1
|
Li HS, Zhang XF, Fu J, Yuan B. Efficacy of microwave ablation vs laparoscopic hepatectomy for primary small liver cancer: A comparative study. World J Gastrointest Surg 2025; 17:101786. [PMID: 40162382 PMCID: PMC11948124 DOI: 10.4240/wjgs.v17.i3.101786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND In-depth comparative investigations in terms of clinical efficacies of liver tumor microwave ablation (MWA) and laparoscopic hepatectomy (LH), which are both important treatment modalities for liver neoplasms, have been limited in patients diagnosed with primary small liver cancer (PSLC). AIM To compare and analyze the clinical efficacy of liver tumor MWA and LH for PSLC. METHODS This study retrospectively analyzed the medical records of 123 patients with PSLC admitted to Xuzhou Central Hospital from January 2015 to November 2022 and categorized them based on treatment modalities into the LH and MWA groups. The LH group, consisting of 61 cases, received LH, and the MWA group, which included 62 cases, underwent liver tumor MWA. Basic data and various perioperative indicators were compared between the two groups, including changes in liver function indicators [alanine aminotransferase (ALT), glutamic aminotransferase (AST), and total bilirubin (TBIL)] pre- and post-treatment, and efficacy and postoperative complications were analyzed. RESULTS No statistically significant difference was observed between the two groups in terms of age, gender, tumor diameter, liver function Child-Pugh classification and number of tumors, body mass index, and educational status (P > 0.05). The overall effective rate was higher in the MWA group than in the LH group (98.39% vs 88.52%) (χ 2 = 4.918, P = 0.027). The MWA group exhibited less operation time, intraoperative bleeding, defecation time, and hospital stay than the LH group (P < 0.05). No difference was found in liver function indicators between the two groups pre-treatment (P > 0.05), and ALT, AST, and TBIL levels decreased in both groups post-treatment, with the MWA group demonstrating lower levels (P < 0.05). The MWA and LH groups exhibited postoperative complication rates of 4.84% and 19.67%, respectively, with statistically significant differences between the two groups (P = 0.012, χ 2 = 6.318). CONCLUSION MWA is more effective in treating PSLC, and it promotes faster postoperative recovery for patients, and more security improves liver function and reduces postoperative complications compared to LH.
Collapse
Affiliation(s)
- Huan-Song Li
- Department of Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| | - Xuan-Feng Zhang
- Department of Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| | - Jun Fu
- Department of Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| | - Bo Yuan
- Department of Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| |
Collapse
|
2
|
Papatheodoridi A, Lekakis V, Chatzigeorgiou A, Papatheodoridis G. The Current Role of Circulating Cell-Free DNA in the Management of Hepatocellular Carcinoma. Cancers (Basel) 2025; 17:1042. [PMID: 40149374 PMCID: PMC11940583 DOI: 10.3390/cancers17061042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Circulating cell-free DNA (cfDNA) has emerged as a compelling candidate of liquid biopsy markers for the diagnosis and prognosis of several cancers. We systematically reviewed data on the role of cfDNA markers in the diagnosis, prognosis and treatment of hepatocellular carcinoma (HCC). Early studies suggested that levels of circulating cfDNA, mitochondrial DNA and cfDNA integrity are higher in patients with HCC than chronic liver diseases. In subsequent studies, methylation changes in circulating tumor DNA (ctDNA) as well as cfDNA fragmentation patterns and circulating nucleosomes were found to offer high sensitivity (>60%) and excellent specificity (>90%) for HCC diagnosis. The predictive role of cfDNA markers and ctDNA has been assessed in a few studies including untreated patients with HCC providing promising results for prediction of survival. However, port-hepatectomy detection of cfDNA/ctDNA markers or copy number variation indicators of cfDNA seem to reflect minimum residual disease and thus a high risk for HCC recurrence. The same markers can be useful for prediction after transarterial chemoembolization, radiofrequency ablation, radiotherapy and even systemic therapies. In conclusion, cfDNA markers can be useful in HCC surveillance, improving early diagnosis rates, as well as for monitoring treatment effectiveness and minimal residual disease post-treatment.
Collapse
Affiliation(s)
- Alkistis Papatheodoridi
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, “Alexandra” General Hospital of Athens, 11528 Athens, Greece;
| | - Vasileios Lekakis
- First Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, 11527 Athens, Greece;
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School of National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - George Papatheodoridis
- First Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, 11527 Athens, Greece;
| |
Collapse
|
3
|
Hu J, Tang H, Jia CC, Zhang XY, Xu Y, Tan JP, Fan J, Jia S, Zhou J. Personalized MRD Assessment in Perisurgical ctDNA for Prognostic Prediction in Hepatocellular Carcinoma. Clin Cancer Res 2025; 31:1047-1056. [PMID: 39526910 DOI: 10.1158/1078-0432.ccr-24-1897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/19/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Detecting residual disease is a critical clinical requirement in the perisurgical management of patients with resectable hepatocellular carcinoma (HCC). Previous studies focused on specific genomic regions exhibiting limited sensitivity and failed to meet the minimal residual disease (MRD) testing threshold. We introduce a next-generation sequencing-based assay, informed by baseline samples, facilitating MRD detection in hepatectomized patients with HCC and offering prognostic predictions. EXPERIMENTAL DESIGN This study involved 88 patients with HCC who underwent surgical resections from January 2016 to May 2016 in Zhongshan Hospital, Fudan University. Tumor and normal tissue samples were collected during surgery, whereas plasma samples were obtained both before surgery and up to 7 days after surgery. Using a next-generation sequencing-based personalized ctDNA assay, we analyzed the MRD in both presurgical and postsurgical blood samples and its correlation with prognosis. RESULTS With a median follow-up period of 80.7 months, our findings demonstrated significant correlations between presurgical ctDNA tumor fractions, postsurgical plasma MRD status, and both recurrence-free survival and overall survival. Postsurgical MRD status emerged as the most significant risk factor for cancer recurrence (HR = 2.162; 95% confidence interval, 1.09-4.30; P = 0.027) compared with other clinical characteristics in multivariate Cox regression analysis. Notably, MRD status showed potential as a prognostic indicator among clinically low-recurrent-risk patients, such as those with Barcelona Clinic Liver Cancer stages 0 to A or China Liver Cancer Staging stages I to II. CONCLUSIONS Evaluating personalized MRD provided crucial prognostic insights into recurrence-free survival and overall survival. It efficiently identified patients at high risk of recurrence, even among those initially perceived as low-risk cases. See related commentary by Pinato et al., p. 955.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/surgery
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/blood
- Carcinoma, Hepatocellular/mortality
- Liver Neoplasms/surgery
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/blood
- Liver Neoplasms/mortality
- Neoplasm, Residual/pathology
- Male
- Female
- Circulating Tumor DNA/genetics
- Circulating Tumor DNA/blood
- Prognosis
- Middle Aged
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/blood
- High-Throughput Nucleotide Sequencing
- Aged
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/blood
- Neoplasm Recurrence, Local/surgery
- Adult
- Hepatectomy
- Follow-Up Studies
- Precision Medicine/methods
Collapse
Affiliation(s)
- Jie Hu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Haoran Tang
- Huidu (Shanghai) Medical Sciences, Ltd., Shanghai, China
| | - Can-Can Jia
- Huidu (Shanghai) Medical Sciences, Ltd., Shanghai, China
| | - Xiang-Yu Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Ying Xu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Jin-Peng Tan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Shidong Jia
- Huidu (Shanghai) Medical Sciences, Ltd., Shanghai, China
| | - Jian Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| |
Collapse
|
4
|
Bruhm DC, Vulpescu NA, Foda ZH, Phallen J, Scharpf RB, Velculescu VE. Genomic and fragmentomic landscapes of cell-free DNA for early cancer detection. Nat Rev Cancer 2025:10.1038/s41568-025-00795-x. [PMID: 40038442 DOI: 10.1038/s41568-025-00795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 03/06/2025]
Abstract
Genomic analyses of cell-free DNA (cfDNA) in plasma are enabling noninvasive blood-based biomarker approaches to cancer detection and disease monitoring. Current approaches for identification of circulating tumour DNA typically use targeted tumour-specific mutations or methylation analyses. An emerging approach is based on the recognition of altered genome-wide cfDNA fragmentation in patients with cancer. Recent studies have revealed a multitude of characteristics that can affect the compendium of cfDNA fragments across the genome, collectively called the 'cfDNA fragmentome'. These changes result from genomic, epigenomic, transcriptomic and chromatin states of an individual and affect the size, position, coverage, mutation, structural and methylation characteristics of cfDNA. Identifying and monitoring these changes has the potential to improve early detection of cancer, especially using highly sensitive multi-feature machine learning approaches that would be amenable to broad use in populations at increased risk. This Review highlights the rapidly evolving field of genome-wide analyses of cfDNA characteristics, their comparison to existing cfDNA methods, and recent related innovations at the intersection of large-scale sequencing and artificial intelligence. As the breadth of clinical applications of cfDNA fragmentome methods have enormous public health implications for cancer screening and personalized approaches for clinical management of patients with cancer, we outline the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Daniel C Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas A Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah H Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
Yu H, Feng M, Liu C, Wang F, Pan S, Sui G, Jing W, Cheng X. CRISPR-Cas12a2-based rapid and sensitive detection system for target nucleic acid. Int J Biol Macromol 2025; 290:138996. [PMID: 39706401 DOI: 10.1016/j.ijbiomac.2024.138996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/24/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Infectious diseases are extremely important public health issues, where the design of effective, rapid, and convenient detection platforms is critical. In this study, we coupled SuCas12a2, a novel Cas12 family RNA-targeting nuclease, with conventional PCR and recombinase polymerase amplification (RPA), respectively, to develop novel detection approaches, named PCR-SuCas12a2 and RPA-SuCas12a2. SuCas12a2 possesses collateral cleavage activity and cuts the additional single-stranded RNA (ssRNA) added to the reaction system once the ternary complex RNA-SuCas12a2-CRISPR RNA (crRNA) is formed. SuCas12a2 is specifically activated, where the cleaved fluorescent-labeled probes release fluorescent signals, with the strength of the fluorescent signal being proportional to the concentration of nucleic acids specifically bound to crRNA. Simultaneous transcription and SuCas12a2 detection can be performed in a single tube by introducing the T7 promoter sequence into the forward primer. Entamoeba histolytica (E. histolytica) and Mycoplasma pneumoniae (M. pneumoniae) were used as proof specimens to evaluate the performance of the platform. PCR-SuCas12a2 has excellent capabilities, including high specificity with no cross-reactivity from other species and ultra-sensitivity that achieves a detection of one copy per reaction for E. histolytica and M. pneumoniae. However, the sensitivity of the RPA-SuCas12a2 assay was 102 copies per reaction, which was inferior to PCR-SuCas12a2. Clinical samples were obtained from suspected infection patients of E. histolytica and M. pneumoniae, and used to evaluate the systems demonstrated 100 % specificity. The technique shows robust performance and suggests great potential for point-of-care testing of other pathogens to facilitate effective management and control of the spread of diseases.
Collapse
Affiliation(s)
- Helin Yu
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Meng Feng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Chuncao Liu
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Feifei Wang
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Shaokun Pan
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Guodong Sui
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200438, China
| | - Wenwen Jing
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China.
| | - Xunjia Cheng
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China.
| |
Collapse
|
6
|
Liu Y, Peng F, Wang S, Jiao H, Zhou K, Guo W, Guo S, Dang M, Zhang H, Zhou W, Guo X, Xing J. Aberrant fragmentomic features of circulating cell-free mitochondrial DNA enable early detection and prognosis prediction of hepatocellular carcinoma. Clin Mol Hepatol 2025; 31:196-212. [PMID: 39406379 PMCID: PMC11791606 DOI: 10.3350/cmh.2024.0527] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND/AIMS Early detection and effective prognosis prediction in patients with hepatocellular carcinoma (HCC) provide an avenue for survival improvement, yet more effective approaches are greatly needed. We sought to develop the detection and prognosis models with ultra-sensitivity and low cost based on fragmentomic features of circulating cell free mtDNA (ccf-mtDNA). METHODS Capture-based mtDNA sequencing was carried out in plasma cell-free DNA samples from 1168 participants, including 571 patients with HCC, 301 patients with chronic hepatitis B or liver cirrhosis (CHB/LC) and 296 healthy controls (HC). RESULTS The systematic analysis revealed significantly aberrant fragmentomic features of ccf-mtDNA in HCC group when compared with CHB/LC and HC groups. Moreover, we constructed a random forest algorithm-based HCC detection model by utilizing ccf-mtDNA fragmentomic features. Both internal and two external validation cohorts demonstrated the excellent capacity of our model in distinguishing early HCC patients from HC and highrisk population with CHB/LC, with AUC exceeding 0.983 and 0.981, sensitivity over 89.6% and 89.61%, and specificity over 98.20% and 95.00%, respectively, greatly surpassing the performance of alpha-fetoprotein (AFP) and mtDNA copy number. We also developed an HCC prognosis prediction model by LASSO-Cox regression to select 20 fragmentomic features, which exhibited exceptional ability in predicting 1-year, 2-year and 3-year survival (AUC=0.8333, 0.8145 and 0.7958 for validation cohort, respectively). CONCLUSION We have developed and validated a high-performing and low-cost approach in a large clinical cohort based on aberrant ccf-mtDNA fragmentomic features with promising clinical translational application for the early detection and prognosis prediction of HCC patients.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Fan Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Siyuan Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Huanmin Jiao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Kaixiang Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Wenjie Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Shanshan Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Miao Dang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Huanqin Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Weizheng Zhou
- Department of General Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Xu Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Jinliang Xing
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
7
|
Li X, Liu T, Bacchiocchi A, Li M, Cheng W, Wittkop T, Mendez FL, Wang Y, Tang P, Yao Q, Bosenberg MW, Sznol M, Yan Q, Faham M, Weng L, Halaban R, Jin H, Hu Z. Ultra-sensitive molecular residual disease detection through whole genome sequencing with single-read error correction. EMBO Mol Med 2024; 16:2188-2209. [PMID: 39164471 PMCID: PMC11393307 DOI: 10.1038/s44321-024-00115-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/06/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
While whole genome sequencing (WGS) of cell-free DNA (cfDNA) holds enormous promise for detection of molecular residual disease (MRD), its performance is limited by WGS error rate. Here we introduce AccuScan, an efficient cfDNA WGS technology that enables genome-wide error correction at single read-level, achieving an error rate of 4.2 × 10-7, which is about two orders of magnitude lower than a read-centric de-noising method. The application of AccuScan to MRD demonstrated analytical sensitivity down to 10-6 circulating variant allele frequency at 99% sample-level specificity. AccuScan showed 90% landmark sensitivity (within 6 weeks after surgery) and 100% specificity for predicting relapse in colorectal cancer. It also showed 67% sensitivity and 100% specificity in esophageal cancer using samples collected within one week after surgery. When AccuScan was applied to monitor immunotherapy in melanoma patients, the circulating tumor DNA (ctDNA) levels and dynamic profiles were consistent with clinical outcomes. Overall, AccuScan provides a highly accurate WGS solution for MRD detection, empowering ctDNA detection at parts per million range without requiring high sample input or personalized reagents.
Collapse
Affiliation(s)
- Xinxing Li
- Department of Gastrointestinal Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, P. R. China
| | - Tao Liu
- Department of Thoracic Surgery, Peking University First Hospital, Beijing, 100034, China
| | | | - Mengxing Li
- Department of Thoracic Surgery, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Wen Cheng
- Department of Thoracic Surgery, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Tobias Wittkop
- Department of Research and Development, AccuraGen Inc, San Jose, CA, 95134, USA
| | - Fernando L Mendez
- Department of Research and Development, AccuraGen Inc, San Jose, CA, 95134, USA
| | - Yingyu Wang
- Department of Research and Development, AccuraGen Inc, San Jose, CA, 95134, USA
| | - Paul Tang
- Department of Research and Development, AccuraGen Inc, San Jose, CA, 95134, USA
| | - Qianqian Yao
- Department of Medical Science, Shanghai YunSheng Medical Laboratory Co., Ltd, Shanghai, 200437, China
| | - Marcus W Bosenberg
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, CT, USA
| | - Mario Sznol
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Qin Yan
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Malek Faham
- Department of Research and Development, AccuraGen Inc, San Jose, CA, 95134, USA
| | - Li Weng
- Department of Research and Development, AccuraGen Inc, San Jose, CA, 95134, USA.
| | - Ruth Halaban
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA.
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
| | - Hai Jin
- Department of Thoracic Surgery, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Zhiqian Hu
- Department of Gastrointestinal Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, P. R. China.
- Department of General Surgery, Changzheng Hospital Naval Medical University, Shanghai, 200003, P. R. China.
| |
Collapse
|
8
|
Jiao Z, Zhang X, Xuan Y, Shi X, Zhang Z, Yu A, Li N, Yang S, He X, Zhao G, Yang R, Chen J, Wu X, Bao H, Wang F, Ren W, Liang H, Chen Q, Wang T. Leveraging cfDNA fragmentomic features in a stacked ensemble model for early detection of esophageal squamous cell carcinoma. Cell Rep Med 2024; 5:101664. [PMID: 39089259 PMCID: PMC11384130 DOI: 10.1016/j.xcrm.2024.101664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/26/2024] [Accepted: 07/08/2024] [Indexed: 08/03/2024]
Abstract
In this study, we develop a stacked ensemble model that utilizes cell-free DNA (cfDNA) fragmentomics for the early detection of esophageal squamous cell carcinoma (ESCC). This model incorporates four distinct fragmentomics features derived from whole-genome sequencing (WGS) and advanced machine learning algorithms for robust analysis. It is validated across both an independent validation cohort and an external cohort to ensure its generalizability and effectiveness. Notably, the model maintains its robustness in low-coverage sequencing environments, demonstrating its potentials in clinical settings with limited sequencing resources. With its remarkable sensitivity and specificity, this approach promises to significantly improve the early diagnosis and management of ESCC. This study represents a substantial step forward in the application of cfDNA fragmentomics in cancer diagnostics, emphasizing the need for further research to fully establish its clinical efficacy.
Collapse
Affiliation(s)
- Zichen Jiao
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China; The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xiaoqiang Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yulong Xuan
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Xiaoming Shi
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Zirui Zhang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Ao Yu
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Ningyou Li
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | | | - Xiaofeng He
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Gefei Zhao
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China
| | - Ruowei Yang
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Jianqun Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | | | - Hua Bao
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Fufeng Wang
- Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Wei Ren
- Department of Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China.
| | - Hongwei Liang
- School of Life Sciences and Technology China Pharmaceutical University, Nanjing, China.
| | - Qihan Chen
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China; The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China; Cancer Center, Faculty of Health Sciences, University of Macau, China; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau, China.
| | - Tao Wang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
9
|
Hou Y, Meng X, Zhou X. Systematically Evaluating Cell-Free DNA Fragmentation Patterns for Cancer Diagnosis and Enhanced Cancer Detection via Integrating Multiple Fragmentation Patterns. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308243. [PMID: 38881520 PMCID: PMC11321639 DOI: 10.1002/advs.202308243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/12/2024] [Indexed: 06/18/2024]
Abstract
Cell-free DNA (cfDNA) fragmentation patterns have immense potential for early cancer detection. However, the definition of fragmentation varies, ranging from the entire genome to specific genomic regions. These patterns have not been systematically compared, impeding broader research and practical implementation. Here, 1382 plasma cfDNA sequencing samples from 8 cancer types are collected. Considering that cfDNA within open chromatin regions is more susceptible to fragmentation, 10 fragmentation patterns within open chromatin regions as features and employed machine learning techniques to evaluate their performance are examined. All fragmentation patterns demonstrated discernible classification capabilities, with the end motif showing the highest diagnostic value for cross-validation. Combining cross and independent validation results revealed that fragmentation patterns that incorporated both fragment length and coverage information exhibited robust predictive capacities. Despite their diagnostic potential, the predictive power of these fragmentation patterns is unstable. To address this limitation, an ensemble classifier via integrating all fragmentation patterns is developed, which demonstrated notable improvements in cancer detection and tissue-of-origin determination. Further functional bioinformatics investigations on significant feature intervals in the model revealed its impressive ability to identify critical regulatory regions involved in cancer pathogenesis.
Collapse
Affiliation(s)
- Yuying Hou
- Hubei Key Laboratory of Agricultural BioinformaticsCollege of InformaticsHuazhong Agricultural UniversityWuhan430070China
| | - Xiang‐Yu Meng
- Hubei Key Laboratory of Agricultural BioinformaticsCollege of InformaticsHuazhong Agricultural UniversityWuhan430070China
- Health Science CenterHubei Minzu UniversityEnshi445000China
- Hubei Provincial Clinical Medical Research Center for NephropathyHubei Minzu UniversityEnshi445000China
| | - Xionghui Zhou
- Hubei Key Laboratory of Agricultural BioinformaticsCollege of InformaticsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Smart Farming for Agricultural AnimalsMinistry of Agriculture and Rural AffairsWuhan430070China
| |
Collapse
|
10
|
Huang C, Qiu Z, Wang M, Ji J, Xiao X, Wang Y, Xu X, Gao Z, Gao C. N-glycan signatures identified in the serum from biliary tract cancer patients: Association with clinical diagnosis and prognosis. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:537-548. [PMID: 38824438 DOI: 10.1002/jhbp.12011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Abstract
BACKGROUND Changes in the expression of genes related to glycosyltransferases may lead to alterations in N-glycan structure abundance, potentially acting as markers for diagnosis and prognosis in biliary tract cancer (BTC). METHODS This study was divided into cross-sectional and longitudinal approaches. The cross-sectional study included 316 BTC and 301 non-BTC. Propensity score matching was applied to adjust for sex and age differences between BTC and non-BTC. Univariate and multivariate logistic regression identified independent risk factors for BTC and constructed the BTC-G model. The ROC curve was used to validate the diagnostic performance of BTC-G. Longitudinal follow-up studies included postoperative (N = 50) and immunotherapy (N = 43) follow-up cohorts. Cox regression analysis identified N-glycan structures impacting BTC prognosis postoperative and immunotherapy, with further confirmation through Kaplan-Meier curves. RESULTS Univariate and multivariate analyses identified Peak3 (OR: 0.790, 95% CI: 0.658-0.949), Peak9 (OR: 1.646, 95% CI: 1.409-1.922), and Peak9p (OR: 2.467, 95% CI: 1.267-4.804) as independent BTC risk factors, leading to the creation of the BTC-G. The ROC curve confirmed that BTC-G performed well in training (AUC: 0.753, 95% CI: 0.703-0.799), validation (AUC: 0.811, 95% CI: 0.740-0.870), and CA19-9 negative cohorts (AUC: 0.717, 95% CI: 0.664-0.767). Cox regression analysis and Kaplan-Meier curves established that Peak12 (HR: 5.578, 95% CI: 1.145-27.170) and Peak11 (HR: 1.104, 95% CI: 0.611-1.994) are independent risk factors for BTC prognosis following surgery and immunotherapy, respectively. CONCLUSIONS Our NGFP technology supplements BTC diagnostics, distinguishing survival and recurrence subtypes for postoperative and immunotherapy, thereby supporting the development of treatment strategies.
Collapse
Affiliation(s)
- Chenjun Huang
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhiquan Qiu
- Department of Biliary Tract Surgery I, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Mengmeng Wang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jun Ji
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Xiao Xiao
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Wang
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuewen Xu
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhiyuan Gao
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunfang Gao
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
11
|
Xu S, Luo J, Tang W, Bao H, Wang J, Chang S, Zou Z, Fan X, Liu Y, Jiang C, Wu X. Detecting pulmonary malignancy against benign nodules using noninvasive cell-free DNA fragmentomics assay. ESMO Open 2024; 9:103595. [PMID: 39088983 PMCID: PMC11345357 DOI: 10.1016/j.esmoop.2024.103595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND Early screening using low-dose computed tomography (LDCT) can reduce mortality caused by non-small-cell lung cancer. However, ∼25% of the 'suspicious' pulmonary nodules identified by LDCT are later confirmed benign through resection surgery, adding to patients' discomfort and the burden on the healthcare system. In this study, we aim to develop a noninvasive liquid biopsy assay for distinguishing pulmonary malignancy from benign yet 'suspicious' lung nodules using cell-free DNA (cfDNA) fragmentomics profiling. METHODS An independent training cohort consisting of 193 patients with malignant nodules and 44 patients with benign nodules was used to construct a machine learning model. Base models using four different fragmentomics profiles were optimized using an automated machine learning approach before being stacked into the final predictive model. An independent validation cohort, including 96 malignant nodules and 22 benign nodules, and an external test cohort, including 58 malignant nodules and 41 benign nodules, were used to assess the performance of the stacked ensemble model. RESULTS Our machine learning models demonstrated excellent performance in detecting patients with malignant nodules. The area under the curves reached 0.857 and 0.860 in the independent validation cohort and the external test cohort, respectively. The validation cohort achieved an excellent specificity (68.2%) at the targeted 90% sensitivity (89.6%). An equivalently good performance was observed while applying the cut-off to the external cohort, which reached a specificity of 63.4% at 89.7% sensitivity. A subgroup analysis for the independent validation cohort showed that the sensitivities for detecting various subgroups of nodule size (<1 cm: 91.7%; 1-3 cm: 88.1%; >3 cm: 100%; unknown: 100%) and smoking history (yes: 88.2%; no: 89.9%) all remained high among the lung cancer group. CONCLUSIONS Our cfDNA fragmentomics assay can provide a noninvasive approach to distinguishing malignant nodules from radiographically suspicious but pathologically benign ones, amending LDCT false positives.
Collapse
Affiliation(s)
- S Xu
- The Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - J Luo
- The Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - W Tang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - H Bao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - J Wang
- The Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - S Chang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Z Zou
- The Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - X Fan
- The Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Y Liu
- The Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - C Jiang
- The Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - X Wu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| |
Collapse
|
12
|
Liu J, Hu D, Lin Y, Chen X, Yang R, Li L, Zhan Y, Bao H, Zang L, Zhu M, Zhu F, Yan J, Zhu D, Zhang H, Xu B, Xu Q. Early detection of uterine corpus endometrial carcinoma utilizing plasma cfDNA fragmentomics. BMC Med 2024; 22:310. [PMID: 39075419 PMCID: PMC11288124 DOI: 10.1186/s12916-024-03531-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Uterine corpus endometrial carcinoma (UCEC) is a prevalent gynecologic malignancy with a favorable prognosis if detected early. However, there is a lack of accurate and reliable early detection tests for UCEC. This study aims to develop a precise and non-invasive diagnostic method for UCEC using circulating cell-free DNA (cfDNA) fragmentomics. METHODS Peripheral blood samples were collected from all participants, and cfDNA was extracted for analysis. Low-coverage whole-genome sequencing was performed to obtain cfDNA fragmentomics data. A robust machine learning model was developed using these features to differentiate between UCEC and healthy conditions. RESULTS The cfDNA fragmentomics-based model showed high predictive power for UCEC detection in training (n = 133; AUC 0.991) and validation cohorts (n = 89; AUC 0.994). The model manifested a specificity of 95.5% and a sensitivity of 98.5% in the training cohort, and a specificity of 95.5% and a sensitivity of 97.8% in the validation cohort. Physiological variables and preanalytical procedures had no significant impact on the classifier's outcomes. In terms of clinical benefit, our model would identify 99% of Chinese UCEC patients at stage I, compared to 21% under standard care, potentially raising the 5-year survival rate from 84 to 95%. CONCLUSION This study presents a novel approach for the early detection of UCEC using cfDNA fragmentomics and machine learning showing promising sensitivity and specificity. Using this model in clinical practice could significantly improve UCEC management and control, enabling early intervention and better patient outcomes. Further optimization and validation of this approach are warranted to establish its clinical utility.
Collapse
Affiliation(s)
- Jing Liu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Dan Hu
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Yibin Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Xiaoxi Chen
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Ruowei Yang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Li Li
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Yanyan Zhan
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - LeLe Zang
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Mingxuan Zhu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Fei Zhu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Junrong Yan
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Dongqin Zhu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Huiqi Zhang
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Benhua Xu
- Department of Radiation, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Qin Xu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China.
| |
Collapse
|
13
|
Liu J, Shen H, Chen K, Li X. Large language model produces high accurate diagnosis of cancer from end-motif profiles of cell-free DNA. Brief Bioinform 2024; 25:bbae430. [PMID: 39222060 PMCID: PMC11367762 DOI: 10.1093/bib/bbae430] [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: 04/15/2024] [Revised: 07/05/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Instruction-tuned large language models (LLMs) demonstrate exceptional ability to align with human intentions. We present an LLM-based model-instruction-tuned LLM for assessment of cancer (iLLMAC)-that can detect cancer using cell-free deoxyribonucleic acid (cfDNA) end-motif profiles. Developed on plasma cfDNA sequencing data from 1135 cancer patients and 1106 controls across three datasets, iLLMAC achieved area under the receiver operating curve (AUROC) of 0.866 [95% confidence interval (CI), 0.773-0.959] for cancer diagnosis and 0.924 (95% CI, 0.841-1.0) for hepatocellular carcinoma (HCC) detection using 16 end-motifs. Performance increased with more motifs, reaching 0.886 (95% CI, 0.794-0.977) and 0.956 (95% CI, 0.89-1.0) for cancer diagnosis and HCC detection, respectively, with 64 end-motifs. On an external-testing set, iLLMAC achieved AUROC of 0.912 (95% CI, 0.849-0.976) for cancer diagnosis and 0.938 (95% CI, 0.885-0.992) for HCC detection with 64 end-motifs, significantly outperforming benchmarked methods. Furthermore, iLLMAC achieved high classification performance on datasets with bisulfite and 5-hydroxymethylcytosine sequencing. Our study highlights the effectiveness of LLM-based instruction-tuning for cfDNA-based cancer detection.
Collapse
Affiliation(s)
- Jilei Liu
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Hongru Shen
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Xiangchun Li
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| |
Collapse
|
14
|
Chen K, Wang J, Jiang L, Zhao F, Zhang R, Wu Z, Wang D, Jiao Y, Xie H, Qu C. A Blood Hepatocellular Carcinoma Signature Recognizes Very Small Tumor Nodules with Metastatic Traits. J Clin Transl Hepatol 2024; 12:551-561. [PMID: 38974959 PMCID: PMC11224907 DOI: 10.14218/jcth.2023.00559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/03/2024] [Accepted: 04/15/2024] [Indexed: 07/09/2024] Open
Abstract
Background and Aims Hepatocellular carcinoma (HCC) cases with small nodules are commonly treated with radiofrequency ablation (RFA), but the recurrence rate remains high. This study aimed to establish a blood signature for identifying HCC with metastatic traits pre-RFA. Methods Data from HCC patients treated between 2010 and 2017 were retrospectively collected. A blood signature for metastatic HCC was established based on blood levels of alpha-fetoprotein and des-γ-carboxy-prothrombin, cell-free DNA (cfDNA) mutations, and methylation changes in target genes in frozen-stored plasma samples that were collected before RFA performance. The HCC blood signature was validated in patients prospectively enrolled in 2021. Results Of 251 HCC patients in the retrospective study, 33.9% experienced recurrence within 1 year post-RFA. The HCC blood signature identified from these patients included des-γ-carboxy-prothrombin ≥40 mAU/mL with cfDNA mutation score, where cfDNA mutations occurred in the genes of TP53, CTNNB1, and TERT promoter. This signature effectively predicted 1-year post-RFA recurrence of HCC with 92% specificity and 91% sensitivity in the retrospective dataset, and with 87% specificity and 76% sensitivity in the prospective dataset (n=32 patients). Among 14 cases in the prospective study with biopsy tissues available, positivity for the HCC blood signature was associated with a higher HCC tissue score and shorter distance between HCC cells and microvasculature. Conclusions This study established an HCC blood signature in pre-RFA blood that potentially reflects HCC with metastatic traits and may be valuable for predicting the disease's early recurrence post-RFA.
Collapse
Affiliation(s)
- Kun Chen
- State Key Lab of Molecular Oncology, Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junxiao Wang
- Senior Department of Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Aerospace Medical Center/Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, China
| | - Liping Jiang
- State Key Lab 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
| | - Fei Zhao
- State Key Lab of Molecular Oncology, Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruochan Zhang
- State Key Lab of Molecular Oncology, Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiyuan Wu
- State Key Lab of Molecular Oncology, Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongmei Wang
- State Key Lab of Molecular Oncology, Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuchen Jiao
- State Key Lab 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
| | - Hui Xie
- Senior Department of Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chunfeng Qu
- State Key Lab of Molecular Oncology, Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Lian S, Lu C, Li F, Yu X, Ai L, Wu B, Gong X, Zhou W, Xie Y, Du Y, Quan W, Wang P, Deng L, Liang X, Zhan J, Yuan Y, Fang F, Liu Z, Ji M, Zheng Z. Circulating DNA genome-wide fragmentation in early detection and disease monitoring of hepatocellular carcinoma. iScience 2024; 27:109701. [PMID: 38680658 PMCID: PMC11053305 DOI: 10.1016/j.isci.2024.109701] [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: 05/16/2023] [Revised: 09/05/2023] [Accepted: 04/05/2024] [Indexed: 05/01/2024] Open
Abstract
Genome-wide circulating cell-free DNA (ccfDNA) fragmentation for cancer detection has been rarely evaluated using blood samples collected before cancer diagnosis. To evaluate ccfDNA fragmentation for detecting early hepatocellular carcinoma (HCC), we first modeled and tested using hospitalized HCC patients and then evaluated in a population-based study. A total of 427 samples were analyzed, including 270 samples collected prior to HCC diagnosis from a population-based study. Our model distinguished hospital HCC patients from controls excellently (area under curve 0.999). A high ccfDNA fragmentation score was highly associated with an advanced tumor stage and a shorter survival. In evaluation, the model showed increasing sensitivities in detecting HCC using 'pre-samples' collected ≥4 years (8.3%), 3-4 years (20.0%), 2-3 years (31.0%), 1-2 years (35.0%), and 0-1 year (36.4%) before diagnosis. These findings suggested ccfDNA fragmentation is sensitive in clinical HCC detection and might be helpful in screening early HCC.
Collapse
Affiliation(s)
- Shifeng Lian
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Sha Tin, Hong Kong SAR of the People’s Republic of China
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Chenyu Lu
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon, Hong Kong SAR of the People’s Republic of China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Research Institute, Shenzhen, People’s Republic of China
| | - Fugui Li
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Xia Yu
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Limei Ai
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Sha Tin, Hong Kong SAR of the People’s Republic of China
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon, Hong Kong SAR of the People’s Republic of China
| | - Biaohua Wu
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Xueyi Gong
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Wenjing Zhou
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon, Hong Kong SAR of the People’s Republic of China
| | - Yulong Xie
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Yun Du
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Wen Quan
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Panpan Wang
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Li Deng
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Xuejun Liang
- Xiaolan Public Health Service Center, Zhongshan, People’s Republic of China
| | - Jiyun Zhan
- Xiaolan Public Health Service Center, Zhongshan, People’s Republic of China
| | - Yong Yuan
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Fang Fang
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zhiwei Liu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Mingfang Ji
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, People’s Republic of China
| | - Zongli Zheng
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Sha Tin, Hong Kong SAR of the People’s Republic of China
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Kowloon, Hong Kong SAR of the People’s Republic of China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Research Institute, Shenzhen, People’s Republic of China
| |
Collapse
|
16
|
Shi X, Guo S, Duan Q, Zhang W, Gao S, Jing W, Jiang G, Kong X, Li P, Li Y, Teng C, Xu X, Chen S, Nian B, Li Z, Zhong C, Yang X, Zhu G, Du Y, Zhang D, Jin G. Detection and characterization of pancreatic and biliary tract cancers using cell-free DNA fragmentomics. J Exp Clin Cancer Res 2024; 43:145. [PMID: 38750539 PMCID: PMC11094938 DOI: 10.1186/s13046-024-03067-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Plasma cell-free DNA (cfDNA) fragmentomics has demonstrated significant differentiation power between cancer patients and healthy individuals, but little is known in pancreatic and biliary tract cancers. The aim of this study is to characterize the cfDNA fragmentomics in biliopancreatic cancers and develop an accurate method for cancer detection. METHODS One hundred forty-seven patients with biliopancreatic cancers and 71 non-cancer volunteers were enrolled, including 55 patients with cholangiocarcinoma, 30 with gallbladder cancer, and 62 with pancreatic cancer. Low-coverage whole-genome sequencing (median coverage: 2.9 ×) was performed on plasma cfDNA. Three cfDNA fragmentomic features, including fragment size, end motif and nucleosome footprint, were subjected to construct a stacked machine learning model for cancer detection. Integration of carbohydrate antigen 19-9 (CA19-9) was explored to improve model performance. RESULTS The stacked model presented robust performance for cancer detection (area under curve (AUC) of 0.978 in the training cohort, and AUC of 0.941 in the validation cohort), and remained consistent even when using extremely low-coverage sequencing depth of 0.5 × (AUC: 0.905). Besides, our method could also help differentiate biliopancreatic cancer subtypes. By integrating the stacked model and CA19-9 to generate the final detection model, a high accuracy in distinguishing biliopancreatic cancers from non-cancer samples with an AUC of 0.995 was achieved. CONCLUSIONS Our model demonstrated ultrasensitivity of plasma cfDNA fragementomics in detecting biliopancreatic cancers, fulfilling the unmet accuracy of widely-used serum biomarker CA19-9, and provided an affordable way for accurate noninvasive biliopancreatic cancer screening in clinical practice.
Collapse
Affiliation(s)
- Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Qiaonan Duan
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Wei Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Wei Jing
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Guojuan Jiang
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Xiangyu Kong
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Penghao Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yikai Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Chuanqi Teng
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiaoya Xu
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Sheng Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Baoning Nian
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Zhikuan Li
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China
| | - Chaoliang Zhong
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiaolu Yang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Guangyu Zhu
- Department of Interventional Radiology and Vascular Surgery, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu Province, 210009, China.
| | - Yiqi Du
- Department of Gastroenterology, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Dadong Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc, 158 Xin Junhuan Road, Pujiang Hi-Tech Park, Shanghai, 201114, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Navy Military Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
17
|
Jiang X, Zhou R, Jiang F, Yan Y, Zhang Z, Wang J. Construction of diagnostic models for the progression of hepatocellular carcinoma using machine learning. Front Oncol 2024; 14:1401496. [PMID: 38812780 PMCID: PMC11133637 DOI: 10.3389/fonc.2024.1401496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Liver cancer is one of the most prevalent forms of cancer worldwide. A significant proportion of patients with hepatocellular carcinoma (HCC) are diagnosed at advanced stages, leading to unfavorable treatment outcomes. Generally, the development of HCC occurs in distinct stages. However, the diagnostic and intervention markers for each stage remain unclear. Therefore, there is an urgent need to explore precise grading methods for HCC. Machine learning has emerged as an effective technique for studying precise tumor diagnosis. In this research, we employed random forest and LightGBM machine learning algorithms for the first time to construct diagnostic models for HCC at various stages of progression. We categorized 118 samples from GSE114564 into three groups: normal liver, precancerous lesion (including chronic hepatitis, liver cirrhosis, dysplastic nodule), and HCC (including early stage HCC and advanced HCC). The LightGBM model exhibited outstanding performance (accuracy = 0.96, precision = 0.96, recall = 0.96, F1-score = 0.95). Similarly, the random forest model also demonstrated good performance (accuracy = 0.83, precision = 0.83, recall = 0.83, F1-score = 0.83). When the progression of HCC was categorized into the most refined six stages: normal liver, chronic hepatitis, liver cirrhosis, dysplastic nodule, early stage HCC, and advanced HCC, the diagnostic model still exhibited high efficacy. Among them, the LightGBM model exhibited good performance (accuracy = 0.71, precision = 0.71, recall = 0.71, F1-score = 0.72). Also, performance of the LightGBM model was superior to that of the random forest model. Overall, we have constructed a diagnostic model for the progression of HCC and identified potential diagnostic characteristic gene for the progression of HCC.
Collapse
Affiliation(s)
- Xin Jiang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Ruilong Zhou
- Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fengle Jiang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Yanan Yan
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Zheting Zhang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Jianmin Wang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| |
Collapse
|
18
|
Guo DZ, Huang A, Wang YC, Zhou S, Wang H, Xing XL, Zhang SY, Cheng JW, Xie KH, Yang QC, Ma CC, Li Q, Chen Y, Su ZX, Fan J, Liu R, Liu XL, Zhou J, Yang XR. Early detection and prognosis evaluation for hepatocellular carcinoma by circulating tumour DNA methylation: A multicentre cohort study. Clin Transl Med 2024; 14:e1652. [PMID: 38741204 DOI: 10.1002/ctm2.1652] [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: 01/27/2024] [Revised: 03/07/2024] [Accepted: 03/21/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Early diagnosis of hepatocellular carcinoma (HCC) can significantly improve patient survival. We aimed to develop a blood-based assay to aid in the diagnosis, detection and prognostic evaluation of HCC. METHODS A three-phase multicentre study was conducted to screen, optimise and validate HCC-specific differentially methylated regions (DMRs) using next-generation sequencing and quantitative methylation-specific PCR (qMSP). RESULTS Genome-wide methylation profiling was conducted to identify DMRs distinguishing HCC tumours from peritumoural tissues and healthy plasmas. The twenty most effective DMRs were verified and incorporated into a multilocus qMSP assay (HepaAiQ). The HepaAiQ model was trained to separate 293 HCC patients (Barcelona Clinic Liver Cancer (BCLC) stage 0/A, 224) from 266 controls including chronic hepatitis B (CHB) or liver cirrhosis (LC) (CHB/LC, 96), benign hepatic lesions (BHL, 23), and healthy controls (HC, 147). The model achieved an area under the curve (AUC) of 0.944 with a sensitivity of 86.0% in HCC and a specificity of 92.1% in controls. Blind validation of the HepaAiQ model in a cohort of 523 participants resulted in an AUC of 0.940 with a sensitivity of 84.4% in 205 HCC cases (BCLC stage 0/A, 167) and a specificity of 90.3% in 318 controls (CHB/LC, 100; BHL, 102; HC, 116). When evaluated in an independent test set, the HepaAiQ model exhibited a sensitivity of 70.8% in 65 HCC patients at BCLC stage 0/A and a specificity of 89.5% in 124 patients with CHB/LC. Moreover, HepaAiQ model was assessed in paired pre- and postoperative plasma samples from 103 HCC patients and correlated with 2-year patient outcomes. Patients with high postoperative HepaAiQ score showed a higher recurrence risk (Hazard ratio, 3.33, p < .001). CONCLUSIONS HepaAiQ, a noninvasive qMSP assay, was developed to accurately measure HCC-specific DMRs and shows great potential for the diagnosis, detection and prognosis of HCC, benefiting at-risk populations.
Collapse
Affiliation(s)
- De-Zhen Guo
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Ao Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Ying-Chao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
| | | | - Hui Wang
- Singlera Genomics Ltd., Shanghai, China
| | - Xiang-Lei Xing
- Biliary Tract Surgery Department IV, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Shi-Yu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Jian-Wen Cheng
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | | | | | | | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Chen
- XiangYa Medical Laboratory, Central South University, Changsha, Hunan, China
| | - Zhi-Xi Su
- Singlera Genomics Ltd., Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Rui Liu
- Singlera Genomics Ltd., Shanghai, China
| | - Xiao-Long Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Xin-Rong Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| |
Collapse
|
19
|
Lin S, Wang S, Xu B. Fragmentation patterns of cell-free DNA and somatic mutations in the urine of metastatic breast cancer patients. J Cancer Res Ther 2024; 20:563-569. [PMID: 38454812 DOI: 10.4103/jcrt.jcrt_1359_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/08/2023] [Indexed: 03/09/2024]
Abstract
BACKGROUND Urinary cell-free deoxyribonucleic acid (DNA) (ucfDNA) holds promise as a biomarker; however, its potential remains largely unexplored. We examined the fragmentation pattern of ucfDNA and identified somatic mutations within urine samples from metastatic breast cancer (MBC) patients. METHODS Urine and blood specimens were collected before treatment from 45 MBC patients and posttreatment urine samples from 16 of the 45 patients at the China National Cancer Center. Somatic mutations and tumor mutational burden (TMB) in the urine and plasma of 10 patients were analyzed by next-generation sequencing (NGS). Fragmentation patterns of cfDNA were displayed using electropherograms. Differences in the extracted amount of cfDNA, length of cfDNA fragments, and TMB between urine and plasma were compared using a Wilcoxon test. RESULTS The fragmentation patterns of ucfDNA were categorized as follows: (1) profile A (n = 26) containing a short peak (100-200 bp) and a long peak (>1500 bp); (2) profile B (n = 8) containing only a long peak; and (3) profile C (n = 11) containing flat pattern. For profile A patients, the short-peaked ucfDNA circulating in the bloodstream was much shorter compared with plasma cfDNA (149 vs. 171 bp, Wilcoxon test, P = 0.023). The fragmentation patterns in lung metastasis patients exhibited a higher propensity toward profile C ( P = 0.002). After treatment, 87.5% of the patients exhibited consistent fragmentation patterns. The concordance rate for somatic mutations in the plasma and urine was 30%, and the median TMB of urine and plasma was not significantly different. CONCLUSIONS This study established a fragmentation pattern for ucfDNA and detected somatic mutations in the urine of MBC patients. These results suggest the potential application of ucfDNA as a biomarker for MBC.
Collapse
Affiliation(s)
- Shaoyan Lin
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Shusen Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
20
|
Lehrich BM, Zhang J, Monga SP, Dhanasekaran R. Battle of the biopsies: Role of tissue and liquid biopsy in hepatocellular carcinoma. J Hepatol 2024; 80:515-530. [PMID: 38104635 PMCID: PMC10923008 DOI: 10.1016/j.jhep.2023.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/27/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
The diagnosis and management of hepatocellular carcinoma (HCC) have improved significantly in recent years. With the introduction of immunotherapy-based combination therapy, there has been a notable expansion in treatment options for patients with unresectable HCC. Simultaneously, innovative molecular tests for early detection and management of HCC are emerging. This progress prompts a key question: as liquid biopsy techniques rise in prominence, will they replace traditional tissue biopsies, or will both techniques remain relevant? Given the ongoing challenges of early HCC detection, including issues with ultrasound sensitivity, accessibility, and patient adherence to surveillance, the evolution of diagnostic techniques is more relevant than ever. Furthermore, the accurate stratification of HCC is limited by the absence of reliable biomarkers which can predict response to therapies. While the advantages of molecular diagnostics are evident, their potential has not yet been fully harnessed, largely because tissue biopsies are not routinely performed for HCC. Liquid biopsies, analysing components such as circulating tumour cells, DNA, and extracellular vesicles, provide a promising alternative, though they are still associated with challenges related to sensitivity, cost, and accessibility. The early results from multi-analyte liquid biopsy panels are promising and suggest they could play a transformative role in HCC detection and management; however, comprehensive clinical validation is still ongoing. In this review, we explore the challenges and potential of both tissue and liquid biopsy, highlighting that these diagnostic methods, while distinct in their approaches, are set to jointly reshape the future of HCC management.
Collapse
Affiliation(s)
- Brandon M Lehrich
- Department of Pathology and Pittsburgh Liver Institute, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Josephine Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Staford, CA, 94303, USA
| | - Satdarshan P Monga
- Department of Pathology and Pittsburgh Liver Institute, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| | - Renumathy Dhanasekaran
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Staford, CA, 94303, USA.
| |
Collapse
|
21
|
Du S, Cao K, Yan Y, Wang Y, Wang Z, Lin D. Developments and current status of cell-free DNA in the early detection and management of hepatocellular carcinoma. J Gastroenterol Hepatol 2024; 39:231-244. [PMID: 37990622 DOI: 10.1111/jgh.16416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023]
Abstract
Nowadays, hepatocellular carcinoma (HCC) is still a major threat to human health globally, with a disappointing prognosis. Regular monitoring of patients at high risk, utilizing abdominal ultrasonography combined with alpha-fetoprotein (AFP) serum analysis, enables the early detection of potentially treatable tumors. However, the approach has limitations due to its lack of sensitivity. Meanwhile, the current standard procedure for obtaining a tumor biopsy in cases of HCC is invasive and lacks the ability to assess the dynamic progression of cancer or account for tumor heterogeneity. Hence, there is a pressing need to develop non-invasive, highly sensitive biomarkers for HCC which can improve the accuracy of early diagnosis, assess treatment response and accurately predict the prognosis. In contrast to the conventional method of tissue biopsy, liquid biopsy offers a non-invasive approach that can be readily repeated. As a liquid biopsy approach, the analysis of cell-free DNA (cfDNA) offers real-time insights that can accurately portray the tumor burden and provide a comprehensive depiction of the genetic profile associated with HCC. In this review, we present a comprehensive summary of the recent research findings pertaining to the significance and potential practicality of cfDNA analysis in the early detection and effective management of HCC.
Collapse
Affiliation(s)
- Sihao Du
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ke Cao
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yadong Yan
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yupeng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenshun Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dongdong Lin
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Li X, Liu T, Bacchiocchi A, Li M, Cheng W, Wittkop T, Mendez F, Wang Y, Tang P, Yao Q, Bosenberg MW, Sznol M, Yan Q, Faham M, Weng L, Halaban R, Jin H, Hu Z. Ultra-sensitive molecular residual disease detection through whole genome sequencing with single-read error correction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.13.24301070. [PMID: 38260271 PMCID: PMC10802755 DOI: 10.1101/2024.01.13.24301070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
While whole genome sequencing (WGS) of cell-free DNA (cfDNA) holds enormous promise for molecular residual disease (MRD) detection, its performance is limited by WGS error rate. Here we introduce AccuScan, an efficient cfDNA WGS technology that enables genome-wide error correction at single read level, achieving an error rate of 4.2×10 -7 , which is about two orders of magnitude lower than a read-centric de-noising method. When applied to MRD detection, AccuScan demonstrated analytical sensitivity down to 10 -6 circulating tumor allele fraction at 99% sample level specificity. In colorectal cancer, AccuScan showed 90% landmark sensitivity for predicting relapse. It also showed robust MRD performance with esophageal cancer using samples collected as early as 1 week after surgery, and predictive value for immunotherapy monitoring with melanoma patients. Overall, AccuScan provides a highly accurate WGS solution for MRD, empowering circulating tumor DNA detection at parts per million range without high sample input nor personalized reagents. One Sentence Summary AccuScan showed remarkable ultra-low limit of detection with a short turnaround time, low sample requirement and a simple workflow for MRD detection.
Collapse
|
23
|
Feng Z, Peng F, Xie F, Liu Y, Zhang H, Ma J, Xing J, Guo X. Comparison of capture-based mtDNA sequencing performance between MGI and illumina sequencing platforms in various sample types. BMC Genomics 2024; 25:41. [PMID: 38191319 PMCID: PMC10773069 DOI: 10.1186/s12864-023-09938-6] [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: 08/09/2023] [Accepted: 12/24/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Mitochondrial genome abnormalities can lead to mitochondrial dysfunction, which in turn affects cellular biology and is closely associated with the development of various diseases. The demand for mitochondrial DNA (mtDNA) sequencing has been increasing, and Illumina and MGI are two commonly used sequencing platforms for capture-based mtDNA sequencing. However, there is currently no systematic comparison of mtDNA sequencing performance between these two platforms. To address this gap, we compared the performance of capture-based mtDNA sequencing between Illumina's NovaSeq 6000 and MGI's DNBSEQ-T7 using tissue, peripheral blood mononuclear cell (PBMC), formalin-fixed paraffin-embedded (FFPE) tissue, plasma, and urine samples. RESULTS Our analysis indicated a high degree of consistency between the two platforms in terms of sequencing quality, GC content, and coverage. In terms of data output, DNBSEQ-T7 showed higher rates of clean data and duplication compared to NovaSeq 6000. Conversely, the amount of mtDNA data obtained by per gigabyte sequencing data was significantly lower in DNBSEQ-T7 compared to NovaSeq 6000. In terms of detection mtDNA copy number, both platforms exhibited good consistency in all sample types. When it comes to detection of mtDNA mutations in tissue, FFPE, and PBMC samples, the two platforms also showed good consistency. However, when detecting mtDNA mutations in plasma and urine samples, significant differenceof themutation number detected was observed between the two platforms. For mtDNA sequencing of plasma and urine samples, a wider range of DNA fragment size distribution was found in NovaSeq 6000 when compared to DNBSEQ-T7. Additionally, two platforms exhibited different characteristics of mtDNA fragment end preference. CONCLUSIONS In summary, the two platforms generally showed good consistency in capture-based mtDNA sequencing. However, it is necessary to consider the data preferences generated by two sequencing platforms when plasma and urine samples were analyzed.
Collapse
Affiliation(s)
- Zehui Feng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Fan Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Fanfan Xie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
- Department of Obstetrics and Gynecology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Liu
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Huanqin Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Jing Ma
- Yanbian University Medical College, Yanji, 133002, China
| | - Jinliang Xing
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xu Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
24
|
Wang AE, Leven EA, Grinspan LT, Villanueva A. Novel biomarkers and strategies for HCC diagnosis and care. Clin Liver Dis (Hoboken) 2024; 23:e0152. [PMID: 38707238 PMCID: PMC11068134 DOI: 10.1097/cld.0000000000000152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/04/2024] [Indexed: 05/07/2024] Open
Affiliation(s)
- Allison E. Wang
- Department of Medicine, Division of Gastroenterology, Mount Sinai Morningside-West, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily A. Leven
- Department of Medicine, Division of Gastroenterology, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren T. Grinspan
- Department of Medicine, Division of Liver Diseases, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Augusto Villanueva
- Department of Medicine, Divisions of Liver Disease and Hematology/Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
25
|
Xing X, Cai L, Ouyang J, Wang F, Li Z, Liu M, Wang Y, Zhou Y, Hu E, Huang C, Wu L, Liu J, Liu X. Proteomics-driven noninvasive screening of circulating serum protein panels for the early diagnosis of hepatocellular carcinoma. Nat Commun 2023; 14:8392. [PMID: 38110372 PMCID: PMC10728065 DOI: 10.1038/s41467-023-44255-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
Early diagnosis of hepatocellular carcinoma (HCC) lacks highly sensitive and specific protein biomarkers. Here, we describe a staged mass spectrometry (MS)-based discovery-verification-validation proteomics workflow to explore serum proteomic biomarkers for HCC early diagnosis in 1002 individuals. Machine learning model determined as P4 panel (HABP2, CD163, AFP and PIVKA-II) clearly distinguish HCC from liver cirrhosis (LC, AUC 0.979, sensitivity 0.925, specificity 0.915) and healthy individuals (HC, AUC 0.992, sensitivity 0.975, specificity 1.000) in an independent validation cohort, outperforming existing clinical prediction strategies. Furthermore, the P4 panel can accurately predict LC to HCC conversion (AUC 0.890, sensitivity 0.909, specificity 0.877) with predicting HCC at a median of 11.4 months prior to imaging in prospective external validation cohorts (No.: Keshen 2018_005_02 and NCT03588442). These results suggest that proteomics-driven serum biomarker discovery provides a valuable reference for the liquid biopsy, and has great potential to improve early diagnosis of HCC.
Collapse
Affiliation(s)
- Xiaohua Xing
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Linsheng Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
- Department of Hepatopancreatobiliary Surgery, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuzhou, 350000, China
| | - Jiahe Ouyang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Fei Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Zongman Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Mingxin Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Yingchao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Yang Zhou
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - En Hu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China
| | - Changli Huang
- Department of Hepatopancreatobiliary Surgery, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuzhou, 350000, China
| | - Liming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China.
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| | - Jingfeng Liu
- Department of Hepatopancreatobiliary Surgery, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuzhou, 350000, China.
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, China.
| |
Collapse
|
26
|
Chen L, Ma R, Luo C, Xie Q, Ning X, Sun K, Meng F, Zhou M, Sun J. Noninvasive early differential diagnosis and progression monitoring of ovarian cancer using the copy number alterations of plasma cell-free DNA. Transl Res 2023; 262:12-24. [PMID: 37499745 DOI: 10.1016/j.trsl.2023.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/20/2023] [Accepted: 07/23/2023] [Indexed: 07/29/2023]
Abstract
Ovarian cancer (OV) is the most lethal gynecological malignancy and requires improved early detection methods and more effective intervention to achieve a better prognosis. The lack of sensitive and noninvasive biomarkers with clinical utility remains a challenge. Here, we conducted a genome-wide copy number variation (CNV) profiling analysis using low-coverage whole genome sequencing (LC-WGS) of plasma cfDNA in patients with nonmalignant and malignant ovarian tumors and identified 10 malignancy-specific and 12 late-stage-specific CNV markers from plasma cfDNA LC-WGS data. Concordance analysis indicated a significant correlation of identified CNV markers between CNV profiles of plasma cfDNA and tissue DNA (Pearson's r = 0.64, P = 0.006 for the TCGA cohort and r = 0.51, P = 0.04 for the Dariush cohort). By leveraging these specific CNV markers and machine learning algorithms, we developed robust predictive models showing excellent performance in distinguishing between malignant and nonmalignant ovarian tumors with F1-scores of 0.90 and ranging from 0.75 to 0.99, and prediction accuracy of 0.89 and ranging from 0.66 to 0.98, respectively, as well as between early- and late-stage ovarian tumors with F1-scores of 0.84 and ranging from 0.61 to 1.00, and prediction accuracy of 0.82 and ranging from 0.63 to 0.96 in our institute cohort and other external validation cohorts. Furthermore, we also discovered and validated certain CNV features associated with survival outcomes and platinum-based chemotherapy response in multicenter cohorts. In conclusion, our study demonstrated the clinical utility of CNV profiling in plasma cfDNA using LC-WGS as a cost-effective and accessible liquid biopsy for OV.
Collapse
Affiliation(s)
- Lu Chen
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, P. R. China; School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou P. R. China
| | - Rong Ma
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Chang Luo
- Department of Birth Control, Red Cross Central Hospital of Harbin, Harbin, P. R. China
| | - Qin Xie
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Xin Ning
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Kaidi Sun
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, P. R. China
| | - Fanling Meng
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, P. R. China.
| | - Meng Zhou
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou P. R. China.
| | - Jie Sun
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou P. R. China.
| |
Collapse
|
27
|
Lu Z, Chen Y, Liu D, Jiao X, Liu C, Wang Y, Zhang Z, Jia K, Gong J, Yang Z, Shen L. The landscape of cancer research and cancer care in China. Nat Med 2023; 29:3022-3032. [PMID: 38087112 DOI: 10.1038/s41591-023-02655-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/19/2023] [Indexed: 12/18/2023]
Abstract
The rising cancer incidence rate in China poses a substantial public health concern, although there have been remarkable improvements in the country's cancer mortality and survival rates. In this Review, we outline the current landscape and future directions of cancer care and research in China. We discuss national screening programs and strategies for cancer detection and delve into the evolving landscape of cancer care, emphasizing the adoption of multidisciplinary, comprehensive treatment and precision oncology. Additionally, we examine changes in drug research and development policies that have enabled approval of new drugs. Finally, we look to the future, highlighting key priorities and identifying gaps. Effectively addressing challenges and seizing opportunities associated with cancer research in China will enable the development of targeted approaches to alleviate the global burden of cancer.
Collapse
Affiliation(s)
- Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xi Jiao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Chang Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yakun Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zizhen Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Keren Jia
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhimin Yang
- National Center for Drug Evaluation, National Medical Products Administration, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
28
|
Liu Y, Jin H, Liu H. Identification of T-cell exhaustion-related gene signature for predicting prognosis in glioblastoma multiforme. J Cell Mol Med 2023; 27:3503-3513. [PMID: 37635346 PMCID: PMC10660619 DOI: 10.1111/jcmm.17927] [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: 03/29/2023] [Revised: 05/16/2023] [Accepted: 05/26/2023] [Indexed: 08/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly malignant primary brain tumour with a poor prognosis in adults. Identifying biomarkers that can aid in the molecular classification and risk stratification of GBM is critical. Here, we conducted a transcriptional profiling analysis of T-cell immunity in the tumour microenvironment of GBM patients and identified two novel T cell exhaustion (TEX)-related GBM subtypes (termed TEX-C1 and TEX-C2) using the consensus clustering. Our multi-omics analysis revealed distinct immunological, molecular and clinical characteristics for these two subtypes. Specifically, the TEX-C1 subtype had higher infiltration levels of immune cells and expressed higher levels of immune checkpoint molecules than the TEX-C2 subtype. Functional analysis revealed that upregulated genes in the TEX-C1 subtype were significantly enriched in immune response and signal transduction pathways, and upregulated genes in the TEX-C2 subtype were predominantly associated with cell fate and nervous system development pathways. Notably, patients with activated T-cell activity status in the TEX-C1 subgroup demonstrated a significantly worse prognosis than those with severe T cell exhaustion status in the TEX-C2 subgroup. Finally, we proposed a machine-learning-derived novel gene signature comprising 12 TEX-related genes (12TexSig) to indicate tumour subtyping. In the TCGA cohort, the 12TexSig demonstrated the ability to accurately predict the prognosis of GBM patients, and this prognostic value was further confirmed in two independent external cohorts. Taken together, our results suggest that the TEX-derived subtyping and gene signature has the potential to serve as a clinically helpful biomarker for guiding the management of GBM patients, pending further prospective validation.
Collapse
Affiliation(s)
- Yue‐hui Liu
- Department of NeurologyAffiliated Hospital of Inner Mongolia Minzu UniversityTongliaoChina
| | - Hong‐quan Jin
- Department of NeurologyAffiliated Hospital of Inner Mongolia Minzu UniversityTongliaoChina
| | - Hai‐ping Liu
- College of Life Science and Food EngineeringInner Mongolia Minzu UniversityTongliaoChina
| |
Collapse
|
29
|
Bie F, Wang Z, Li Y, Guo W, Hong Y, Han T, Lv F, Yang S, Li S, Li X, Nie P, Xu S, Zang R, Zhang M, Song P, Feng F, Duan J, Bai G, Li Y, Huai Q, Zhou B, Huang YS, Chen W, Tan F, Gao S. Multimodal analysis of cell-free DNA whole-methylome sequencing for cancer detection and localization. Nat Commun 2023; 14:6042. [PMID: 37758728 PMCID: PMC10533817 DOI: 10.1038/s41467-023-41774-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Multimodal epigenetic characterization of cell-free DNA (cfDNA) could improve the performance of blood-based early cancer detection. However, integrative profiling of cfDNA methylome and fragmentome has been technologically challenging. Here, we adapt an enzyme-mediated methylation sequencing method for comprehensive analysis of genome-wide cfDNA methylation, fragmentation, and copy number alteration (CNA) characteristics for enhanced cancer detection. We apply this method to plasma samples of 497 healthy controls and 780 patients of seven cancer types and develop an ensemble classifier by incorporating methylation, fragmentation, and CNA features. In the test cohort, our approach achieves an area under the curve value of 0.966 for overall cancer detection. Detection sensitivity for early-stage patients achieves 73% at 99% specificity. Finally, we demonstrate the feasibility to accurately localize the origin of cancer signals with combined methylation and fragmentation profiling of tissue-specific accessible chromatin regions. Overall, this proof-of-concept study provides a technical platform to utilize multimodal cfDNA features for improved cancer detection.
Collapse
Grants
- This work was supported by the National Key R&D Program of China (2021YFC2500900, Shugeng Gao), CAMS Initiative for Innovative Medicine (2021-I2M-1-015, Shugeng Gao), Central Health Research Key Projects (2022ZD17, Shugeng Gao).
- This work was supported by the National Key R&D Program of China (2021YFC2500400, Weizhi Chen).
- This work was supported by the CAMS Initiative for Innovative Medicine (2021-I2M-1-015, Fengwei Tan), CAMS Innovation Fund for Medical Sciences (2021-I2M-1-061, Fengwei Tan), and National Natural Science Foundation of China (81871885, Fengwei Tan).
Collapse
Affiliation(s)
- Fenglong Bie
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhijie Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yulong Li
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuanyuan Hong
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Tiancheng Han
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Fang Lv
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Shunli Yang
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Suxing Li
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Xi Li
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Peiyao Nie
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Shun Xu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - Ruochuan Zang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Moyan Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Feiyue Feng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianchun Duan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guangyu Bai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuan Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qilin Huai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bolun Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yu S Huang
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Weizhi Chen
- Genecast Biotechnology Co., Ltd., Wuxi, 214105, Jiangsu, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
30
|
Hu Y, Zhang X, Li Q, Zhou Q, Fang D, Lu Y. An immune and epigenetics-related scoring model and drug candidate prediction for hepatic carcinogenesis via dynamic network biomarker analysis and connectivity mapping. Comput Struct Biotechnol J 2023; 21:4619-4633. [PMID: 37817777 PMCID: PMC10561057 DOI: 10.1016/j.csbj.2023.09.030] [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: 07/31/2023] [Revised: 09/18/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high mortality. This study aimed to build a prognostic signature for HCC patients based on immune-related genes (IRGs) and epigenetics-related genes (EPGs). RNA-seq data from Gene Expression Omnibus were used for dynamic network biomarker (DNB) analysis to identify 56 candidate IRG-EPG-DNBs and their first-neighbor genes. These genes were screened using LASSO-Cox regression analysis to finally obtain five candidate genes-RNF2, YBX1, EZH2, CAD, and PSMD1-which constituted the prognostic signature panel. According to this panel, patients in The Cancer Genome Atlas and International Cancer Genome Consortium were divided into high- and low-risk groups. The prognosis, clinicopathological features, and immune cell infiltration significantly differed between the two risk groups. The prognostic ability of the signature panel and expression profiling were further validated using online databases. We used an independent cohort of patients to validate the expression profiles of the five genes using reverse transcription-PCR. CMap and CellMiner predicted four small molecule drug-protein pairs based on the five prognostic genes. Of them, two market drugs approved by the Food and Drug Administration (AT-13387 and KU-55933) have emerged as candidates for HCC study. This new signature panel may serve as a potential prognostic marker, engendering the possibility of novel personalized therapy with classification of HCC patients.
Collapse
Affiliation(s)
- Yuting Hu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xingli Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qingya Li
- Henan University of Chinese Medicine, Henan 450046, China
| | - Qianmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dongdong Fang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiyu Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
31
|
Lin PC, Hsu WY, Lee PY, Hsu SH, Chiou SS. Insights into Hepatocellular Carcinoma in Patients with Thalassemia: From Pathophysiology to Novel Therapies. Int J Mol Sci 2023; 24:12654. [PMID: 37628834 PMCID: PMC10454908 DOI: 10.3390/ijms241612654] [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: 06/26/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Thalassemia is a heterogeneous congenital hemoglobinopathy common in the Mediterranean region, Middle East, Indian subcontinent, and Southeast Asia with increasing incidence in Northern Europe and North America due to immigration. Iron overloading is one of the major long-term complications in patients with thalassemia and can lead to organ damage and carcinogenesis. Hepatocellular carcinoma (HCC) is one of the most common malignancies in both transfusion-dependent thalassemia (TDT) and non-transfusion-dependent thalassemia (NTDT). The incidence of HCC in patients with thalassemia has increased over time, as better chelation therapy confers a sufficiently long lifespan for the development of HCC. The mechanisms of iron-overloading-associated HCC development include the increased reactive oxygen species (ROS), inflammation cytokines, dysregulated hepcidin, and ferroportin metabolism. The treatment of HCC in patients with thalassemia was basically similar to those in general population. However, due to the younger age of HCC onset in thalassemia, regular surveillance for HCC development is mandatory in TDT and NTDT. Other supplemental therapies and experiences of novel treatments for HCC in the thalassemia population were also reviewed in this article.
Collapse
Affiliation(s)
- Pei-Chin Lin
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807378, Taiwan; (P.-C.L.); (W.-Y.H.); (P.-Y.L.)
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Wan-Yi Hsu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807378, Taiwan; (P.-C.L.); (W.-Y.H.); (P.-Y.L.)
| | - Po-Yi Lee
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807378, Taiwan; (P.-C.L.); (W.-Y.H.); (P.-Y.L.)
| | - Shih-Hsien Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Shyh-Shin Chiou
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807378, Taiwan; (P.-C.L.); (W.-Y.H.); (P.-Y.L.)
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Division of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807378, Taiwan
| |
Collapse
|
32
|
Su S, Xuan Y, Fan X, Bao H, Tang H, Lv X, Ren W, Chen F, Shao Y, Wang T, Wang L. Testing the generalizability of cfDNA fragmentomic features across different studies for cancer early detection. Genomics 2023; 115:110662. [PMID: 37270068 DOI: 10.1016/j.ygeno.2023.110662] [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: 02/07/2023] [Revised: 05/12/2023] [Accepted: 05/27/2023] [Indexed: 06/05/2023]
Abstract
cfDNA fragmentomic features have been used in cancer detection models; however, the generalizability of the models needs to be tested. We proposed a type of cfDNA fragmentomic feature named chromosomal arm-level fragment size distribution (ARM-FSD), evaluated and compared its performance and generalizability for lung cancer and pan-cancer detection with existing cfDNA fragmentomic features (as reference) by using cohorts from different institutions. The ARM-FSD lung cancer model outperformed the reference model by ∼10% when being tested by two external cohorts (AUC: 0.97 vs. 0.86; 0.87 vs. 0.76). For pan-cancer detection, the performance of the ARM-FSD based model is consistently higher than the reference (AUC: 0.88 vs. 0.75, 0.98 vs. 0.63) in a pan-cancer and a lung cancer external validation cohort, indicating that ARM-FSD model produces stable performance across multiple cohorts. Our study reveals ARM-FSD based models have a higher generalizability, and highlights the necessity of cross-study validation for predictive model development.
Collapse
Affiliation(s)
- Shu Su
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Medical School of Nanjing University & Clinical Center Institute of Nanjing University, Nanjing, China
| | - Yulong Xuan
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiaojun Fan
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing 210032, Jiangsu, China
| | - Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing 210032, Jiangsu, China
| | - Haimeng Tang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing 210032, Jiangsu, China
| | - Xin Lv
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Medical School of Nanjing University & Clinical Center Institute of Nanjing University, Nanjing, China
| | - Wei Ren
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Medical School of Nanjing University & Clinical Center Institute of Nanjing University, Nanjing, China
| | - Fangjun Chen
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Medical School of Nanjing University & Clinical Center Institute of Nanjing University, Nanjing, China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing 210032, Jiangsu, China; School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Tao Wang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Lifeng Wang
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Medical School of Nanjing University & Clinical Center Institute of Nanjing University, Nanjing, China.
| |
Collapse
|
33
|
Lee YT, Fujiwara N, Yang JD, Hoshida Y. Risk stratification and early detection biomarkers for precision HCC screening. Hepatology 2023; 78:319-362. [PMID: 36082510 PMCID: PMC9995677 DOI: 10.1002/hep.32779] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 12/08/2022]
Abstract
Hepatocellular carcinoma (HCC) mortality remains high primarily due to late diagnosis as a consequence of failed early detection. Professional societies recommend semi-annual HCC screening in at-risk patients with chronic liver disease to increase the likelihood of curative treatment receipt and improve survival. However, recent dynamic shift of HCC etiologies from viral to metabolic liver diseases has significantly increased the potential target population for the screening, whereas annual incidence rate has become substantially lower. Thus, with the contemporary HCC etiologies, the traditional screening approach might not be practical and cost-effective. HCC screening consists of (i) definition of rational at-risk population, and subsequent (ii) repeated application of early detection tests to the population at regular intervals. The suboptimal performance of the currently available HCC screening tests highlights an urgent need for new modalities and strategies to improve early HCC detection. In this review, we overview recent developments of clinical, molecular, and imaging-based tools to address the current challenge, and discuss conceptual framework and approaches of their clinical translation and implementation. These encouraging progresses are expected to transform the current "one-size-fits-all" HCC screening into individualized precision approaches to early HCC detection and ultimately improve the poor HCC prognosis in the foreseeable future.
Collapse
Affiliation(s)
- Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California
| | - Naoto Fujiwara
- Liver Tumor Translational Research Program, Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California; Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, Los Angeles, California; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yujin Hoshida
- Liver Tumor Translational Research Program, Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
34
|
Wang S, Xia Z, You J, Gu X, Meng F, Chen P, Tang W, Bao H, Zhang J, Wu X, Shao Y, Wang J, Zuo X, Xu L, Yin R. Enhanced Detection of Landmark Minimal Residual Disease in Lung Cancer Using Cell-free DNA Fragmentomics. CANCER RESEARCH COMMUNICATIONS 2023; 3:933-942. [PMID: 37377889 PMCID: PMC10228550 DOI: 10.1158/2767-9764.crc-22-0363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/07/2022] [Accepted: 05/09/2023] [Indexed: 06/29/2023]
Abstract
Currently, approximately 30%-55% of the patients with non-small cell lung cancer (NSCLC) develop recurrence due to minimal residual disease (MRD) after receiving surgical resection of the tumor. This study aims to develop an ultrasensitive and affordable fragmentomic assay for MRD detection in patients with NSCLC. A total of 87 patients with NSCLC, who received curative surgical resections (23 patients relapsed during follow-up), enrolled in this study. A total of 163 plasma samples, collected at 7 days and 6 months postsurgical, were used for both whole-genome sequencing (WGS) and targeted sequencing. WGS-based cell-free DNA (cfDNA) fragment profile was used to fit regularized Cox regression models, and leave-one-out cross-validation was further used to evaluate models' performance. The models showed excellent performances in detecting patients with a high risk of recurrence. At 7 days postsurgical, the high-risk patients detected by our model showed an increased risk of 4.6 times, while the risk increased to 8.3 times at 6 months postsurgical. These fragmentomics determined higher risk compared with the targeted sequencing-based circulating mutations both at 7 days and 6 months postsurgical. The overall sensitivity for detecting patients with recurrence reached 78.3% while using both fragmentomics and mutation results from 7 days and 6 months postsurgical, which increased from the 43.5% sensitivity by using only the circulating mutations. The fragmentomics showed great sensitivity in predicting patient recurrence compared with the traditional circulating mutation, especially after the surgery for early-stage NSCLC, therefore exhibiting great potential to guide adjuvant therapeutics. Significance The circulating tumor DNA mutation-based approach shows limited performance in MRD detection, especially for landmark MRD detection at an early-stage cancer after surgery. Here, we describe a cfDNA fragmentomics-based method in MRD detection of resectable NSCLC using WGS, and the cfDNA fragmentomics showed a great sensitivity in predicting prognosis.
Collapse
Affiliation(s)
- Siwei Wang
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
| | - Zhijun Xia
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
| | - Jing You
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
| | - Xiaolan Gu
- Department of Anesthesiology, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
| | - Fanchen Meng
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
| | - Peng Chen
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
| | - Wanxiangfu Tang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, P.R. China
| | - Hua Bao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, P.R. China
| | - Jingyuan Zhang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, P.R. China
| | - Xue Wu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, P.R. China
| | - Yang Shao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, P.R. China
| | - Jie Wang
- Department of Science and Technology, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
- Biobank of Lung Cancer, Jiangsu Biobank of Clinical Resources, Nanjing, P.R. China
| | - Xianglin Zuo
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, P.R. China
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
- Department of Science and Technology, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P.R. China
- Biobank of Lung Cancer, Jiangsu Biobank of Clinical Resources, Nanjing, P.R. China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
35
|
Hu S, Liu Y, Yang Q, Chen L, Chai H, Xiao M, Qi C, Qiu W. Liquid biopsy using cell-free DNA in the early diagnosis of hepatocellular carcinoma. Invest New Drugs 2023:10.1007/s10637-023-01363-6. [PMID: 37099161 DOI: 10.1007/s10637-023-01363-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 04/17/2023] [Indexed: 04/27/2023]
Abstract
Hepatocellular carcinoma ranks fourth in cancer-related causes of death worldwide and second in China. Patients with hepatocellular carcinoma (HCC) at the early stage have a better prognosis compared to HCC patients at the late stage. Therefore, early screening for HCC is critical for clinical treatment decisions and improving the prognosis of patients. Ultrasound (US), computed tomography (CT), and serum alpha fetoprotein (AFP) have been used to screen HCC, but HCC is still difficult to be diagnosed in the early stage due to the low sensitivity of the above methods. It is urgent to find a method with high sensitivity and specificity for the early diagnosis of HCC. Liquid biopsy is a noninvasive detection method using blood or other bodily fluids. Cell-free DNA (cfDNA) and circulating tumor DNA (ctDNA) are important biomarkers for liquid biopsy. Recently, HCC screening methods using the application of cfDNA and ctDNA have become the hot spot of early HCC diagnostics. In this mini review, we summarize the latest research progress of liquid biopsy based on blood cfDNA in early screening of HCC.
Collapse
Affiliation(s)
- Shiqi Hu
- The oncology department, Xiangtan Central Hospital, Hunan, China
| | - Yaqin Liu
- The Medical Department, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Qidong Yang
- The Medical Department, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Lin Chen
- The Medical Department, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Huizi Chai
- The Medical Department, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Mingzhe Xiao
- The Medical Department, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Chuang Qi
- The Medical Department, The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Wei Qiu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, 130000, Jilin, China.
| |
Collapse
|
36
|
Brockley LJ, Souza VGP, Forder A, Pewarchuk ME, Erkan M, Telkar N, Benard K, Trejo J, Stewart MD, Stewart GL, Reis PP, Lam WL, Martinez VD. Sequence-Based Platforms for Discovering Biomarkers in Liquid Biopsy of Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:2275. [PMID: 37190212 PMCID: PMC10136462 DOI: 10.3390/cancers15082275] [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: 03/07/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Lung cancer detection and monitoring are hampered by a lack of sensitive biomarkers, which results in diagnosis at late stages and difficulty in tracking response to treatment. Recent developments have established liquid biopsies as promising non-invasive methods for detecting biomarkers in lung cancer patients. With concurrent advances in high-throughput sequencing technologies and bioinformatics tools, new approaches for biomarker discovery have emerged. In this article, we survey established and emerging biomarker discovery methods using nucleic acid materials derived from bodily fluids in the context of lung cancer. We introduce nucleic acid biomarkers extracted from liquid biopsies and outline biological sources and methods of isolation. We discuss next-generation sequencing (NGS) platforms commonly used to identify novel biomarkers and describe how these have been applied to liquid biopsy. We highlight emerging biomarker discovery methods, including applications of long-read sequencing, fragmentomics, whole-genome amplification methods for single-cell analysis, and whole-genome methylation assays. Finally, we discuss advanced bioinformatics tools, describing methods for processing NGS data, as well as recently developed software tailored for liquid biopsy biomarker detection, which holds promise for early diagnosis of lung cancer.
Collapse
Affiliation(s)
- Liam J. Brockley
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Vanessa G. P. Souza
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
- Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
| | - Aisling Forder
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Michelle E. Pewarchuk
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Melis Erkan
- Department of Pathology and Laboratory Medicine, IWK Health Centre, Halifax, NS B3K 6R8, Canada;
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
| | - Nikita Telkar
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
- British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Katya Benard
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Jessica Trejo
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Matt D. Stewart
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Greg L. Stewart
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Patricia P. Reis
- Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Wan L. Lam
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.P.S.); (A.F.); (M.E.P.); (N.T.); (K.B.); (J.T.); (M.D.S.); (G.L.S.); (W.L.L.)
| | - Victor D. Martinez
- Department of Pathology and Laboratory Medicine, IWK Health Centre, Halifax, NS B3K 6R8, Canada;
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
37
|
Moser T, Kühberger S, Lazzeri I, Vlachos G, Heitzer E. Bridging biological cfDNA features and machine learning approaches. Trends Genet 2023; 39:285-307. [PMID: 36792446 DOI: 10.1016/j.tig.2023.01.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 02/15/2023]
Abstract
Liquid biopsies (LBs), particularly using circulating tumor DNA (ctDNA), are expected to revolutionize precision oncology and blood-based cancer screening. Recent technological improvements, in combination with the ever-growing understanding of cell-free DNA (cfDNA) biology, are enabling the detection of tumor-specific changes with extremely high resolution and new analysis concepts beyond genetic alterations, including methylomics, fragmentomics, and nucleosomics. The interrogation of a large number of markers and the high complexity of data render traditional correlation methods insufficient. In this regard, machine learning (ML) algorithms are increasingly being used to decipher disease- and tissue-specific signals from cfDNA. Here, we review recent insights into biological ctDNA features and how these are incorporated into sophisticated ML applications.
Collapse
Affiliation(s)
- Tina Moser
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria
| | - Stefan Kühberger
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria
| | - Isaac Lazzeri
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria
| | - Georgios Vlachos
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria.
| |
Collapse
|
38
|
Tan WY, Sharma A, Das P, Ahuja N. Early Detection of Cancers in the Era of Precision Oncology. Curr Opin Oncol 2023; 35:115-124. [PMID: 36721896 DOI: 10.1097/cco.0000000000000931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW The increasing global incidence of cancer demands innovative cancer detection modalities. The current population-based early cancer detection approaches focus on several major types of cancers (breast, prostate, cervical, lung and colon) at their early stages, however, they generally do not target high-risk individuals at precancerous stages. RECENT FINDINGS Some cancers, such as pancreatic cancer, are challenging to detect in their early stages. Therefore, there is a pressing need for improved, accessible, noninvasive, and cost-effective early detection methods. Harnessing cell-free-based biomarker-driven strategies paves a new era of precision diagnosis for multicancer early detection. The majority of these tests are in the early stages and expensive, but these approaches are expected to become cost sensitive in the near future. SUMMARY This review provides an overview of early cancer detection strategies, highlighting the methods, challenges, and issues to be addressed to revolutionize and improve global early cancer detection.
Collapse
Affiliation(s)
| | - Anup Sharma
- Yale School of Medicine, Department of Surgery
| | | | - Nita Ahuja
- Yale School of Medicine, Department of Surgery
- Yale School of Medicine, Department of Pathology
- Yale School of Medicine, Biological and Biomedical Sciences Program (BBS), Yale University, New Haven, Connecticut, USA
| |
Collapse
|
39
|
Labiano I, Huerta AE, Arrazubi V, Hernandez-Garcia I, Mata E, Gomez D, Arasanz H, Vera R, Alsina M. State of the Art: ctDNA in Upper Gastrointestinal Malignancies. Cancers (Basel) 2023; 15:1379. [PMID: 36900172 PMCID: PMC10000247 DOI: 10.3390/cancers15051379] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Circulating tumor DNA (ctDNA) has emerged as a promising non-invasive source to characterize genetic alterations related to the tumor. Upper gastrointestinal cancers, including gastroesophageal adenocarcinoma (GEC), biliary tract cancer (BTC) and pancreatic ductal adenocarcinoma (PADC) are poor prognostic malignancies, usually diagnosed at advanced stages when no longer amenable to surgical resection and show a poor prognosis even for resected patients. In this sense, ctDNA has emerged as a promising non-invasive tool with different applications, from early diagnosis to molecular characterization and follow-up of tumor genomic evolution. In this manuscript, novel advances in the field of ctDNA analysis in upper gastrointestinal tumors are presented and discussed. Overall, ctDNA analyses can help in early diagnosis, outperforming current diagnostic approaches. Detection of ctDNA prior to surgery or active treatment is also a prognostic marker that associates with worse survival, while ctDNA detection after surgery is indicative of minimal residual disease, anticipating in some cases the imaging-based detection of progression. In the advanced setting, ctDNA analyses characterize the genetic landscape of the tumor and identify patients for targeted-therapy approaches, and studies show variable concordance levels with tissue-based genetic testing. In this line, several studies also show that ctDNA serves to follow responses to active therapy, especially in targeted approaches, where it can detect multiple resistance mechanisms. Unfortunately, current studies are still limited and observational. Future prospective multi-center and interventional studies, carefully designed to assess the value of ctDNA to help clinical decision-making, will shed light on the real applicability of ctDNA in upper gastrointestinal tumor management. This manuscript presents a review of the evidence available in this field up to date.
Collapse
Affiliation(s)
- Ibone Labiano
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
| | - Ana Elsa Huerta
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
| | - Virginia Arrazubi
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Irene Hernandez-Garcia
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Elena Mata
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - David Gomez
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Hugo Arasanz
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Ruth Vera
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Maria Alsina
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| |
Collapse
|
40
|
Pan A, Truong TN, Su YH, Dao DY. Circulating Biomarkers for the Early Diagnosis and Management of Hepatocellular Carcinoma with Potential Application in Resource-Limited Settings. Diagnostics (Basel) 2023; 13:676. [PMID: 36832164 PMCID: PMC9954913 DOI: 10.3390/diagnostics13040676] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is among the world's third most lethal cancers. In resource-limited settings (RLS), up to 70% of HCCs are diagnosed with limited curative treatments at an advanced symptomatic stage. Even when HCC is detected early and resection surgery is offered, the post-operative recurrence rate after resection exceeds 70% in five years, of which about 50% occur within two years of surgery. There are no specific biomarkers addressing the surveillance of HCC recurrence due to the limited sensitivity of the available methods. The primary goal in the early diagnosis and management of HCC is to cure disease and improve survival, respectively. Circulating biomarkers can be used as screening, diagnostic, prognostic, and predictive biomarkers to achieve the primary goal of HCC. In this review, we highlighted key circulating blood- or urine-based HCC biomarkers and considered their potential applications in resource-limited settings, where the unmet medical needs of HCC are disproportionately highly significant.
Collapse
Affiliation(s)
- Annabelle Pan
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Thai N. Truong
- Department of Internal Medicine, Campus in Thanh Hoa, Hanoi Medical University, Thanh Hoa 40000, Vietnam
| | - Ying-Hsiu Su
- Department of Translational Medical Science, The Baruch S. Blumberg Institute, Doylestown, PA 18902, USA
| | - Doan Y Dao
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Center of Excellence for Liver Disease in Vietnam, Johns Hopkins University of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
41
|
Qi T, Pan M, Shi H, Wang L, Bai Y, Ge Q. Cell-Free DNA Fragmentomics: The Novel Promising Biomarker. Int J Mol Sci 2023; 24:1503. [PMID: 36675018 PMCID: PMC9866579 DOI: 10.3390/ijms24021503] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
Cell-free DNA molecules are released into the plasma via apoptotic or necrotic events and active release mechanisms, which carry the genetic and epigenetic information of its origin tissues. However, cfDNA is the mixture of various cell fragments, and the efficient enrichment of cfDNA fragments with diagnostic value remains a great challenge for application in the clinical setting. Evidence from recent years shows that cfDNA fragmentomics' characteristics differ in normal and diseased individuals without the need to distinguish the source of the cfDNA fragments, which makes it a promising novel biomarker. Moreover, cfDNA fragmentomics can identify tissue origins by inferring epigenetic information. Thus, further insights into the fragmentomics of plasma cfDNA shed light on the origin and fragmentation mechanisms of cfDNA during physiological and pathological processes in diseases and enhance our ability to take the advantage of plasma cfDNA as a molecular diagnostic tool. In this review, we focus on the cfDNA fragment characteristics and its potential application, such as fragment length, end motifs, jagged ends, preferred end coordinates, as well as nucleosome footprints, open chromatin region, and gene expression inferred by the cfDNA fragmentation pattern across the genome. Furthermore, we summarize the methods for deducing the tissue of origin by cfDNA fragmentomics.
Collapse
Affiliation(s)
- Ting Qi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Pan
- School of Medicine, Southeast University, Nanjing 210097, China
| | - Huajuan Shi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Liangying Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yunfei Bai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Qinyu Ge
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
42
|
Zhang D, Huo L, Pan Y, Yang Z, Zeng H, Wang X, Chen J, Wang J, Zhang Y, Zhou Z, Chen M, Hu D. A Systemic Inflammation Response Score for Prognostic Prediction in Hepatocellular Carcinoma Patients After Hepatectomy. J Inflamm Res 2022; 15:6869-6881. [PMID: 36600994 PMCID: PMC9807220 DOI: 10.2147/jir.s397375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Purpose To investigate the value of preoperative systemic inflammation response (SIRS) score in predicting the prognosis of hepatocellular carcinoma (HCC) after hepatectomy. Patients and Methods The study analyzed 1001 patients with pathologically proven HCC who received curative resection at Sun Yat-sen University Cancer Center between March 2016 and May 2020. Patients were randomly divided into a training cohort (n = 751) and a validation cohort (n = 250). Clinicopathological characteristics were collected retrospectively. The SIRS score formula was based on the results of a multivariate cox analysis of hematological inflammation indexes in the training cohort. Then, a nomogram consisting of the SIRS score was constructed and the calibration plot, areas under the receiver operating characteristic (AUC) curve, and decision curve analysis (DCA) showed good predictive ability. Results Univariate and multivariate cox analysis revealed that the SIRS score is an independent prognostic factor for OS in HCC patients. A higher SIRS score was associated with a larger maximum lesion diameter, poor tumor differentiation, a greater possibility of vascular invasion, and a more advanced cancer stage. When the nomogram was used to predict 1-year, 3-year, and 5-year survival rates, the AUC in the training cohort was 0.763, 0.712, and 0.687, respectively; In the validation cohort, it was 0.715, 0.648, and 0.614, respectively. The AUC of this nomogram showed significantly better predictive performance than those of commonly used staging systems. Conclusion The preoperative SIRS score has good efficacy in predicting the prognosis of HCC patients undergoing hepatectomy, and nomograms based on the SIRS score can potentially guide individualized follow-up and adjuvant therapy.
Collapse
Affiliation(s)
- Deyao Zhang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Lanqing Huo
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Yangxun Pan
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Zhenyun Yang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Huilan Zeng
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Xin Wang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Jinbin Chen
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Juncheng Wang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Yaojun Zhang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Zhongguo Zhou
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Minshan Chen
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Correspondence: Minshan Chen; Dandan Hu, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510000, People’s Republic of China, Tel +86 13902241061; +86 18676630499, Fax +86 8734-3115; +86 8734-3115, Email ;
| | - Dandan Hu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China,Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| |
Collapse
|
43
|
He W, Xiao Y, Yan S, Zhu Y, Ren S. Cell-free DNA in the management of prostate cancer: Current status and future prospective. Asian J Urol 2022. [PMID: 37538150 PMCID: PMC10394290 DOI: 10.1016/j.ajur.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective With the escalating prevalence of prostate cancer (PCa) in China, there is an urgent demand for novel diagnostic and therapeutic approaches. Extensive investigations have been conducted on the clinical implementation of circulating free DNA (cfDNA) in PCa. This review aims to provide a comprehensive overview of the present state of cfDNA as a biomarker for PCa and to examine its merits and obstacles for future clinical utilization. Methods Relevant peer-reviewed manuscripts on cfDNA as a PCa marker were evaluated by PubMed search (2010-2022) to evaluate the roles of cfDNA in PCa diagnosis, prognosis, and prediction, respectively. Results cfDNA is primarily released from cells undergoing necrosis and apoptosis, allowing for non-invasive insight into the genomic, transcriptomic, and epigenomic alterations within various PCa disease states. Next-generation sequencing, among other detection methods, enables the assessment of cfDNA abundance, mutation status, fragment characteristics, and epigenetic modifications. Multidimensional analysis based on cfDNA can facilitate early detection of PCa, risk stratification, and treatment monitoring. However, standardization of cfDNA detection methods is still required to expedite its clinical application. Conclusion cfDNA provides a non-invasive, rapid, and repeatable means of acquiring multidimensional information from PCa patients, which can aid in guiding clinical decisions and enhancing patient management. Overcoming the application barriers of cfDNA necessitates increased data sharing and international collaboration.
Collapse
|
44
|
Improved anti-hepatocellular carcinoma effect by enhanced Co-delivery of Tim-3 siRNA and sorafenib via multiple pH triggered drug-eluting nanoparticles. Mater Today Bio 2022; 16:100350. [PMID: 35856043 PMCID: PMC9287642 DOI: 10.1016/j.mtbio.2022.100350] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/15/2022] [Accepted: 07/01/2022] [Indexed: 11/23/2022] Open
Abstract
Effective systemic treatment for hepatocellular carcinoma (HCC) remains urgently needed. Sorafenib is the first FDA-approved systemic treatment for HCC. However, individual HCC patents’ response to sorafenib varies greatly. How to enhance the anti-HCC effect of sorafenib is still a significant challenge. T cell immunoglobulin mucin-3 (Tim-3) is a newly identified immune checkpoint molecule and a promising target for HCC treatment. Herein, we developed a novel pH-triggered drug-eluting nanoparticle (CC@SR&SF@PP) for simultaneously delivery of Tim-3 siRNA and sorafenib to HCC in situ. By a single emulsification method, a representative HCC targeted-therapeutic drug sorafenib (SF) was encapsulated into the pH-triggered positive-charged mPEG5K-PAE10K (PP) nanoparticles, followed by condensing of negative-charged Tim-3 siRNA. Then, carboxymethyl chitosan (CMCS), an amphoteric polysaccharide with negative charge in the physiological pH and positive charge in the acidic environment of the tumor, was eventually adsorbed onto the surface of nanoparticles. This co-delivery nanoparticle rapidly and specifically accumulated in the tumor site of the liver and enhanced the targeted, specific and multiple release of siRNA and sorafenib. Enhanced Tim-3 siRNA transfected into tumor cells can not only directly inhibit the growth of tumor cells by knock down the expression Tim-3, but also induce the immune response and enhance the recruitment of cytotoxic T cells to kill tumor cells. The following pH-triggered sorafenib release from SF@PP NPs greatly inhibited the tumor proliferation and angiogenesis, resulting in remarkable tumor growth inhibition in a mouse hepatoma 22 (H22) orthotopic tumor model. Thus, co-delivery of Tim-3 siRNA and sorafenib via this novel pH triggered drug-eluting nanoparticle enhances their anti-tumor efficacy. We expect that such combination treatment strategy will have great potential in future clinical applications.
Collapse
|
45
|
Schlosser S, Tümen D, Volz B, Neumeyer K, Egler N, Kunst C, Tews HC, Schmid S, Kandulski A, Müller M, Gülow K. HCC biomarkers - state of the old and outlook to future promising biomarkers and their potential in everyday clinical practice. Front Oncol 2022; 12:1016952. [PMID: 36518320 PMCID: PMC9742592 DOI: 10.3389/fonc.2022.1016952] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/04/2022] [Indexed: 08/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and deadly tumors worldwide. Management of HCC depends on reliable biomarkers for screening, diagnosis, and monitoring of the disease, as well as predicting response towards therapy and safety. To date, imaging has been the established standard technique in the diagnosis and follow-up of HCC. However, imaging techniques have their limitations, especially in the early detection of HCC. Therefore, there is an urgent need for reliable, non/minimal invasive biomarkers. To date, alpha-fetoprotein (AFP) is the only serum biomarker used in clinical practice for the management of HCC. However, AFP is of relatively rather low quality in terms of specificity and sensitivity. Liquid biopsies as a source for biomarkers have become the focus of clinical research. Our review highlights alternative biomarkers derived from liquid biopsies, including circulating tumor cells, proteins, circulating nucleic acids, and exosomes, and their potential for clinical application. Using defined combinations of different biomarkers will open new perspectives for diagnosing, treating, and monitoring HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Karsten Gülow
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
46
|
Bao H, Chen X, Xiao Q, Yang S, Wu S, Wang X, Wu X, Ding K, Shao Y. Associations of genome-wide cell-free DNA fragmentation profiles with blood biochemical and hematological parameters in healthy individuals. Genomics 2022; 114:110504. [PMID: 36257481 DOI: 10.1016/j.ygeno.2022.110504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 01/15/2023]
Abstract
Cell-free DNA (cfDNA), as a non-invasive approach, has been introduced in a wide range of applications, including cancer diagnosis/ monitoring, prenatal testing, and transplantation monitoring. Yet, studies of cfDNA fragmentomics in physiological conditions are lacking. In this study, we aim to explore the correlation of fragmentation patterns of cfDNA with blood biochemical and hematological parameters in healthy individuals. We addressed the impact of physiological variables and abnormal blood biochemical and hematological parameters on cfDNA fragment size distribution. We also figured and validated that hematological inflammation markers, including leukocyte, lymphocyte, neutrophil, and platelet distribution width as well as aspartate transaminase levels were significantly correlated with the genome-wide cfDNA fragmentation pattern. Our findings suggest that cfDNA fragmentation profiles were associated with physiological parameters related to cardiovascular risk factors, inflammatory response and hepatocyte injury, which may provide insights for further research on the potential role of cfDNA fragmentation in diagnosis and monitor of several disease.
Collapse
Affiliation(s)
- Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xiaoxi Chen
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Qian Xiao
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shanshan Yang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Shuyu Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xiaonan Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China; School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
47
|
Sun X, Feng W, Cui P, Ruan R, Ma W, Han Z, Sun J, Pan Y, Zhu J, Zhong X, Li J, Ma M, Hu R, Lv M, Huang Q, Zhang W, Feng M, Zhuang X, Huang B, Zhou X. Detection and monitoring of HBV-related hepatocellular carcinoma from plasma cfDNA fragmentation profiles. Genomics 2022; 114:110502. [PMID: 36220554 DOI: 10.1016/j.ygeno.2022.110502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 01/15/2023]
Abstract
Most hepatocellular carcinomas (HCCs) are associated with hepatitis B virus infection (HBV) in China. Early detection of HCC can significantly improve prognosis but is not yet fully clinically feasible. This study aims to develop methods for detecting HCC and studying the carcinogenesis of HBV using plasma cell-free DNA (cfDNA) whole-genome sequencing (WGS) data. Low coverage WGS was performed for 452 participants, including healthy individuals, hepatitis B patients, cirrhosis patients, and HCC patients. Then the sequencing data were processed using various machine learning models based on cfDNA fragmentation profiles for cancer detection. Our best model achieved a sensitivity of 87.10% and a specificity of 88.37%, and it showed an increased sensitivity with higher BCLC stages of HCC. Overall, this study proves the potential of a non-invasive assay based on cfDNA fragmentation profiles for the detection and prognosis of HCC and provides preliminary data on the carcinogenic mechanism of HBV.
Collapse
Affiliation(s)
- Xinfeng Sun
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Wenxing Feng
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Pin Cui
- Shenzhen Rapha Biotechnology Incorporate, Shenzhen 518118, China
| | - Ruyun Ruan
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China
| | - Wenfeng Ma
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Zhiyi Han
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Jialing Sun
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Yuanke Pan
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China
| | - Jinxin Zhu
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China
| | - Xin Zhong
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Jing Li
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Mengqing Ma
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Rui Hu
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Minling Lv
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Qi Huang
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Wei Zhang
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| | - Mingji Feng
- Shenzhen Rapha Biotechnology Incorporate, Shenzhen 518118, China
| | - Xintao Zhuang
- Shenzhen Rapha Biotechnology Incorporate, Shenzhen 518118, China
| | - Bingding Huang
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China.
| | - Xiaozhou Zhou
- Department of Liver Disease, the fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen 518033, China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China.
| |
Collapse
|
48
|
Agopian VG, Yang JD, Zhu Y, You S, Tseng HR. Early detection of primary liver cancer using plasma cell-free DNA fragmentomics: Do all the pieces come together? Hepatology 2022; 76:289-291. [PMID: 35124841 DOI: 10.1002/hep.32396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 12/08/2022]
Affiliation(s)
- Vatche G Agopian
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yazhen Zhu
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA.,Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, Los Angeles, California, USA
| | - Sungyong You
- Division of Cancer Biology and Therapeutics, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hsian-Rong Tseng
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA.,Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, Los Angeles, California, USA
| |
Collapse
|
49
|
Jiang ST, Zhang JW, Lu X, Xu YY. Letter to the editor: Discussion on application scenario of cell-free DNA fragmentomics in primary liver cancer. Hepatology 2022; 76:E44. [PMID: 35334134 DOI: 10.1002/hep.32477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 12/08/2022]
Affiliation(s)
- Shi-Tao Jiang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun-Wei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Yao Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
50
|
Ueno M, Takeda H, Takai A, Seno H. Risk factors and diagnostic biomarkers for nonalcoholic fatty liver disease-associated hepatocellular carcinoma: Current evidence and future perspectives. World J Gastroenterol 2022; 28:3410-3421. [PMID: 36158261 PMCID: PMC9346451 DOI: 10.3748/wjg.v28.i27.3410] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/24/2022] [Accepted: 06/16/2022] [Indexed: 02/06/2023] Open
Abstract
High rates of excessive calorie intake diets and sedentary lifestyles have led to a global increase in nonalcoholic fatty liver disease (NAFLD). As a result, this condition has recently become one of the leading causes of hepatocellular carcinoma (HCC). Furthermore, the incidence of NAFLD-associated HCC (NAFLD-HCC) is expected to increase in the near future. Advanced liver fibrosis is the most common risk factor for NAFLD-HCC. However, up to 50% of NAFLD-HCC cases develop without underlying liver cirrhosis. Epidemiological studies have revealed many other risk factors for this condition; including diabetes, other metabolic traits, obesity, old age, male sex, Hispanic ethnicity, mild alcohol intake, and elevated liver enzymes. Specific gene variants, such as single-nucleotide polymorphisms of patatin-like phospholipase domain 3, transmembrane 6 superfamily member 2, and membrane-bound O-acyl-transferase domain-containing 7, are also associated with an increased risk of HCC in patients with NAFLD. This clinical and genetic information should be interpreted together for accurate risk prediction. Alpha-fetoprotein (AFP) is the only biomarker currently recommended for HCC screening. However, it is not sufficiently sensitive in addressing this diagnostic challenge. The GALAD score can be calculated based on sex, age, lectin-bound AFP, AFP, and des-carboxyprothrombin and is reported to show better diagnostic performance for HCC. In addition, emerging studies on genetic and epigenetic biomarkers have also yielded promising diagnostic potential. However, further research is needed to establish an effective surveillance program for the early diagnosis of NAFLD-HCC.
Collapse
Affiliation(s)
- Masayuki Ueno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Haruhiko Takeda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Atsushi Takai
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| |
Collapse
|