1
|
Afridi WA, Picos SH, Bark JM, Stamoudis DAF, Vasani S, Irwin D, Fielding D, Punyadeera C. Minimally invasive biomarkers for triaging lung nodules-challenges and future perspectives. Cancer Metastasis Rev 2025; 44:29. [PMID: 39888565 PMCID: PMC11785609 DOI: 10.1007/s10555-025-10247-5] [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: 11/05/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
CT chest scans are commonly performed worldwide, either in routine clinical practice for a wide range of indications or as part of lung cancer screening programs. Many of these scans detect lung nodules, which are small, rounded opacities measuring 8-30 mm. While the concern about nodules is that they may represent early lung cancer, in screening programs, only 1% of such nodules turn out to be cancer. This leads to a series of complex decisions and, at times, unnecessary biopsies for nodules that are ultimately determined to be benign. Additionally, patients may be anxious about the status of detected lung nodules. The high rate of false positive lung nodule detections has driven advancements in biomarker-based research aimed at triaging lung nodules (benign versus malignant) to identify truly malignant nodules better. Biomarkers found in biofluids and breath hold promise owing to their minimally invasive sampling methods, ease of use, and cost-effectiveness. Although several biomarkers have demonstrated clinical utility, their sensitivity and specificity are still relatively low. Combining multiple biomarkers could enhance the characterisation of small pulmonary nodules by addressing the limitations of individual biomarkers. This approach may help reduce unnecessary invasive procedures and accelerate diagnosis in the future. This review offers a thorough overview of emerging minimally invasive biomarkers for triaging lung nodules, emphasising key challenges and proposing potential solutions for biomarker-based nodule differentiation. It focuses on diagnosis rather than screening, analysing research published primarily in the past five years with some exceptions. The incorporation of biomarkers into clinical practice will facilitate the early detection of malignant nodules, leading to timely interventions and improved outcomes. Further efforts are needed to increase the cost-effectiveness and practicality of many of these applications in clinical settings. However, the range of technologies is advancing rapidly, and they may soon be implemented in clinics in the near future.
Collapse
Affiliation(s)
- Waqar Ahmed Afridi
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia
- Virtual University of Pakistan, Islamabad, 44000, Pakistan
| | - Samandra Hernandez Picos
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia
| | - Juliana Muller Bark
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia
| | - Danyelle Assis Ferreira Stamoudis
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia
| | - Sarju Vasani
- Department of Otolaryngology, Royal Brisbane and Women's Hospital, Herston, 4006, Australia
| | - Darryl Irwin
- The Agena Biosciences, Bowen Hills, Brisbane, 4006, Australia
| | - David Fielding
- The Royal Brisbane and Women's Hospital, Herston, Brisbane, 4006, Australia
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational Laboratory, Institute for Biomedicine and Glycomics (IBG), Griffith University, Brisbane, 4111, Australia.
| |
Collapse
|
2
|
Ding T, Hao S, Wang Z, Zhang W, Zhang G. Analysis of risk factors for pneumothorax after particle implantation in the treatment of advanced lung cancer after surgery and establishment of a nomogram prediction model. Front Med (Lausanne) 2024; 11:1428456. [PMID: 39478816 PMCID: PMC11521879 DOI: 10.3389/fmed.2024.1428456] [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: 05/06/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024] Open
Abstract
Objective To analyze the risk factors for pneumothorax after particle implantation in the treatment of advanced lung cancer and to construct and validate a nomogram prediction model. Methods A retrospective analysis was conducted on 148 patients who underwent 125I particle implantation for advanced lung cancer at the *** from December 2022 to December 2023. Potential risk factors were identified using univariate logistic regression analysis, followed by a multivariate logistic regression analysis to evaluate the predictive factors for pneumothorax. Interaction effects between variables were studied and incorporated into the model construction. ROC curves and nomograms were generated for visualization. Calibration analysis was performed, and the corresponding net benefit was calculated to adjust the predictive model. Results Among the 148 patients, 58 (39.19%) experienced pneumothorax, with a mean age of 62.5 (55.25, 70) years. Multivariate analysis showed that the angle between the puncture needle and the pleura < 50° (P = 0.002, OR: 3.908, CI: 1.621-9.422), preoperative CT suggesting emphysema (P = 0.002, OR: 3.798, CI: 1.600-9.016), atelectasis (P = 0.009, OR: 3.156, CI: 1.331-7.481), and lesion located in the left lung fissure (P = 0.008, OR: 4.675, CI: 14.683) were independent risk factors for pneumothorax after particle implantation in the treatment of advanced lung cancer. Preoperative CT suggesting lesions in the left lung fissure or suggesting emphysema had a significant impact in the nomogram, with probabilities of pneumothorax occurrence at 40% and 38%, respectively. The predictive AUC for the above four risk factors for pneumothorax after particle implantation in the treatment of lung adenocarcinoma was 0.837 (95% CI: 0.767-0.908). When the Youden index was 0.59, the sensitivity was 85.56%, specificity was 74.13%, accuracy was 81.01%, positive predictive value was 83.69%, and negative predictive value was 76.78%. Conclusion The angle between the puncture needle and the pleura < 50°, preoperative CT suggesting emphysema, atelectasis, and lesion located in the left lung fissure are independent risk factors for pneumothorax after particle implantation in the treatment of advanced lung cancer. Preoperative planning of the puncture path should avoid lung bullae, interlobar fissures, areas of severe emphysema, and atelectasis.
Collapse
Affiliation(s)
| | | | | | | | - Guoxu Zhang
- Northern Theater Command General Hospital, Shenyang, China
| |
Collapse
|
3
|
Jin Y, Lu R, Liu F, Jiang G, Wang R, Zheng M. DNA methylation analysis in plasma for early diagnosis in lung adenocarcinoma. Medicine (Baltimore) 2024; 103:e38867. [PMID: 38996143 PMCID: PMC11245223 DOI: 10.1097/md.0000000000038867] [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: 05/12/2023] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) represents the most prevalent type of lung cancer. SHOX2 and RASSF1A methylation have been identified as important biomarkers for diagnosis and prognosis of lung cancer. Bronchoalveolar lavage fluid (BALF) exhibits good specificity and sensitivity in diagnosing pulmonary diseases, but its acquisition is challenging and may cause discomfort to patients. In clinical, plasma samples are more convenient to obtain than BALF; however, there is little research on the concurrent detection of SHOX2 and RASSF1A methylation in plasma. This study aims to assess the diagnostic value of a combined promoter methylation assay for SHOX2 and RASSF1A in early-stage LUAD using plasma samples. METHODS BALF and blood samples were obtained from 36 early-stage LUAD patients, with a control group of nineteen non-tumor individuals. The promoter methylation levels of SHOX2 and RASSF1A in all subjects were assessed using the human SHOX2 and RASSF1A gene methylation kit. RESULTS The methylation detection rate of SHOX2 and RASSF1A in plasma was 61.11%, slightly lower than that in BALF (66.7%). The Chi-square test revealed no significant difference in the methylation rate between BALF and plasma (P > 0.05). The area under the receiver operating characteristic (ROC) curve analysis for blood was 0.806 (95% CI, 0.677 to 0.900), while for BALF it was 0.781 (95% CI, 0.649 to 0.881). Additionally, we conducted an analysis on the correlation between SHOX2 and RASSF1A methylation levels in plasma with gender, age, tumor differentiation, pathologic classification, and other clinicopathological variables; however, no significant correlations were observed. CONCLUSIONS Measurement of SHOX2 and RASSF1A methylation for early diagnosis of LUAD can be achieved with high sensitivity and specificity by using plasma as a substitute for BALF samples.
Collapse
Affiliation(s)
- Yulin Jin
- Department of Thoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Rongguo Lu
- Department of Thoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Feng Liu
- Department of Thoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Guanyu Jiang
- Department of Thoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Ruixin Wang
- Department of Thoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Mingfeng Zheng
- Department of Thoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
4
|
Phuong NA, Dao TT, Pham PB, Nguyen UD, Nguyen BV, Ho TH. Novel Semi-Nested Real-Time PCR Assay Leveraging Extendable Blocking Probes for Improved SHOX2 Methylation Analysis in Lung Cancer. Biomolecules 2024; 14:729. [PMID: 38927132 PMCID: PMC11201985 DOI: 10.3390/biom14060729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Lung cancer is the leading cause of cancer deaths globally, necessitating effective early detection methods. Traditional diagnostics like low-dose computed tomography (LDCT) often yield high false positive rates. SHOX2 gene methylation has emerged as a promising biomarker. This study aimed to develop and validate a novel semi-nested real-time PCR assay enhancing sensitivity and specificity for detecting SHOX2 methylation using extendable blocking probes (ExBPs). The assay integrates a semi-nested PCR approach with ExBPs, enhancing the detection of low-abundance methylated SHOX2 DNA amidst unmethylated sequences. It was tested on spiked samples with varied methylation levels and on clinical samples from lung cancer patients and individuals with benign lung conditions. The assay detected methylated SHOX2 DNA down to 0.01%. Clinical evaluations confirmed its ability to effectively differentiate between lung cancer patients and those with benign conditions, demonstrating enhanced sensitivity and specificity. The use of ExBPs minimized non-target sequence amplification, crucial for reducing false positives. The novel semi-nested real-time PCR assay offers a cost-effective, highly sensitive, and specific method for detecting SHOX2 methylation, enhancing early lung cancer detection and monitoring, particularly valuable in resource-limited settings.
Collapse
Affiliation(s)
- Ngoc Anh Phuong
- Vietnam National Lung Hospital, 463 Hoang Hoa Tham, Ba Dinh, Hanoi 10000, Vietnam;
- Department of Genomics and Cytogenetics, Institute of Biomedicine and Pharmacy (IBP), Vietnam Military Medical University, No. 222 Phung Hung, Ha Dong, Hanoi 10000, Vietnam
| | - Trang Thuy Dao
- Department of Genomics and Cytogenetics, Institute of Biomedicine and Pharmacy (IBP), Vietnam Military Medical University, No. 222 Phung Hung, Ha Dong, Hanoi 10000, Vietnam
| | - Phuong Bich Pham
- Department of Genomics and Cytogenetics, Institute of Biomedicine and Pharmacy (IBP), Vietnam Military Medical University, No. 222 Phung Hung, Ha Dong, Hanoi 10000, Vietnam
| | - Ung Dinh Nguyen
- Department of Genomics and Cytogenetics, Institute of Biomedicine and Pharmacy (IBP), Vietnam Military Medical University, No. 222 Phung Hung, Ha Dong, Hanoi 10000, Vietnam
| | - Ba Van Nguyen
- Oncology Center, 103 Military Hospital, Vietnam Military Medical University, Hanoi 10000, Vietnam
| | - Tho Huu Ho
- Department of Genomics and Cytogenetics, Institute of Biomedicine and Pharmacy (IBP), Vietnam Military Medical University, No. 222 Phung Hung, Ha Dong, Hanoi 10000, Vietnam
| |
Collapse
|
5
|
Xie B, Dong W, He F, Peng F, Zhang H, Wang W. The Combination of SHOX2 and RASSF1A DNA Methylation Had a Diagnostic Value in Pulmonary Nodules and Early Lung Cancer. Oncology 2024; 102:759-774. [PMID: 38262380 DOI: 10.1159/000534275] [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: 02/06/2023] [Accepted: 09/19/2023] [Indexed: 01/25/2024]
Abstract
INTRODUCTION The study explored the effects of SHOX2 and RASSF1A DNA methylation in lung cancer (LC). METHOD Bronchoalveolar lavage fluid (BALF) samples as well as LC and normal adjacent tissues were collected from 72 LC patients and 35 patients with benign pulmonary nodules. Quantitative analysis of SHOX2 and RASSF1A DNA methylation was performed in benign pulmonary nodules and different stages of LC. The diagnostic value of SHOX2 and RASSF1A DNA methylation in LC and benign pulmonary nodules was determined by receiver operating characteristics analysis. Gain/loss-of-function experiments were constructed in LC cells and mouse models of xenograft and pulmonary nodule metastasis. The levels of SHOX2 and transfer-associated genes were tested through quantitative reverse transcription polymerase chain reaction and Western blot. Malignant phenotype of LC cells was assessed by functional experiment. The tumor volume and weight of mice in xenograft models were measured. Pulmonary nodule metastasis was determined through HE staining assay. 5-azacytidine appeared as a positive control drug. RESULT SHOX2 DNA methylation or RASSF1A DNA methylation had diagnostic efficiency in pulmonary nodules and early LC, with the two combined having better diagnostic value. SHOX2 expression was upregulated in LC. Similar to 5-azacytidine, SHOX2 knockdown inhibited LC cell viability, migration, and invasion in vitro as well as restrained LC tumorigenesis and pulmonary nodule metastasis in vivo, whereas overexpressed SHOX2 had the opposite effects. CONCLUSION The combination of SHOX2 and RASSF1A DNA methylation had a diagnostic value in pulmonary nodules and early LC. SHOX2 positively modulated the tumorigenesis and metastasis of LC by regulating DNA methylation processes.
Collapse
Affiliation(s)
- Bin Xie
- Department of Respiratory Medicine, Yue Bei People's Hospital, Shaoguan, China
| | - Wenyan Dong
- Department of Geriatric Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fengping He
- Central Laboratory, Yue Bei People's Hospital, Shaoguan, China
| | - Feng Peng
- Department of Respiratory Medicine, Yue Bei People's Hospital, Shaoguan, China
| | - Honghua Zhang
- Department of Respiratory Medicine, Yue Bei People's Hospital, Shaoguan, China
| | - Wei Wang
- Medical Integration and Practice Center of Shandong University, Jinan, China
| |
Collapse
|
6
|
Borg M, Wen SWC, Andersen RF, Timm S, Hansen TF, Hilberg O. Methylated Circulating Tumor DNA in Blood as a Tool for Diagnosing Lung Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2023; 15:3959. [PMID: 37568774 PMCID: PMC10417522 DOI: 10.3390/cancers15153959] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/24/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths, and early detection is crucial for improving patient outcomes. Current screening methods using computed tomography have limitations, prompting interest in non-invasive diagnostic tools such as methylated circulating tumor DNA (ctDNA). The PRISMA guidelines for systematic reviews were followed. The electronic databases MEDLINE, Embase, Web of Science, and Cochrane Library were systematically searched for articles. The search string contained three main topics: Lung cancer, blood, and methylated ctDNA. The extraction of data and quality assessment were carried out independently by the reviewers. In total, 33 studies were eligible for inclusion in this systematic review and meta-analysis. The most frequently studied genes were SHOX2, RASSF1A, and APC. The sensitivity and specificity of methylated ctDNA varied across studies, with a summary sensitivity estimate of 46.9% and a summary specificity estimate of 92.9%. The area under the hierarchical summary receiver operating characteristics curve was 0.81. The included studies were generally of acceptable quality, although they lacked information in certain areas. The risk of publication bias was not significant. Based on the findings, methylated ctDNA in blood shows potential as a rule-in tool for lung cancer diagnosis but requires further research, possibly in combination with other biomarkers.
Collapse
Affiliation(s)
- Morten Borg
- Department of Medicine, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (M.B.)
| | - Sara Witting Christensen Wen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Rikke Fredslund Andersen
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Signe Timm
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Ole Hilberg
- Department of Medicine, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (M.B.)
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
7
|
DNA Methylation Analysis of the SHOX2 and RASSF1A Panel Using Cell-Free DNA in the Diagnosis of Malignant Pleural Effusion. JOURNAL OF ONCOLOGY 2023; 2023:5888844. [PMID: 36691467 PMCID: PMC9867579 DOI: 10.1155/2023/5888844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/16/2023]
Abstract
Objectives The differential diagnosis of pleural effusion (PE) is a common but major challenge in clinical practice. This study aimed to establish a strategy based on a PE-cell-free DNA (cfDNA) methylation detection system for the differential diagnosis of malignant pleural effusion (MPE) and benign pleural effusion (BPE). Methods A total of 104 patients with PE were enrolled in this study, among which 50 patients had MPE, 9 malignant tumor patients had PE of indefinite causes, and the other 45 patients were classified as benign controls. The methylation status of short stature homeobox 2 (SHOX2) and RAS association domain family 1, isoform A (RASSF1A) was detected using PE-cfDNA specimens by real-time fluorescence quantitative PCR. Total methylation (TM) was defined as the combination of the methylation levels of SHOX2 and RASSF1A. The electrochemiluminescence immunoassay was applied to evaluate the levels of multiple serum tumor markers. Results The PE-cfDNA methylation status of either SHOX2 or RASSF1A was much higher in MPE samples than in benign controls. The combination of SHOX2 and RASSF1A methylation in PE yielded a diagnostic sensitivity of 96% and a specificity of 100%, respectively. When compared with the corresponding serum tumor marker detection results, TM showed the highest diagnostic efficiency (AUC = 0.985). Furthermore, the combination of the SHOX2 and RASSF1A methylation panels using PE-cfDNA could apparently improve the differential diagnostic efficacy of BPE and MPE and could help compensate for the deficiency of cytology. Conclusions Our results indicated that SHOX2 and RASSF1A methylation panel detection could accurately classify BPE and MPE diseases and showed better diagnostic performance than traditional serum parameters. The SHOX2 and RASSF1A methylation detection of PE-cfDNA could be a potentially effective complementary tool for cytology in the process of differential diagnosis. In summary, PE-cfDNA could be used as a promising non-invasive analyte for the auxiliary diagnosis of MPE.
Collapse
|
8
|
Yang B, Fan Y, Liang R, Wu Y, Gu A. Identification of a prognostic six-immune-gene signature and a nomogram model for uveal melanoma. BMC Ophthalmol 2023; 23:2. [PMID: 36597071 PMCID: PMC9809105 DOI: 10.1186/s12886-022-02723-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/01/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND To identify an immune-related prognostic signature and find potential therapeutic targets for uveal melanoma. METHODS The RNA-sequencing data obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. The prognostic six-immune-gene signature was constructed through least absolute shrinkage and selection operator and multi-variate Cox regression analyses. Functional enrichment analysis and single sample GSEA were carried out. In addition, a nomogram model established by integrating clinical variables and this signature risk score was also constructed and evaluated. RESULTS We obtained 130 prognostic immune genes, and six of them were selected to construct a prognostic signature in the TCGA uveal melanoma dataset. Patients were classified into high-risk and low-risk groups according to a median risk score of this signature. High-risk group patients had poorer overall survival in comparison to the patients in the low-risk group (p < 0.001). These findings were further validated in two external GEO datasets. A nomogram model proved to be a good classifier for uveal melanoma by combining this signature. Both functional enrichment analysis and single sample GSEA analysis verified that this signature was truly correlated with immune system. In addition, in vitro cell experiments results demonstrated the consistent trend of our computational findings. CONCLUSION Our newly identified six-immune-gene signature and a nomogram model could be used as meaningful prognostic biomarkers, which might provide uveal melanoma patients with individualized clinical prognosis prediction and potential novel treatment targets.
Collapse
Affiliation(s)
- Binghua Yang
- grid.413405.70000 0004 1808 0686Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, 510182 Guangdong China
| | - Yuxia Fan
- grid.417234.70000 0004 1808 3203Department of Ophthalmology, Gansu Provincial Hospital, Lanzhou, 730000 Gansu China
| | - Renlong Liang
- grid.413405.70000 0004 1808 0686Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, 510182 Guangdong China
| | - Yi Wu
- grid.413405.70000 0004 1808 0686Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, 510182 Guangdong China
| | - Aiping Gu
- grid.413405.70000 0004 1808 0686Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, 510182 Guangdong China ,grid.413405.70000 0004 1808 0686Department of Ophthalmology, Guangdong Second Provincial General Hospital, No. 466 Xin’gangzhong Road, Haizhu, 510317 Guangzhou China
| |
Collapse
|
9
|
Blood-based DNA methylation signatures in cancer: A systematic review. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166583. [PMID: 36270476 DOI: 10.1016/j.bbadis.2022.166583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/30/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022]
Abstract
DNA methylation profiles are in dynamic equilibrium via the initiation of methylation, maintenance of methylation and demethylation, which control gene expression and chromosome stability. Changes in DNA methylation patterns play important roles in carcinogenesis and primarily manifests as hypomethylation of the entire genome and the hypermethylation of individual loci. These changes may be reflected in blood-based DNA, which provides a non-invasive means for cancer monitoring. Previous blood-based DNA detection objects primarily included circulating tumor DNA/cell-free DNA (ctDNA/cfDNA), circulating tumor cells (CTCs) and exosomes. Researchers gradually found that methylation changes in peripheral blood mononuclear cells (PBMCs) also reflected the presence of tumors. Blood-based DNA methylation is widely used in early diagnosis, prognosis prediction, dynamic monitoring after treatment and other fields of clinical research on cancer. The reversible methylation of genes also makes them important therapeutic targets. The present paper summarizes the changes in DNA methylation in cancer based on existing research and focuses on the characteristics of the detection objects of blood-based DNA, including ctDNA/cfDNA, CTCs, exosomes and PBMCs, and their application in clinical research.
Collapse
|
10
|
Zeng S, Lin C, Huang Y. miR-375 Combined with SHOX2 Methylation has Higher Diagnostic Efficacy for Non-Small-Cell Lung Cancer. Mol Biotechnol 2022:10.1007/s12033-022-00604-y. [DOI: 10.1007/s12033-022-00604-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 11/07/2022] [Indexed: 12/05/2022]
|
11
|
Markou Α, Londra D, Tserpeli V, Kollias Ι, Tsaroucha E, Vamvakaris I, Potaris K, Pateras I, Kotsakis Α, Georgoulias V, Lianidou Ε. DNA methylation analysis of tumor suppressor genes in liquid biopsy components of early stage NSCLC: a promising tool for early detection. Clin Epigenetics 2022; 14:61. [PMID: 35538556 PMCID: PMC9092693 DOI: 10.1186/s13148-022-01283-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/27/2022] [Indexed: 12/02/2022] Open
Abstract
Purpose Circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) analysis represents a liquid biopsy approach for real-time monitoring of tumor evolution. DNA methylation is considered to be an early event in the process of cancer development and progression. The aim of the present study was to evaluate whether detection of DNA methylation of selected tumor suppressor genes in CTC and matched ctDNA provides prognostic information in early stage NSCLC. Experimental design The methylation status of five selected gene promoters (APC, RASSFIA1, FOXA1, SLFN11, SHOX2) was examined by highly specific and sensitive real-time methylation specific PCR assays in: (a) a training group of 35 primary tumors and their corresponding adjacent non-cancerous tissues of early stage NSCLC patients, (b) a validation group of 22 primary tumor tissues (FFPEs) and 42 peripheral blood samples of early stage NSCLC patients. gDNA was isolated from FFPEs, CTCs (size-based enriched by Parsortix; Angle and plasma, and (c) a control group of healthy blood donors (n = 12). Results All five gene promoters tested were highly methylated in the training group; methylation of SHOX2 promoter in primary tumors was associated with unfavorable outcome. RASSFIA and APC were found methylated in plasma-cfDNA samples at 14.3% and 11.9%, respectively, whereas in the corresponding CTCs SLFN11 and APC promoters were methylated in 7.1%. The incidence of relapses was higher in patients with a) promoter methylation of APC and SLFN11 in plasma-cfDNA (P = 0.037 and P = 0.042 respectively) and b) at least one detected methylated gene promoter in CTC or plasma-cfDNA (P = 0.015). Conclusions DNA methylation of these five gene promoters was significantly lower in CTCs and plasma-cfDNA than in the primary tumors. Combination of DNA methylation analysis in CTC and plasma-cfDNA was associated with worse DFI of NSCLC patients. Additional studies are required to validate our findings in a large cohort of early stage NSCLC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01283-x.
Collapse
Affiliation(s)
- Α Markou
- Analysis of Circulating Tumor Cells (ACTC) Lab, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece.
| | - D Londra
- Analysis of Circulating Tumor Cells (ACTC) Lab, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - V Tserpeli
- Analysis of Circulating Tumor Cells (ACTC) Lab, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Ι Kollias
- Analysis of Circulating Tumor Cells (ACTC) Lab, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - E Tsaroucha
- 8th Department of Pulmonary Diseases, 'Sotiria' General Hospital for Chest Diseases, Athens, Greece
| | - I Vamvakaris
- 8th Department of Pulmonary Diseases, 'Sotiria' General Hospital for Chest Diseases, Athens, Greece
| | - K Potaris
- 8th Department of Pulmonary Diseases, 'Sotiria' General Hospital for Chest Diseases, Athens, Greece
| | - I Pateras
- Laboratory of Histology-Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Α Kotsakis
- Department of Medical Oncology, University General Hospital of Larissa, Thessaly, Greece
| | - V Georgoulias
- First Department of Medical Oncology, Metropolitan General Hospital of Athens, Cholargos, Greece
| | - Ε Lianidou
- Analysis of Circulating Tumor Cells (ACTC) Lab, Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|