Hiroshima, Japan
| | - Koichi Koizumi
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Souichi Yanamoto
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
26
|
Zhang C, Li N, Zhang P, Jiang Z, Cheng Y, Li H, Pang Z. Advancing precision and personalized breast cancer treatment through multi-omics technologies. Am J Cancer Res 2024; 14:5614-5627. [PMID: 39803662 PMCID: PMC11711544 DOI: 10.62347/mwnz5609] [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: 07/03/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
Breast cancer is the most common malignant tumour in women, with more than 685,000 women dying of breast cancer each year. The heterogeneity of breast cancer complicates both treatment and diagnosis. Traditional methods based on histopathology and hormone receptor status are now no longer sufficient. Recently, advances in multi-omics techniques, including genomic, proteomic, and transcriptomic analyses, have deepened our understanding of breast cancer. Combining these approaches allows for precise molecular subtyping, which is essential for the detection of key mutations, protein interactions and gene expression patterns that are highly relevant to different therapeutic strategies. Genomic analyses have been effectively identifying key mutations in cancer. Meanwhile, proteomics and transcriptomics complement by identifying new therapeutic targets and elucidating gene expression dynamics. Integrating multi-omics and conventional diagnostics improves tumour characterisation and enables prognostic accuracy comparable to established standards and treatment response. Existing and emerging technologies enable real-time enhanced tumour follow-up and data analysis through liquid biopsy and artificial intelligence, respectively. Despite these clinical implementation challenges, multi-omics including clinical phenotyping offers significant potential for precision breast cancer treatment. This article describes recent advances in molecular subtyping and multi-omics technologies that are driving key innovations to optimise patient outcomes and further develop personalised medicine in the context of breast cancer care.
Collapse
Affiliation(s)
- Chenlu Zhang
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Nan Li
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
- Heilongjiang Provincial Center for Prevention and Treatment of Cerebral Palsy in Children Postdoctoral Research WorkstationNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Pengxia Zhang
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Zhimei Jiang
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Yichao Cheng
- School of Rehabilitation Medicine, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Huiqing Li
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Zhenfei Pang
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| |
Collapse
|
27
|
Li MX, Hu S, Lei HH, Yuan M, Li X, Hou WK, Huang XJ, Xiao BW, Yu TX, Zhang XH, Wu XT, Jing WQ, Lee HJ, Li JJ, Fu D, Zhang LM, Yan W. Tumor-derived miR-9-5p-loaded EVs regulate cholesterol homeostasis to promote breast cancer liver metastasis in mice. Nat Commun 2024; 15:10539. [PMID: 39627188 PMCID: PMC11615374 DOI: 10.1038/s41467-024-54706-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 11/15/2024] [Indexed: 12/06/2024] Open
Abstract
Cancer cells secrete extracellular vesicles (EV) encapsulating bioactive cargoes to facilitate inter-organ communication in vivo and are emerging as critical mediators of tumor progression and metastasis, a condition which is often accompanied by a dysregulated cholesterol metabolism. Whether EVs are involved in the control of cholesterol homeostasis during tumor metastasis is still undefined and warrant further investigation. Here, we find that breast cancer-derived exosomal miR-9-5p induces the expression of HMGCR and CH25H, two enzymes involved in cholesterol synthesis and the conversion of 25-hydroxycholesterol from cholesterol by targeting INSIG1, INSIG2 and ATF3 genes in the liver. Notably, in vivo miR-9-5p antagomir treatment and genetic CH25H ablation prevents tumor metastasis in a mouse model of breast cancer. Thus, our findings reveal the regulatory mechanism of tumor-derived miR-9-5p in liver metastasis by linking oxysterol metabolism and Kupffer cell polarization, shedding light on future applications for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Mei-Xin Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Sheng Hu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - He-Hua Lei
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences (CAS), Wuhan, Hubei, 430064, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Meng Yuan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xu Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Wen-Kui Hou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xiang-Jie Huang
- College of Biomedical Engineering and Instrument Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Bing-Wen Xiao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Teng-Xiang Yu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xiao-Hui Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xiao-Ting Wu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Wen-Qiang Jing
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Hyeon-Jeong Lee
- College of Biomedical Engineering and Instrument Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Juan-Juan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Da Fu
- General Surgery, Ruijin Hospital & Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Li-Min Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences (CAS), Wuhan, Hubei, 430064, China.
- University of Chinese Academy of Sciences, 100864, Beijing, China.
| | - Wei Yan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China.
| |
Collapse
|
28
|
Ma L, Guo H, Zhao Y, Liu Z, Wang C, Bu J, Sun T, Wei J. Liquid biopsy in cancer current: status, challenges and future prospects. Signal Transduct Target Ther 2024; 9:336. [PMID: 39617822 PMCID: PMC11609310 DOI: 10.1038/s41392-024-02021-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/10/2024] [Accepted: 10/14/2024] [Indexed: 12/06/2024] Open
Abstract
Cancer has a high mortality rate across the globe, and tissue biopsy remains the gold standard for tumor diagnosis due to its high level of laboratory standardization, good consistency of results, relatively stable samples, and high accuracy of results. However, there are still many limitations and drawbacks in the application of tissue biopsy in tumor. The emergence of liquid biopsy provides new ideas for early diagnosis and prognosis of tumor. Compared with tissue biopsy, liquid biopsy has many advantages in the diagnosis and treatment of various types of cancer, including non-invasive, quickly and so on. Currently, the application of liquid biopsy in tumor detection has received widely attention. It is now undergoing rapid progress, and it holds significant potential for future applications. Around now, liquid biopsies encompass several components such as circulating tumor cells, circulating tumor DNA, exosomes, microRNA, circulating RNA, tumor platelets, and tumor endothelial cells. In addition, advances in the identification of liquid biopsy indicators have significantly enhanced the possibility of utilizing liquid biopsies in clinical settings. In this review, we will discuss the application, advantages and challenges of liquid biopsy in some common tumors from the perspective of diverse systems of tumors, and look forward to its future development prospects in the field of cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China.
| | - Huiling Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Yunxiang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhibo Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Chenran Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Jiahao Bu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China.
| | - Jianwei Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
29
|
Nafar S, Hosseini K, Shokrgozar N, Farahmandi AY, Alamdari-Palangi V, Saber Sichani A, Fallahi J. An Investigation into Cell-Free DNA in Different Common Cancers. Mol Biotechnol 2024; 66:3462-3474. [PMID: 38071680 DOI: 10.1007/s12033-023-00976-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 10/23/2023] [Indexed: 11/15/2024]
Abstract
Diagnosis is the most important step in different diseases, especially in cancers and blood malignancies. There are different methods in order to better diagnose of cancer, but many of them are invasive and also, some of them are not useful for immediate diagnosis. Cell-free DNA (cfDNA) or liquid biopsy easily accessible in peripheral blood is one of the non-invasive prognostic biomarkers in various areas of cancer management. In fact, amounts of cfDNA in serum or plasma can be used for diagnosis. In this review, we have considered some cancers such as hepatocellular carcinoma, lung cancer, breast cancer, and hematologic malignancies to compare the various methods of cfDNA diagnosis.
Collapse
Affiliation(s)
- Samira Nafar
- Medical Genetic Department, Shiraz University of Medical Science, Shiraz, Iran
| | - Kamran Hosseini
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Shokrgozar
- Hematology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | | | - Vahab Alamdari-Palangi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Saber Sichani
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
30
|
Richard M, Moreau R, Croyal M, Mathiot L, Frénel J, Campone M, Dupont A, Gavard J, André‐Grégoire G, Guével L. Monitoring concentration and lipid signature of plasma extracellular vesicles from HR + metastatic breast cancer patients under CDK4/6 inhibitors treatment. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70013. [PMID: 39691590 PMCID: PMC11650302 DOI: 10.1002/jex2.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/05/2024] [Accepted: 09/20/2024] [Indexed: 12/19/2024]
Abstract
Extracellular vesicles (EVs) are cell-derived small membrane structures that transport various molecules. They have emerged as potential circulating biomarkers for monitoring responses to cancer therapies. This study aimed to comprehensively characterize plasma-carried EVs in hormone receptor-positive (HR+) metastatic breast cancer (MBC) patients treated with first-line CDK4/6 inhibitors (iCDK4/6) combined with endocrine therapy. MBC patients were classified into three groups based on their response to therapy: resistant, intermediate or sensitive. In a prospective cohort, we monitored the concentration of circulating EVs, analyzed their lipid signature and correlated these factors with treatment response. To facilitate the translation of EV research to clinical practice, we established a three-step procedure: (1) EVs were isolated from plasma using semi-automatized size exclusion chromatography (SEC); (2) EV concentration, termed vesiclemia, was determined by drop counting via interferometric light microscopy (ILM); and (3) EV lipid composition was analyzed by mass spectrometry. ILM-based vesiclemia values were highly fluctuating upon iCDK4/6 treatment, while early increase associated with accelerated progression. Of note, vesiclemia remained a steady parameter over a 1-year period in age-matched healthy women. Additionally, analysis of the EV cargo unveiled a distinct sphingolipid profile, characterized by increased levels of ceramides and sphingomyelins in resistant patients within the first 2 months of treatment. Based on 16 sphingolipid species, sensitive and resistant patients were correctly classified with an overall accuracy of 82%. This specific sphingolipid pattern was exclusively discernible within EVs, and not in plasma, highlighting the significance of EVs in the early prediction of individual responses to iCDK4/6 and disease progression. Overall, this study provides insights of the longitudinal characterization of plasma-borne EVs in both a healthy group and HR+ MBC patients under iCDK4/6 therapies. Combined vesiclemia and EV sphingolipid profile emphasize the promising potential of EVs as non-invasive biomarkers for monitoring early treatment response.
Collapse
Affiliation(s)
- Mathilde Richard
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
| | - Rosalie Moreau
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
| | - Mikaël Croyal
- Nantes Université, CHU Nantes, CNRS, INSERMNantesFrance
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556NantesFrance
- CRNH‐Ouest Mass Spectrometry Core FacilityNantesFrance
| | - Laurent Mathiot
- Institut de Cancérologie de l'Ouest (ICO), Site Rene GauducheauSaint HerblainFrance
| | | | - Mario Campone
- Institut de Cancérologie de l'Ouest (ICO), Site Rene GauducheauSaint HerblainFrance
| | - Aurélien Dupont
- SFR UMS CNRS 3480, INSERM 018, Biosit biologie, santé, innovation technologiqueRennesFrance
| | - Julie Gavard
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
- Institut de Cancérologie de l'Ouest (ICO), Site Rene GauducheauSaint HerblainFrance
| | - Gwennan André‐Grégoire
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
- Institut de Cancérologie de l'Ouest (ICO), Site Rene GauducheauSaint HerblainFrance
| | - Laëtitia Guével
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
| |
Collapse
|
31
|
Izhar M, Thakur A, Park DJ, Chang SD. Ultrasound mediated blood-brain barrier opening increases brain tumor biomarkers: A review of preclinical and clinical trials. THE JOURNAL OF LIQUID BIOPSY 2024; 6:100277. [PMID: 40027314 PMCID: PMC11863884 DOI: 10.1016/j.jlb.2024.100277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 03/05/2025]
Abstract
The diagnosis of brain tumors typically relies on magnetic resonance imaging (MRI), computed tomography (CT), and invasive procedures like biopsies or surgical resection for confirmation and genetic profiling. However, these methods have limitations, especially in distinguishing treatment effects like pseudo-progression from actual tumor progression, and repeated biopsies pose risks. Liquid biopsy (LB) offers a non-invasive alternative, detecting tumor-derived biomarkers in blood and cerebrospinal fluid (CSF). Despite its potential, the low concentration of brain tumor biomarkers in blood due to the blood-brain barrier (BBB), limits the clinical utility of LB. MRI-guided focused ultrasound (MRgFUS) combined with microbubbles provides a novel solution by temporarily disrupting the BBB, facilitating the passage of therapeutic agents, and enabling tumor biomarker detection. This technique, termed "sonobiopsy," enables non-invasive biomarker collection for liquid biopsy, potentially improving brain tumor diagnosis and monitoring.
Collapse
Affiliation(s)
- Muhammad Izhar
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Abhimanyu Thakur
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David J. Park
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven D. Chang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
32
|
Pearce H, Chang YC, Javitt MC, Datta J, Pimentel A, Bialick S, Hosein PJ, Alessandrino F. ctDNA in the reading room: A guide for radiologists. Eur J Radiol 2024; 181:111796. [PMID: 39461058 DOI: 10.1016/j.ejrad.2024.111796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/02/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
Liquid biopsy with sequencing of circulating tumor DNA (ctDNA) is a minimally invasive method for sampling body fluids and offers a promising alternative to tissue biopsies that involve greater risks, costs, and time. ctDNA not only identifies actionable targets by revealing unique molecular signatures in cancer, but also may assess treatment response, treatment resistance and progression, and recurrence. Imaging correlates of these applications are already being identified and utilized for various solid tumors. Radiologists have new challenges in interpreting oncologic imaging. Given their integral role in cancer surveillance, they must become familiar with the importance of ctDNA in detecting recurrence and minimal residual disease, measuring treatment response, predicting survival and metastatic patterns, and identifying new molecular therapeutic targets. In this review, we provide an overview of ctDNA testing, and a snapshot of current clinical guidelines from the National Comprehensive Cancer Network and the European Society of Molecular Oncology on the use of ctDNA in lung, breast, colorectal, pancreatic, and hepatobiliary cancers. For each cancer type, we also highlight current research applications of ctDNA that are relevant to the field of diagnostic radiology.
Collapse
Affiliation(s)
- Hayes Pearce
- University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA
| | - Yu-Cherng Chang
- Department of Radiology, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA.
| | - Marcia C Javitt
- Division of Abdominal Imaging, Department of Radiology, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA
| | - Jashodeep Datta
- Division of Surgical Oncology, Department of Surgery, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA
| | - Agustin Pimentel
- Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA; Division of Medical Oncology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA
| | - Steven Bialick
- Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA; Division of Medical Oncology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA
| | - Peter J Hosein
- Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA; Division of Medical Oncology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA
| | - Francesco Alessandrino
- Division of Abdominal Imaging, Department of Radiology, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, Miami, FL, USA
| |
Collapse
|
33
|
Rong RY, Shen YK, Wu SN, Xu SH, Hu JY, Zou J, He L, Chen C, Kang M, Ying P, Wei H, Ling Q, Ge QM, Lou Y, Shao Y. Prediction model for ocular metastasis of breast cancer: machine learning model development and interpretation study. BMC Cancer 2024; 24:1472. [PMID: 39614215 DOI: 10.1186/s12885-024-12928-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 09/10/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is caused by the uncontrolled proliferation of breast epithelial cells followed by malignant transformation, and it has the highest incidence among female malignant tumors. The metastasis of BC occurs through direct and lymphatic spread. Although ocular metastasis is relatively rare, it is a good indicator of a worse prognosis. We used machine learning (ML) to establish a model to analyze the risk factors of BC eye metastasis. METHODS The clinical data of 2225 patients with BC from 2003 to 2019 were collected and randomly classified into the training and test sets using a ratio of 7:3. Based on the presence or absence of eye metastasis, the patients with BC were classified into the ocular metastasis (OM) and non-ocular metastasis (NOM) groups. Univariate and multivariate logistic regression analyses and least absolute shrinkage and selection operator (LASSO) were conducted. We used six ML algorithms to establish a predictive BC model and used 10-fold cross-validation for internal verification. The area under the receiver operating characteristic (ROC) curve was used to evaluate the predictive ability of the model. In addition, we established a web hazard calculator depending on the best-performing model to facilitate its clinical application. Shapley additive interpretation (SHAP) was used to determine the risk factors and the interpretability of the black box model. RESULTS Univariate logistic regression analysis showed that histopathology (other types), axillary lymph node metastasis (ALNM) (> 4), Ca2+, total cholesterol (TC), low-density lipoprotein (LDL), apolipoprotein A (ApoA), carcinoembryonic antigen (CEA), carbohydrate antigen (CA) 125, CA153, CA199, alkaline phosphatase (ALP), and hemoglobin (Hb) were risk factors for BC eye metastasis. Multivariate logistic regression analysis showed that CA153, ApoA, and LDL were hazardous components for BC eye metastasis. LASSO showed that ALNM, LDL, CA125, Hb, ALP, and CA199 were the first six key variables that were useful for the diagnosis of ocular metastasis in breast cancer. Bootstrapped aggregation (BAG) demonstrated the discriminative ability (area under ROC curve [AUC] = 0.992, accuracy = 0.953, sensitivity = 0.987). Based on this, we applied the BAG machine learning model to build an online web computing system to help clinicians assist in determining the risk of BC eye metastasis. In addition, two typical cases are analyzed to determine the interpretability of the model. CONCLUSION We used ML to establish a risk prediction model for BC ocular metastasis, and BAG showed the greatest performance. The model can predict the risk of OM in patients with BC, facilitate early and timely diagnosis and treatment, and reduce the burden on society.
Collapse
Affiliation(s)
- Ru-Yi Rong
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Yan-Kun Shen
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200030, China
| | - Shi-Nan Wu
- School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - San-Hua Xu
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jin-Yu Hu
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jie Zou
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Liangqi He
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Cheng Chen
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Min Kang
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ping Ying
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Hong Wei
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qian Ling
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qian-Ming Ge
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yan Lou
- School of Medical Information and Engineering, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Yi Shao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
34
|
Tegeler CM, Hartkopf AD, Banys-Paluchowski M, Krawczyk N, Fehm T, Jaeger BAS. Circulating Tumor DNA in Early and Metastatic Breast Cance-Current Role and What Is Coming Next. Cancers (Basel) 2024; 16:3919. [PMID: 39682108 DOI: 10.3390/cancers16233919] [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: 09/15/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 12/18/2024] Open
Abstract
The progress that has been made in recent years in relation to liquid biopsies in general and circulating tumor DNA (ctDNA) in particular can be seen as groundbreaking for the future of breast cancer treatment, monitoring and early detection. Cell-free DNA (cfDNA) consists of circulating DNA fragments released by various cell types into the bloodstream. A portion of this cfDNA, known as ctDNA, originates from malignant cells and carries specific genetic mutations. Analysis of ctDNA provides a minimally invasive method for diagnosis, monitoring response to therapy, and detecting the emergence of resistance. Several methods are available for the analysis of ctDNA, each with distinct advantages and limitations. Quantitative polymerase chain reaction is a well-established technique widely used due to its high sensitivity and specificity, particularly for detecting known mutations. In addition to the detection of individual mutations, multigene analyses were developed that could detect several mutations at once, including rarer mutations. These methods are complementary and can be used strategically depending on the clinical question. In the context of metastatic breast cancer, ctDNA holds particular promise as it allows for the dynamic monitoring of tumor evolution. Through ctDNA analysis, mutations in the ESR1 or PIK3CA genes, which are associated with therapy resistance, can be identified. This enables the early adjustment of treatment and has the potential to significantly enhance clinical outcome. The application of ctDNA in early breast cancer is an ongoing investigation. In (neo)adjuvant settings, there is preliminary data indicating that ctDNA can be used for therapy monitoring and risk stratification to decide on post-neoadjuvant strategies. In the monitoring of aftercare, the detection of ctDNA appears to be several months ahead of routine imaging. However, the feasibility of implementing this approach in a clinical setting remains to be seen. While the use of ctDNA as a screening method for the asymptomatic population would be highly advantageous due to its minimally invasive nature, the available data on its clinical benefit are still insufficient. Nevertheless, ctDNA represents the most promising avenue for fulfilling this potential future need.
Collapse
Affiliation(s)
- Christian Martin Tegeler
- Department of Obstetrics and Gynecology, University Hospital Tübingen, 72076 Tübingen, Germany
- Department of Peptide-Based Immunotherapy, Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Andreas Daniel Hartkopf
- Department of Obstetrics and Gynecology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Maggie Banys-Paluchowski
- Department of Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Campus Luebeck, 23538 Luebeck, Germany
| | - Natalia Krawczyk
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, 40225 Duesseldorf, Germany
- Center for Integrated Oncology (CIO) ABCD, 40225 Duesseldorf, Germany
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, 40225 Duesseldorf, Germany
- Center for Integrated Oncology (CIO) ABCD, 40225 Duesseldorf, Germany
| | - Bernadette Anna Sophia Jaeger
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, 40225 Duesseldorf, Germany
- Center for Integrated Oncology (CIO) ABCD, 40225 Duesseldorf, Germany
| |
Collapse
|
35
|
Wyatt AW, Litiere S, Bidard FC, Cabel L, Dyrskjøt L, Karlovich CA, Pantel K, Petrie J, Philip R, Andrews HS, Vellanki PJ, Tolmeijer SH, Villalobos Alberu X, Alfano C, Bogaerts J, Calvo E, Chen AP, Toledo RA, de Vries EGE, Seymour L, Laurie SA, Garralda E. Plasma ctDNA as a Treatment Response Biomarker in Metastatic Cancers: Evaluation by the RECIST Working Group. Clin Cancer Res 2024; 30:5034-5041. [PMID: 39269996 DOI: 10.1158/1078-0432.ccr-24-1883] [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: 06/17/2024] [Revised: 08/08/2024] [Accepted: 09/13/2024] [Indexed: 09/15/2024]
Abstract
Early indicators of metastatic cancer response to therapy are important for evaluating new drugs and stopping ineffective treatment. The RECIST guidelines based on repeat cancer imaging are widely adopted in clinical trials, are used to identify active regimens that may change practice, and contribute to regulatory approvals. However, these criteria do not provide insight before 6 to 12 weeks of treatment and typically require that patients have measurable disease. Recent data suggest that measuring on-treatment changes in the amount or proportion of ctDNA in peripheral blood plasma may accurately identify responding and nonresponding cancers at earlier time points. Over the past year, the RECIST working group has evaluated current evidence for plasma ctDNA kinetics as a treatment response biomarker in metastatic cancers and early endpoint in clinical trials to identify areas of focus for future research and validation. Here, we outline the requirement for large standardized trial datasets, greater scrutiny of optimal ctDNA collection time points and assay thresholds, and consideration of regulatory body guidelines and patient opinions. In particular, clinically meaningful changes in plasma ctDNA abundance are likely to differ by cancer type and therapy class and must be assessed before ctDNA can be considered a potential pan-cancer response evaluation biomarker. Despite the need for additional data, minimally invasive on-treatment ctDNA measurements hold promise to build upon existing response assessments such as RECIST and offer opportunities for developing novel early endpoints for modern clinical trials.
Collapse
Affiliation(s)
- Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Genome Sciences Centre and Clinical Cancer Genomics Program, BC Cancer, Vancouver, British Columbia, Canada
| | - Saskia Litiere
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Francois-Clément Bidard
- Department of Medical Oncology, Institut Curie, Université Versailles Saint-Quentin, Université Paris-Saclay, Saint-Cloud, France
| | - Luc Cabel
- Department of Medical Oncology, Institut Curie, Université Versailles Saint-Quentin, Université Paris-Saclay, Saint-Cloud, France
| | - Lars Dyrskjøt
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Chris A Karlovich
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Klaus Pantel
- Department of Tumor Biology, Center for Experimental Medicine, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joan Petrie
- Canadian Cancer Trials Group, Kingston, Ontario, Canada
| | - Reena Philip
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland
| | | | - Paz J Vellanki
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Sofie H Tolmeijer
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Christian Alfano
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Jan Bogaerts
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | | | - Elisabeth G E de Vries
- University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Lesley Seymour
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Scott A Laurie
- Division of Medical Oncology, The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Elena Garralda
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
36
|
Giovino C, Subasri V, Telfer F, Malkin D. New Paradigms in the Clinical Management of Li-Fraumeni Syndrome. Cold Spring Harb Perspect Med 2024; 14:a041584. [PMID: 38692744 PMCID: PMC11529854 DOI: 10.1101/cshperspect.a041584] [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: 05/03/2024]
Abstract
Approximately 8.5%-16.2% of childhood cancers are associated with a pathogenic/likely pathogenic germline variant-a prevalence that is likely to rise with improvements in phenotype recognition, sequencing, and variant validation. One highly informative, classical hereditary cancer predisposition syndrome is Li-Fraumeni syndrome (LFS), associated with germline variants in the TP53 tumor suppressor gene, and a >90% cumulative lifetime cancer risk. In seeking to improve outcomes for young LFS patients, we must improve the specificity and sensitivity of existing cancer surveillance programs and explore how to complement early detection strategies with pharmacology-based risk-reduction interventions. Here, we describe novel precision screening technologies and clinical strategies for cancer risk reduction. In particular, we summarize the biomarkers for early diagnosis and risk stratification of LFS patients from birth, noninvasive and machine learning-based cancer screening, and drugs that have shown the potential to be repurposed for cancer prevention.
Collapse
Affiliation(s)
- Camilla Giovino
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Vallijah Subasri
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Frank Telfer
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - David Malkin
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1L7, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Division of Hematology-Oncology, The Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|
37
|
Burdon AJ, Baird RD, Jaki T. Adaptive enrichment trial designs using joint modelling of longitudinal and time-to-event data. Stat Methods Med Res 2024; 33:2098-2114. [PMID: 39410878 DOI: 10.1177/09622802241287711] [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] [Indexed: 11/20/2024]
Abstract
Adaptive enrichment allows for pre-defined patient subgroups of interest to be investigated throughout the course of a clinical trial. These designs have gained attention in recent years because of their potential to shorten the trial's duration and identify effective therapies tailored to specific patient groups. We describe enrichment trials which consider long-term time-to-event outcomes but also incorporate additional short-term information from routinely collected longitudinal biomarkers. These methods are suitable for use in the setting where the trajectory of the biomarker may differ between subgroups and it is believed that the long-term endpoint is influenced by treatment, subgroup and biomarker. Methods are most promising when the majority of patients have biomarker measurements for at least two time points. We implement joint modelling of longitudinal and time-to-event data to define subgroup selection and stopping criteria and we show that the familywise error rate is protected in the strong sense. To assess the results, we perform a simulation study and find that, compared to the study where longitudinal biomarker observations are ignored, incorporating biomarker information leads to increases in power and the (sub)population which truly benefits from the experimental treatment being enriched with higher probability at the interim analysis. The investigations are motivated by a trial for the treatment of metastatic breast cancer and the parameter values for the simulation study are informed using real-world data where repeated circulating tumour DNA measurements and HER2 statuses are available for each patient and are used as our longitudinal data and subgroup identifiers, respectively.
Collapse
Affiliation(s)
| | - Richard D Baird
- Department of Oncology, Cancer Research UK, Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Thomas Jaki
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Department of Machine Learning and Data Science, University of Regensburg, Regensburg, Bayern, Germany
| |
Collapse
|
38
|
Ge Q, Zhang ZY, Li SN, Ma JQ, Zhao Z. Liquid biopsy: Comprehensive overview of circulating tumor DNA (Review). Oncol Lett 2024; 28:548. [PMID: 39319213 PMCID: PMC11420644 DOI: 10.3892/ol.2024.14681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024] Open
Abstract
Traditional tumor diagnosis methods rely on tissue biopsy, which can be invasive and unsuitable for long-term monitoring of tumor dynamics. The advent of liquid biopsy has notably improved the overall management of patients with cancer. Liquid biopsy techniques primarily involve detection of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). The present review focuses on ctDNA because of its significance in tumor diagnosis, monitoring and treatment. The use of ctDNA-based liquid biopsy offers several advantages, including non-invasive or minimally invasive collection methods, the ability to conduct repeated assessment and comprehensive insights into tumor biology. It serves crucial roles in disease management by facilitating screening of high-risk patients, dynamically monitoring therapeutic responses and diagnosis. Furthermore, ctDNA can be used to demonstrate pseudo-progression, monitor postoperative tumor status and guide adaptive treatment plans. The present study provides a comprehensive review of ctDNA, exploring its origins, metabolism, detection methods, clinical role and the current challenges associated with its application.
Collapse
Affiliation(s)
- Qian Ge
- Graduate School, Xi'an Medical University, Xi'an, Shaanxi 710000, P.R. China
| | - Zhi-Yun Zhang
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, P.R. China
| | - Suo-Ni Li
- Department of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Jie-Qun Ma
- Department of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Zheng Zhao
- Department of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710000, P.R. China
| |
Collapse
|
39
|
Darville-O’Quinn P, Gokgoz N, Tsoi KM, Andrulis IL, Wunder JS. Investigating the Use of Circulating Tumor DNA for Sarcoma Management. J Clin Med 2024; 13:6539. [PMID: 39518682 PMCID: PMC11545914 DOI: 10.3390/jcm13216539] [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: 10/01/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Sarcomas are a heterogeneous group of cancers, many with high rates of recurrence and metastasis, leading to significant morbidity and mortality. Due to a lack of early diagnostic biomarkers, by the time recurrent disease can be clinically detected, it is often extensive and difficult to treat. Here, we sought to investigate methods of detecting ctDNA in sarcoma patient plasma to potentially monitor disease recurrence, progression, and response to treatment. Methods: Whole-exome sequencing of matched tumor and blood samples revealed patient-specific mutations, which were used to develop personalized assays to detect ctDNA in patient plasma. Since ctDNA is present in extremely low quantities, detection requires highly sensitive methodologies. Droplet digital PCR is highly sensitive; however, it is limited in that it can only be used to target one tumor variant at a time. Therefore, a protocol combining multiplex PCR and targeted amplicon sequencing was developed. Results: ddPCR was successfully able to detect tumor-specific mutations in plasma, confirming the presence of ctDNA in sarcoma patients. Multiplex PCR followed by amplicon sequencing was able to detect multiple tumor variants simultaneously, although it was not as sensitive as ddPCR. Additionally, ctDNA was detected in patient plasma collected at two different time points. Conclusions: This work demonstrates that although there is a lack of recurrent biomarkers, personalized assays detecting ctDNA have the potential to be used to monitor disease progression in sarcoma.
Collapse
Affiliation(s)
- Paige Darville-O’Quinn
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada; (P.D.-O.); (N.G.); (K.M.T.)
| | - Nalan Gokgoz
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada; (P.D.-O.); (N.G.); (K.M.T.)
| | - Kim M. Tsoi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada; (P.D.-O.); (N.G.); (K.M.T.)
- Musculoskeletal Oncology Unit, Sinai Health System, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Irene L. Andrulis
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada; (P.D.-O.); (N.G.); (K.M.T.)
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jay S. Wunder
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada; (P.D.-O.); (N.G.); (K.M.T.)
- Musculoskeletal Oncology Unit, Sinai Health System, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
40
|
Kinane DF, Gabert J, Xynopoulos G, Guzeldemir‐Akcakanat E. Strategic approaches in oral squamous cell carcinoma diagnostics using liquid biopsy. Periodontol 2000 2024; 96:316-328. [PMID: 38676371 PMCID: PMC11579816 DOI: 10.1111/prd.12567] [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: 11/01/2023] [Revised: 02/06/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024]
Abstract
Liquid biopsy is a noninvasive diagnostic technique used for monitoring cancer utilizing specific genetic biomarkers present in bodily fluids, such as blood, saliva, or urine. These analyses employ multiple biomolecular sources including circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and exosomes (that contain DNA fragments) to detect genetic biomarkers that can predict, disclose, and/or monitor cancers. Levels of these biomarkers can inform on the presence of cancer, its genetic characteristics, and its potential treatment response and also provide predictive genetic predisposition information for specific cancers including oral squamous cell carcinomas (OSCC). Liquid biopsies can aid cancer management as they offer real-time dynamic information on the response to say chemotherapy or radiotherapy and recurrence following surgical excision. Unlike traditional tissue biopsies, which are invasive with a degree of morbidity and require specific tumor location sampling, liquid biopsies are noninvasive and can be repeated frequently. For oral squamous cell carcinoma, on which this review focuses, liquid biopsy of blood or saliva can be valuable in predicting susceptibility, providing early detection, and monitoring the disease's progression and response to therapy. This review gives a general narrative overview of the technology, its current medical usage, and advantages and disadvantages compared with current techniques and discusses a range of current potential biomarkers for disclosing OSCC and predicting its risk. Oral squamous cell carcinoma is all too often detected in the late stages. In future, liquid biopsy may provide an effective screening process such that cancers including OSCC will be detected in the early stages rather than later when prognosis is poor and morbidity and debilitation are greater. In this screening process, periodontists and hygienists have a critical role in that they are adept in examining mucosa, they see patients with shared risk factors for periodontitis and OSCC, namely smoking and poor oral hygiene, and they see patients frequently such that OSCC examinations should be a routine part of the recall visit. With this additional screening manpower, oral medicine and oral surgery colleagues will detect OSCC earlier and this coupled with new techniques such as liquid biopsy may greatly decrease global morbidity in OSCC.
Collapse
Affiliation(s)
- Denis F. Kinane
- Department of Periodontology, Dental SchoolUniversity BernBernSwitzerland
- ExpressTestCignpost Diagnostics Ltd.FarnboroughUnited Kingdom
| | | | | | - Esra Guzeldemir‐Akcakanat
- Department of Periodontology, Faculty of DentistryKocaeli UniversityİzmitTurkey
- College of Dental MedicineQU Health, Qatar UniversityQatarQatar
| |
Collapse
|
41
|
Fu SW, Tang C, Tan X, Srivastava S. Liquid biopsy for early cancer detection: technological revolutions and clinical dilemma. Expert Rev Mol Diagn 2024; 24:937-955. [PMID: 39360748 DOI: 10.1080/14737159.2024.2408744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Liquid biopsy is an innovative advancement in oncology, offering a noninvasive method for early cancer detection and monitoring by analyzing circulating tumor cells, DNA, RNA, and other biomarkers in bodily fluids. This technique has the potential to revolutionize precision oncology by providing real-time analysis of tumor dynamics, enabling early detection, monitoring treatment responses, and tailoring personalized therapies based on the molecular profiles of individual patients. AREAS COVERED In this review, the authors discuss current methodologies, technological challenges, and clinical applications of liquid biopsy. This includes advancements in detecting minimal residual disease, tracking tumor evolution, and combining liquid biopsy with other diagnostic modalities for precision oncology. Key areas explored are the sensitivity, specificity, and integration of multi-omics, AI, ML, and LLM technologies. EXPERT OPINION Liquid biopsy holds great potential to revolutionize cancer care through early detection and personalized treatment strategies. However, its success depends on overcoming technological and clinical hurdles, such as ensuring high sensitivity and specificity, interpreting results amidst tumor heterogeneity, and making tests accessible and affordable. Continued innovation and collaboration are crucial to fully realize the potential of liquid biopsy in improving early cancer detection, treatment, and monitoring.
Collapse
Affiliation(s)
- Sidney W Fu
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Cong Tang
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Xiaohui Tan
- Division of LS Research, LSBioscience, LLC, Frederick, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
42
|
Lee SB, Kim JW, Kim HG, Hwang SH, Kim KJ, Lee JH, Seo J, Kang M, Jung EH, Suh KJ, Kim SH, Kim JW, Kim YJ, Kim JH, Kwon NJ, Lee KW. Longitudinal Comparative Analysis of Circulating Tumor DNA and Matched Tumor Tissue DNA in Patients with Metastatic Colorectal Cancer Receiving Palliative First-Line Systemic Anti-Cancer Therapy. Cancer Res Treat 2024; 56:1171-1182. [PMID: 38697850 PMCID: PMC11491242 DOI: 10.4143/crt.2024.016] [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: 01/04/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024] Open
Abstract
PURPOSE This study aimed to compare tumor tissue DNA (ttDNA) and circulating tumor DNA (ctDNA) to explore the clinical applicability of ctDNA and to better understand clonal evolution in patients with metastatic colorectal cancer undergoing palliative first-line systemic therapy. MATERIALS AND METHODS We performed targeted sequencing analysis of 88 cancer-associated genes using germline DNA, ctDNA at baseline (baseline-ctDNA), and ctDNA at progressive disease (PD-ctDNA). The results were compared with ttDNA data. RESULTS Among 208 consecutively enrolled patients, we selected 84 (41 males; median age, 59 years; range, 35 to 90 years) with all four sample types available. A total of 202 driver mutations were found in 34 genes. ttDNA exhibited the highest mutation frequency (n=232), followed by baseline-ctDNA (n=155) and PD-ctDNA (n=117). Sequencing ctDNA alongside ttDNA revealed additional mutations in 40 patients (47.6%). PD-ctDNA detected 13 novel mutations in 10 patients (11.9%) compared to ttDNA and baseline-ctDNA. Notably, seven mutations in five patients (6.0%) were missense or nonsense mutations in APC, TP53, SMAD4, and CDH1 genes. In baseline-ctDNA, higher maximal variant allele frequency (VAF) values (p=0.010) and higher VAF values of APC (p=0.012), TP53 (p=0.012), and KRAS (p=0.005) mutations were significantly associated with worse overall survival. CONCLUSION While ttDNA remains more sensitive than ctDNA, our ctDNA platform demonstrated validity and potential value when ttDNA was unavailable. Post-treatment analysis of PD-ctDNA unveiled new pathogenic mutations, signifying cancer's clonal evolution. Additionally, baseline-ctDNA's VAF values were prognostic after treatment.
Collapse
Affiliation(s)
| | - Ji-Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | | | - Sung-Hyun Hwang
- Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kui-Jin Kim
- Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ju Hyun Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
- Department of Statistics, Hankuk University of Foreign Studies, Yongin, Korea
| | - Jeongmin Seo
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Minsu Kang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Eun Hee Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Koung Jin Suh
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Se Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Yu Jung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | | | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
43
|
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
|
44
|
Ambriz-Barrera F, Rojas-Jiménez E, Díaz-Velásquez CE, De-La-Cruz-Montoya AH, Martínez-Gregorio H, Ruiz-De-La-Cruz M, Huertas A, Montealegre AL, Castro-Rojas C, Acosta G, Vaca-Paniagua F, Perdomo S. Mutational spectrum of breast cancer by shallow whole-genome sequencing of cfDNA and tumor gene panel analysis. PLoS One 2024; 19:e0308176. [PMID: 39264897 PMCID: PMC11392417 DOI: 10.1371/journal.pone.0308176] [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: 01/21/2024] [Accepted: 07/17/2024] [Indexed: 09/14/2024] Open
Abstract
Breast cancer (BC) has different molecular subgroups related to different risks and treatments. Tumor biopsies for BC detection are invasive and may not reflect tumor heterogeneity. Liquid biopsies have become relevant because they might overcome these limitations. We rationalize that liquid cfDNA biopsies through shallow whole genome sequencing (sWGS) could improve the detection of tumor alterations, complementing the genomic profiling. We evaluated the feasibility to detect somatic copy number alterations (SCNAs) in BC using shallow whole genome sequencing (sWGS) in cfDNA from archived samples from National Cancer Institute of Colombia patients. We sequenced tumor tissues from 38 BC patients with different molecular subtypes using a gene panel of 176 genes significantly mutated in cancer, and by liquid biopsies using sWGS on 20 paired samples to detect SCNAs and compare with the tumor samples. We identified an extensive intertumoral heterogeneity between the molecular subtypes of BC, with a mean tumor load of 602 mutations in the gene panel of tumor tissues. There was a 12.3% of concordance in deletions in the cfDNA-tumor pairs considering only the genes covered by the panel encompassing seven genes: BRCA1, CDK12, NF1, MAP2K4, NCOR1, TP53, and KEAP1 in three patients. This study shows the feasibility to complement the genomic analysis of tumor tissue biopsies to detect SCNA in BC using sWGS in cfDNA, providing a wider identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Fernando Ambriz-Barrera
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, México
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, México
| | - Ernesto Rojas-Jiménez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, México
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, México
| | - Clara Estela Díaz-Velásquez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, México
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, México
| | - Aldo Hugo De-La-Cruz-Montoya
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, México
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, México
| | - Héctor Martínez-Gregorio
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, México
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, México
| | - Miguel Ruiz-De-La-Cruz
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, México
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, México
| | - Antonio Huertas
- Terry Fox National Tumor Bank, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Ana Lorena Montealegre
- Nutrition, Genetics and Metabolism Research Group, Faculty of Medicine, Universidad El Bosque, Bogotá, Colombia
| | - Carlos Castro-Rojas
- Nutrition, Genetics and Metabolism Research Group, Faculty of Medicine, Universidad El Bosque, Bogotá, Colombia
| | - Gabriela Acosta
- Nutrition, Genetics and Metabolism Research Group, Faculty of Medicine, Universidad El Bosque, Bogotá, Colombia
| | - Felipe Vaca-Paniagua
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla, México
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, México
| | - Sandra Perdomo
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| |
Collapse
|
45
|
Chrenková E, Študentová H, Holá K, Kahounová Z, Hendrychová R, Souček K, Bouchal J. Castration-resistant prostate cancer monitoring by cell-free circulating biomarkers. Front Oncol 2024; 14:1394292. [PMID: 39319053 PMCID: PMC11420116 DOI: 10.3389/fonc.2024.1394292] [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: 03/01/2024] [Accepted: 08/23/2024] [Indexed: 09/26/2024] Open
Abstract
Background Prostate cancer is the second leading cause of male cancer-related deaths in Western countries, which is predominantly attributed to the metastatic castration-resistant stage of the disease (CRPC). There is an urgent need for better prognostic and predictive biomarkers, particularly for androgen receptor targeted agents and taxanes. Methods We have searched the PubMed database for original articles and meta-analyses providing information on blood-based markers for castration-resistant prostate cancer monitoring, risk group stratification and prediction of therapy response. Results The molecular markers are discussed along with the standard clinical parameters, such as prostate specific antigen, lactate dehydrogenase or C-reactive protein. Androgen receptor (AR) alterations are commonly associated with progression to CRPC. These include amplification of AR and its enhancer, point mutations and splice variants. Among DNA methylations, a novel 5-hydroxymethylcytosine activation marker of TOP2A and EZH2 has been identified for the aggressive disease. miR-375 is currently the most promising candidate among non-coding RNAs and sphingolipid analysis has recently emerged as a novel approach. Conclusions The promising biomarkers have the potential to improve the care of metastatic prostate cancer patients, however, they need further validation for routine implementation.
Collapse
Affiliation(s)
- Eva Chrenková
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| | - Hana Študentová
- Department of Oncology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| | - Kateřina Holá
- Department of Oncology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| | - Zuzana Kahounová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia
| | - Romana Hendrychová
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| |
Collapse
|
46
|
Hsieh RW, Symonds LK, Siu J, Cohen SA. Identification of circulating tumor DNA as a biomarker for diagnosis and response to therapies in cancer patients. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 391:43-93. [PMID: 39939078 DOI: 10.1016/bs.ircmb.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
The sampling of circulating biomarkers provides an opportunity for non-invasive evaluation and monitoring of cancer activity. In modern day practice, this has typically been in the form of circulating tumor DNA (ctDNA) detected in plasma. The field of ctDNA has been a burgeoning technology, with prominent applications for blood-based cancer screening and in disease status assessment, especially after curative-intent surgery to evaluate for minimal residual disease (MRD). Clinical applications for the latter show an incredibly high sensitivity in certain cancer types with a need for additional studies to determine how much clinical decision-making should be adapted based on ctDNA results and which cancer types, stages, and treatments are best informed by ctDNA results. This chapter provides an overview of ctDNA detection as tool for cancer screening, detecting MRD, and/or molecularly characterizing a cancer, highlighting the rapidly amassing research as a prognostic biomarker and emerging data on ctDNA as a predictive biomarker.
Collapse
Affiliation(s)
- Ronan W Hsieh
- Division of Hematology/Oncology, University of Washington, Seattle, WA, United States; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Lynn K Symonds
- Division of Hematology/Oncology, University of Washington, Seattle, WA, United States; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Jason Siu
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States
| | - Stacey A Cohen
- Division of Hematology/Oncology, University of Washington, Seattle, WA, United States; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States.
| |
Collapse
|
47
|
Huang FF, Di XF, Bai MH. Analysis of urine cell-free DNA in bladder cancer diagnosis by emerging bioactive technologies and materials. Front Bioeng Biotechnol 2024; 12:1458362. [PMID: 39295845 PMCID: PMC11408225 DOI: 10.3389/fbioe.2024.1458362] [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: 07/02/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
Urinary cell-free DNA (UcfDNA) is gaining recognition as an important biomarker for diagnosing bladder cancer. UcfDNA contains tumor derived DNA sequences, making it a viable candidate for non-invasive early detection, diagnosis, and surveillance of bladder cancer. The quantification and qualification of UcfDNA have demonstrated high sensitivity and specificity in the molecular characterization of bladder cancer. However, precise analysis of UcfDNA for clinical bladder cancer diagnosis remains challenging. This review summarizes the history of UcfDNA discovery, its biological properties, and the quantitative and qualitative evaluations of UcfDNA for its clinical significance and utility in bladder cancer patients, emphasizing the critical role of UcfDNA in bladder cancer diagnosis. Emerging bioactive technologies and materials currently offer promising tools for multiple UcfDNA analysis, aiming to achieve more precise and efficient capture of UcfDNA, thereby significantly enhancing diagnostic accuracy. This review also highlights breakthroughs in detection technologies and substrates with the potential to revolutionize bladder cancer diagnosis in clinic.
Collapse
Affiliation(s)
- Fei-Fei Huang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiao-Fei Di
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Mo-Han Bai
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
48
|
Dickinson K, Sharma A, Agnihotram RKV, Altuntur S, Park M, Meterissian S, Burnier JV. Circulating Tumor DNA and Survival in Metastatic Breast Cancer: A Systematic Review and Meta-Analysis. JAMA Netw Open 2024; 7:e2431722. [PMID: 39235812 PMCID: PMC11378006 DOI: 10.1001/jamanetworkopen.2024.31722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Importance Metastatic breast cancer (MBC) poses a substantial clinical challenge despite advancements in diagnosis and treatment. While tissue biopsies offer a static snapshot of disease, liquid biopsy-through detection of circulating tumor DNA (ctDNA)-provides minimally invasive, real-time insight into tumor biology. Objective To determine the association between ctDNA and survival outcomes in patients with MBC. Data Sources An electronic search was performed in 5 databases (CINAHL, Cochrane Library, Embase, Medline, and Web of Science) and included all articles published from inception until October 23, 2023. Study Selection To be included in the meta-analysis, studies had to (1) include women diagnosed with MBC; (2) report baseline plasma ctDNA data; and (3) report overall survival, progression-free survival, or disease-free survival with associated hazards ratios. Data Extraction and Synthesis Titles and abstracts were screened independently by 2 authors. Data were pooled using a random-effects model. This study adhered to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guideline, and quality was assessed using the Newcastle-Ottawa Scale. Main Outcomes and Measures The primary study outcome was the association between detection of specific genomic alterations in ctDNA with survival outcomes. Secondary objectives were associations of study methodology with survival. Results Of 3162 articles reviewed, 37 met the inclusion criteria and reported data from 4264 female patients aged 20 to 94 years. Aggregated analysis revealed a significant association between ctDNA detection and worse survival (hazard ratio, 1.40; 95% CI, 1.22-1.58). Subgroup analysis identified significant associations of TP53 and ESR1 alterations with worse survival (hazard ratios, 1.58 [95% CI, 1.34-1.81] and 1.28 [95% CI, 0.96-1.60], respectively), while PIK3CA alterations were not associated with survival outcomes. Stratifying by detection method, ctDNA detection through next-generation sequencing and digital polymerase chain reaction was associated with worse survival (hazard ratios, 1.48 [95% CI, 1.22-1.74] and 1.28 [95% CI, 1.05-1.50], respectively). Conclusions and Relevance In this systematic review and meta-analysis, detection of specific genomic alterations in ctDNA was associated with worse overall, progression-free, and disease-free survival, suggesting its potential as a prognostic biomarker in MBC. These results may help guide the design of future studies to determine the actionability of ctDNA findings.
Collapse
Affiliation(s)
- Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Archi Sharma
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Selin Altuntur
- McConnell Resource Centre Medical Library, McGill University Health Centre, Montreal, Quebec, Canada
| | - Morag Park
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Sarkis Meterissian
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
- Department of Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
49
|
Ou X, Gao G, Habaz IA, Wang Y. Mechanisms of resistance to tyrosine kinase inhibitor-targeted therapy and overcoming strategies. MedComm (Beijing) 2024; 5:e694. [PMID: 39184861 PMCID: PMC11344283 DOI: 10.1002/mco2.694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
Tyrosine kinase inhibitor (TKI)-targeted therapy has revolutionized cancer treatment by selectively blocking specific signaling pathways crucial for tumor growth, offering improved outcomes with fewer side effects compared with conventional chemotherapy. However, despite their initial effectiveness, resistance to TKIs remains a significant challenge in clinical practice. Understanding the mechanisms underlying TKI resistance is paramount for improving patient outcomes and developing more effective treatment strategies. In this review, we explored various mechanisms contributing to TKI resistance, including on-target mechanisms and off-target mechanisms, as well as changes in the tumor histology and tumor microenvironment (intrinsic mechanisms). Additionally, we summarized current therapeutic approaches aiming at circumventing TKI resistance, including the development of next-generation TKIs and combination therapies. We also discussed emerging strategies such as the use of dual-targeted antibodies and PROteolysis Targeting Chimeras. Furthermore, we explored future directions in TKI-targeted therapy, including the methods for detecting and monitoring drug resistance during treatment, identification of novel targets, exploration of dual-acting kinase inhibitors, application of nanotechnologies in targeted therapy, and so on. Overall, this review provides a comprehensive overview of the challenges and opportunities in TKI-targeted therapy, aiming to advance our understanding of resistance mechanisms and guide the development of more effective therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
- Xuejin Ou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Ge Gao
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China HospitalSichuan UniversityChengduChina
| | - Inbar A. Habaz
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
50
|
Yoo TKR, Lee JY, Park H, Cho WK, Jeon S, Jun HR, Lee SB, Chung IY, Kim HJ, Ko BS, Lee JW, Son BH, Ahn SH, Jeong JH, Kim JE, Ahn JH, Jung KH, Kim SB, Lee HJ, Gong G, Kim J, Chun SM. Longitudinal dynamics of circulating tumor DNA for treatment monitoring in patients with breast cancer recurrence. Sci Rep 2024; 14:20252. [PMID: 39215119 PMCID: PMC11364657 DOI: 10.1038/s41598-024-70887-5] [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: 01/09/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The prevalence and dynamics of circulating tumor DNA (ctDNA) in patients with breast cancer recurrence or de novo metastatic cancer were examined in a retrospective analysis of a prospective observational cohort. Twenty-three recurrent/metastatic breast cancer cases (8 locoregional, 15 distant metastasis) were enrolled, and sequential plasma samples were obtained. Anchor mutations were selected from the target sequencing of each patient's primary and/or metastatic tumor. An in-house developed assay (UHS assay) was employed for a tumor-informed ctDNA assay during treatment and follow-up. A median of three (range 1-5) anchor mutations per case were applied for ctDNA detection. ctDNA was detected in 14 (63.6%, 14/22) cases at the time of enrollment and 18 (78.5%, 18/23) cases during follow-up. More anchor mutations and higher tumor burden were significantly related to higher ctDNA positive rates (p-value 0.036, 0.043, respectively). The mean enriched variant allele frequency (eVAF) at each time point was significantly higher for stable or progressive disease responses (ANOVA test p-value < 0.001). Eight patients showed an increase in their ctDNA eVAF prior to clinical progression with a mean lead time of 6.2 months (range 1.5-11 months). ctDNA dynamics measured using personalized assay reflected the clinical course of breast cancer recurrence.
Collapse
Affiliation(s)
- Tae-Kyung Robyn Yoo
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji-Young Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Hwan Park
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Whi-Kyung Cho
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seyeon Jeon
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Ha Ra Jun
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Sae Byul Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Il Yong Chung
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Jeong Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Beom Seok Ko
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jong Won Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byung Ho Son
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sei-Hyun Ahn
- Department of Surgery, Ewha Womens University Mokdong Hospital, Seoul, Republic of Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong Eun Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin-Hee Ahn
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jisun Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Sung-Min Chun
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea.
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|