1
|
Fang Z, Cao A, Huang Z, Jin X, Zhang Z, Cao Y, Zhao J, Zuo X. Nanosized Membrane Fusion-Triggered Three-Dimensional DNA Walker for Subtype-Specific Analysis of Breast Cancer Extracellular Vesicles. NANO LETTERS 2025; 25:5512-5519. [PMID: 40129113 DOI: 10.1021/acs.nanolett.5c01221] [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: 03/26/2025]
Abstract
Extracellular vesicles (EVs) are highly attractive in cancer diagnosis, owing to their ability to reflect the molecular characteristics of the source cells. Herein, we engineer a DNA-equipped liposome nanoprobe for developing a nanosized membrane fusion-triggered three-dimensional (3D) DNA walker for the analysis of breast cancer EVs. Specifically, liposome nanoprobes efficiently fuse with target EVs by recognizing surface-expressed epidermal growth factor receptors, thereby creating 3D tracks for the DNA walker. Subsequently, walking probes targeting human epidermal growth factor receptor-2 (HER-2) were recruited onto the fused vesicles to manipulate the walker, eventually generating considerable electrochemical signals. Results reveal a good linearity between the electrochemical output and the target EV concentration and also prove the ability of the membrane fusion-triggered DNA walker to differentiate HER-2-positive breast cancer patients and monitor the disease progression. Taken together, this work presents an effective approach for the early and subtype-specific diagnosis of breast cancer.
Collapse
Affiliation(s)
- Zhikai Fang
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Sciences, Shanghai University, Shanghai 200444, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ayong Cao
- Key Laboratory of Breast Cancer, Department of Breast Surgery Fudan University Shanghai Cancer Center, Shanghai 201321, China
| | - Zichen Huang
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xi Jin
- Key Laboratory of Breast Cancer, Department of Breast Surgery Fudan University Shanghai Cancer Center, Shanghai 201321, China
| | - Zhan Zhang
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Ya Cao
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jing Zhao
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
2
|
Lehrer EJ, Khunsriraksakul C, Garrett S, Trifiletti DM, Sheehan JP, Guckenberger M, Louie AV, Siva S, Ost P, Goodman KA, Dawson LA, Tchelebi LT, Yang JT, Showalter TN, Park HS, Spratt DE, Kishan AU, Gupta GP, Shah C, Fanti S, Calais J, Wang M, Schmitz K, Liu D, Abraham JA, Dess RT, Buvat I, Solomon B, Zaorsky NG. Future directions in the evaluation and management of newly diagnosed metastatic cancer. Crit Rev Oncol Hematol 2025; 208:104631. [PMID: 39864534 DOI: 10.1016/j.critrevonc.2025.104631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 01/28/2025] Open
Abstract
There is much debate regarding optimal selection in patients with metastatic cancer who should undergo local treatment (surgery or radiation treatment) to the primary tumor and/or metastases. Additionally, the optimal treatment of newly diagnosed metastatic cancer is largely unclear. Current prognostication systems to best inform these clinical scenarios are limited, as all metastatic patients are grouped together as having Stage IV disease without further incorporation of patient and disease-specific covariates that significantly impact patient outcomes. Therefore, improving current prognostic scoring systems and incorporation of these covariates is essential to best individualize treatment for patients with metastatic cancer. In this narrative review article, we provide a detailed review of prognostication systems that can be used for both the site of metastasis and primary site to best tailor treatment in these patients. Additionally, we discuss the incorporation and ongoing developments in radiographic, genomic, and biostatistical techniques that can be used as prognostication tools.
Collapse
Affiliation(s)
- Eric J Lehrer
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA.
| | | | - Sara Garrett
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, USA
| | | | - Jason P Sheehan
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, USA
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Alexander V Louie
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Shankar Siva
- Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, GZA Ziekenhuizen, Wilrijk, Belgium; Department of Human Structure and Repair, Ghent University, Belgium, Iridium Network, GZA Ziekenhuizen, Wilrijk, Belgium
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura A Dawson
- Department of Radiation Oncology, University of Toronto, Ontario, Canada
| | | | - Jonathan T Yang
- Department of Radiation Oncology, New York University School of Medicine, New York, NY, USA
| | - Timothy N Showalter
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA, USA
| | - Henry S Park
- Department of Therapeutic Radiology, Yale University, New Haven, CT, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Amar U Kishan
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA
| | - Gaorav P Gupta
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Chirag Shah
- Department of Radiation Oncology, Allegheny Health Network, Pittsburgh, PA, USA
| | - Stefano Fanti
- Department of Nuclear Medicine, IRCCS AOU di Bologna, Italy
| | - Jeremie Calais
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Ming Wang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kathryn Schmitz
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dajiang Liu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - John A Abraham
- Rothman Institute, Thomas Jefferson University, Philadelphia, PA, USA
| | - Robert T Dess
- Department of Radiation Oncology, University of Michigan, MI, USA
| | - Irène Buvat
- Laboratory of Translational Imaging in Oncology, Institut Curie, Inserm, PSL University, Orsay, France
| | - Benjamin Solomon
- Department of Medical Oncology, University of Melbourne- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
3
|
Kierans AS, Lutfi A, Afghan MK, Khan S, Javaid S, Currie BM, Rocca J, Samstein B, Golden E, Popa E, Hissong E, Kasi PM. Spectrum of Findings Seen in Patients With IDH1/2-Mutant Cholangiocarcinoma. Int J Surg Pathol 2025; 33:417-425. [PMID: 39314068 DOI: 10.1177/10668969241271397] [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: 09/25/2024]
Abstract
BackgroundCholangiocarcinoma-with a growing incidence rate and poor prognosis-is not an uncommon cancer. Molecular profiling can reveal actionable aberrations in at least a third of the tumors. This is especially so in the case of intrahepatic cholangiocarcinoma (ICC), where mutations in the isocitrate dehydrogenase 1 and 2 genes (IDH1/2) make up 15%-20% of these tumors. IDH1/2 mutant ICC is a rare disease that has not been adequately reported. To expand the spectrum of findings seen in these patients, we present a single institution case series.Methods and resultsWe descriptively characterize the clinical, radiological, and histopathological findings of 12 such patients. IDH1/2 mutant ICC was found in elderly women, with two-thirds of patients having additional co-mutations. Anecdotally, patients who did receive systemic and/or locoregional therapies had long-term durable outcomes.ConclusionOur findings indicate an increasing need to personalize an approach for these patients with specific molecular alterations. With the advent of the IDH1 inhibitor ivosidenib and other inhibitors in this space, IDH1/2 mutation have both prognostic and predictive value. Our series builds upon the patterns and findings seen in these patients.
Collapse
Affiliation(s)
| | - Areeb Lutfi
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Maaz Khan Afghan
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sahrish Khan
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sana Javaid
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Brian Michael Currie
- Department of Vascular and Interventional Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Juan Rocca
- Division of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Benjamin Samstein
- Division of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Encouse Golden
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Elizabeta Popa
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Erika Hissong
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Pashtoon Murtaza Kasi
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
4
|
Zhou Y, Wang R, Zeng M, Liu S. Circulating tumor DNA: a revolutionary approach for early detection and personalized treatment of bladder cancer. Front Pharmacol 2025; 16:1551219. [PMID: 40191434 PMCID: PMC11968738 DOI: 10.3389/fphar.2025.1551219] [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: 12/25/2024] [Accepted: 02/25/2025] [Indexed: 04/09/2025] Open
Abstract
Bladder cancer is a malignant tumor with a high global incidence and recurrence rate. Traditional diagnostic methods, such as cystoscopy and urine cytology, have limitations in sensitivity and specificity, particularly in detecting low-grade bladder cancer. Circulating tumor DNA (ctDNA) offers a non-invasive alternative, reflecting tumor genetic characteristics through blood samples. It demonstrates high sensitivity and repeatability, making it a promising tool for early detection, recurrence monitoring, and treatment evaluation. Clinical studies have shown that ctDNA not only detects tumor burden but also captures dynamic tumor mutations, aiding in personalized treatment strategies. Despite its potential, clinical implementation of ctDNA faces challenges, including optimization of detection techniques, standardization, and the cost of testing. This paper explores the role of ctDNA in advancing bladder cancer diagnosis and treatment, with a focus on refining its clinical application and guiding future research toward improved patient outcomes.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Rongzhong Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Mingtang Zeng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Sijia Liu
- West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Andersen L, Birkbak NJ. Advancing circulating tumor DNA detection: using whole-genome sequencing to power minimal residual disease monitoring in breast cancer. Ann Oncol 2025:S0923-7534(25)00114-0. [PMID: 40120761 DOI: 10.1016/j.annonc.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Affiliation(s)
- L Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Bioinformatics Research Center (BiRC), Aarhus University, Aarhus, Denmark
| | - N J Birkbak
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Bioinformatics Research Center (BiRC), Aarhus University, Aarhus, Denmark.
| |
Collapse
|
6
|
Montalti R, Pepe F, Cassese G, Russo G, Malapelle U, Carlomagno C, Giglio MC, Falco G, Alagia M, De Simone G, Geboes K, Troncone G, Troisi RI, Rompianesi G. Prognostic role of postoperative persistence of ctDNA molecular signature after liver resection for colorectal liver metastases: Preliminary results from a prospective study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109706. [PMID: 40009912 DOI: 10.1016/j.ejso.2025.109706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
INTRODUCTION Liver resection represents the cornerstone treatment for Colo-Rectal Liver Metastases (CRLM), but it is still followed by a high recurrence rate. The identification of a valid and reproducible predictor of recurrence can play a key role in correct and timely management of the disease. In this scenario, liquid biopsy may represent an integrative, less invasive, and easily manageable tool to clinically stratify colorectal cancer patients.We aimed to investigate the prognostic role of persistent circulating tumor DNA (ctDNA) signature on CRLM recurrence after liver resection. METHODS A series of 51 consecutive patients undergoing liver resection for CRLM were prospectively enrolled between January 2020 and March 2023. Patients underwent liquid biopsy from peripheral blood samples before and after surgery. Other risk factors for disease recurrence were also investigated. RESULTS Twenty-nine patients were found to be positive for one of the clinically relevant molecular alterations detected by NGS, at preoperative (T0) ctDNA analysis. At univariate analysis, total tumor size bigger than 60 mm (p = 0.046, HR 2.486) and postoperative persistence of ctDNA molecular signature (p = 0.024, HR 2.831) were significantly associated with recurrence. Multivariable Cox Regression analysis showed that only postoperative persistence of ctDNA molecular signature was associated with recurrence (p = 0.027, HR 2.766). Similarly, 1-3 yr DFS was significantly lower in the subgroup with persistence of molecular signature at liquid biopsy (100-49.5 % vs 57.1-21.4 %, p = 0.018). CONCLUSION Postoperative persistence of ctDNA molecular signature could represent a useful tool to predict recurrence after liver resection for CRLM patients.
Collapse
Affiliation(s)
- Roberto Montalti
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy; Department of Public Health, Federico II University, 80131, Naples, Italy.
| | - Francesco Pepe
- Department of Public Health, Federico II University, 80131, Naples, Italy
| | - Gianluca Cassese
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, Federico II University, 80131, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, Federico II University, 80131, Naples, Italy
| | - Chiara Carlomagno
- Department of Clinical Medicine and Surgery, Division of Oncology, University Federico II, Naples, Italy
| | - Mariano Cesare Giglio
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Giacinto Falco
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Mariantonietta Alagia
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Giuseppe De Simone
- Department of Anesthesiology, Federico II University Hospital, Naples, Italy
| | - Karen Geboes
- Oncology Centrum, Ghent University Hospital, Belgium
| | - Giancarlo Troncone
- Department of Public Health, Federico II University, 80131, Naples, Italy
| | - Roberto Ivan Troisi
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| | - Gianluca Rompianesi
- Department of Clinical Medicine and Surgery, Division of HBP, Minimally Invasive and Robotic Surgery, Transplantation Service, Federico II University Hospital, 80131, Naples, Italy
| |
Collapse
|
7
|
Guo W, Chen W, Zhang J, Li M, Huang H, Wang Q, Fei X, Huang J, Zheng T, Fan H, Wang Y, Gu H, Ding G, Chen Y. High-throughput methylation sequencing reveals novel biomarkers for the early detection of renal cell carcinoma. BMC Cancer 2025; 25:96. [PMID: 39819319 PMCID: PMC11737265 DOI: 10.1186/s12885-024-13380-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025] Open
Abstract
PURPOSE Renal cell carcinoma (RCC) is a common malignancy, with patients frequently diagnosed at an advanced stage due to the absence of sufficiently sensitive detection technologies, significantly compromising patient survival and quality of life. Advances in cell-free DNA (cfDNA) methylation profiling using liquid biopsies offer a promising non-invasive diagnostic option, but robust biomarkers for early detection are current not available. This study aimed to identify methylation biomarkers for RCC and establish a DNA methylation signature-based prognostic model for this disease. METHODS High-throughput methylation sequencing was performed on peripheral blood samples obtained from 49 primarily Stage I RCC patients and 44 healthy controls. Comparative analysis and Least Absolute Shrinkage and Selection Operator (LASSO) regression methods were employed to identify RCC methylation signatures.Subsequently, methylation markers-based diagnostic and prognostic models for RCC were independently trained and validated using random forest and Cox regression methodologies, respectively. RESULTS Comparative analysis revealed 864 differentially methylated CpG islands (DMCGIs), 96.3% of which were hypermethylated. Using a training set from The Cancer Genome Atlas (TCGA) dataset of 443 early-stage RCC tumors and matched normal tissues, we applied LASSO regression and identified 23 methylation signatures. We then constructed a random forest-based diagnostic model for early-stage RCC and validated the model using two independent datasets: a TCGA set of 460 RCC tumors and controls, and a blood sample set from our study of 15 RCC cases and 29 healthy controls. For Stage I RCC tissue, the model showed excellent discrimination (AUC-ROC: 0.999, sensitivity: 98.5%, specificity: 100%). Blood sample validation also yielded commendable results (AUC-ROC: 0.852, sensitivity: 73.9%, specificity: 89.7%). Further analysis using Cox regression identified 7 of the 23 DMCGIs as prognostic markers for RCC, allowing the development of a prognostic model with strong predictive power for 1-, 3-, and 5-year survival (AUC-ROC > 0.7). CONCLUSIONS Our findings highlight the critical role of hypermethylation in RCC etiology and progression, and present these identified biomarkers as promising candidates for diagnostic and prognostic applications.
Collapse
Affiliation(s)
- Wenhao Guo
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
- Department of Urology, Shaoxing Branch of Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Shaoxing, 312000, Zhejiang Province, China
| | - Weiwu Chen
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
- School of Medicine, Zhejiang University, Hangzhou, 310011, Zhejiang Province, China
| | - Jie Zhang
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
| | - Mingzhe Li
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
| | - Hongyuan Huang
- Department of Urology, Jinjiang Municipal Hospital, Quanzhou, 362000, Fujian Province, China
| | - Qian Wang
- Hangzhou Shengting Medical Technology Co., Ltd., Hangzhou, 310018, Zhejiang Province, China
| | - Xiaoyi Fei
- Hangzhou Shengting Medical Technology Co., Ltd., Hangzhou, 310018, Zhejiang Province, China
| | - Jian Huang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui Province, China
| | - Tongning Zheng
- Department of Urology, Ningbo Zhenhai People's Hospital, Ningbo, 315202, Zhejiang Province, China
| | - Haobo Fan
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
- School of Medicine, Zhejiang University, Hangzhou, 310011, Zhejiang Province, China
| | - Yunfei Wang
- Hangzhou Shengting Medical Technology Co., Ltd., Hangzhou, 310018, Zhejiang Province, China
| | - Hongcang Gu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui Province, China.
| | - Guoqing Ding
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China.
| | - Yicheng Chen
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China.
| |
Collapse
|
8
|
Cao Y, Xia J, Li L, Zeng Y, Zhao J, Li G. Electrochemical Biosensors for Cancer Diagnosis: Multitarget Analysis to Present Molecular Characteristics of Tumor Heterogeneity. JACS AU 2024; 4:4655-4672. [PMID: 39735934 PMCID: PMC11672140 DOI: 10.1021/jacsau.4c00989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/31/2024]
Abstract
Electrochemical biosensors are gaining attention as powerful tools in cancer diagnosis, particularly in liquid biopsy, due to their high efficiency, rapid response, exceptional sensitivity, and specificity. However, the complexity of intra- and intertumor heterogeneity, with variations in genetic and protein expression profiles and epigenetic modifications, makes electrochemical biosensors susceptible to false-positive or false-negative diagnostic outcomes. To address this challenge, there is growing interest in simultaneously analyzing multiple biomarkers to reveal molecular characteristics of tumor heterogeneity for precise cancer diagnosis. In this Perspective, we highlight recent advancements in utilizing electrochemical biosensors for cancer diagnosis, with a specific emphasis on the multitarget analysis of cancer biomarkers including tumor-associated nucleic acids, tumor protein markers, extracellular vesicles, and tumor cells. These biosensors hold significant promise for improving precision in early cancer diagnosis and monitoring, as well as potentially offering new insights into personalized cancer management.
Collapse
Affiliation(s)
- Ya Cao
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jianan Xia
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Lijuan Li
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yujing Zeng
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jing Zhao
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Genxi Li
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
9
|
Li H, Zhao P, Tian L, Lu Y, Wang X, Shao W, Cheng Y. Advances in biomarkers for immunotherapy in small-cell lung cancer. Front Immunol 2024; 15:1490590. [PMID: 39723215 PMCID: PMC11668642 DOI: 10.3389/fimmu.2024.1490590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
Small-cell lung cancer (SCLC) is a refractory cancer with rapid growth and high aggressiveness. Extensive-stage SCLC is initially sensitive to chemotherapy; however, drug resistance and recurrence occur rapidly, resulting in a poor survival outcome due to lack of subsequently efficient therapy. The emergence of immune checkpoint inhibitors (ICIs) generated a new landscape of SCLC treatment and significantly prolonged the survival of patients. However, the unselected immunotherapy restrains both beneficiary population and responsive period in SCLC compared to the other tumors. The complex tumor origin, high heterogeneity, and immunosuppressive microenvironment may disturb the value of conventional biomarkers in SCLC including programmed cell death 1 ligand 1 and tumor mutation burden. Transcriptional regulator-based subtypes of SCLC are current research hotspot, revealing that Y (I) subtype can benefit from ICIs. Additionally, molecules related to immune microenvironment, immunogenicity, epigenetics, and SCLC itself also indicated the therapeutic benefits of ICIs, becoming potential predictive biomarkers. In this review, we discussed the advances of biomarkers for prediction and prognosis of immunotherapy, promising directions in the future, and provide reference and options for precision immunotherapy and survival improvement in patients with SCLC.
Collapse
Affiliation(s)
- Hui Li
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Peiyan Zhao
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Lin Tian
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
| | - Yuanhua Lu
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
| | - Xinyue Wang
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
| | - Wenjun Shao
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
| | - Ying Cheng
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| |
Collapse
|
10
|
Allen-Taylor D, Boro G, Cabato P, Mai C, Nguyen K, Rijal G. Staphylococcus epidermidis biofilm in inflammatory breast cancer and its treatment strategies. Biofilm 2024; 8:100220. [PMID: 39318870 PMCID: PMC11420492 DOI: 10.1016/j.bioflm.2024.100220] [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: 08/06/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Bacterial biofilms represent a significant challenge in both clinical and industrial settings because of their robust nature and resistance to antimicrobials. Biofilms are formed by microorganisms that produce an exopolysaccharide matrix, protecting function and supporting for nutrients. Among the various bacterial species capable of forming biofilms, Staphylococcus epidermidis, a commensal organism found on human skin and mucous membranes, has emerged as a prominent opportunistic pathogen, when introduced into the body via medical devices, such as catheters, prosthetic joints, and heart valves. The formation of biofilms by S. epidermidis on these surfaces facilitates colonization and provides protection against host immune responses and antibiotic therapies, leading to persistent and difficult-to-treat infections. The possible involvement of biofilms for breast oncogenesis has recently created the curiosity. This paper therefore delves into S. epidermidis biofilm involvement in breast cancer. S. epidermidis biofilms can create a sustained inflammatory environment through their metabolites and can break DNA in breast tissue, promoting cellular proliferation, angiogenesis, and genetic instability. Preventing biofilm formation primarily involves preventing bacterial proliferation using prophylactic measures and sterilization of medical devices and equipment. In cancer treatment, common modalities include chemotherapy, surgery, immunotherapy, alkylating agents, and various anticancer drugs. Understanding the relationship between anticancer drugs and bacterial biofilms is crucial, especially for those undergoing cancer treatment who may be at increased risk of bacterial infections, for improving patient outcomes. By elucidating these interactions, strategies to prevent or disrupt biofilm formation, thereby reducing the incidence of infections associated with medical devices and implants, can be identified.
Collapse
Affiliation(s)
- D. Allen-Taylor
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - G. Boro
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - P.M. Cabato
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - C. Mai
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - K. Nguyen
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - G. Rijal
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| |
Collapse
|
11
|
Liu S, Zhu Y, Chen Y, Wang Y, Zhang D, Zhang J, Wang Y, Zhang A, Hu Q, Liu A. Circulating Tumor DNA Combining with Imaging Analysis for Lesion Detection of Langerhans Cell Histiocytosis in Children. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1449. [PMID: 39767878 PMCID: PMC11675047 DOI: 10.3390/children11121449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND The detection of mutations from circulating tumor DNA (ctDNA) represents a promising enrichment technique. In this retrospective study, the significance of ctDNA and imaging in Langerhans cell histiocytosis (LCH) monitoring was first examined, and the broader role of ctDNA in monitoring LCH was additionally explored. METHODS First, data visualization and survival analysis models were used to generalize the concordance between cfBRAFV600E molecular response and radiographic response on clinical outcomes. Next, the molecular response of cfBRAFV600E was observed from a dynamic perspective. A comparative analysis was then conducted between cfBRAFV600E and ltBRAFV600E status, examining their relationship to clinical manifestations and prognosis of LCH. RESULTS Eventually, 119 participants were enrolled in this trial between 2019 and 2023. Progression-free survival (PFS) was significantly shorter in patients with both radiologic and cfDNA molecular progression (17.67 versus 24.67 months, p < 0.05) compared to those without. A critical cfBRAFV600E value of 0.03% has been determined for the first time. Both cfBRAFV600E and ltBRAFV600E mutations were associated with a higher proportion of children under 3 years of age, skin and spleen involvement, and a lower 3-year PFS rate. In contrast to ltBRAFV600E, cfBRAFV600E was linked to a higher proportion of risk organ invasion LCH (52% vs. 27.9%, p < 0.05) and a better therapeutic response at the sixth week (24% vs. 4.7%, p < 0.05). Furthermore, in patients with risk organ invasion-LCH and multisystem-LCH subtypes, cfBRAFV600E was associated with a significantly lower 3-year PFS. CONCLUSIONS In summary, these findings enhanced and supplemented the implications of ctDNA and imaging analysis application in children with LCH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Aiguo Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
12
|
Koh DM. Liquid Biopsy and Imaging Are Complementary for Assessing Tumor Burden. Radiology 2024; 313:e242525. [PMID: 39589252 DOI: 10.1148/radiol.242525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Affiliation(s)
- Dow-Mu Koh
- From the Department of Radiology, The Royal Marsden NHS Foundation Trust, Downs Rd, Sutton SM2 5PT, United Kingdom
| |
Collapse
|
13
|
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
|
14
|
Jiao J, Zeng D, Wu Y, Li C, Mo T. Programmable and ultra-efficient Argonaute protein-mediated nucleic acid tests: A review. Int J Biol Macromol 2024; 278:134755. [PMID: 39147338 DOI: 10.1016/j.ijbiomac.2024.134755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
With the attributes of high sensitivity, single-base resolution, multiplex detection capability, and programmability upon nucleic acid recognition, Argonaute (Ago)-based biosensing assays are increasingly recognized as one of the most promising tools for precise identification and quantification of target analytes. Employed as highly specific sequence recognition elements of these robust diagnostic methods, Agos are revolutionizing how nucleic acid targets are detected. A systematic and comprehensive summary of this emerging and rapid-advancing technology is necessary to give play to the potential of Ago-based biosensing assays. The structure and function of Agos were briefly overviewed at the beginning of the work, followed by a review of the recent advancements in employing Agos sensing for detecting various targets with a comprehensive analysis such as viruses, tumor biomarkers, pathogens, mycoplasma, and parasite. The significance and benefits of these platforms were then deliberated. In addition, the authors shared subjective viewpoints on the existing challenges and offered relevant guidance for the future progress of Agos assays. Finally, the future research outlook regarding Ago-based sensing in this field was also outlined. As such, this review is expected to offer valuable information and fresh perspectives for a broader group of researchers.
Collapse
Affiliation(s)
- Jinlong Jiao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Dandan Zeng
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yafang Wu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Chentao Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Tianlu Mo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
15
|
Qiu SF, Zhang QZ, Wu ZY, Liu MZ, Ding Q, Sun FM, Wang Y, Yang HX, Zheng L, Chen X, Wu L, Bai J, Liu JF, Chen CB. Establishment and validation of circulating cell-free DNA signatures for nasopharyngeal carcinoma detection. EBioMedicine 2024; 108:105321. [PMID: 39265506 PMCID: PMC11416236 DOI: 10.1016/j.ebiom.2024.105321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Early detection of nasopharyngeal carcinoma (NPC) poses a significant challenge. The absence of highly sensitive and specific diagnostic biomarkers for nasopharyngeal carcinoma contributes to the unfavourable prognosis of NPC patients. Here, we aimed to establish a non-invasive approach for detecting NPC using circulating cell-free DNA (cfDNA). METHODS We investigated the potential of next-generation sequencing (NGS) of peripheral blood cells as a diagnostic tool for NPC. We collected data on genome-wide nucleosome footprint (NF), 5'-end motifs, fragmentation patterns, CNV information, and EBV content from 553 Chinese subjects, including 234 NPC patients and 319 healthy individuals. Through case-control analysis, we developed a diagnostic model for NPC, and validated its detection capability. FINDINGS Our findings revealed that the frequencies of NF, fragmentation, and motifs were significantly higher in NPC patients compared to healthy controls. We developed an NPC score based on these parameters that accurately distinguished NPC from non-NPC cases according to the American Joint Committee on Cancer staging system from non-NPC (validation set: area under curve (AUC) = 99.9% (95% CI: 99.8%-100%), se: 98.15%, sp: 100%). This model showed superior performance over plasma EBV DNA. Additionally, the NPC score effectively differentiated between NPC patients and healthy controls, even after clinical treatment. Furthermore, the NPC score was found to be independent of potential confounders such as age, sex, or TNM stage. INTERPRETATION We have developed and verified a non-invasive approach with substantial potential for clinical application in detecting NPC. FUNDING A full list of funding bodies that contributed to this study can be found in Funding section.
Collapse
Affiliation(s)
- Su-Fang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China
| | | | - Zi-Yi Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Ming-Zhu Liu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Qin Ding
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China
| | - Fu-Ming Sun
- Berry Oncology Corporation, Beijing, 100102, China
| | - Yin Wang
- Berry Oncology Corporation, Beijing, 100102, China; Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, 350200, China
| | - Han-Xuan Yang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China
| | - Lu Zheng
- Berry Oncology Corporation, Beijing, 100102, China
| | - Xin Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Lin Wu
- Berry Oncology Corporation, Beijing, 100102, China
| | - Jian Bai
- Berry Oncology Corporation, Beijing, 100102, China; Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, 350200, China.
| | - Jing-Feng Liu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, China.
| | - Chuan-Ben Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China; Fujian Provincial Key Laboratory of Tumor Biotherapy Fuzhou, 350200, China.
| |
Collapse
|
16
|
Powles T, Chang YH, Yamamoto Y, Munoz J, Reyes-Cosmelli F, Peer A, Cohen G, Yu EY, Lorch A, Bavle A, Homet Moreno B, Markensohn J, Edmondson M, Chen C, Cristescu R, Peña C, Lunceford J, Gunduz S. Pembrolizumab for advanced urothelial carcinoma: exploratory ctDNA biomarker analyses of the KEYNOTE-361 phase 3 trial. Nat Med 2024; 30:2508-2516. [PMID: 38823511 PMCID: PMC11405267 DOI: 10.1038/s41591-024-03091-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
Circulating tumor DNA (ctDNA) is emerging as a potential biomarker in early-stage urothelial cancer, but its utility in metastatic disease remains unknown. In the phase 3 KEYNOTE-361 study, pembrolizumab with and without chemotherapy was compared with chemotherapy alone in patients with metastatic urothelial cancer. The study did not meet prespecified efficacy thresholds for statistical significance. To identify potential biomarkers of response, we retrospectively evaluated the association of pre- and posttreatment ctDNA with clinical outcomes in a subset of patients who received pembrolizumab (n = 130) or chemotherapy (n = 130) in KEYNOTE-361. Baseline ctDNA was associated with best overall response (BOR; P = 0.009), progression-free survival (P < 0.001) and overall survival (OS; P < 0.001) for pembrolizumab but not for chemotherapy (all; P > 0.05). Chemotherapy induced larger ctDNA decreases from baseline to treatment cycle 2 than pembrolizumab; however, change with pembrolizumab (n = 87) was more associated with BOR (P = 4.39 × 10-5) and OS (P = 7.07 × 10-5) than chemotherapy (n = 102; BOR: P = 1.01 × 10-4; OS: P = 0.018). Tumor tissue-informed versions of ctDNA change metrics were most associated with clinical outcomes but did not show a statistically significant independent value for explaining OS beyond radiographic change by RECIST v.1.1 when jointly modeled (pembrolizumab P = 0.364; chemotherapy P = 0.823). These results suggest distinct patterns in early ctDNA changes with immunotherapy and chemotherapy and differences in their association with long-term outcomes, which provide preliminary insights into the utility of liquid biopsies for treatment monitoring in metastatic urothelial cancer. Clinical trial registration: NCT02853305 .
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/therapeutic use
- Circulating Tumor DNA/blood
- Circulating Tumor DNA/genetics
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/blood
- Female
- Male
- Aged
- Middle Aged
- Retrospective Studies
- Antineoplastic Agents, Immunological/therapeutic use
- Urologic Neoplasms/drug therapy
- Urologic Neoplasms/genetics
- Urologic Neoplasms/pathology
- Urologic Neoplasms/blood
- Carcinoma, Transitional Cell/drug therapy
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/blood
- Carcinoma, Transitional Cell/pathology
- Carcinoma, Transitional Cell/mortality
- Treatment Outcome
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/blood
- Urinary Bladder Neoplasms/pathology
- Progression-Free Survival
Collapse
Affiliation(s)
- Thomas Powles
- Barts Cancer Institute, Queen Mary University of London, London, UK.
| | | | | | - Jose Munoz
- Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | | | | | - Graham Cohen
- Mary Potter Oncology Centre, Gauteng, South Africa
| | - Evan Y Yu
- Fred Hutchinson Cancer Center and University of Washington, Seattle, WA, USA
| | - Anja Lorch
- Universitätsspital Zürich, Zürich, Switzerland
- University Hospital Düsseldorf, Düsseldorf, Germany
| | | | | | | | | | - Cai Chen
- Merck & Co. Inc., Rahway, NJ, USA
| | | | | | | | - Seyda Gunduz
- Istinye University Liv Hospital, Istanbul, Turkey
| |
Collapse
|
17
|
Guo G, Zhang Z, Zhang J, Wang D, Xu S, Liu G, Gao Y, Mei J, Yan Z, Zhao R, Wang M, Li T, Bu X. Predicting recurrent glioblastoma clinical outcome to immune checkpoint inhibition and low-dose bevacizumab with tumor in situ fluid circulating tumor DNA analysis. Cancer Immunol Immunother 2024; 73:193. [PMID: 39105794 PMCID: PMC11303371 DOI: 10.1007/s00262-024-03774-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/05/2024] [Indexed: 08/07/2024]
Abstract
OBJECTIVE Most recurrent glioblastoma (rGBM) patients do not benefit from immune checkpoint inhibition, emphasizing the necessity for response biomarkers. This study evaluates whether tumor in situ fluid (TISF) circulating tumor DNA (ctDNA) could serve as a biomarker for response to low-dose bevacizumab (Bev) plus anti-PD-1 therapy in rGBM patients, aiming to enhance systemic responses to immunotherapy. METHODS In this phase II trial, 32 GBM patients with first recurrence after standard therapy were enrolled and then received tislelizumab plus low-dose Bev each cycle. TISF samples were analyzed for ctDNA using a 551-gene panel before each treatment. RESULTS The median progression-free survival (mPFS) and overall survival (mOS) were 8.2 months (95% CI, 5.2-11.1) and 14.3 months (95% CI, 6.5-22.1), respectively. The 12-month OS was 43.8%, and the objective response rate was 56.3%. Patients with more than 20% reduction in the mutant allele fraction and tumor mutational burden after treatment were significantly associated with better prognosis compared to baseline TISF-ctDNA. Among detectable gene mutations, patients with MUC16 mutation, EGFR mutation & amplification, SRSF2 amplification, and H3F3B amplification were significantly associated with worse prognosis. CONCLUSIONS Low-dose Bev plus anti-PD-1 therapy significantly improves OS in rGBM patients, offering guiding significance for future individualized treatment strategies. TISF-ctDNA can monitor rGBM patients' response to combination therapy and guide treatment. CLINICAL TRIAL REGISTRATION This trial is registered with ClinicalTrials.gov, NCT05540275.
Collapse
Affiliation(s)
- Guangzhong Guo
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Ziyue Zhang
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Jiubing Zhang
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Dayang Wang
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Sensen Xu
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Guanzheng Liu
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Yushuai Gao
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Jie Mei
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Zhaoyue Yan
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Ruijiao Zhao
- Department of Pathology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Meiyun Wang
- Department of Radiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Tianxiao Li
- Henan Provincial Neurointerventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, China
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Xingyao Bu
- Department of Neurosurgery, Juha International Center for Neurosurgery, Glioma Clinical Diagnosis and Treatment Center of Henan Province, Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
18
|
Schenker M, Burotto M, Richardet M, Ciuleanu TE, Gonçalves A, Steeghs N, Schoffski P, Ascierto PA, Maio M, Lugowska I, Lupinacci L, Leary A, Delord JP, Grasselli J, Tan DSP, Friedmann J, Vuky J, Tschaika M, Konduru S, Vemula SV, Slepetis R, Kollia G, Pacius M, Duong Q, Huang N, Doshi P, Baden J, Di Nicola M. Randomized, open-label, phase 2 study of nivolumab plus ipilimumab or nivolumab monotherapy in patients with advanced or metastatic solid tumors of high tumor mutational burden. J Immunother Cancer 2024; 12:e008872. [PMID: 39107131 PMCID: PMC11308901 DOI: 10.1136/jitc-2024-008872] [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] [Accepted: 06/14/2024] [Indexed: 08/09/2024] Open
Abstract
BACKGROUND Checkpoint inhibitor therapy has demonstrated overall survival benefit in multiple tumor types. Tumor mutational burden (TMB) is a predictive biomarker for response to immunotherapies. This study evaluated the efficacy of nivolumab+ipilimumab in multiple tumor types based on TMB status evaluated using either tumor tissue (tTMB) or circulating tumor DNA in the blood (bTMB). PATIENTS AND METHODS Patients with metastatic or unresectable solid tumors with high (≥10 mutations per megabase) tTMB (tTMB-H) and/or bTMB (bTMB-H) who were refractory to standard therapies were randomized 2:1 to receive nivolumab+ipilimumab or nivolumab monotherapy in an open-label, phase 2 study (CheckMate 848; NCT03668119). tTMB and bTMB were determined by the Foundation Medicine FoundationOne® CDx test and bTMB Clinical Trial Assay, respectively. The dual primary endpoints were objective response rate (ORR) in patients with tTMB-H and/or bTMB-H tumors treated with nivolumab+ipilimumab. RESULTS In total, 201 patients refractory to standard therapies were randomized: 135 had tTMB-H and 125 had bTMB-H; 82 patients had dual tTMB-H/bTMB-H. In patients with tTMB-H, ORR was 38.6% (95% CI 28.4% to 49.6%) with nivolumab+ipilimumab and 29.8% (95% CI 17.3% to 44.9%) with nivolumab monotherapy. In patients with bTMB-H, ORR was 22.5% (95% CI 13.9% to 33.2%) with nivolumab+ipilimumab and 15.6% (95% CI 6.5% to 29.5%) with nivolumab monotherapy. Early and durable responses to treatment with nivolumab+ipilimumab were seen in patients with tTMB-H or bTMB-H. The safety profile of nivolumab+ipilimumab was manageable, with no new safety signals. CONCLUSIONS Patients with metastatic or unresectable solid tumors with TMB-H, as determined by tissue biopsy or by blood sample when tissue biopsy is unavailable, who have no other treatment options, may benefit from nivolumab+ipilimumab. TRIAL REGISTRATION NUMBER NCT03668119.
Collapse
Affiliation(s)
- Michael Schenker
- Sf Nectarie Oncology Center and University of Medicine and Pharmacy, Craiova, Romania
| | | | - Martin Richardet
- Fundación Richardet Longo, Instituto Oncológico de Córdoba, Córdoba, Argentina
| | - Tudor-Eliade Ciuleanu
- Department of Oncology, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
- Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Patrick Schoffski
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Michele Maio
- Department of Oncology, University of Siena and Center for Immuno-Oncology, Siena, Italy
| | - Iwona Lugowska
- Department of Early Phase Clinical Trials, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Alexandra Leary
- Université Paris-Saclay and Institut Gustave‑Roussy, Villejuif, France
| | - Jean-Pierre Delord
- Department of Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Toulouse, France
| | - Julieta Grasselli
- Center for Medical Education and Clinical Research (CEMIC) University Hospital, Buenos Aires, Argentina
| | - David S P Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
- Cancer Science Institute, National University of Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore
| | - Jennifer Friedmann
- Segal Cancer Center, Jewish General Hospital, Montreal, Québec, Canada
- Rossy Cancer Network, McGill University, Montreal, Québec, Canada
| | - Jacqueline Vuky
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | | | | | | | | | | | | | - Quyen Duong
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Ning Huang
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Parul Doshi
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Massimo Di Nicola
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
19
|
Sahin IH, Yanes R, Saridogan T, Holder-Murray J, Dasari AN. The Role of Circulating Tumor DNA for Management of Patients With Rectal Cancer: Challenges and Opportunities. Cancer J 2024; 30:290-296. [PMID: 39042781 DOI: 10.1097/ppo.0000000000000731] [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: 07/25/2024]
Abstract
ABSTRACT Recently, organ preservation with total neoadjuvant therapy resulted in substantial progress in the management of locally advanced rectal cancer (LARC). The PROSPECT trial showed noninferiority of de-escalation of radiotherapy for patients with low-risk LARC who do not need abdominoperineal resection. Although these escalation and de-escalation approaches offer more personalized therapeutic approaches, the current state of care for patients with rectal cancer is far from individualized management. Circulating tumor DNA (ctDNA) is known to be one of the most powerful prognostic factors for early relapse and has been investigated in several interventional clinical trials to offer more precise treatment algorithms. In this review article, we discuss recent updates from studies examining the role of ctDNA for the prediction of treatment response and recurrence for patients with rectal cancer. We also elaborate on the future potential use of ctDNA in treatment escalation and de-escalation approaches for more personalized therapeutic interventions.
Collapse
Affiliation(s)
- Ibrahim Halil Sahin
- From the Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Rami Yanes
- From the Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | | | - Arvind N Dasari
- Department of Gastrointestinal Oncology, The University of Texas MD Anderson Cancer Center, Houston TX
| |
Collapse
|
20
|
Wang Y, Wang W, Zhang T, Yang Y, Wang J, Li C, Xu X, Wu Y, Jiang Y, Duan J, Wang L, Bi N. Dynamic bTMB combined with residual ctDNA improves survival prediction in locally advanced NSCLC patients with chemoradiotherapy and consolidation immunotherapy. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:177-187. [PMID: 39282582 PMCID: PMC11390629 DOI: 10.1016/j.jncc.2024.01.008] [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: 11/21/2023] [Revised: 01/21/2024] [Accepted: 01/30/2024] [Indexed: 09/19/2024] Open
Abstract
Background Liquid biopsy-based biomarkers, including circulating tumor DNA (ctDNA) and blood tumor mutational burden (bTMB), are recognized as promising predictors of prognoses and responses to immune checkpoint inhibitors (ICIs), despite insufficient sensitivity of single biomarker detection. This research aims to determine whether the combinatorial utility of longitudinal ctDNA with bTMB analysis could improve the prognostic and predictive effects. Methods This prospective two-center cohort trial, consisting of discovery and validation datasets, enrolled unresectable locally advanced non-small-cell lung cancer (LA-NSCLC) patients and assigned them to chemoradiotherapy (CRT) or CRT + consolidation ICI cohorts from 2018 to 2022. Blood specimens were collected pretreatment, 4 weeks post-CRT, and at progression to assess bTMB and ctDNA using 486-gene next-generation sequencing. Dynamic ∆bTMB was calculated as post-CRT bTMB minus baseline bTMB levels. Decision curve analyses were performed to calculate Concordance index (C-index). Results One hundred twenty-eight patients were enrolled. In the discovery dataset (n = 73), patients treated with CRT and consolidation ICI had significantly longer overall survival (OS; median not reached [NR] vs 20.2 months; P < 0.001) and progression-free survival (PFS; median 25.2 vs 11.4 months; P = 0.011) than those without ICI. Longitudinal analysis demonstrated a significant decrease in ctDNA abundance post-CRT (P < 0.001) but a relative increase with disease progression. Post-CRT detectable residual ctDNA correlated with significantly shorter OS (median 18.3 months vs NR; P = 0.001) and PFS (median 7.3 vs 25.2 months; P < 0.001). For patients with residual ctDNA, consolidation ICI brought significantly greater OS (median NR vs 14.8 months; P = 0.005) and PFS (median 13.8 vs 6.2 months; P = 0.028) benefit, but no significant difference for patients with ctDNA clearance. Dynamic ∆bTMB was predictive of prognosis. Patients with residual ctDNA and increased ∆bTMB (∆bTMB > 0) had significantly worse OS (median 9.0 vs 23.0 months vs NR; P < 0.001) and PFS (median 3.4 vs 7.3 vs 25.2 months; P < 0.001). The combinatorial model integrating post-CRT ctDNA with ∆bTMB had optimal predictive effects on OS (C-index = 0.723) and PFS (C-index = 0.693), outperforming individual features. In the independent validation set, we confirmed residual ctDNA predicted poorer PFS (median 50.8 vs 14.3 months; P = 0.026) but identified more consolidation ICI benefit (median NR vs 8.3 months; P = 0.039). The combined model exhibited a stable predictive advantage (C-index = 0.742 for PFS). Conclusions The multiparameter assay integrating qualitative residual ctDNA testing with quantitative ∆bTMB dynamics improves patient prognostic risk stratification and efficacy predictions, allowing for personalized consolidation therapy for LA-NSCLC.
Collapse
Affiliation(s)
- Yu Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenqing Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Zhang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Yang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianyang Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Canjun Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Xu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuqi Wu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Jiang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinghao Duan
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luhua Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- 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, China
| |
Collapse
|
21
|
Gao L, Medford A, Spring L, Bar Y, Hu B, Jimenez R, Isakoff SJ, Bardia A, Peppercorn J. Searching for the "Holy Grail" of breast cancer recurrence risk: a narrative review of the hunt for a better biomarker and the promise of circulating tumor DNA (ctDNA). Breast Cancer Res Treat 2024; 205:211-226. [PMID: 38355821 DOI: 10.1007/s10549-024-07253-6] [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: 08/14/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND This paper is a narrative review of a major clinical challenge at the heart of breast cancer care: determining which patients are at risk of recurrence, which require systemic therapy, and which remain at risk in the survivorship phase of care despite initial therapy. METHODS We review the literature on prognostic and predictive biomarkers in breast cancer with a focus on detection of minimal residual disease. RESULTS While we have many tools to estimate and refine risk that are used to individualize local and systemic therapy, we know that we continue to over treat many patients and undertreat others. Many patients also experience what is, at least in hindsight, needless fear of recurrence. In this review, we frame this dilemma for the practicing breast oncologist and discuss the search for what we term the "holy grail" of breast cancer evaluation: the ideal biomarker of residual distant disease. We review the history of attempts to address this problem and the up-to-date science on biomarkers, circulating tumor cells and circulating tumor DNA (ctDNA). CONCLUSION This review suggests that the emerging promise of ctDNA may help resolve a crticical dilemma at the heart of breast cancer care, and improve prognostication, treatment selection, and outcomes for patients with breast cancer.
Collapse
Affiliation(s)
- Lucy Gao
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Arielle Medford
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Laura Spring
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yael Bar
- Massachusetts General Hospital, Boston, MA, USA
| | - Bonnie Hu
- Massachusetts General Hospital, Boston, MA, USA
| | - Rachel Jimenez
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Steven J Isakoff
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aditya Bardia
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jeffrey Peppercorn
- Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Liu H, Chen J, Huang Y, Zhang Y, Ni Y, Xu N, Zhao F, Tang Y, Liu H, Sun G, Shen P, Liu Z, Huang J, Liao B, Zeng H. Prognostic significance of circulating tumor DNA in urothelial carcinoma: a systematic review and meta-analysis. Int J Surg 2024; 110:3923-3936. [PMID: 38573063 PMCID: PMC11175790 DOI: 10.1097/js9.0000000000001372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) has emerged as a noninvasive technique that provides valuable insights into molecular profiles and tumor disease management. This study aimed to evaluate the prognostic significance of circulating tumor DNA (ctDNA) in urothelial carcinoma (UC) through a systematic review and meta-analysis. METHODS A comprehensive search was conducted in MEDLINE, EMBASE, and the Cochrane Library from the inception to December 2023. Studies investigating the prognostic value of ctDNA in UC were included. Hazard ratios (HRs) of disease-free survival (DFS) and overall survival (OS) were extracted. Overall meta-analysis and subgroup exploration stratified by metastatic status, ctDNA sampling time, treatment type, and detection method was performed using the R software (version 4.2.2). RESULTS A total of 16 studies with 1725 patients were included. Fourteen studies assessed the association between baseline ctDNA status and patient outcomes. Patients with elevated ctDNA levels exhibited significantly worse DFS (HR=6.26; 95% CI: 3.71-10.58, P <0.001) and OS (HR=4.23; 95% CI: 2.72-6.57, P <0.001) regardless of metastatic status, ctDNA sampling time, treatment type, and detection methods. Six studies evaluated the prognostic value of ctDNA dynamics in UC. Patients who showed a decrease or clearance in ctDNA levels during treatment or observation demonstrated more favorable DFS (HR=0.26, 95% CI: 0.17-0.41, P <0.001) and OS (HR=0.21, 95% CI: 0.11-0.38, P <0.001) compared to those who did not. The association remained consistent across the subgroup analysis based on metastatic status and detection methods. In the immune checkpoint inhibitor-treated setting, both lower baseline ctDNA level and ctDNA decrease during the treatment were significantly associated with more favorable oncologic outcomes. Furthermore, specific gene mutations such as FGFR3 identified in ctDNA also demonstrated predictive value in UC patients. CONCLUSION This meta-analysis demonstrates a strong association of ctDNA status and its dynamic change with survival outcomes in UC, suggesting substantial clinical utility of ctDNA testing in prognosis prediction and decision making in this setting.
Collapse
Affiliation(s)
- Haoyang Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Yuchen Huang
- Department of Cardiothoracic Surgery, West China Hospital, Sichuan University
| | - Yaowen Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Yuchao Ni
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Nanwei Xu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Fengnian Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Yanfeng Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Haolin Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Jin Huang
- Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Banghua Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| |
Collapse
|
23
|
Wang L, Wen X, Yang Y, Hu Z, Jiang J, Duan L, Liao X, He Y, Liu Y, Wang J, Liang Z, Zhu X, Liu Q, Liu T, Luo D. CRISPR/Cas13a-based supersensitive circulating tumor DNA assay for detecting EGFR mutations in plasma. Commun Biol 2024; 7:657. [PMID: 38806596 PMCID: PMC11133305 DOI: 10.1038/s42003-024-06368-2] [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: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
Despite recent technological advancements in cell tumor DNA (ctDNA) mutation detection, challenges persist in identifying low-frequency mutations due to inadequate sensitivity and coverage of current procedures. Herein, we introduce a super-sensitivity and specificity technique for detecting ctDNA mutations, named HiCASE. The method utilizes PCR-based CRISPR, coupled with the restriction enzyme. In this work, HiCASE focuses on testing a series of EGFR mutations to provide enhanced detection technology for non-small cell lung cancer (NSCLC), enabling a detection sensitivity of 0.01% with 40 ng cell free DNA standard. When applied to a panel of 140 plasma samples from 120 NSCLC patients, HiCASE exhibits 88.1% clinical sensitivity and 100% specificity with 40 μL of plasma, higher than ddPCR and Super-ARMS assay. In addition, HiCASE can also clearly distinguish T790M/C797S mutations in different positions at a 1% variant allele frequency, offering valuable guidance for drug utilization. Indeed, the established HiCASE assay shows potential for clinical applications.
Collapse
Affiliation(s)
- Li Wang
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, PR China
| | - Xiaosha Wen
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, PR China
- Shenzhen University Medical School, Shenzhen, 518060, PR China
| | - Yang Yang
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, PR China
- Shenzhen University Medical School, Shenzhen, 518060, PR China
| | - Zheng Hu
- Translational Medicine Institute, the First People's Hospital of Chenzhou Affiliated to University of South China, Chenzhou, 423000, PR China
| | - Jing Jiang
- Translational Medicine Institute, the First People's Hospital of Chenzhou Affiliated to University of South China, Chenzhou, 423000, PR China
| | - Lili Duan
- Translational Medicine Institute, the First People's Hospital of Chenzhou Affiliated to University of South China, Chenzhou, 423000, PR China
| | - Xiaofen Liao
- Translational Medicine Institute, the First People's Hospital of Chenzhou Affiliated to University of South China, Chenzhou, 423000, PR China
| | - Yan He
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, PR China
- Shenzhen University Medical School, Shenzhen, 518060, PR China
| | - Yaru Liu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, PR China
- Shenzhen University Medical School, Shenzhen, 518060, PR China
| | - Jing Wang
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, PR China
- Shenzhen University Medical School, Shenzhen, 518060, PR China
| | - Zhikun Liang
- Research Institute, DAAN Gene Co., Ltd., Guangzhou, 510665, PR China
| | - Xiaoya Zhu
- Research Institute, DAAN Gene Co., Ltd., Guangzhou, 510665, PR China
| | - Quan Liu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, PR China.
- Shenzhen University Medical School, Shenzhen, 518060, PR China.
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, PR China.
| | - Dixian Luo
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, PR China.
- Shenzhen University Medical School, Shenzhen, 518060, PR China.
| |
Collapse
|
24
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [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: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
25
|
Morgensztern D. The expanding role of circulating tumor DNA in resectable non-small cell lung cancer. Cancer 2024; 130:1730-1732. [PMID: 38421941 DOI: 10.1002/cncr.35268] [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: 03/02/2024]
Abstract
Despite treatment with curative intent, many patients with non–small cell lung cancer develop tumor relapse. The detection of minimal residual disease with circulating tumor DNA may allow for a more personalized treatment, which may potentially allow patients with a high risk for relapse to receive more aggressive therapy and those with a low risk for relapse to avoid overtreatment.
Collapse
|
26
|
Luo X, Li J, Huang G, Xie F, He Z, Zeng X, Tian H, Liu Y, Fu W, Yang X. Metal-Graphene Hybrid Terahertz Metasurfaces for Circulating Tumor DNA Detection Based on Dual Signal Amplification. ACS Sens 2024; 9:2122-2133. [PMID: 38602840 DOI: 10.1021/acssensors.4c00168] [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: 04/13/2024]
Abstract
Terahertz (THz) spectroscopy has impressive capability for label-free biosensing, but its utility in clinical laboratories is rarely reported due to often unsatisfactory detection performances. Here, we fabricated metal-graphene hybrid THz metasurfaces (MSs) for the sensitive and enzyme-free detection of circulating tumor DNA (ctDNA) in pancreatic cancer plasma samples. The feasibility and mechanism of the enhanced effects of a graphene bridge across the MS and amplified by gold nanoparticles (AuNPs) were investigated experimentally and theoretically. The AuNPs serve to boost charge injection in the graphene film and result in producing a remarkable change in the graded transmissivity index to THz radiation of the MS resonators. Assay design utilizes this feature and a cascade hybridization chain reaction initiated on magnetic beads in the presence of target ctDNA to achieve dual signal amplification (chemical and optical). In addition to demonstrating subfemtomolar detection sensitivity and single-nucleotide mismatch selectivity, the proposed method showed remarkable capability to discriminate between pancreatic cancer patients and healthy individuals by recognizing and quantifying targeted ctDNAs. The introduction of graphene to the metasurface produces an improved sensitivity of 2 orders of magnitude for ctDNA detection. This is the first study to report the combined application of graphene and AuNPs in biosensing by THz spectroscopic resonators and provides a combined identification scheme to detect and discriminate different biological analytes, including nucleic acids, proteins, and various biomarkers.
Collapse
Affiliation(s)
- Xizi Luo
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jining Li
- Institute of Laser and Optoelectronics, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
| | - Guorong Huang
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Fengxin Xie
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhe He
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiaojun Zeng
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Huiyan Tian
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yu Liu
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Weiling Fu
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiang Yang
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
27
|
Eturi A, Bhasin A, Zarrabi KK, Tester WJ. Predictive and Prognostic Biomarkers and Tumor Antigens for Targeted Therapy in Urothelial Carcinoma. Molecules 2024; 29:1896. [PMID: 38675715 PMCID: PMC11054340 DOI: 10.3390/molecules29081896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/01/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Urothelial carcinoma (UC) is the fourth most prevalent cancer amongst males worldwide. While patients with non-muscle-invasive disease have a favorable prognosis, 25% of UC patients present with locally advanced disease which is associated with a 10-15% 5-year survival rate and poor overall prognosis. Muscle-invasive bladder cancer (MIBC) is associated with about 50% 5 year survival when treated by radical cystectomy or trimodality therapy; stage IV disease is associated with 10-15% 5 year survival. Current therapeutic modalities for MIBC include neoadjuvant chemotherapy, surgery and/or chemoradiation, although patients with relapsed or refractory disease have a poor prognosis. However, the rapid success of immuno-oncology in various hematologic and solid malignancies offers new targets with tremendous therapeutic potential in UC. Historically, there were no predictive biomarkers to guide the clinical management and treatment of UC, and biomarker development was an unmet need. However, recent and ongoing clinical trials have identified several promising tumor biomarkers that have the potential to serve as predictive or prognostic tools in UC. This review provides a comprehensive summary of emerging biomarkers and molecular tumor targets including programmed death ligand 1 (PD-L1), epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), fibroblast growth factor receptor (FGFR), DNA damage response and repair (DDR) mutations, poly (ADP-ribose) polymerase (PARP) expression and circulating tumor DNA (ctDNA), as well as their clinical utility in UC. We also evaluate recent advancements in precision oncology in UC, while illustrating limiting factors and challenges related to the clinical application of these biomarkers in clinical practice.
Collapse
Affiliation(s)
- Aditya Eturi
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (K.K.Z.); (W.J.T.)
| | - Amman Bhasin
- Department of Internal Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Kevin K. Zarrabi
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (K.K.Z.); (W.J.T.)
| | - William J. Tester
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (K.K.Z.); (W.J.T.)
| |
Collapse
|
28
|
Aquino IMC, Pascut D. Liquid biopsy: New opportunities for precision medicine in hepatocellular carcinoma care. Ann Hepatol 2024; 29:101176. [PMID: 37972709 DOI: 10.1016/j.aohep.2023.101176] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
Liquid biopsy, specifically the analysis of circulating tumor DNA (ctDNA), offers a non-invasive approach for hepatocellular carcinoma (HCC) diagnosis and management. However, its implementation in the clinical setting is difficult due to challenges such as low ctDNA yield and difficulty in understanding the mutation signals from background noise. This review highlights the crucial role of artificial intelligence (AI) in addressing these limitations and in improving discoveries in the field of liquid biopsy for HCC care. Combining AI with liquid biopsy data can offer a promising future for the discovery of novel biomarkers and an AI-powered clinical decision support system (CDSS) can turn liquid biopsy into an important tool for personalized management of HCC. Despite the current challenges, the integration of AI shows promise to significantly improve patient outcomes and revolutionize the field of oncology.
Collapse
Affiliation(s)
- Inah Marie C Aquino
- College of Medicine, University of the Philippines Manila, Ermita, Manila, Metro Manila 1000, Philippines; Liver Cancer Unit, Fondazione Italiana Fegato - ONLUS, Basovizza, Trieste 34149, Italy
| | - Devis Pascut
- Liver Cancer Unit, Fondazione Italiana Fegato - ONLUS, Basovizza, Trieste 34149, Italy.
| |
Collapse
|
29
|
Sun Y, He S, Peng Y, Liu M, Xu D. A novel label-free capillary electrophoresis LED-induced fluorescence platform based on catalytic hairpin assembly for sensitive detection of multiple circulating tumor DNA. Analyst 2024; 149:1548-1556. [PMID: 38284430 DOI: 10.1039/d3an01993d] [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: 01/30/2024]
Abstract
Circulating tumor DNA (ctDNA) is a highly promising biomarker for the early diagnosis and treatment of gastric cancer (GC). However, there is still a lack of effective and practical ctDNA detection methods. In this work, a simple and economical capillary non-gel sieving electrophoresis-LED induced fluorescence detection (NGCE-LEDIF) platform coupled with catalytic hairpin assembly (CHA) as the signal amplification strategy is proposed for quantitative detection of PIK3CA E542K and TP53 (two types of ctDNA associated with GC). We have reasonably designed two pairs of programmable oligonucleotide hairpin probes for PIK3CA E542K and TP53. Using a one-pot reaction, the presence of ctDNA triggers the cyclic amplification of CHA, forming numerous thermodynamically stable H1/H2 double-strands. The H1/H2 double-stranded DNA catalyzed by PIK3CA E542K and TP53 can be easily separated by NGCE due to their different lengths, enabling simultaneous detection of both ctDNAs. Under optimal experimental conditions, the detection limits of this strategy for detecting GC-related biomarkers PIK3CA E542K and TP53 are 20.35 pM and 19.61 pM, respectively, and can achieve 730-fold signal amplification. This strategy has a good recovery in the serum matrix. The results of this study show that this strategy has significant advantages such as high selectivity, a simple process, no special instruments and equipment, no need for fluorescence modification of hairpin probes in advance, high automation, low cost, and minimal sample consumption. This provides a powerful method for the detection of trace cancer biomarkers in the serum matrix with good application prospects.
Collapse
Affiliation(s)
- Yanyan Sun
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, No 163, Xianlin Avenue, Nanjing, 210023, PR China.
| | - Si He
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, No 163, Xianlin Avenue, Nanjing, 210023, PR China.
| | - Yufei Peng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, No 163, Xianlin Avenue, Nanjing, 210023, PR China.
| | - Min Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, No 163, Xianlin Avenue, Nanjing, 210023, PR China.
| | - Danke Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, No 163, Xianlin Avenue, Nanjing, 210023, PR China.
| |
Collapse
|
30
|
Walmsley CS, Jonsson P, Cheng ML, McBride S, Kaeser C, Vargas HA, Laudone V, Taylor BS, Kappagantula R, Baez P, Richards AL, Noronha AM, Perera D, Berger M, Solit DB, Iacobuzio-Donahue CA, Scher HI, Donoghue MTA, Abida W, Schram AM. Convergent evolution of BRCA2 reversion mutations under therapeutic pressure by PARP inhibition and platinum chemotherapy. NPJ Precis Oncol 2024; 8:34. [PMID: 38355834 PMCID: PMC10866935 DOI: 10.1038/s41698-024-00526-9] [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: 06/20/2022] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
Reversion mutations that restore wild-type function of the BRCA gene have been described as a key mechanism of resistance to Poly(ADP-ribose) polymerase (PARP) inhibitor therapy in BRCA-associated cancers. Here, we report a case of a patient with metastatic castration-resistant prostate cancer (mCRPC) with a germline BRCA2 mutation who developed acquired resistance to PARP inhibition. Extensive genomic interrogation of cell-free DNA (cfDNA) and tissue at baseline, post-progression, and postmortem revealed ten unique BRCA2 reversion mutations across ten sites. While several of the reversion mutations were private to a specific site, nine out of ten tumors contained at least one mutation, suggesting a powerful clonal selection for reversion mutations in the presence of therapeutic pressure by PARP inhibition. Variable cfDNA shed was seen across tumor sites, emphasizing a potential shortcoming of cfDNA monitoring for PARPi resistance. This report provides a genomic portrait of the temporal and spatial heterogeneity of prostate cancer under the selective pressure of a PARP inhibition and exposes limitations in the current strategies for detection of reversion mutations.
Collapse
Grants
- P30 CA008748 NCI NIH HHS
- Grant funding from ASCO Conquer Cancer Foundation CDA, NCI P30CA008748 CCITLA, Memorial Sloan Kettering Cancer Center Support Grant (P30 CA008748).
- WA has received honoraria from Roche, Medscape, Aptitude Health, Clinical Education Alliance, OncLive/MJH Life Sciences, touchIME, Pfizer, and the MedNet. WA has also received advisory board compensation from Clovis Oncology, ORIC pharmaceuticals, Daiichi Sankyo, AstraZeneca/MedImmune, Pfizer and Laekna Therapeutics, and research funding from AstraZeneca, Zenith Epigenetics, Clovis Oncology, ORIC Pharmaceuticals, Epizyme, Nuvation Bio, Merus, and Transthera.
Collapse
Affiliation(s)
- Charlotte S Walmsley
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Philip Jonsson
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Michael L Cheng
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Sean McBride
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | | | - Vincent Laudone
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | | | - Priscilla Baez
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | | | - Dilmi Perera
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Michael Berger
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - David B Solit
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | - Howard I Scher
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | - Wassim Abida
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Alison M Schram
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| |
Collapse
|
31
|
Lee KS, Lee CK, Kwon SS, Kwon WS, Park S, Lee ST, Choi JR, Rha SY, Shin S. Clinical relevance of clonal hematopoiesis and its interference in cell-free DNA profiling of patients with gastric cancer. Clin Chem Lab Med 2024; 62:178-186. [PMID: 37435889 DOI: 10.1515/cclm-2023-0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/02/2023] [Indexed: 07/13/2023]
Abstract
OBJECTIVES Clonal hematopoiesis (CH) is a condition in which healthy individuals have somatic mutations in hematopoietic stem cells. It has been reported with increased risk of hematologic malignancy and cardiovascular disease in the general population, but studies of Korean populations with comorbid disease entities are scarce. METHODS White blood cells (WBCs) from patients with gastric cancer (GC) (n=121) were analyzed using a DNA-based targeted (531 genes) panel with customized pipeline designed to detect single nucleotide variants and small indels with low-allele-frequency of ≥0.2 %. We defined significant CH variants as having variant allele frequency (VAF) ≥2 % among variants found in WBCs. Matched cell-free DNA (cfDNA) samples were also analyzed with the same pipeline to investigate the false-positive results caused by WBC variants in cfDNA profiling. RESULTS Significant CH variants were detected in 29.8 % of patients and were associated with age and male sex. The number of CH variants was associated with a history of anti-cancer therapy and age. DNMT3A and TET2 were recurrently mutated. Overall survival rate of treatment-naïve patients with stage IV GC was higher in those with CH, but Cox regression showed no significant association after adjustment for age, sex, anti-cancer therapy, and smoking history. In addition, we analyzed the potential interference of WBC variants in plasma cell-free DNA testing, which has attracted interest as a complementary method for tissue biopsy. Results showed that 37.0 % (47/127) of plasma specimens harbored at least one WBC variant. VAFs of interfering WBC variants in the plasma and WBC were correlated, and WBC variants with VAF ≥4 % in WBC were frequently detected in plasma with the same VAF. CONCLUSIONS This study revealed the clinical impact of CH in Korean patients and suggests the potential for its interference in cfDNA tests.
Collapse
Affiliation(s)
- Kwang Seob Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soon Sung Kwon
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woo Sun Kwon
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sejung Park
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Dxome, Seoul, Republic of Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Dxome, Seoul, Republic of Korea
| | - Sun Young Rha
- Dxome, Seoul, Republic of Korea
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
32
|
Ma W, Wu H, Chen Y, Xu H, Jiang J, Du B, Wan M, Ma X, Chen X, Lin L, Su X, Bao X, Shen Y, Xu N, Ruan J, Jiang H, Ding Y. New techniques to identify the tissue of origin for cancer of unknown primary in the era of precision medicine: progress and challenges. Brief Bioinform 2024; 25:bbae028. [PMID: 38343328 PMCID: PMC10859692 DOI: 10.1093/bib/bbae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 12/10/2023] [Accepted: 01/11/2024] [Indexed: 02/15/2024] Open
Abstract
Despite a standardized diagnostic examination, cancer of unknown primary (CUP) is a rare metastatic malignancy with an unidentified tissue of origin (TOO). Patients diagnosed with CUP are typically treated with empiric chemotherapy, although their prognosis is worse than those with metastatic cancer of a known origin. TOO identification of CUP has been employed in precision medicine, and subsequent site-specific therapy is clinically helpful. For example, molecular profiling, including genomic profiling, gene expression profiling, epigenetics and proteins, has facilitated TOO identification. Moreover, machine learning has improved identification accuracy, and non-invasive methods, such as liquid biopsy and image omics, are gaining momentum. However, the heterogeneity in prediction accuracy, sample requirements and technical fundamentals among the various techniques is noteworthy. Accordingly, we systematically reviewed the development and limitations of novel TOO identification methods, compared their pros and cons and assessed their potential clinical usefulness. Our study may help patients shift from empirical to customized care and improve their prognoses.
Collapse
Affiliation(s)
- Wenyuan Ma
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Wu
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiran Chen
- Department of Surgical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongxia Xu
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Junjie Jiang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bang Du
- Real Doctor AI Research Centre, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Mingyu Wan
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolu Ma
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Chen
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Lin
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinhui Su
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuanwen Bao
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifei Shen
- Department of Laboratory Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiping Jiang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongfeng Ding
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
33
|
Nicolò E, Tarantino P, D’Ecclesiis O, Antonarelli G, Boscolo Bielo L, Marra A, Gandini S, Crimini E, Giugliano F, Zagami P, Corti C, Trapani D, Morganti S, Criscitiello C, Locatelli M, Belli C, Esposito A, Minchella I, Cristofanilli M, Tolaney SM, Curigliano G. Baseline Tumor Size as Prognostic Index in Patients With Advanced Solid Tumors Receiving Experimental Targeted Agents. Oncologist 2024; 29:75-83. [PMID: 37548439 PMCID: PMC10769799 DOI: 10.1093/oncolo/oyad212] [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: 03/05/2023] [Accepted: 06/30/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Baseline tumor size (BTS) has been associated with outcomes in patients with cancer treated with immunotherapy. However, the prognostic impact of BTS on patients receiving targeted therapies (TTs) remains undetermined. METHODS We reviewed data of patients with advanced solid tumors consecutively treated within early-phase clinical trials at our institution from 01/2014 to 04/2021. Treatments were categorized as immunotherapy-based or TT-based (biomarker-matched or not). BTS was calculated as the sum of RECIST1.1 baseline target lesions. RESULTS A total of 444 patients were eligible; the median BTS was 69 mm (IQR 40-100). OS was significantly longer for patients with BTS lower versus higher than the median (16.6 vs. 8.2 months, P < .001), including among those receiving immunotherapy (12 vs. 7.5 months, P = .005). Among patients receiving TT, lower BTS was associated with longer PFS (4.7 vs. 3.1 months, P = .002) and OS (20.5 vs. 9.9 months, P < .001) as compared to high BTS. However, such association was only significant among patients receiving biomarker-matched TT, with longer PFS (6.2 vs. 3.3 months, P < .001) and OS (21.2 vs. 6.7 months, P < .001) in the low-BTS subgroup, despite a similar ORR (28% vs. 22%, P = .57). BTS was not prognostic among patients receiving unmatched TT, with similar PFS (3.7 vs. 4.4 months, P = .30), OS (19.3 vs. 11.8 months, P = .20), and ORR (33% vs. 28%, P = .78) in the 2 BTS groups. Multivariate analysis confirmed that BTS was independently associated with PFS (P = .03) and OS (P < .001) but not with ORR (P = .11). CONCLUSIONS Higher BTS is associated with worse survival outcomes among patients receiving biomarker-matched, but not biomarker-unmatched TT.
Collapse
Affiliation(s)
- Eleonora Nicolò
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Paolo Tarantino
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Oriana D’Ecclesiis
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Gabriele Antonarelli
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Antonio Marra
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
| | - Sara Gandini
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Edoardo Crimini
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Federica Giugliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Paola Zagami
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Chiara Corti
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
| | - Stefania Morganti
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Marzia Locatelli
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
| | - Carmen Belli
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
| | - Angela Esposito
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
| | - Ida Minchella
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
| | - Massimo Cristofanilli
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Sara M Tolaney
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
34
|
Fang J, Yuan C, Luo X, He Z, Fu W. A Thermus thermophilus argonaute-coupling exponential amplification assay for ultrarapid analysis of circulating tumor DNA. Talanta 2024; 266:125034. [PMID: 37597338 DOI: 10.1016/j.talanta.2023.125034] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/21/2023]
Abstract
Circulating tumor DNA (ctDNA) is a noninvasive biomarker for liquid biopsy with important clinical and biological information, but existing detection techniques are expensive, complex and quite time-consuming. Here, we report an ultrarapid, sensitive and simple method, which we term Thermus thermophilus argonaute-coupling exponential amplification reaction (TtAgo-CEAR), that selectively amplifies mutated ctDNA. Aiming at seven Kirsten rat sarcoma-2 virus (KRAS) point mutations, the present strategy allows for easy detection with attomolar sensitivity and single-nucleotide specificity within as little as 16 min without prior PCR amplification. We also demonstrate that TtAgo-coupling assay is easily adaptable to Terahertz spectroscopy-based and lateral-flow-based readout. We show that the detected ctDNA concentrations by mouse models can respond to the variations of disease burden in serum samples. It is envisioned that this TtAgo-CEAR approach has great potential for rapid diagnosis and monitoring of diverse malignant tumors.
Collapse
Affiliation(s)
- Jie Fang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Changjing Yuan
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xizi Luo
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhe He
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Weiling Fu
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
35
|
Gao Y, Zhou N, Liu J. Ovarian Cancer Diagnosis and Prognosis Based on Cell-Free DNA Methylation. Cancer Control 2024; 31:10732748241255548. [PMID: 38764160 PMCID: PMC11104031 DOI: 10.1177/10732748241255548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/12/2024] [Accepted: 04/30/2024] [Indexed: 05/21/2024] Open
Abstract
Background: Ovarian cancer stands as the deadliest malignant tumor within the female reproductive tract. As a result of the absence of effective diagnostic and monitoring markers, 75% of ovarian cancer cases are diagnosed at a late stage, leading to a mere 50% survival rate within five years. The advancement of molecular biology is essential for accurate diagnosis and treatment of ovarian cancer. Methods: A review of several randomized clinical trials, focusing on the ovarian cancer, was undertaken. The advancement of molecular biology and diagnostic methods related to accurate diagnosis and treatment of ovarian cancer were examined. Results: Liquid biopsy is an innovative method of detecting malignant tumors that has gained increasing attention over the past few years. Cell-free DNA assay-based liquid biopsies show potential in delineating tumor status heterogeneity and tracking tumor recurrence. DNA methylation influences a multitude of biological functions and diseases, especially during the initial phases of cancer. The cell-free DNA methylation profiling system has emerged as a sensitive and non-invasive technique for identifying and detecting the biological origins of cancer. It holds promise as a biomarker, enabling early screening, recurrence monitoring, and prognostic evaluation of cancer. Conclusions: This review evaluates recent advancements and challenges associated with cell-free DNA methylation analysis for the diagnosis, prognosis monitoring, and assessment of therapeutic responses in the management of ovarian cancers, aiming to offer guidance for precise diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Yajuan Gao
- Department of Gynecology and Obstetrics, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Nanyang Zhou
- Department of Traditional Chinese Medicine, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| | - Jie Liu
- Department of Gynecology and Obstetrics, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, China
| |
Collapse
|
36
|
Levkova M, Chervenkov T, Angelova L, Dzenkov D. Oxford Nanopore Technology and its Application in Liquid Biopsies. Curr Genomics 2023; 24:337-344. [PMID: 38327653 PMCID: PMC10845067 DOI: 10.2174/0113892029286632231127055733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 02/09/2024] Open
Abstract
Advanced medical technologies are transforming the future of healthcare, in particular, the screening and detection of molecular-genetic changes in patients suspected of having a neoplasm. They are based on the assumption that neoplasms release small amounts of various neoplasm-specific molecules, such as tumor DNA, called circulating DNA (cirDNA), into the extracellular space and subsequently into the blood. The detection of tumor-specific molecules and specific molecular changes in body fluids in a noninvasive or minimally invasive approach is known as "liquid biopsy." The aim of this review is to summarize the current knowledge of the application of ONT for analyzing circulating DNA in the field of liquid biopsies among cancer patients. Databases were searched using the keywords "nanopore" and "liquid biopsy" and by applying strict inclusion criteria. This technique can be used for the detection of neoplastic disease, including metastases, guiding precision therapy, and monitoring its effects. There are many challenges, however, for the successful implementation of this technology into the clinical practice. The first one is the low amount of tumor-specific molecules in the body fluids. Secondly, a tumor molecular signature should be discriminated from benign conditions like clonal hematopoiesis of unknown significance. Oxford Nanopore Technology (ONT) is a third-generation sequencing technology that seems particularly promising to complete these tasks. It offers rapid sequencing thanks to its ability to detect changes in the density of the electric current passing through nanopores. Even though ONT still needs validation technology, it is a promising approach for early diagnosis, therapy guidance, and monitoring of different neoplasms based on analyzing the cirDNA.
Collapse
Affiliation(s)
- Mariya Levkova
- Department of Medical Genetics, Medical University Varna, Marin Drinov Str 55, Varna, 9000, Bulgaria
- Laboratory of Medical Genetics, St. Marina Hospital, Hristo Smirnenski Blv 1, Varna, 9000, Bulgaria
| | - Trifon Chervenkov
- Department of Medical Genetics, Medical University Varna, Marin Drinov Str 55, Varna, 9000, Bulgaria
- Laboratory of Clinical immunology, St. Marina Hospital, Hristo Smirnenski Blv 1, Varna, 9000, Bulgaria
| | - Lyudmila Angelova
- Department of Medical Genetics, Medical University Varna, Marin Drinov Str 55, Varna, 9000, Bulgaria
| | - Deyan Dzenkov
- Department of General and Clinical Pathology, Forensic Medicine and Deontology, Division of General and Clinical Pathology, Medical University Varna, Marin Drinov Str 55, Varna, 9000, Bulgaria
| |
Collapse
|
37
|
Prasanth BK, Alkhowaiter S, Sawarkar G, Dharshini BD, R Baskaran A. Unlocking Early Cancer Detection: Exploring Biomarkers, Circulating DNA, and Innovative Technological Approaches. Cureus 2023; 15:e51090. [PMID: 38274938 PMCID: PMC10808885 DOI: 10.7759/cureus.51090] [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] [Accepted: 12/25/2023] [Indexed: 01/27/2024] Open
Abstract
Research and development improvements in early cancer diagnosis have had a significant positive impact on health. In the treatment and prevention of cancer, early detection is essential. In this context, biomarkers are essential because they offer important information on the state of cells at any particular time. Cells go through unique changes when they shift from a healthy condition to a malignant state, changes that appropriate biomarkers may pick up. Recent advancements have been made to identify and characterize circulating cancer-specific mutations in cell-free circulating DNA derived from tumors and tumor cells. A patient's delay between the time they first detect symptoms and the time they contact a doctor has been noted for many cancer forms. The tumor's location and features significantly impact the presentation of symptoms judged appropriate for early diagnosis. Lack of knowledge of the severity of the symptoms may be one cause for this delay. Our review is largely focused on the ongoing developments of early diagnosis in the study of biomarkers, circulating DNA for diagnosis, the biology of early challenges, early symptoms, liquid biopsies, detectable by imaging, established tumor markers, plasma DNA technologies, gender differences, and artificial intelligence (AI) in diagnosis. This review aims to determine and evaluate Indicators for detecting early cancer, assessing medical conditions, and evaluating potential risks. For Individuals with a heightened likelihood of developing cancer or who have already been diagnosed, early identification is crucial for enhancing prognosis and raising the likelihood of effective treatment.
Collapse
Affiliation(s)
- B Krishna Prasanth
- Department of Community Medicine, Sree Balaji Medical College and Hospital, Bharath Institute of Higher Education and Research, Chennai, IND
| | - Saad Alkhowaiter
- Department of Gastroenterology, College of Medicine, King Khalid University Hospital, Riyadh, SAU
| | - Gaurav Sawarkar
- Rachana Sharir, Mahatma Gandhi Ayurveda College, Hospital and Research Centre, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - B Divya Dharshini
- Department of Biochemistry, Government Medical College, Khammam, Telangana, IND
| | - Ajay R Baskaran
- Department of Psychiatry, National Health Service, Shrewsbury, GBR
| |
Collapse
|
38
|
Li C, Shao J, Li P, Feng J, Li J, Wang C. Circulating tumor DNA as liquid biopsy in lung cancer: Biological characteristics and clinical integration. Cancer Lett 2023; 577:216365. [PMID: 37634743 DOI: 10.1016/j.canlet.2023.216365] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Lung cancer maintains high morbidity and mortality rate globally despite significant advancements in diagnosis and treatment in the era of precision medicine. Pathological analysis of tumor tissue, the current gold standard for lung cancer diagnosis, is intrusive and intrinsically confined to evaluating the limited amount of tissues that could be physically extracted. However, tissue biopsy has several limitations, including the invasiveness of the procedure and difficulty in obtaining samples for patients at advanced stages., there Additionally,has been no major breakthrough in tumor biomarkers with high specificity and sensitivity, particularly for early-stage lung cancer. Liquid biopsy has been considered a feasible auxiliary tool for tearly dianosis, evaluating treatment responses and monitoring prognosis of lung cancer. Circulating tumor DNA (ctDNA), an ideal biomarker of liquid biopsy, has emerged as one of the most reliable tools for monitoring tumor processes at molecular levels. Herein, this review focuses on tumor heterogeneity to elucidate the superiority of liquid biopsy and retrospectively discussdeciphersolution. We systematically elaborate ctDNA biological characteristics, introduce methods for ctDNA detection, and discuss the current role of plasma ctDNA in lung cancer management. Finally, we summarize the drawbacks of ctDNA analysis and highlight its potential clinical application in lung cancer.
Collapse
Affiliation(s)
- Changshu Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Shao
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Peiyi Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaming Feng
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
39
|
Jiang Y, Wang C, Zhou S. Artificial intelligence-based risk stratification, accurate diagnosis and treatment prediction in gynecologic oncology. Semin Cancer Biol 2023; 96:82-99. [PMID: 37783319 DOI: 10.1016/j.semcancer.2023.09.005] [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: 12/17/2022] [Revised: 08/27/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
As data-driven science, artificial intelligence (AI) has paved a promising path toward an evolving health system teeming with thrilling opportunities for precision oncology. Notwithstanding the tremendous success of oncological AI in such fields as lung carcinoma, breast tumor and brain malignancy, less attention has been devoted to investigating the influence of AI on gynecologic oncology. Hereby, this review sheds light on the ever-increasing contribution of state-of-the-art AI techniques to the refined risk stratification and whole-course management of patients with gynecologic tumors, in particular, cervical, ovarian and endometrial cancer, centering on information and features extracted from clinical data (electronic health records), cancer imaging including radiological imaging, colposcopic images, cytological and histopathological digital images, and molecular profiling (genomics, transcriptomics, metabolomics and so forth). However, there are still noteworthy challenges beyond performance validation. Thus, this work further describes the limitations and challenges faced in the real-word implementation of AI models, as well as potential solutions to address these issues.
Collapse
Affiliation(s)
- Yuting Jiang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China; Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengdi Wang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China; Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China; Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
40
|
Diaz PM, Leehans A, Ravishankar P, Daily A. Multiomic Approaches for Cancer Biomarker Discovery in Liquid Biopsies: Advances and Challenges. Biomark Insights 2023; 18:11772719231204508. [PMID: 37846373 PMCID: PMC10576933 DOI: 10.1177/11772719231204508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023] Open
Abstract
Cancer is a complex and heterogeneous disease that poses a significant threat to global health. Early diagnosis and treatment are critical for improving patient outcomes, and the use of liquid biopsies has emerged as a promising approach for cancer detection and monitoring. Traditionally, cancer diagnosis has relied on invasive tissue biopsies, the collection of which can prove challenging for patients and the results of which may not always provide accurate results due to tumor heterogeneity. Liquid biopsies have gained increasing attention as they provide a non-invasive and accessible source of cancer biomarkers, which can be used to diagnose cancer, monitor treatment response, and detect relapse. The integration of -omics technologies, such as proteomics, genomics, and metabolomics, has further enhanced the capabilities of liquid biopsies by introducing precision oncology and enabling the tailoring of treatment for individual patients based on their unique tumor biology. In this review, we will discuss the challenges and advances in the field of cancer liquid biopsies and the integration of -omics technologies for different types of liquid biopsies, including blood, tear, urine, sweat, saliva, and cerebrospinal fluid.
Collapse
Affiliation(s)
- Paola Monterroso Diaz
- Namida Lab Inc., Fayetteville, AR, USA
- University of Arkansas, Department of Biomedical Engineering, Fayetteville, AR, USA
| | | | | | | |
Collapse
|
41
|
Zhang C, Li Z, Liu J, Liu C, Zhang H, Lee WG, Yao C, Guo H, Xu F. Synthetic Gene Circuit-Based Assay with Multilevel Switch Enables Background-Free and Absolute Quantification of Circulating Tumor DNA. RESEARCH (WASHINGTON, D.C.) 2023; 6:0217. [PMID: 37789988 PMCID: PMC10543738 DOI: 10.34133/research.0217] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/02/2023] [Indexed: 10/05/2023]
Abstract
Circulating tumor DNA (ctDNA) detection has found widespread applications in tumor diagnostics and treatment, where the key is to obtain accurate quantification of ctDNA. However, this remains challenging due to the issue of background noise associated with existing assays. In this work, we developed a synthetic gene circuit-based assay with multilevel switch (termed CATCH) for background-free and absolute quantification of ctDNA. The multilevel switch combining a small transcription activating RNA and a toehold switch was designed to simultaneously regulate transcription and translation processes in gene circuit to eliminate background noise. Moreover, such a multilevel switch-based gene circuit was integrated with a Cas9 nickase H840A (Cas9n) recognizer and a molecular beacon reporter to form CATCH for ctDNA detection. The CATCH can be implemented in one-pot reaction at 35 °C with virtually no background noise, and achieve robust absolute quantification of ctDNA when integrated with a digital chip (i.e., digital CATCH). Finally, we validated the clinical capability of CATCH by detecting drug-resistant ctDNA mutations from the plasma of 76 non-small cell lung cancer (NSCLC) patients, showing satisfying clinical sensitivity and specificity. We envision that the simple and robust CATCH would be a powerful tool for next-generation ctDNA detection.
Collapse
Affiliation(s)
- Chao Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi’an 710049, P.R. China
| | - Zedong Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi’an 710049, P.R. China
- TFX Group-Xi'an Jiaotong University Institute of Life Health, Xi'an 710049, P.R. China
| | - Jie Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi’an 710049, P.R. China
| | - Chang Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi’an 710049, P.R. China
| | - Haoqing Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi’an 710049, P.R. China
| | - Won Gu Lee
- Department of Mechanical Engineering,
Kyung Hee University, Yongin 17104, Republic of Korea
| | - Chunyan Yao
- Department of Transfusion Medicine, Southwest Hospital,
Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi’an 710049, P.R. China
| |
Collapse
|
42
|
Blanco-García L, Ruano Y, Blanco Martínez-Illescas R, Cubo R, Jiménez Sánchez P, Sánchez-Arévalo Lobo VJ, Riveiro Falkenbach E, Ortiz Romero P, Garrido MC, Rodríguez Peralto JL. pTERT C250T mutation: A potential biomarker of poor prognosis in metastatic melanoma. Heliyon 2023; 9:e18953. [PMID: 37609429 PMCID: PMC10440525 DOI: 10.1016/j.heliyon.2023.e18953] [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: 12/29/2022] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
Melanoma is the most aggressive form of skin cancer and the leading cause of death from cutaneous tumors. Several studies have associated alterations in the TERT promoter region (pTERT) with gene overexpression, aggressiveness and poor prognosis of the disease. The aim of this study was to clarify the role of pTERT molecular status in paired samples of primary melanoma and metastasis using tissue and plasma to establish a correlation with disease progression and survival. A total of 88 FFPE tissue samples from 53 patients with advanced melanoma were analyzed. Of these, 35 had paired samples. We also examined cfDNA samples from plasma of 25 patients. We detected a good correlation between primary tumors and metastases in pTERT mutation and methylation status. We were also able to identify pTERT mutations in plasma samples that correlated with mutational status in tissue samples. Interestingly, the C250T mutation was associated with worse survival and higher TERT mRNA expression, compared to the other most common mutation: C228T. In addition, hyper-methylation of the promoter region seems to be related to the progression of pTERT wild type (WT) patients. These results suggest that TERT gene alterations plays an important role during tumor progression, with the detection of the C250T mutation in tissue and plasma as a potential biomarker of poor prognosis in patients with advanced melanoma.
Collapse
Affiliation(s)
| | - Yolanda Ruano
- Research Institute 12 de Octubre Hospital, Madrid, Spain
| | - Raquel Blanco Martínez-Illescas
- Research Institute 12 de Octubre Hospital, Madrid, Spain
- Biosanitary Research Institute, Faculty of Experimental Sciences, Francisco de Vitoria University, Pozuelo de Alarcón, Madrid, Spain
| | - Rocío Cubo
- Research Institute 12 de Octubre Hospital, Madrid, Spain
| | - Paula Jiménez Sánchez
- Research Institute 12 de Octubre Hospital, Madrid, Spain
- Biosanitary Research Institute, Faculty of Experimental Sciences, Francisco de Vitoria University, Pozuelo de Alarcón, Madrid, Spain
| | - Víctor J. Sánchez-Arévalo Lobo
- Research Institute 12 de Octubre Hospital, Madrid, Spain
- Biosanitary Research Institute, Faculty of Experimental Sciences, Francisco de Vitoria University, Pozuelo de Alarcón, Madrid, Spain
| | | | - Pablo Ortiz Romero
- Department of Dermatology, 12 de Octubre University Hospital, Madrid, Spain
| | - María C. Garrido
- Department of Pathology, 12 de Octubre University Hospital, Madrid, Spain
- Complutense University of Madrid; Madrid, Spain
| | - José L. Rodríguez Peralto
- Department of Pathology, 12 de Octubre University Hospital, Madrid, Spain
- Complutense University of Madrid; Madrid, Spain
| |
Collapse
|
43
|
Wang XY, Zhang R, Han JH, Chen SQ, Zhao FL, Chen H, Lin J, Fan J, Zhu WW, Lu L, Chen JH. Early Circulating Tumor DNA Dynamics Predict Neoadjuvant Therapy Response and Recurrence in Colorectal Liver Metastases: A Prospective Study. Ann Surg Oncol 2023; 30:5252-5263. [PMID: 37202570 DOI: 10.1245/s10434-023-13604-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/20/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND For patients with colorectal liver metastases (CRLM) who receive neoadjuvant therapy (NAT), reliable indicators that can early and accurately predict treatment response are lacking. This study was conducted to prospectively investigate the potential of early circulating tumor DNA (ctDNA) dynamics as a precise predictor of NAT response and recurrence in CRLM. METHODS This study prospectively enrolled 34 patients with CRLM who received NAT, with blood samples collected and subjected to deep targeted panel sequencing at two time points: 1 day before the first and the second cycles of NAT. Correlations of ctDNA mean variant allele frequency (mVAF) dynamics and treatment response were assessed. The performance of early ctDNA dynamics in predicting treatment response was assessed and compared with those of carcinoembryonic antigen (CEA) and cancer antigen 19-9 (CA19-9). RESULTS The baseline ctDNA mVAF was significantly associated with pre-NAT tumor diameter (r = 0.65; P < 0.0001). After one cycle of NAT, the ctDNA mVAF declined remarkably (P < 0.0001). The dynamic change in ctDNA mVAF of 50% or more was significantly correlated with better NAT responses. The discriminatory capacity of ctDNA mVAF changes was superior to that of CEA or CA19-9 in predicting radiologic response (area under the curve [AUC], 0.90 vs 0.71 vs 0.61) and pathologic tumor regression grade (AUC, 0.83 vs 0.64 vs 0.67). The early changes in ctDNA mVAF but not CEA or CA19-9 were an independent indicator of recurrence-free survival (RFS) (hazard ratio, 4.0; P = 0.023). CONCLUSIONS For CRLM patients receiving NAT, an early ctDNA change is a superior predictor of treatment response and recurrence compared with conventional tumor markers.
Collapse
Affiliation(s)
- Xiang-Yu Wang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Rui Zhang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia-Hao Han
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Shi-Qing Chen
- Department of Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Fei-Long Zhao
- Department of Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Hui Chen
- Department of Medical Affairs, 3D Medicines Inc, Shanghai, China
| | - Jing Lin
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Fan
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wen-Wei Zhu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Lu Lu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jin-Hong Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
44
|
Xia T, Fang C, Chen Y. Advances in application of circulating tumor DNA in ovarian cancer. Funct Integr Genomics 2023; 23:250. [PMID: 37479960 DOI: 10.1007/s10142-023-01181-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/23/2023]
Abstract
Ovarian cancer is the third most common gynecologic cancer worldwide and has the highest mortality rate among gynecologic cancers. Identifying timely and effective biomarkers at different stages of the disease is the key to improve the prognosis of ovarian cancer patients. Circulating tumor DNA (ctDNA) is a fragment of free DNA produced by tumor cells in the blood. Current techniques for detecting ctDNA mainly include quantitative polymerase chain reaction (PCR), targeted next-generation sequencing (NGS), and non-targeted NGS (such as whole exon or whole genome sequencing). As a non-invasive liquid biopsy technique, ctDNA has a good application prospect in the ovarian cancer diagnosis, monitoring of treatment response and efficacy evaluation, detection of reverse mutation and related medication guidance, and prognosis evaluation. This article reviews the advances in application of ctDNA in ovarian cancer.
Collapse
Affiliation(s)
- Ting Xia
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Chenyan Fang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yaqing Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
45
|
Sato Y, Okamoto K, Kawano Y, Kasai A, Kawaguchi T, Sagawa T, Sogabe M, Miyamoto H, Takayama T. Novel Biomarkers of Gastric Cancer: Current Research and Future Perspectives. J Clin Med 2023; 12:4646. [PMID: 37510761 PMCID: PMC10380533 DOI: 10.3390/jcm12144646] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Gastric cancer is a heterogeneous disease with diverse histological and genomic subtypes, making it difficult to demonstrate treatment efficacy in clinical trials. However, recent efforts have been made to identify molecular biomarkers with prognostic and predictive implications to better understand the broad heterogeneity of gastric cancer and develop effective targeted therapies for it. HER2 overexpression, HER2/neu amplification, MSI-H, and PD-L1+ are predictive biomarkers in gastric cancer, and a growing number of clinical trials based on novel biomarkers have demonstrated the efficacy of targeted therapies alone or in combination with conventional chemotherapy. Enrichment design clinical trials of targeted therapies against FGFR2b and claudin 18.2 have demonstrated efficacy in unresectable advanced gastric cancer. Nonetheless, it is essential to continuously validate promising molecular biomarkers and introduce them into clinical practice to optimize treatment selection and improve patient outcomes. In this review, we focused on established (PD-L1, HER2, MSI) and emerging biomarkers (FGFR2, CLDN18.2) in gastric cancer, their clinical significance, detection methods, limitations, and molecular agents that target these biomarkers.
Collapse
Affiliation(s)
- Yasushi Sato
- Department of Community Medicine for Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, Tokushima 770-8503, Japan
| | - Koichi Okamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, Tokushima 770-8503, Japan
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, Tokushima 770-8503, Japan
| | - Akinari Kasai
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, Tokushima 770-8503, Japan
| | - Tomoyuki Kawaguchi
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, Tokushima 770-8503, Japan
| | - Tamotsu Sagawa
- Department of Gastroenterology, Hokkaido Cancer Center, Sapporo 060-0042, Japan
| | - Masahiro Sogabe
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, Tokushima 770-8503, Japan
| | - Hiroshi Miyamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, Tokushima 770-8503, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Medical Science, Tokushima 770-8503, Japan
| |
Collapse
|
46
|
Verner EL, Jackson JB, Severson E, Valkenburg KC, Greer AE, Riley DR, Sausen M, Maddox C, McGregor PM, Karandikar A, Hastings SB, Previs RA, Reddy VP, Jensen TJ, Ramkissoon SH. Validation of the Labcorp Plasma Focus Test to Facilitate Precision Oncology Through Cell-Free DNA Genomic Profiling of Solid Tumors. J Mol Diagn 2023; 25:477-489. [PMID: 37068734 DOI: 10.1016/j.jmoldx.2023.03.008] [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: 12/24/2022] [Revised: 02/28/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023] Open
Abstract
Genomic profiling is critical for precision oncology to guide treatment decisions. Liquid biopsy testing is a complementary approach to tissue testing, particularly when tissue is not readily available. The Labcorp Plasma Focus test is a circulating cell-free DNA genomic profiling test that identifies actionable variants in solid cancers, including non-small-cell lung, colorectal, melanoma, breast, esophageal, gastroesophageal junction, and gastric cancers. This study highlights the analytical validation of the test, including accuracy compared with orthogonal methods, as well as sensitivity, specificity, precision, reproducibility, and repeatability. Concordance with orthogonal methods showed percent positive agreement of 98.7%, 89.3%, and 96.2% for single nucleotide variants (SNVs), insertion/deletions (indels), and copy number amplifications (CNAs), respectively, and 100.0% for translocations and microsatellite instability (MSI). Analytical sensitivity revealed a median limit of detection of 0.7% and 0.6% for SNVs and indels, 1.4-fold for CNAs, 0.5% variant allele frequency for translocations, and 0.6% for MSI. Specificity was >99% for SNVs/indels and 100% for CNAs, translocations, and MSI. Average positive agreement from precision, reproducibility, and repeatability experiments was 97.5% and 88.9% for SNVs/indels and CNAs, and 100% for translocations and MSI. Taken together, these data show that the Labcorp Plasma Focus test is a highly accurate, sensitive, and specific approach for cell-free DNA genomic profiling to supplement tissue testing and inform treatment decisions.
Collapse
Affiliation(s)
- Ellen L Verner
- Personal Genome Diagnostics (PGDx), Baltimore, Maryland.
| | | | - Eric Severson
- Enterprise Oncology, Labcorp, Durham, North Carolina
| | | | - Amy E Greer
- Personal Genome Diagnostics (PGDx), Baltimore, Maryland
| | - David R Riley
- Personal Genome Diagnostics (PGDx), Baltimore, Maryland
| | - Mark Sausen
- Personal Genome Diagnostics (PGDx), Baltimore, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Raei N, Safaralizadeh R, Latifi-Navid S. Clinical application of circulating tumor DNA in metastatic cancers. Expert Rev Mol Diagn 2023; 23:1209-1220. [PMID: 37797209 DOI: 10.1080/14737159.2023.2268008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Advances in genomics have facilitated the application of cell-free DNA (cfDNA) and circulating tumor DNA (ctDNA) in phase II and phase III clinical trials. The various mutations of cfDNA/ctDNA have been correlated with clinical features. Advances in next-generation sequencing (NGS) and digital droplet PCR have paved the way for identifying cfDNA/ctDNA mutations. AREAS COVERED Herein, the biology of ctDNA and its function in clinical application in metastasis, which may lead to improved clinical management of metastatic cancer patients, are comprehensively reviewed. EXPERT OPINION Metastatic cancer ctDNA shows the greatest frequency of mutations in TP53, HER-2, KRAS, and EGFR genes (alteration frequency of > 50%). Therefore, identifying key mutations frequently present in metastatic cancers can help identify patients with pre-malignant tumors before cancer progression. Studying ctDNA can help determine the prognosis and select appropriate treatments for affected patients. Nevertheless, the obstacles to detecting and analyzing ctDNA should be addressed before translation into routine practice. Also, more clinical trials should be conducted to study the significance of ctDNA in commonly diagnosed malignancies. Given the recent advances in personalized anti-neoplastic treatments, further studies are needed to detect a panel of ctDNA and patient-specific ctDNA for various cancers.
Collapse
Affiliation(s)
- Negin Raei
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Saeid Latifi-Navid
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| |
Collapse
|
48
|
Xia J, Zhang J, Xiong Y, Zhao J, Zhou Y, Jiang T, Zhu J. Circulating tumor DNA minimal residual disease in clinical practice of non-small cell lung cancer. Expert Rev Mol Diagn 2023; 23:913-924. [PMID: 37702546 DOI: 10.1080/14737159.2023.2252334] [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: 05/03/2023] [Accepted: 08/23/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION The advance of diagnostics and treatments has greatly improved the prognosis of non-small cell lung cancer (NSCLC) patients. However, relapse and metastasis are still common problems encountered by NSCLC patients who have achieved complete remission. Therefore, overcoming the challenge of relapse and metastasis is particularly important for improving the prognosis of NSCLC patients. Research has shown that minimal residual disease (MRD) was a potential source of tumor relapse and metastasis, and circulating tumor DNA (ctDNA) MRD has obvious advantages in predicting the relapse and metastasis of NSCLC and evaluating treatment effectiveness. Therefore, dynamic monitoring of MRD is of great significance for NSCLC patient management strategies. AREAS COVERED We have reviewed articles related to NSCLC MRD included in PubMed and describes the biological significance and historical context of MRD research, reasons for using ctDNA to evaluate MRD, and potential value and challenges of ctDNA MRD in assessing relapse and metastasis of NSCLC, ultimately guiding clinical therapeutic strategies and management. EXPERT OPINION The standardized scope of ctDNA MRD detection for NSCLC requires more clinical research evidence to minimize study differences, making it possible to include in the clinical staging as a reliable indicator.
Collapse
Affiliation(s)
- Jinghua Xia
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jiao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jinbo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yinxi Zhou
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jianfei Zhu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
49
|
Hu A, Li K, Zheng H, Rao H, Zhang T, Li B. Survival Analysis of 159 Patients With Advanced Non-Small-Cell Lung Cancer Resistant to First-Generation Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor. Clin Med Insights Oncol 2023; 17:11795549231176398. [PMID: 37378392 PMCID: PMC10291855 DOI: 10.1177/11795549231176398] [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: 02/25/2023] [Accepted: 04/28/2023] [Indexed: 06/29/2023] Open
Abstract
Background Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) have limited or no response in some certain patients of non-small-cell lung cancer (NSCLC). However, real-world survival analyses comparing clinical data and EGFR-plasma mutation are still lacking. Methods In total, 159 patients with advanced NSCLC resistant to first-generation EGFR-TKIs were included for consecutive blood sampling in this study. Super-amplification refractory mutation system (Super-ARMS) was used to detect EGFR-plasma mutations and correlations between survival and circulating tumor DNA (ctDNA) were analyzed. Results Among 159 eligible patients, the T790M mutation was detected in 27.0% (43/159) of patients. The median progression-free survival (mPFS) was 10.7 months in all patients. Survival analysis revealed that patients with the T790M mutation had shorter progression-free survival (PFS) than those with the T790M wild-type (10.6 months vs 10.8 months, P = .038). Patients who cleared EGFR-plasma mutation had prolonged PFS compared with those with nonclearing EGFR-plasma mutation (11.6 months vs 9.0 months, P = .001). Cox multivariate analysis showed that the nonclearance of EGFR-plasma mutations was an independent risk factor for PFS (RR = 1.745, 95% CI: [1.184, 2.571], P = .005). The T790M mutation was associated with nonclearance of the EGFR-plasma mutation (χ2 = 10.407, P = .001). Conclusion Patients with advanced NSCLC who were resistant to the first-generation EGFR-TKI had a prolonged PFS with clearance of EGFR-plasma mutation. Those nonclearers were more likely to harbor T790M mutations in plasma.
Collapse
Affiliation(s)
- Aimin Hu
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Kun Li
- Department of Pathology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hua Zheng
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Haitao Rao
- Department of Medical Records, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Tongmei Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Baolan Li
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
50
|
Lin CF, Chen ZW, Kang FP, Hu JF, Huang L, Liao CY, Lai JL, Huang Y, Wang ZW, Tian YF, Chen S. Analyzing molecular typing and clinical application of immunogenic cell death-related genes in hepatocellular carcinoma. BMC Cancer 2023; 23:522. [PMID: 37291495 PMCID: PMC10249577 DOI: 10.1186/s12885-023-10992-2] [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: 02/08/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is considered one of the most common cancers, characterized by low early detection and high mortality rates, and is a global health challenge. Immunogenic cell death (ICD) is defined as a specific type of regulated cell death (RCD) capable of reshaping the tumor immune microenvironment by releasing danger signals that trigger immune responses, which would contribute to immunotherapy. METHODS The ICD gene sets were collected from the literature. We collected expression data and clinical information from public databases for the HCC samples in our study. Data processing and mapping were performed using R software to analyze the differences in biological characteristics between different subgroups. The expression of the ICD representative gene in clinical specimens was assessed by immunohistochemistry, and the role of the representative gene in HCC was evaluated by various in vitro assays, including qRT-PCR, colony formation, and CCK8 assay. Lasso-Cox regression was used to screen prognosis-related genes, and an ICD-related risk model (ICDRM) was constructed. To improve the clinical value of ICDRM, Nomograms and calibration curves were created to predict survival probabilities. Finally, the critical gene of ICDRM was further investigated through pan-cancer analysis and single-cell analysis. RESULTS We identified two ICD clusters that differed significantly in terms of survival, biological function, and immune infiltration. As well as assessing the immune microenvironment of tumors in HCC patients, we demonstrate that ICDRM can differentiate ICD clusters and predict the prognosis and effectiveness of therapy. High-risk subpopulations are characterized by high TMB, suppressed immunity, and poor survival and response to immunotherapy, whereas the opposite is true for low-risk subpopulations. CONCLUSIONS This study reveals the potential impact of ICDRM on the tumor microenvironment (TME), immune infiltration, and prognosis of HCC patients, but also a potential tool for predicting prognosis.
Collapse
Affiliation(s)
- Cai-Feng Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Zhi-Wen Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Feng-Ping Kang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Jian-Fei Hu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Long Huang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Cheng-Yu Liao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Jian-Lin Lai
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Yi Huang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Zu-Wei Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China.
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, 350001, China.
| | - Yi-Feng Tian
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China.
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China.
- Department of Hepatobiliary Surgery, Shengli Clinical Medical College of Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, PR China.
| | - Shi Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian Province, People's Republic of China.
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, People's Republic of China.
- Department of Hepatobiliary Surgery, Shengli Clinical Medical College of Fujian Medical University, No. 134, East Street, Fuzhou, 350001, Fujian Province, PR China.
| |
Collapse
|