1
|
Wei W, Wang Y, Ouyang R, Wang T, Chen R, Yuan X, Wang F, Wu S, Hou H. Machine Learning for Early Discrimination Between Lung Cancer and Benign Nodules Using Routine Clinical and Laboratory Data. Ann Surg Oncol 2024; 31:7738-7749. [PMID: 39014163 DOI: 10.1245/s10434-024-15762-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/24/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Lung cancer poses a global health threat necessitating early detection and precise staging for improved patient outcomes. This study focuses on developing and validating a machine learning-based risk model for early lung cancer screening and staging, using routine clinical data. METHODS Two medical center, observational, retrospective studies were conducted, involving 2312 lung cancer patients and 653 patients with benign nodules. Machine learning techniques, including differential analysis and feature selection, were employed to identify key factors for modeling. The study focused on variables such as nodule density, carcinoembryonic antigen (CEA), age, and lifestyle habits. The Logistic Regression model was utilized for early diagnoses, and the XGBoost model was utilized for staging based on selected features. RESULTS For early diagnoses, the Logistic Regression model achieved an area under the curve (AUC) of 0.716 (95% confidence interval [CI] 0.607-0.826), with 0.703 sensitivity and 0.654 specificity. The XGBoost model excelled in distinguishing late-stage from early-stage lung cancer, exhibiting an AUC of 0.913 (95% CI 0.862-0.963), with 0.909 sensitivity and 0.814 specificity. These findings highlight the model's potential for enhancing diagnostic accuracy and staging in lung cancer. CONCLUSION This study introduces a novel machine learning-based risk model for early lung cancer screening and staging, leveraging routine clinical information and laboratory data. The model shows promise in enhancing accuracy, mitigating overdiagnosis, and improving patient outcomes.
Collapse
Affiliation(s)
- Wei Wei
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renren Ouyang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rujia Chen
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Yuan
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shiji Wu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Hongyan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Chen J, Ming M, Huang S, Wei X, Wu J, Zhou S, Ling Z. AI-enhanced diagnostic model for pulmonary nodule classification. Front Oncol 2024; 14:1417753. [PMID: 39281372 PMCID: PMC11393475 DOI: 10.3389/fonc.2024.1417753] [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: 04/15/2024] [Accepted: 07/29/2024] [Indexed: 09/18/2024] Open
Abstract
Background The identification of benign and malignant pulmonary nodules (BPN and MPN) can significantly reduce mortality. However, a reliable and validated diagnostic model for clinical decision-making is still lacking. Methods Enzyme-linked immunosorbent assay and electro chemiluminescent immunoassay were utilized to determine the serum concentrations of 7AABs (p53, GAGE7, PGP9.5, CAGE, MAGEA1, SOX2, GBU4-5), and 4TTMs (CYFR21, CEA, NSE and SCC) in 260 participants (72 BPNs and 188 early-stage MPNs), respectively. The malignancy probability was calculated using Artificial intelligence pulmonary nodule auxiliary diagnosis system, or Mayo model. Along with age, sex, smoking history and nodule size, 18 variables were enrolled for model development. Baseline comparison, univariate ROC analysis, variable correlation analysis, lasso regression, univariate and stepwise logistic regression, and decision curve analysis (DCA) was used to reduce and screen variables. A nomogram and DCA were built for model construction and clinical use. Training (60%) and validation (40%) cohorts were used to for model validation. Results Age, CYFRA21_1, AI, PGP9.5, GAGE7, and GBU4_5 was screened out from 18 variables and utilized to establish the regression model for identifying BPN and early-stage MPN, as well as nomogram and DCA for clinical practical use. The AUC of the nomogram in the training and validation cohorts were 0.884 and 0.820, respectively. Moreover, the calibration curve showed high coherence between the predicted and actual probability. Conclusion This diagnostic model and DCA could provide evidence for upgrading or maintaining the current clinical decision based on malignancy probability stratification. It enables low and moderate risk or ambiguous patients to benefit from more precise clinical decision stratification, more timely detection of malignant nodules, and early treatment.
Collapse
Affiliation(s)
- Jifei Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangxi Medical University, Key Laboratory of Biological Molecular Medicine Research (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Moyu Ming
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Shuangping Huang
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Xuan Wei
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Jinyan Wu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Sufang Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangxi Medical University, Key Laboratory of Biological Molecular Medicine Research (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zhougui Ling
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|
3
|
Montero-Calle A, Garranzo-Asensio M, Moreno-Casbas MT, Campuzano S, Barderas R. Autoantibodies in cancer: a systematic review of their clinical role in the most prevalent cancers. Front Immunol 2024; 15:1455602. [PMID: 39234247 PMCID: PMC11371560 DOI: 10.3389/fimmu.2024.1455602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Although blood autoantibodies were initially associated with autoimmune diseases, multiple evidence have been accumulated showing their presence in many types of cancer. This has opened their use in clinics, since cancer autoantibodies might be useful for early detection, prognosis, and monitoring of cancer patients. In this review, we discuss the different techniques available for their discovery and validation. Additionally, we discuss here in detail those autoantibody panels verified in at least two different reports that should be more likely to be specific of each of the four most incident cancers. We also report the recent developed kits for breast and lung cancer detection mostly based on autoantibodies and the identification of novel therapeutic targets because of the screening of the cancer humoral immune response. Finally, we discuss unsolved issues that still need to be addressed for the implementation of cancer autoantibodies in clinical routine for cancer diagnosis, prognosis, and/or monitoring.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Maria Teresa Moreno-Casbas
- Investén-isciii, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network for Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Susana Campuzano
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network for Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Mohamed E, García Martínez DJ, Hosseini MS, Yoong SQ, Fletcher D, Hart S, Guinn BA. Identification of biomarkers for the early detection of non-small cell lung cancer: a systematic review and meta-analysis. Carcinogenesis 2024; 45:1-22. [PMID: 38066655 DOI: 10.1093/carcin/bgad091] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 02/13/2024] Open
Abstract
Lung cancer (LC) causes few symptoms in the earliest stages, leading to one of the highest mortality rates among cancers. Low-dose computerised tomography (LDCT) is used to screen high-risk individuals, reducing the mortality rate by 20%. However, LDCT results in a high number of false positives and is associated with unnecessary follow-up and cost. Biomarkers with high sensitivities and specificities could assist in the early detection of LC, especially in patients with high-risk features. Carcinoembryonic antigen (CEA), cytokeratin 19 fragments and cancer antigen 125 have been found to be highly expressed during the later stages of LC but have low sensitivity in the earliest stages. We determined the best biomarkers for the early diagnosis of LC, using a systematic review of eight databases. We identified 98 articles that focussed on the identification and assessment of diagnostic biomarkers and achieved a pooled area under curve of 0.85 (95% CI 0.82-0.088), indicating that the diagnostic performance of these biomarkers when combined was excellent. Of the studies, 30 focussed on single/antigen panels, 22 on autoantibodies, 31 on miRNA and RNA panels, and 15 suggested the use of circulating DNA combined with CEA or neuron-specific enolase (NSE) for early LC detection. Verification of blood biomarkers with high sensitivities (Ciz1, exoGCC2, ITGA2B), high specificities (CYFR21-1, antiHE4, OPNV) or both (HSP90α, CEA) along with miR-15b and miR-27b/miR-21 from sputum may improve early LC detection. Further assessment is needed using appropriate sample sizes, control groups that include patients with non-malignant conditions, and standardised cut-off levels for each biomarker.
Collapse
Affiliation(s)
- Eithar Mohamed
- Centre for Biomedicine, Hull York Medical School, University of Hull, Kingston-upon-Hull, HU6 7RX, UK
| | - Daniel J García Martínez
- Department of Biotechnology, Pozuelo de Alarcón, University Francisco De Vitoria, Madrid, 28223, Spain
| | - Mohammad-Salar Hosseini
- Research Centre for Evidence-Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Si Qi Yoong
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Daniel Fletcher
- Centre for Biomedicine, Hull York Medical School, University of Hull, Kingston-upon-Hull, HU6 7RX, UK
| | - Simon Hart
- Respiratory Medicine, Hull York Medical School, University of Hull, Kingston-upon-Hull, HU6 7RX, UK
| | - Barbara-Ann Guinn
- Centre for Biomedicine, Hull York Medical School, University of Hull, Kingston-upon-Hull, HU6 7RX, UK
| |
Collapse
|
5
|
Li T, Wang P, Sun G, Zou Y, Cheng Y, Wang H, Lu Y, Shi J, Wang K, Zhang Q, Ye H. hccTAAb Atlas: An Integrated Knowledge Database for Tumor-Associated Autoantibodies in Hepatocellular Carcinoma. J Proteome Res 2024; 23:728-737. [PMID: 38156953 DOI: 10.1021/acs.jproteome.3c00579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Tumor-associated autoantibodies (TAAbs) have demonstrated potential as biomarkers for cancer detection. However, the understanding of their role in hepatocellular carcinoma (HCC) remains limited. In this study, we aimed to systematically collect and standardize information about these TAAbs and establish a comprehensive database as a platform for in-depth research. A total of 170 TAAbs were identified from published papers retrieved from PubMed, Web of Science, and Embase. Following normative reannotation, these TAAbs were referred to as 162 official symbols. The hccTAAb (tumor-associated autoantibodies in hepatocellular carcinoma) atlas was developed using the R Shiny framework and incorporating literature-based and multiomics data sets. This comprehensive online resource provides key information such as sensitivity, specificity, and additional details such as official symbols, official full names, UniProt, NCBI, HPA, neXtProt, and aliases through hyperlinks. Additionally, hccTAAb offers six analytical modules for visualizing expression profiles, survival analysis, immune infiltration, similarity analysis, DNA methylation, and DNA mutation analysis. Overall, the hccTAAb Atlas provides valuable insights into the mechanisms underlying TAAb and has the potential to enhance the diagnosis and treatment of HCC using autoantibodies. The hccTAAb Atlas is freely accessible at https://nscc.v.zzu.edu.cn/hccTAAb/.
Collapse
Affiliation(s)
- Tiandong Li
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Peng Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Guiying Sun
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Yuanlin Zou
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Yifan Cheng
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Han Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Yin Lu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Jianxiang Shi
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Keyan Wang
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Qiang Zhang
- School of Nursing and Health, Zhengzhou University, Zhengzhou 450001, China
| | - Hua Ye
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Tumor Epidemiology & State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
6
|
van den Broek D, Groen HJM. Screening approaches for lung cancer by blood-based biomarkers: Challenges and opportunities. Tumour Biol 2024; 46:S65-S80. [PMID: 37393461 DOI: 10.3233/tub-230004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2023] Open
Abstract
Lung cancer (LC) is one of the leading causes for cancer-related deaths in the world, accounting for 28% of all cancer deaths in Europe. Screening for lung cancer can enable earlier detection of LC and reduce lung cancer mortality as was demonstrated in several large image-based screening studies such as the NELSON and the NLST. Based on these studies, screening is recommended in the US and in the UK a targeted lung health check program was initiated. In Europe lung cancer screening (LCS) has not been implemented due to limited data on cost-effectiveness in the different health care systems and questions on for example the selection of high-risk individuals, adherence to screening, management of indeterminate nodules, and risk of overdiagnosis. Liquid biomarkers are considered to have a high potential to address these questions by supporting pre- and post- Low Dose CT (LDCT) risk-assessment thereby improving the overall efficacy of LCS. A wide variety of biomarkers, including cfDNA, miRNA, proteins and inflammatory markers have been studied in the context of LCS. Despite the available data, biomarkers are currently not implemented or evaluated in screening studies or screening programs. As a result, it remains an open question which biomarker will actually improve a LCS program and do this against acceptable costs. In this paper we discuss the current status of different promising biomarkers and the challenges and opportunities of blood-based biomarkers in the context of lung cancer screening.
Collapse
Affiliation(s)
- Daniel van den Broek
- Department of laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
7
|
Li T, Xia J, Yun H, Sun G, Shen Y, Wang P, Shi J, Wang K, Yang H, Ye H. A novel autoantibody signatures for enhanced clinical diagnosis of pancreatic ductal adenocarcinoma. Cancer Cell Int 2023; 23:273. [PMID: 37974212 PMCID: PMC10655307 DOI: 10.1186/s12935-023-03107-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease that requires precise diagnosis for effective treatment. However, the diagnostic value of carbohydrate antigen 19 - 9 (CA19-9) is limited. Therefore, this study aims to identify novel tumor-associated autoantibodies (TAAbs) for PDAC diagnosis. METHODS A three-phase strategy comprising discovery, test, and validation was implemented. HuProt™ Human Proteome Microarray v3.1 was used to screen potential TAAbs in 49 samples. Subsequently, the levels of potential TAAbs were evaluated in 477 samples via enzyme-linked immunosorbent assay (ELISA) in PDAC, benign pancreatic diseases (BPD), and normal control (NC), followed by the construction of a diagnostic model. RESULTS In the discovery phase, protein microarrays identified 167 candidate TAAbs. Based on bioinformatics analysis, fifteen tumor-associated antigens (TAAs) were selected for further validation using ELISA. Ten TAAbs exhibited differentially expressed in PDAC patients in the test phase (P < 0.05), with an area under the curve (AUC) ranging from 0.61 to 0.76. An immunodiagnostic model including three TAAbs (anti-HEXB, anti-TXLNA, anti-SLAMF6) was then developed, demonstrating AUCs of 0.81 (58.0% sensitivity, 86.0% specificity) and 0.78 (55.71% sensitivity, 87.14% specificity) for distinguishing PDAC from NC. Additionally, the model yielded AUCs of 0.80 (58.0% sensitivity, 86.25% specificity) and 0.83 (55.71% sensitivity, 100% specificity) for distinguishing PDAC from BPD in the test and validation phases, respectively. Notably, the combination of the immunodiagnostic model with CA19-9 resulted in an increased positive rate of PDAC to 92.91%. CONCLUSION The immunodiagnostic model may offer a novel serological detection method for PDAC diagnosis, providing valuable insights into the development of effective diagnostic biomarkers.
Collapse
Affiliation(s)
- Tiandong Li
- College of Public Health, Zhengzhou University, 450001, Zhengzhou, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Junfen Xia
- Office of Health Care, The Third Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Huan Yun
- Zhengzhou University, 450001, Zhengzhou, Henan Province, China
| | - Guiying Sun
- College of Public Health, Zhengzhou University, 450001, Zhengzhou, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Yajing Shen
- College of Public Health, Zhengzhou University, 450001, Zhengzhou, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Peng Wang
- College of Public Health, Zhengzhou University, 450001, Zhengzhou, Henan Province, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Jianxiang Shi
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 450052, Zhengzhou, Henan Province, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Keyan Wang
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 450052, Zhengzhou, Henan Province, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Hongwei Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan Province, China
| | - Hua Ye
- College of Public Health, Zhengzhou University, 450001, Zhengzhou, Henan Province, China.
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 450052, Zhengzhou, Henan Province, China.
| |
Collapse
|
8
|
Abstract
Current lung cancer screening protocols use low-dose computed tomography scans in selected high-risk individuals. Unfortunately, utilization is low, and the rate of false-positive screens is high. Peripheral biomarkers carry meaningful promise in diagnosing and monitoring cancer with added potential advantages reducing invasive procedures and improving turnaround time. Herein, the use of such blood-based assays is considered as an adjunct to further utilization and accuracy of lung cancer screening.
Collapse
Affiliation(s)
- Nathaniel Deboever
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Edwin J Ostrin
- Department of General Internal Medicine, Pulmonary Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Jing K, Zhao H, Cai J, Chen L, Zheng P, Ouyang L, Li G, Wang R. The presence of autoantibodies is associated with improved overall survival in lung cancer patients. Front Oncol 2023; 13:1234847. [PMID: 37799460 PMCID: PMC10547871 DOI: 10.3389/fonc.2023.1234847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/21/2023] [Indexed: 10/07/2023] Open
Abstract
Objective Autoantibodies have been reported to be associated with cancers. As a biomarker, autoantibodies have been widely used in the early screening of lung cancer. However, the correlation between autoantibodies and the prognosis of lung cancer patients is poorly understood, especially in the Asian population. This retrospective study investigated the association between the presence of autoantibodies and outcomes in patients with lung cancer. Methods A total of 264 patients diagnosed with lung cancer were tested for autoantibodies in Henan Provincial People's Hospital from January 2017 to June 2022. The general clinical data of these patients were collected, and after screening out those who met the exclusion criteria, 151 patients were finally included in the study. The Cox proportional hazards model was used to analyze the effect of autoantibodies on the outcomes of patients with lung cancer. The Kaplan-Meier curve was used to analyze the relationship between autoantibodies and the overall survival of patients with lung cancer. Results Compared to lung cancer patients without autoantibodies, those with autoantibodies had an associated reduced risk of death (HRs: 0.45, 95% CIs 0.27~0.77), independent of gender, age, smoking history, pathological type, and pathological stage of lung cancer. Additionally, the association was found to be more significant by subgroup analysis in male patients, younger patients, and patients with small cell lung cancer. Furthermore, lung cancer patients with autoantibodies had significantly longer survival time than those without autoantibodies. Conclusion The presence of autoantibodies is an independent indicator of good prognosis in patients with lung cancer, providing a new biomarker for prognostic evaluation in patients with lung cancer.
Collapse
Affiliation(s)
- Keying Jing
- Henan University People's Hospital, Department of Clinical Laboratory, Henan Provincial People’s Hospital, Henan University, Zhengzhou, Henan, China
| | - Huijuan Zhao
- Basic Medical College, Henan University of Science and Technology, Luoyang, Henan, China
| | - Jun Cai
- Henan University People's Hospital, Department of Clinical Laboratory, Henan Provincial People’s Hospital, Henan University, Zhengzhou, Henan, China
| | - Lianlian Chen
- Henan Hospital of Integrated Chinese and Western Medicine, Zhengzhou, Henan, China
| | - Peiming Zheng
- Henan University People's Hospital, Department of Clinical Laboratory, Henan Provincial People’s Hospital, Henan University, Zhengzhou, Henan, China
| | - Libo Ouyang
- Henan University People's Hospital, Department of Clinical Laboratory, Henan Provincial People’s Hospital, Henan University, Zhengzhou, Henan, China
| | - Gang Li
- Henan University People's Hospital, Department of Clinical Laboratory, Henan Provincial People’s Hospital, Henan University, Zhengzhou, Henan, China
| | - Rong Wang
- Henan University People's Hospital, Department of Clinical Laboratory, Henan Provincial People’s Hospital, Henan University, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Feng X, Wu WYY, Onwuka JU, Haider Z, Alcala K, Smith-Byrne K, Zahed H, Guida F, Wang R, Bassett JK, Stevens V, Wang Y, Weinstein S, Freedman ND, Chen C, Tinker L, Nøst TH, Koh WP, Muller D, Colorado-Yohar SM, Tumino R, Hung RJ, Amos CI, Lin X, Zhang X, Arslan AA, Sánchez MJ, Sørgjerd EP, Severi G, Hveem K, Brennan P, Langhammer A, Milne RL, Yuan JM, Melin B, Johansson M, Robbins HA, Johansson M. Lung cancer risk discrimination of prediagnostic proteomics measurements compared with existing prediction tools. J Natl Cancer Inst 2023; 115:1050-1059. [PMID: 37260165 PMCID: PMC10483263 DOI: 10.1093/jnci/djad071] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/05/2023] [Accepted: 04/08/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND We sought to develop a proteomics-based risk model for lung cancer and evaluate its risk-discriminatory performance in comparison with a smoking-based risk model (PLCOm2012) and a commercially available autoantibody biomarker test. METHODS We designed a case-control study nested in 6 prospective cohorts, including 624 lung cancer participants who donated blood samples at most 3 years prior to lung cancer diagnosis and 624 smoking-matched cancer free participants who were assayed for 302 proteins. We used 470 case-control pairs from 4 cohorts to select proteins and train a protein-based risk model. We subsequently used 154 case-control pairs from 2 cohorts to compare the risk-discriminatory performance of the protein-based model with that of the Early Cancer Detection Test (EarlyCDT)-Lung and the PLCOm2012 model using receiver operating characteristics analysis and by estimating models' sensitivity. All tests were 2-sided. RESULTS The area under the curve for the protein-based risk model in the validation sample was 0.75 (95% confidence interval [CI] = 0.70 to 0.81) compared with 0.64 (95% CI = 0.57 to 0.70) for the PLCOm2012 model (Pdifference = .001). The EarlyCDT-Lung had a sensitivity of 14% (95% CI = 8.2% to 19%) and a specificity of 86% (95% CI = 81% to 92%) for incident lung cancer. At the same specificity of 86%, the sensitivity for the protein-based risk model was estimated at 49% (95% CI = 41% to 57%) and 30% (95% CI = 23% to 37%) for the PLCOm2012 model. CONCLUSION Circulating proteins showed promise in predicting incident lung cancer and outperformed a standard risk prediction model and the commercialized EarlyCDT-Lung.
Collapse
Affiliation(s)
- Xiaoshuang Feng
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Wendy Yi-Ying Wu
- Department of Radiation Sciences, Oncology, Umea University, Umea, Sweden
| | | | - Zahra Haider
- Department of Radiation Sciences, Oncology, Umea University, Umea, Sweden
| | - Karine Alcala
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | | | - Hana Zahed
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Florence Guida
- Environment and Lifestyle Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Renwei Wang
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julie K Bassett
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Victoria Stevens
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Ying Wang
- American Cancer Society, Atlanta, GA, USA
| | - Stephanie Weinstein
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Neal D Freedman
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Chu Chen
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lesley Tinker
- Women’s Health Initiative Clinical Coordinating Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Therese Haugdahl Nøst
- Department of Community Medicine, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
| | - Woon-Puay Koh
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - David Muller
- Division of Genetic Medicine, Imperial College London School of Public Health, London, UK
| | - Sandra M Colorado-Yohar
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Research Group on Demography and Health, National Faculty of Public Health, University of Antioquia, Medellín, Colombia
| | - Rosario Tumino
- Hyblean Association for Epidemiological Research, AIRE ONLUS Ragusa, Ragusa, Italy
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Statistics, Harvard University, Cambridge, MA, USA
| | - Xuehong Zhang
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Alan A Arslan
- Department of Population Health, New York University School of Medicine, New York, NY, USA
| | - Maria-Jose Sánchez
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain
- Instituto de Investigación Biosanitaria ib, Granada, Spain
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
| | - Elin Pettersen Sørgjerd
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
| | | | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
| | - Paul Brennan
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Arnulf Langhammer
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beatrice Melin
- Department of Radiation Sciences, Oncology, Umea University, Umea, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umea University, Umea, Sweden
| | - Hilary A Robbins
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Mattias Johansson
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
11
|
Wu WYY, Haider Z, Feng X, Heath AK, Tjønneland A, Agudo A, Masala G, Robbins HA, Huerta MJ, Guevara M, Schulze MB, Rodriguez-Barranco M, Vineis P, Tumino R, Kaaks R, Fortner RT, Sieri S, Panico S, Nøst TH, Sandanger TM, Braaten T, Johansson M, Melin B, Johansson M. Assessment of the EarlyCDT-Lung test as an early biomarker of lung cancer in ever-smokers: A retrospective nested case-control study in two prospective cohorts. Int J Cancer 2023; 152:2002-2010. [PMID: 36305647 PMCID: PMC10157531 DOI: 10.1002/ijc.34340] [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: 06/28/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/11/2022]
Abstract
The EarlyCDT-Lung test is a blood-based autoantibody assay intended to identify high-risk individuals for low-dose computed tomography lung cancer screening. However, there is a paucity of evidence on the performance of the EarlyCDT-Lung test in ever-smokers. We conducted a nested case-control study within two prospective cohorts to evaluate the risk-discriminatory performance of the EarlyCDT-Lung test using prediagnostic blood samples from 154 future lung cancer cases and 154 matched controls. Cases were selected from those who had ever smoked and had a prediagnostic blood sample <3 years prior to diagnosis. Conditional logistic regression was used to estimate the association between EarlyCDT-Lung test results and lung cancer risk. Sensitivity and specificity of the EarlyCDT-Lung test were calculated in all subjects and subgroups based on age, smoking history, lung cancer stage, sample collection time before diagnosis and year of sample collection. The overall lung cancer odds ratios were 0.89 (95% CI: 0.34-2.30) for a moderate risk EarlyCDT-Lung test result and 1.09 (95% CI: 0.48-2.47) for a high-risk test result compared to no significant test result. The overall sensitivity was 8.4% (95% CI: 4.6-14) and overall specificity was 92% (95% CI: 87-96) when considering a high-risk result as positive. Stratified analysis indicated higher sensitivity (17%, 95% CI: 7.2-32.1) in subjects with blood drawn up to 1 year prior to diagnosis. In conclusion, our study does not support a role of the EarlyCDT-Lung test in identifying the high-risk subjects in ever-smokers for lung cancer screening in the EPIC and NSHDS cohorts.
Collapse
Affiliation(s)
- Wendy Yi-Ying Wu
- Department of Radiation Sciences, Oncology, Umeå University, Sweden
| | - Zahra Haider
- Department of Radiation Sciences, Oncology, Umeå University, Sweden
| | - Xiaoshuang Feng
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Alicia K. Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Anne Tjønneland
- Diet, Cancer and Health, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Denmark
| | - Antonio Agudo
- Unit of Nutrition and Cancer, Catalan Institute of Oncology - ICO, L’Hospitalet de Llobregat, Spain
- Nutrition and Cancer Group; Epidemiology, Public Health, Cancer Prevention and Palliative Care Program; Bellvitge Biomedical Research Institute - IDIBELL, L’Hospitalet de Llobregat, Spain
| | - Giovanna Masala
- Institute for cancer research, prevention and clinical network (ISPRO) Florence, Italy
| | - Hilary A. Robbins
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - María-José Huerta
- Department of Epidemiology, Murcia Regional Health Council, Murcia, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| | - Marcela Guevara
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Navarra Public Health Institute, 31003 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Matthias B. Schulze
- German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Miguel Rodriguez-Barranco
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Escuela Andaluza de Salud Pública (EASP), 18011 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Paolo Vineis
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, UK
| | - Rosario Tumino
- Hyblean Association for Epidemiological Research, AIRE ONLUS Ragusa, Italy
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DFKZ), Heidelberg, Germany
- Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Renée T. Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano Via Venezian, Milan, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia Federico II Universioty, Naples, Italy
| | - Therese Haugdahl Nøst
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Torkjel M. Sandanger
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Tonje Braaten
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Mattias Johansson
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Beatrice Melin
- Department of Radiation Sciences, Oncology, Umeå University, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, Sweden
| |
Collapse
|
12
|
Auger C, Moudgalya H, Neely MR, Stephan JT, Tarhoni I, Gerard D, Basu S, Fhied CL, Abdelkader A, Vargas M, Hu S, Hulett T, Liptay MJ, Shah P, Seder CW, Borgia JA. Development of a Novel Circulating Autoantibody Biomarker Panel for the Identification of Patients with 'Actionable' Pulmonary Nodules. Cancers (Basel) 2023; 15:2259. [PMID: 37190187 PMCID: PMC10136536 DOI: 10.3390/cancers15082259] [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: 03/11/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Due to poor compliance and uptake of LDCT screening among high-risk populations, lung cancer is often diagnosed in advanced stages where treatment is rarely curative. Based upon the American College of Radiology's Lung Imaging and Reporting Data System (Lung-RADS) 80-90% of patients screened will have clinically "non-actionable" nodules (Lung-RADS 1 or 2), and those harboring larger, clinically "actionable" nodules (Lung-RADS 3 or 4) have a significantly greater risk of lung cancer. The development of a companion diagnostic method capable of identifying patients likely to have a clinically actionable nodule identified during LDCT is anticipated to improve accessibility and uptake of the paradigm and improve early detection rates. Using protein microarrays, we identified 501 circulating targets with differential immunoreactivities against cohorts characterized as possessing either actionable (n = 42) or non-actionable (n = 20) solid pulmonary nodules, per Lung-RADS guidelines. Quantitative assays were assembled on the Luminex platform for the 26 most promising targets. These assays were used to measure serum autoantibody levels in 841 patients, consisting of benign (BN; n = 101), early-stage non-small cell lung cancer (NSCLC; n = 245), other early-stage malignancies within the lung (n = 29), and individuals meeting United States Preventative Screening Task Force (USPSTF) screening inclusion criteria with both actionable (n = 87) and non-actionable radiologic findings (n = 379). These 841 patients were randomly split into three cohorts: Training, Validation 1, and Validation 2. Of the 26 candidate biomarkers tested, 17 differentiated patients with actionable nodules from those with non-actionable nodules. A random forest model consisting of six autoantibody (Annexin 2, DCD, MID1IP1, PNMA1, TAF10, ZNF696) biomarkers was developed to optimize our classification performance; it possessed a positive predictive value (PPV) of 61.4%/61.0% and negative predictive value (NPV) of 95.7%/83.9% against Validation cohorts 1 and 2, respectively. This panel may improve patient selection methods for lung cancer screening, serving to greatly reduce the futile screening rate while also improving accessibility to the paradigm for underserved populations.
Collapse
Affiliation(s)
- Claire Auger
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Hita Moudgalya
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Matthew R. Neely
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jeremy T. Stephan
- Rush University Medical College, Rush University Medical Center, Chicago, IL 60612, USA
| | - Imad Tarhoni
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
| | - David Gerard
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sanjib Basu
- Division of Medical Oncology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Cristina L. Fhied
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ahmed Abdelkader
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
| | | | - Shaohui Hu
- CDI Laboratories, Mayagüez, PR 00680, USA
| | | | - Michael J. Liptay
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Palmi Shah
- Department of Diagnostic Radiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Christopher W. Seder
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jeffrey A. Borgia
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Casagrande GMS, Silva MDO, Reis RM, Leal LF. Liquid Biopsy for Lung Cancer: Up-to-Date and Perspectives for Screening Programs. Int J Mol Sci 2023; 24:2505. [PMID: 36768828 PMCID: PMC9917347 DOI: 10.3390/ijms24032505] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 01/31/2023] Open
Abstract
Lung cancer is the deadliest cancer worldwide. Tissue biopsy is currently employed for the diagnosis and molecular stratification of lung cancer. Liquid biopsy is a minimally invasive approach to determine biomarkers from body fluids, such as blood, urine, sputum, and saliva. Tumor cells release cfDNA, ctDNA, exosomes, miRNAs, circRNAs, CTCs, and DNA methylated fragments, among others, which can be successfully used as biomarkers for diagnosis, prognosis, and prediction of treatment response. Predictive biomarkers are well-established for managing lung cancer, and liquid biopsy options have emerged in the last few years. Currently, detecting EGFR p.(Tyr790Met) mutation in plasma samples from lung cancer patients has been used for predicting response and monitoring tyrosine kinase inhibitors (TKi)-treated patients with lung cancer. In addition, many efforts continue to bring more sensitive technologies to improve the detection of clinically relevant biomarkers for lung cancer. Moreover, liquid biopsy can dramatically decrease the turnaround time for laboratory reports, accelerating the beginning of treatment and improving the overall survival of lung cancer patients. Herein, we summarized all available and emerging approaches of liquid biopsy-techniques, molecules, and sample type-for lung cancer.
Collapse
Affiliation(s)
| | - Marcela de Oliveira Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331 Rua Antenor Duarte Vilela, Barretos 14784-400, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331 Rua Antenor Duarte Vilela, Barretos 14784-400, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Letícia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331 Rua Antenor Duarte Vilela, Barretos 14784-400, Brazil
- Barretos School of Medicine Dr. Paulo Prata—FACISB, Barretos 14785-002, Brazil
| |
Collapse
|
14
|
Smok-Kalwat J, Mertowska P, Mertowski S, Smolak K, Kozińska A, Koszałka F, Kwaśniewski W, Grywalska E, Góźdź S. The Importance of the Immune System and Molecular Cell Signaling Pathways in the Pathogenesis and Progression of Lung Cancer. Int J Mol Sci 2023; 24:1506. [PMID: 36675020 PMCID: PMC9861992 DOI: 10.3390/ijms24021506] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Lung cancer is a disease that in recent years has become one of the greatest threats to modern society. Every year there are more and more new cases and the percentage of deaths caused by this type of cancer increases. Despite many studies, scientists are still looking for answers regarding the mechanisms of lung cancer development and progression, with particular emphasis on the role of the immune system. The aim of this literature review was to present the importance of disorders of the immune system and the accompanying changes at the level of cell signaling in the pathogenesis of lung cancer. The collected results showed that in the process of immunopathogenesis of almost all subtypes of lung cancer, changes in the tumor microenvironment, deregulation of immune checkpoints and abnormalities in cell signaling pathways are involved, which contribute to the multistage and multifaceted carcinogenesis of this type of cancer. We, therefore, suggest that in future studies, researchers should focus on a detailed analysis of tumor microenvironmental immune checkpoints, and to validate their validity, perform genetic polymorphism analyses in a wide range of patients and healthy individuals to determine the genetic susceptibility to lung cancer development. In addition, further research related to the analysis of the tumor microenvironment; immune system disorders, with a particular emphasis on immunological checkpoints and genetic differences may contribute to the development of new personalized therapies that improve the prognosis of patients.
Collapse
Affiliation(s)
- Jolanta Smok-Kalwat
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Konrad Smolak
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Aleksandra Kozińska
- Student Research Group of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Filip Koszałka
- Student Research Group of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Wojciech Kwaśniewski
- Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland
| | - Stanisław Góźdź
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| |
Collapse
|
15
|
Xu Y, Zhang W, Xia T, Liu Y, Bi Z, Guo L, Xie W, Xiang Y, Xu Z, Yu Z, Li Y, Bai L. Diagnostic value of tumor-associated autoantibodies panel in combination with traditional tumor markers for lung cancer. Front Oncol 2023; 13:1022331. [PMID: 36874112 PMCID: PMC9975551 DOI: 10.3389/fonc.2023.1022331] [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: 08/18/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Introduction The diagnostic value of 7 tumor-associated autoantibodies (AABs) including p53, PGP9.5, SOX2, GAGE7, GBU4-5, MEGEA1, and CAGE for the detection of lung cancer has shown inconsistency in several studies. This study aimed to confirm the diagnostic value of 7AABs and to explore whether the diagnostic value would be improved by combining them with 7 traditional tumor-associated antigens (CEA, NSE, CA125, SCC, CA15-3, pro-GRP, and CYFRA21-1) in clinical settings. Methods The plasma levels of 7-AABs were detected by enzyme-linked immunosorbent assay (ELISA) in 533 lung cancer cases and 454 controls. The 7 tumor antigens (7-TAs) were measured by Electrochemiluminescence immunoassay with Cobas 6000 (Roche, Basel, Switzerland). Results The positive rate of 7-AABs in the lung cancer group (64.00%) was significantly higher than that of healthy controls (47.90%). The 7-AABs panel was able to discriminate lung cancer from controls with a specificity of 51.50%. After combining the 7-AABs with 7-TAs, the sensitivity showed a significantly enhancement compared with 7AABs panel alone (92.09% vs 63.21%). In patients with resectable lung cancer, the combination of 7-AABs and 7-TAs improved the sensitivity from 63.52% to 97.42. Discussion In conclusion, our study found that the diagnostic value of 7-AABs was enhanced when combined with 7-TAs. This combined panel could be used as promising biomarker to detect resectable lung cancer in clinical settings.
Collapse
Affiliation(s)
- Yu Xu
- Department of Respiratory and Critical Care Medicine, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenjing Zhang
- Department of Respiratory and Critical Care Medicine, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Tingting Xia
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuliang Liu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhoukui Bi
- Department of Respiratory and Critical Care Medicine, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Liang Guo
- Department of Respiratory and Critical Care Medicine, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Weijia Xie
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ying Xiang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhi Xu
- Department of Respiratory and Critical Care Medicine, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zubin Yu
- Department of Thoracic Surgery, North-Kuanren General Hospital, Chongqing, China.,Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yafei Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Bai
- Department of Respiratory and Critical Care Medicine, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
16
|
Duarte A, Corbett M, Melton H, Harden M, Palmer S, Soares M, Simmonds M. EarlyCDT Lung blood test for risk classification of solid pulmonary nodules: systematic review and economic evaluation. Health Technol Assess 2022; 26:1-184. [PMID: 36534989 PMCID: PMC9791464 DOI: 10.3310/ijfm4802] [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] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND EarlyCDT Lung (Oncimmune Holdings plc, Nottingham, UK) is a blood test to assess malignancy risk in people with solid pulmonary nodules. It measures the presence of seven lung cancer-associated autoantibodies. Elevated levels of these autoantibodies may indicate malignant disease. The results of the test might be used to modify the risk of malignancy estimated by existing risk calculators, including the Brock and Herder models. OBJECTIVES The objectives were to determine the diagnostic accuracy, clinical effectiveness and cost-effectiveness of EarlyCDT Lung; and to develop a conceptual model and identify evidence requirements for a robust cost-effectiveness analysis. DATA SOURCES MEDLINE (including Epub Ahead of Print, In-Process & Other Non-Indexed Citations, Ovid MEDLINE Daily and Ovid MEDLINE), EMBASE, Cochrane Central Register of Controlled Trials, Science Citation Index, EconLit, Cochrane Database of Systematic Reviews, Database of Abstracts of Reviews of Effects, Health Technology Assessment database, NHS Economic Evaluation Database ( NHS EED ) and the international Health Technology Assessment database were searched on 8 March 2021. REVIEW METHODS A systematic review was performed of evidence on EarlyCDT Lung, including diagnostic accuracy, clinical effectiveness and cost-effectiveness. Study quality was assessed with the quality assessment of diagnostic accuracy studies-2 tool. Evidence on other components of the pulmonary nodule diagnostic pathway (computerised tomography surveillance, Brock risk, Herder risk, positron emission tomography-computerised tomography and biopsy) was also reviewed. When feasible, bivariate meta-analyses of diagnostic accuracy were performed. Clinical outcomes were synthesised narratively. A simulation study investigated the clinical impact of using EarlyCDT Lung. Additional reviews of cost-effectiveness studies evaluated (1) other diagnostic strategies for lung cancer and (2) screening approaches for lung cancer. A conceptual model was developed. RESULTS A total of 47 clinical publications on EarlyCDT Lung were identified, but only five cohorts (695 patients) reported diagnostic accuracy data on patients with pulmonary nodules. All cohorts were small or at high risk of bias. EarlyCDT Lung on its own was found to have poor diagnostic accuracy, with a summary sensitivity of 20.2% (95% confidence interval 10.5% to 35.5%) and specificity of 92.2% (95% confidence interval 86.2% to 95.8%). This sensitivity was substantially lower than that estimated by the manufacturer (41.3%). No evidence on the clinical impact of EarlyCDT Lung was identified. The simulation study suggested that EarlyCDT Lung might potentially have some benefit when considering intermediate risk nodules (10-70% risk) after Herder risk analysis. Two cost-effectiveness studies on EarlyCDT Lung for pulmonary nodules were identified; none was considered suitable to inform the current decision problem. The conceptualisation process identified three core components for a future cost-effectiveness assessment of EarlyCDT Lung: (1) the features of the subpopulations and relevant heterogeneity, (2) the way EarlyCDT Lung test results affect subsequent clinical management decisions and (3) how changes in these decisions can affect outcomes. All reviewed studies linked earlier diagnosis to stage progression and stage shift to final outcomes, but evidence on these components was sparse. LIMITATIONS The evidence on EarlyCDT Lung among patients with pulmonary nodules was very limited, preventing meta-analyses and economic analyses. CONCLUSIONS The evidence on EarlyCDT Lung among patients with pulmonary nodules is insufficient to draw any firm conclusions as to its diagnostic accuracy or clinical or economic value. FUTURE WORK Prospective cohort studies, in which EarlyCDT Lung is used among patients with identified pulmonary nodules, are required to support a future assessment of the clinical and economic value of this test. Studies should investigate the diagnostic accuracy and clinical impact of EarlyCDT Lung in combination with Brock and Herder risk assessments. A well-designed cost-effectiveness study is also required, integrating emerging relevant evidence with the recommendations in this report. STUDY REGISTRATION This study is registered as PROSPERO CRD42021242248. FUNDING This project was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 26, No. 49. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Ana Duarte
- Centre for Health Economics, University of York, York UK
| | - Mark Corbett
- Centre for Reviews and Dissemination, University of York, York UK
| | - Hollie Melton
- Centre for Reviews and Dissemination, University of York, York UK
| | - Melissa Harden
- Centre for Reviews and Dissemination, University of York, York UK
| | - Stephen Palmer
- Centre for Health Economics, University of York, York UK
| | - Marta Soares
- Centre for Health Economics, University of York, York UK
| | - Mark Simmonds
- Centre for Reviews and Dissemination, University of York, York UK
| |
Collapse
|
17
|
Padinharayil H, Varghese J, John MC, Rajanikant GK, Wilson CM, Al-Yozbaki M, Renu K, Dewanjee S, Sanyal R, Dey A, Mukherjee AG, Wanjari UR, Gopalakrishnan AV, George A. Non-small cell lung carcinoma (NSCLC): Implications on molecular pathology and advances in early diagnostics and therapeutics. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
18
|
Li C, Wang H, Jiang Y, Fu W, Liu X, Zhong R, Cheng B, Zhu F, Xiang Y, He J, Liang W. Advances in lung cancer screening and early detection. Cancer Biol Med 2022; 19:j.issn.2095-3941.2021.0690. [PMID: 35535966 PMCID: PMC9196057 DOI: 10.20892/j.issn.2095-3941.2021.0690] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/03/2022] [Indexed: 11/18/2022] Open
Abstract
Lung cancer is associated with a heavy cancer-related burden in terms of patients' physical and mental health worldwide. Two randomized controlled trials, the US-National Lung Screening Trial (NLST) and Nederlands-Leuvens Longkanker Screenings Onderzoek (NELSON), indicated that low-dose CT (LDCT) screening results in a statistically significant decrease in mortality in patients with lung cancer, LDCT has become the standard approach for lung cancer screening. However, many issues in lung cancer screening remain unresolved, such as the screening criteria, high false-positive rate, and radiation exposure. This review first summarizes recent studies on lung cancer screening from the US, Europe, and Asia, and discusses risk-based selection for screening and the related issues. Second, an overview of novel techniques for the differential diagnosis of pulmonary nodules, including artificial intelligence and molecular biomarker-based screening, is presented. Third, current explorations of strategies for suspected malignancy are summarized. Overall, this review aims to help clinicians understand recent progress in lung cancer screening and alleviate the burden of lung cancer.
Collapse
Affiliation(s)
- Caichen Li
- Department of Thoracic Oncology and Surgery, the First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
- Dongguan Affiliated Hospital of Southern Medical University, Dongguan People Hospital, Dongguan 523059, China
| | - Huiting Wang
- Department of Thoracic Oncology and Surgery, the First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
- Dongguan Affiliated Hospital of Southern Medical University, Dongguan People Hospital, Dongguan 523059, China
| | - Yu Jiang
- Dongguan Affiliated Hospital of Southern Medical University, Dongguan People Hospital, Dongguan 523059, China
| | - Wenhai Fu
- Department of Thoracic Oncology and Surgery, the First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Xiwen Liu
- Dongguan Affiliated Hospital of Southern Medical University, Dongguan People Hospital, Dongguan 523059, China
| | - Ran Zhong
- Department of Thoracic Oncology and Surgery, the First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
- Dongguan Affiliated Hospital of Southern Medical University, Dongguan People Hospital, Dongguan 523059, China
| | - Bo Cheng
- Dongguan Affiliated Hospital of Southern Medical University, Dongguan People Hospital, Dongguan 523059, China
| | - Feng Zhu
- Department of Internal Medicine, Detroit Medical Center Sinai-Grace Hospital, Detroit, Michigan 48235, USA
| | - Yang Xiang
- Department of Thoracic Oncology and Surgery, the First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
| | - Jianxing He
- Dongguan Affiliated Hospital of Southern Medical University, Dongguan People Hospital, Dongguan 523059, China
- Department of Thoracic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou 510515, China
| | - Wenhua Liang
- Department of Thoracic Oncology and Surgery, the First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510120, China
- Dongguan Affiliated Hospital of Southern Medical University, Dongguan People Hospital, Dongguan 523059, China
- Department of Oncology, the First People’s Hospital of Zhaoqing, Zhaoqing 526020, China
| |
Collapse
|
19
|
Xu L, Chang N, Yang T, Lang Y, Zhang Y, Che Y, Xi H, Zhang W, Song Q, Zhou Y, Yang X, Yang J, Qu S, Zhang J. Development of Diagnosis Model for Early Lung Nodules Based on a Seven Autoantibodies Panel and Imaging Features. Front Oncol 2022; 12:883543. [PMID: 35530343 PMCID: PMC9069812 DOI: 10.3389/fonc.2022.883543] [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: 02/25/2022] [Accepted: 03/15/2022] [Indexed: 11/30/2022] Open
Abstract
Background There is increasing incidence of pulmonary nodules due to the promotion and popularization of low-dose computed tomography (LDCT) screening for potential populations with suspected lung cancer. However, a high rate of false-positive and concern of radiation-related cancer risk of repeated CT scanning remains a major obstacle to its wide application. Here, we aimed to investigate the clinical value of a non-invasive and simple test, named the seven autoantibodies (7-AABs) assay (P53, PGP9.5, SOX2, GAGE7, GUB4-5, MAGEA1, and CAGE), in distinguishing malignant pulmonary diseases from benign ones in routine clinical practice, and construct a neural network diagnostic model with the development of machine learning methods. Method A total of 933 patients with lung diseases and 744 with lung nodules were identified. The serum levels of the 7-AABs were tested by an enzyme-linked Immunosorbent assay (ELISA). The primary goal was to assess the sensitivity and specificity of the 7-AABs panel in the detection of lung cancer. ROC curves were used to estimate the diagnosis potential of the 7-AABs in different groups. Next, we constructed a machine learning model based on the 7-AABs and imaging features to evaluate the diagnostic efficacy in lung nodules. Results The serum levels of all 7-AABs in the malignant lung diseases group were significantly higher than that in the benign group. The sensitivity and specificity of the 7-AABs panel test were 60.7% and 81.5% in the whole group, and 59.7% and 81.1% in cases with early lung nodules. Comparing to the 7-AABs panel test alone, the neural network model improved the AUC from 0.748 to 0.96 in patients with pulmonary nodules. Conclusion The 7-AABs panel may be a promising method for early detection of lung cancer, and we constructed a new diagnostic model with better efficiency to distinguish malignant lung nodules from benign nodules which could be used in clinical practice.
Collapse
Affiliation(s)
- Leidi Xu
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Ning Chang
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Tingyi Yang
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yuxiang Lang
- National Science Library, Chinese Academy of Sciences, Beijing, China
| | - Yong Zhang
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yinggang Che
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Hangtian Xi
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Weiqi Zhang
- Department of Radiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | | | - Ying Zhou
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xuemin Yang
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Juanli Yang
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Shuoyao Qu
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jian Zhang
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
20
|
Belousov PV. The Autoantibodies against Tumor-Associated Antigens as Potential Blood-Based Biomarkers in Thyroid Neoplasia: Rationales, Opportunities and Challenges. Biomedicines 2022; 10:biomedicines10020468. [PMID: 35203677 PMCID: PMC8962333 DOI: 10.3390/biomedicines10020468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 11/24/2022] Open
Abstract
The Autoantibodies targeting Tumor-Associated Antigens (TAA-AAbs) emerge as a result of a variety of tumor-related immunogenic stimuli and may be regarded as the eyewitnesses to the anti-tumor immune response. TAA-AAbs may be readily detected in peripheral blood to unveil the presence of a particular TAA-expressing tumor, and a fair number of TAAs eliciting the tumor-associated autoantibody response have been identified. The potential of TAA-AAbs as tumor biomarkers has been extensively studied in many human malignancies with a major influence on public health; however, tumors of the endocrine system, and, in particular, the well-differentiated follicular cell-derived thyroid neoplasms, remain understudied in this context. This review provides a detailed perspective on and legitimate rationales for the potential use of TAA-AAbs in thyroid neoplasia, with particular reference to the already established diagnostic implications of the TAA-AAbs in human cancer, to the windows for improvement and diagnostic niches in the current workup strategies in nodular thyroid disease and differentiated thyroid cancer that TAA-AAbs may successfully occupy, as well as to the proof-of-concept studies demonstrating the usefulness of TAA-AAbs in thyroid oncology, particularly for the pre-surgical discrimination between tumors of different malignant potential in the context of the indeterminate results of the fine-needle aspiration cytology.
Collapse
Affiliation(s)
- Pavel V. Belousov
- National Center for Personalized Medicine of Endocrine Diseases, National Medical Research Center for Endocrinology, Ministry of Health of the Russian Federation, 117036 Moscow, Russia; or
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
21
|
Ding Z, Wang N, Ji N, Chen ZS. Proteomics technologies for cancer liquid biopsies. Mol Cancer 2022; 21:53. [PMID: 35168611 PMCID: PMC8845389 DOI: 10.1186/s12943-022-01526-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/31/2022] [Indexed: 02/07/2023] Open
Abstract
Alterations in DNAs could not reveal what happened in proteins. The accumulated alterations of DNAs would change the manifestation of proteins. Therefore, as is the case in cancer liquid biopsies, deep proteome profiling will likely provide invaluable and clinically relevant information in real-time throughout all stages of cancer progression. However, due to the great complexity of proteomes in liquid biopsy samples and the limitations of proteomic technologies compared to high-plex sequencing technologies, proteomic discoveries have yet lagged behind their counterpart, genomic technologies. Therefore, novel protein technologies are in urgent demand to fulfill the goals set out for biomarker discovery in cancer liquid biopsies.Notably, conventional and innovative technologies are being rapidly developed for proteomic analysis in cancer liquid biopsies. These advances have greatly facilitated early detection, diagnosis, prognosis, and monitoring of cancer evolution, adapted or adopted in response to therapeutic interventions. In this paper, we review the high-plex proteomics technologies that are capable of measuring at least hundreds of proteins simultaneously from liquid biopsy samples, ranging from traditional technologies based on mass spectrometry (MS) and antibody/antigen arrays to innovative technologies based on aptamer, proximity extension assay (PEA), and reverse phase protein arrays (RPPA).
Collapse
Affiliation(s)
- Zhiyong Ding
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Gangxing 3rd Rd, High-Tech and Innovation Zone, Bldg. 2, Rm. 2201, Jinan City, Shandong Province 250101 P. R. China
| | - Nan Wang
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Gangxing 3rd Rd, High-Tech and Innovation Zone, Bldg. 2, Rm. 2201, Jinan City, Shandong Province 250101 P. R. China
| | - Ning Ji
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for Biotechnology, St. John’s University, 8000 Utopia Parkway, Queens, New York, 11439 USA
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for Biotechnology, St. John’s University, 8000 Utopia Parkway, Queens, New York, 11439 USA
| |
Collapse
|
22
|
García-Río F, Alcázar-Navarrete B, Castillo-Villegas D, Cilloniz C, García-Ortega A, Leiro-Fernández V, Lojo-Rodriguez I, Padilla-Galo A, Quezada-Loaiza CA, Rodriguez-Portal JA, Sánchez-de-la-Torre M, Sibila O, Martínez-García MA. [Translated article] Biological Biomarkers in Respiratory Diseases. ARCHIVOS DE BRONCONEUMOLOGÍA 2022. [DOI: 10.1016/j.arbres.2022.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
23
|
Zhang X, Li J, Wang Y, Liu M, Liu F, Zhang X, Pei L, Wang T, Jiang D, Wang X, Zhang J, Dai L. A Diagnostic Model With IgM Autoantibodies and Carcinoembryonic Antigen for Early Detection of Lung Adenocarcinoma. Front Immunol 2022; 12:728853. [PMID: 35140701 PMCID: PMC8818794 DOI: 10.3389/fimmu.2021.728853] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Immunoglobulin M (IgM) autoantibodies, as the early appearing antibodies in humoral immunity when stimulated by antigens, might be excellent biomarkers for the early detection of lung cancer (LC). We aimed to develop a multi-analyte integrative model combining IgM autoantibodies and a traditional tumor biomarker that could be a valuable and powerful auxiliary diagnostic tool and might improve the accuracy of early detection of lung adenocarcinoma (LUAD). A customized protein array based on cancer driver genes was constructed and applied in the discovery cohort consisting of 68 LUAD patients and 68 normal controls (NCs); 31 differentially expressed IgM autoantibodies were identified. The top 5 candidate IgM autoantibodies [based on the area under the receiver operating characteristic curve (AUC) ranking], namely, TSHR, ERBB2, survivin, PIK3CA, and JAK2, were validated in the validation cohort using enzyme-linked immunosorbent assay (ELISA), which included 147 LUAD samples, 72 lung squamous cell carcinoma (LUSC) samples, 44 small cell lung carcinoma (SCLC) samples, and 147 NCs. These indicators presented diagnostic capacity for LUAD, with AUCs of 0.599, 0.613, 0.579, 0.601, and 0.633, respectively (p < 0.05). However, none of them showed a significant difference between the SCLC and NC groups, and only the IgM autoantibody against JAK2 showed a higher expression in LUSC than in NC (p = 0.046). Through logistic regression analysis, with the five IgM autoantibodies and carcinoembryonic antigen (CEA), one diagnostic model was constructed for LUAD. The model yielded an AUC of 0.827 (sensitivity = 56.63%, specificity = 93.98%). The diagnostic efficiency was superior to that of either CEA (AUC = 0.692) or IgM autoantibodies alone (AUC = 0.698). Notably, the accuracy of this model in early-stage LUAD reached 83.02%. In conclusion, we discovered and identified five novel IgM indicators and developed a multi-analyte model combining IgM autoantibodies and CEA, which could be a valuable and powerful auxiliary diagnostic tool and might improve the accuracy of early detection of LUAD.
Collapse
Affiliation(s)
- Xue Zhang
- Henan Institute of Medical and Pharmaceutical Sciences & School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Jiaqi Li
- Henan Institute of Medical and Pharmaceutical Sciences & School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Yulin Wang
- Henan Institute of Medical and Pharmaceutical Sciences & School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Man Liu
- Henan Institute of Medical and Pharmaceutical Sciences & School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Fenghui Liu
- Department of Respiratory and Sleep Medicine in the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Lu Pei
- Department of Clinical Laboratory, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, China
| | - Tingting Wang
- Department of Clinical Laboratory, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Di Jiang
- Henan Institute of Medical and Pharmaceutical Sciences & School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Xiao Wang
- Henan Institute of Medical and Pharmaceutical Sciences & School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Jianying Zhang
- Henan Institute of Medical and Pharmaceutical Sciences & School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences & School of Basic Medical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- *Correspondence: Liping Dai,
| |
Collapse
|
24
|
Garcia-Rio F, Alcázar B, Castillo D, Cilloniz C, García-Ortega A, Leiro-Fernández V, Lojo-Rodriguez I, Padilla A, Quezada CA, Rodriguez-Portal JA, Sánchez-de-la-Torre M, Sibila O, Martinez-Garcia MA. Biomarcadores biológicos en las enfermedades respiratorias. Arch Bronconeumol 2022; 58:323-333. [DOI: 10.1016/j.arbres.2022.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 11/26/2022]
|
25
|
Assessment of background levels of autoantibodies as a prognostic marker for severe SARS-CoV-2 infection. J Circ Biomark 2022; 11:24-27. [PMID: 35517714 PMCID: PMC9069225 DOI: 10.33393/jcb.2022.2337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/20/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Patients with more severe forms of SARS-CoV-2 exhibit activation of immunological cascades. Participants (current or ex-smokers with at least 20 years pack history) in a trial (Early Diagnosis of Lung Cancer, Scotland [ECLS]) of autoantibody detection to predict lung cancer risk had seven autoantibodies measured 5 years before the pandemic. This study compared the response to Covid infection in study participants who tested positive and negative to antibodies to tumour-associated antigens: p53, NY-ESO-1, CAGE, GBU4-5, HuD, MAGE A4 and SOX2. Methods: Autoantibody data from the ECLS study was deterministically linked to the EAVE II database, a national, real-time prospective cohort using Scotland’s health data infrastructure, to describe the epidemiology of SARS-CoV-2 infection, patterns of healthcare use and outcomes. The strength of associations was explored using a network algorithm for exact contingency table significance testing by permutation. Results: There were no significant differences discerned between SARS-CoV-2 test results and EarlyCDT-Lung test results (p = 0.734). An additional analysis of intensive care unit (ICU) admissions detected no significant differences between those who tested positive and negative. Subgroup analyses showed no difference in COVID-19 positivity or death rates amongst those diagnosed with chronic obstructive pulmonary disease (COPD) with positive and negative EarlyCDT results. Conclusions: This hypothesis-generating study demonstrated no clinically valuable or statistically significant associations between EarlyCDT positivity in 2013-15 and the likelihood of SARS-CoV-2 positivity in 2020, ICU admission or death in all participants (current or ex-smokers with at least 20 years pack history) or in those with COPD or lung cancer.
Collapse
|
26
|
Tan Q, Dai L, Wang Y, Liu S, Liang T, Luo R, Wang S, Lou N, Chen H, Zhou Y, Zhong Q, Yang J, Xing P, Hu X, Liu Y, Zhou S, Yao J, Wu D, Zhang Z, Tang L, Yu X, Han X, Shi Y. Anti-PD1/PDL1 IgG subclass distribution in ten cancer types and anti-PD1 IgG4 as biomarker for the long time survival in NSCLC with anti-PD1 therapy. Cancer Immunol Immunother 2021; 71:1681-1691. [PMID: 34817638 DOI: 10.1007/s00262-021-03106-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/28/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Antibodies targeting programmed cell death-1(PD1) and its ligand (PDL1) have revolutionized cancer therapy. However, little is known about the preexisted anti-PD1/PDL1 autoantibodies (AAbs) distribution in multiple cancer types, nor is their potential biomarker role for anti-PD1 therapy. METHOD Plasma anti-PD1/PDL1 AAb IgG and subclasses (IgG1-4) were detected by enzyme-linked immune sorbent assay (ELISA) in 190 cancer patients, covering 10 cancer types (lung, breast, esophageal, colorectal, liver, prostatic, cervical, ovarian, gastric cancers and lymphoma), the comprehensive correlation of AAbs with multiple clinical parameters was analyzed. We further tested these AAbs in 76 non-small cell lung cancer (NSCLC) samples receiving anti-PD1 therapy, the association of AAbs level with survival was analyzed and validated in an independent cohort (n = 32). RESULTS Anti-PD1/PDL1 AAb IgG were globally detected in 10 types of cancer patients. IgG1 and IgG2 were the major subtypes for anti-PD1/PDL1 AAbs. Correlation analysis revealed a distinct landscape between various cancer types. The random forest model indicated that IgG4 subtype was mostly associated with cancer. In discovery cohort of 76 NSCLC patients, high anti-PD1 IgG4 was associated with a reduced overall survival (OS, p = 0.019), not progression-free survival (PFS, p = 0.088). The negative association of anti-PD1 IgG4 with OS was validated in 32 NSCLC patients (p = 0.032). CONCLUSION This study reports for the first time the distribution of preexisted anti-PD1/PDL1 AAb IgG and subclasses across 10 cancer types. Moreover, the anti-PD1 AAb IgG4 subclass was identified to associate with OS, which may serve as a potential biomarker for anti-PD1 therapeutic survival benefit in NSCLC patients.
Collapse
Affiliation(s)
- Qiaoyun Tan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Liyuan Dai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Yanrong Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Shuxia Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Te Liang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Rongrong Luo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Shasha Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Ning Lou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Haizhu Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Yu Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Qiaofeng Zhong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Jianliang Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Xingsheng Hu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Shengyu Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Jiarui Yao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Di Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Zhishang Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100032, China.
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China.
| |
Collapse
|
27
|
Toumazis I, Erdogan SA, Bastani M, Leung A, Plevritis SK. A Cost-Effectiveness Analysis of Lung Cancer Screening With Low-Dose Computed Tomography and a Diagnostic Biomarker. JNCI Cancer Spectr 2021; 5:pkab081. [PMID: 34738073 PMCID: PMC8564700 DOI: 10.1093/jncics/pkab081] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/12/2021] [Accepted: 08/20/2021] [Indexed: 12/17/2022] Open
Abstract
Background The Lung Computed Tomography Screening Reporting and Data System (Lung-RADS) reduces the false-positive rate of lung cancer screening but introduces prolonged periods of uncertainty for indeterminate findings. We assess the cost-effectiveness of a screening program that assesses indeterminate findings earlier via a hypothetical diagnostic biomarker introduced in place of Lung-RADS 3 and 4A guidelines. Methods We evaluated the performance of the US Preventive Services Task Force (USPSTF) recommendations on lung cancer screening with and without a hypothetical noninvasive diagnostic biomarker using a validated microsimulation model. The diagnostic biomarker assesses the malignancy of indeterminate nodules, replacing Lung-RADS 3 and 4A guidelines, and is characterized by a varying sensitivity profile that depends on nodules' size, specificity, and cost. We tested the robustness of our findings through univariate sensitivity analyses. Results A lung cancer screening program per the USPSTF guidelines that incorporates a diagnostic biomarker with at least medium sensitivity profile and 90% specificity, that costs $250 or less, is cost-effective with an incremental cost-effectiveness ratio lower than $100 000 per quality-adjusted life year, and improves lung cancer-specific mortality reduction while requiring fewer screening exams than the USPSTF guidelines with Lung-RADS. A screening program with a biomarker costing $750 or more is not cost-effective. The health benefits accrued and costs associated with the screening program are sensitive to the disutility of indeterminate findings and specificity of the biomarker, respectively. Conclusions Lung cancer screening that incorporates a diagnostic biomarker, in place of Lung-RADS 3 and 4A guidelines, could improve the cost-effectiveness of the screening program and warrants further investigation.
Collapse
Affiliation(s)
- Iakovos Toumazis
- Departments of Biomedical Data Science and Radiology, Stanford University, Stanford, CA, USA
| | - S Ayca Erdogan
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Mehrad Bastani
- Departments of Biomedical Data Science and Radiology, Stanford University, Stanford, CA, USA
| | - Ann Leung
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Sylvia K Plevritis
- Departments of Biomedical Data Science and Radiology, Stanford University, Stanford, CA, USA
| |
Collapse
|
28
|
Wang M, Liu F, Pan Y, Xu R, Li F, Liu A, Yang H, Duan L, Shen L, Wu Q, Liu Y, Liu M, Liu Z, Hu Z, Chen H, Cai H, He Z, Ke Y. Tumor-associated autoantibodies in ESCC screening: Detecting prevalent early-stage malignancy or predicting future cancer risk? EBioMedicine 2021; 73:103674. [PMID: 34753106 PMCID: PMC8586741 DOI: 10.1016/j.ebiom.2021.103674] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND To assess potential roles for tumor-associated autoantibodies (TAAs) in esophageal squamous cell carcinoma (ESCC) screening: detecting early-stage malignancy, and predicting future cancer risk. METHOD Thirteen candidate autoantibodies identified in previous literatures were measured using multiplex serological assays in sera from cases and matched controls nested in two population-level screening cohorts in China. To evaluate the role of TAAs in detecting prevalent esophageal malignant lesions, an identification set (150 cases vs. 560 controls) and an external validation set (34 cases vs. 121 controls) were established with pre-screening sera collected ≤ 12 months prior to screening-related diagnosis. To explore the role of TAAs in predicting future ESCC risk, an exploration set (105 cases vs. 416 controls) with pre-diagnostic sera collected > 12 months before clinical diagnosis was established. Two models, the questionnaire-based model and full model additionally incorporating TAA markers, were constructed. Area under the receiver operating characteristic curve (AUC) and net reclassification improvement (NRI) were calculated to compare the performance of the two models. FINDINGS In the identification set, NY-ESO-1 (OR=2·12, 95% CI=1·02-4·40) and STIP1 (OR=1·83, 95% CI=1·10-3·05) were positively associated with higher risk of esophageal malignancy. Elevated MMP-7 was associated with higher risk of malignancy in females (ORfemale=5·07, 95% CI=1·30-19·71). The estimates in validation set were consistent with these results, but were close to null in exploration set. Integration of selected TAAs improved the performance of questionnaire-based models in detecting prevalent esophageal malignancy (female: AUCfull model=0·745, 95% CI=0·675-0·814, AUCquestionnaire-based model=0·658, 95% CI=0·585-0·732, NRI=0·604, P<0·0001; male: AUCfull model=0·662, 95% CI=0·596-0·728, AUCquestionnaire-based model=0·619, 95% CI=0·548-0·690, NRI=0·357, P=0·0028). This improvement was also seen in validation set, but was not similarly effective in distinguishing long-term incident cases from healthy controls. INTERPRETATION Serological autoantibodies against NY-ESO-1, STIP1, and MMP-7 perform well in detecting early-stage esophageal malignancy, but are less effective in predicting future ESCC risks. FUNDING This work was supported by the National Science & Technology Fundamental Resources Investigation Program of China (2019FY101102), the National Natural Science Foundation of China (82073626), the National Key R&D Program of China (2016YFC0901404), the Beijing-Tianjin-Hebei Basic Research Cooperation Project (J200016), the Digestive Medical Coordinated Development Center of Beijing Hospitals Authority (XXZ0204), and the Natural Science Foundation of Beijing Municipality (7182033).
Collapse
Affiliation(s)
- Minmin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Fangfang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Yaqi Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Ruiping Xu
- Anyang Cancer Hospital, Anyang, Henan Province, P.R. China
| | - Fenglei Li
- Hua County People's Hospital, Anyang, Henan Province, P.R. China
| | - Anxiang Liu
- Endoscopy center, Anyang Cancer Hospital, Anyang, Henan Province, P.R. China
| | - Haijun Yang
- Department of pathology, Anyang Cancer Hospital, Anyang, Henan Province, P.R. China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing, P.R. China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Qi Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Endoscopy Center, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Ying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Mengfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Zhen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Zhe Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Huanyu Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Hong Cai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China
| | - Zhonghu He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| | - Yang Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, P.R. China.
| |
Collapse
|
29
|
Abstract
This study aimed to evaluate the diagnostic efficacy of seven autoantibodies in all lung cancer, lung adenocarcinoma, lung squamous cell carcinoma and early-stage lung cancer patients. ELISA testing of a seven autoantibody panel was performed on 386 lung cancer patients and 238 normal controls. The sensitivity and specificity of each autoantibody were analyzed using the receiver operating characteristic curve analysis. The diagnostic efficacy of a combination of these seven autoantibodies was evaluated by binary logistic regression. The results indicated that six of the seven autoantibodies (p53, SOX2, GAGE7, GBU4-5, MAGEA1 and CAGE) had high specificity and low sensitivity, while PGP9.5 had high sensitivity and low specificity. Further analysis showed that all seven autoantibodies had better diagnostic value in lung squamous cell carcinoma patients when compared to lung adenocarcinoma or all lung cancer patients. Logistic regression showed that a combination of the seven autoantibodies resulted in more reliable detection of lung cancer than any individual autoantibody in early-stage lung cancer (sensitivity/specificity: 47.8%/81.4%, areas under the curve: 0.764, 95% confidence interval: 0.718-0.811). Additionally, this panel had a better sensitivity of 56.5% for detection of lung squamous cell carcinoma than for all lung cancer (50.1%) or adenocarcinoma (51.7%) (P < 0.05). Our results indicated that the seven autoantibody panel could be used for early lung cancer detection, and it had better sensitivity in diagnosis of lung squamous cell carcinoma.
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Lung cancer remains the leading cause of cancer-related death in the United States, with poor overall 5-year survival. Early detection and diagnosis are key to survival as demonstrated in lung cancer screening trials. However, with increasing implementation of screening guidelines and use of computed tomography, there has been a sharp rise in the incidence of indeterminate pulmonary nodules (IPNs). Risk stratification of IPNs, particularly those in the intermediate-risk category, remains challenging in clinical practice. Individual risk factors, imaging characteristics, biomarkers, and prediction models are currently used to assist in risk stratifying patients, but such strategies remain suboptimal. This review focuses on established risk stratification methods, current areas of research, and future directions. RECENT FINDINGS The multitude of yearly incidental and screening-detected IPNs, its management-related healthcare costs, and risk of invasive procedures provides a strong rationale for risk stratification efforts. The development of new molecular and imaging biomarkers to discriminate benign from malignant lung nodules shows great promise. Yet, risk stratification methods need integration into the diagnostic workflow and await validation in prospective, biomarker-driven clinical trials. SUMMARY Novel biomarkers and new imaging analysis, including radiomics and deep-learning methods, have been developed to optimize the risk stratification of IPNs. While promising, additional validation and clinical studies are needed before they can be part of routine clinical practice.
Collapse
Affiliation(s)
- Rafael Paez
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center
| | - Michael N Kammer
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center
- Department of Chemistry, Vanderbilt University
| | - Pierre Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center
- Pulmonary and Critical Care Section, Medical Service, Tennessee Valley Healthcare System, Nashville Campus, Nashville, Tennessee, USA
| |
Collapse
|
31
|
Borg M, Wen SWC, Nederby L, Hansen TF, Jakobsen A, Andersen RF, Weinreich UM, Hilberg O. Performance of the EarlyCDT® Lung test in detection of lung cancer and pulmonary metastases in a high-risk cohort. Lung Cancer 2021; 158:85-90. [PMID: 34130044 DOI: 10.1016/j.lungcan.2021.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Early detection of lung cancer is pivotal for an optimal prognosis. CT screening is currently implemented in USA. To decrease the amount of CT scans, the application of a blood-based biomarker as part of screening criteria is desirable. MATERIALS AND METHODS The EarlyCDT® Lung test was performed in a high-risk cohort composed 246 patients referred from their GP on suspicion of lung cancer. Blood samples were taken at first visit and patients underwent diagnostic workup on suspicion of lung cancer resulting in either a malignant diagnosis or ruled out cancer. Sensitivity and specificity of the EarlyCDT® Lung were calculated in the cohort and subgroups based on age, smoking history, sex and lung cancer stage. RESULTS Overall sensitivity in the cohort was 33 % for lung cancer and 31 % for primary lung cancer and lung metastases combined. Sensitivity in age groups was 11 % (60 years or below), 31 % (61-75 years) and 55 % (>75 years). In patients with at least 10 tobacco pack years, sensitivity was 33 % while the sensitivity in patients with at least 50 tobacco pack years was 44 %. The assay sensitivity in stage I-II lung cancer patients was 21 %, while this was 40 % in stage III-IV lung cancer patients. In a subgroup of patients that met current CT screening criteria (age 55-80 years and minimum 30 tobacco pack years) the sensitivity was 37 %. CONCLUSION The rationale of screening for lung cancer is to find patients in an early and resectable stage. However, the EarlyCDT® Lung test performed best in elderly, late stage lung cancer patients with a heavy smoking history. Based on these results, the current study finds insufficient sensitivity of the EarlyCDT® Lung test to be used as part of inclusion criteria in a low-dose CT program for detection of lung cancer.
Collapse
Affiliation(s)
- Morten Borg
- Department of Respiratory Diseases, Aalborg University Hospital, Mølleparkvej 4, 9000 Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, 9000 Aalborg, Denmark.
| | - Sara W C Wen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 3, 5000 Odense C, Denmark.
| | - Line Nederby
- Department of Clinical Biochemistry, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark.
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 3, 5000 Odense C, Denmark.
| | - Anders Jakobsen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 3, 5000 Odense C, Denmark.
| | - Rikke Fredslund Andersen
- Department of Clinical Biochemistry, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark.
| | - Ulla Møller Weinreich
- Department of Respiratory Diseases, Aalborg University Hospital, Mølleparkvej 4, 9000 Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, 9000 Aalborg, Denmark.
| | - Ole Hilberg
- Institute of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 3, 5000 Odense C, Denmark; Department of Internal Medicine, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark.
| |
Collapse
|
32
|
The Value of a Seven-Autoantibody Panel Combined with the Mayo Model in the Differential Diagnosis of Pulmonary Nodules. DISEASE MARKERS 2021; 2021:6677823. [PMID: 33688380 PMCID: PMC7914080 DOI: 10.1155/2021/6677823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/26/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022]
Abstract
Background Identifying malignant pulmonary nodules and detecting early-stage lung cancer (LC) could reduce mortality. This study investigated the clinical value of a seven-autoantibody (7-AAB) panel in combination with the Mayo model for the early detection of LC and distinguishing benign from malignant pulmonary nodules (MPNs). Methods The concentrations of the elements of a 7-AAB panel were quantitated by enzyme-linked immunosorbent assay (ELISA) in 806 participants. The probability of MPNs was calculated using the Mayo predictive model. The performances of the 7-AAB panel and the Mayo model were analyzed by receiver operating characteristic (ROC) analyses, and the difference between groups was evaluated by chi-square tests (χ2). Results The combined area under the ROC curve (AUC) for all 7 AABs was higher than that of a single one. The sensitivities of the 7-AAB panel were 67.5% in the stage I-II LC patients and 60.3% in the stage III-IV patients, with a specificity of 89.6% for the healthy controls and 83.1% for benign lung disease patients. The detection rate of the 7-AAB panel in the early-stage LC patients was higher than that of traditional tumor markers. The AUC of the 7-AAB panel in combination with the Mayo model was higher than that of the 7-AAB panel alone or the Mayo model alone in distinguishing MPN from benign nodules. For early-stage MPN, the sensitivity and specificity of the combination were 93.5% and 58.0%, respectively. For advanced-stage MPN, the sensitivity and specificity of the combination were 91.4% and 72.8%, respectively. The combination of the 7-AAB panel with the Mayo model significantly improved the detection rate of MPN, but the positive predictive value (PPV) and the specificity were not improved when compared with either the 7-AAB panel alone or the Mayo model alone. Conclusion Our study confirmed the clinical value of the 7-AAB panel for the early detection of lung cancer and in combination with the Mayo model could be used to distinguish benign from malignant pulmonary nodules.
Collapse
|
33
|
de Jonge H, Iamele L, Maggi M, Pessino G, Scotti C. Anti-Cancer Auto-Antibodies: Roles, Applications and Open Issues. Cancers (Basel) 2021; 13:813. [PMID: 33672007 PMCID: PMC7919283 DOI: 10.3390/cancers13040813] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Auto-antibodies are classically associated with autoimmune diseases, where they are an integral part of diagnostic panels. However, recent evidence is accumulating on the presence of auto-antibodies against single or selected panels of auto-antigens in many types of cancer. Auto-antibodies might initially represent an epiphenomenon derived from the inflammatory environment induced by the tumor. However, their effect on tumor evolution can be crucial, as is discussed in this paper. It has been demonstrated that some of these auto-antibodies can be used for early detection and cancer staging, as well as for monitoring of cancer regression during treatment and follow up. Interestingly, certain auto-antibodies were found to promote cancer progression and metastasis, while others contribute to the body's defense against it. Moreover, auto-antibodies are of a polyclonal nature, which means that often several antibodies are involved in the response to a single tumor antigen. Dissection of these antibody specificities is now possible, allowing their identification at the genetic, structural, and epitope levels. In this review, we report the evidence available on the presence of auto-antibodies in the main cancer types and discuss some of the open issues that still need to be addressed by the research community.
Collapse
Affiliation(s)
| | | | | | | | - Claudia Scotti
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy; (H.d.J.); (L.I.); (M.M.); (G.P.)
| |
Collapse
|
34
|
Rodríguez M, Ajona D, Seijo LM, Sanz J, Valencia K, Corral J, Mesa-Guzmán M, Pío R, Calvo A, Lozano MD, Zulueta JJ, Montuenga LM. Molecular biomarkers in early stage lung cancer. Transl Lung Cancer Res 2021; 10:1165-1185. [PMID: 33718054 PMCID: PMC7947407 DOI: 10.21037/tlcr-20-750] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Low dose computed tomography (LDCT) screening, together with the recent advances in targeted and immunotherapies, have shown to improve non-small cell lung cancer (NSCLC) survival. Furthermore, screening has increased the number of early stage-detected tumors, allowing for surgical resection and multimodality treatments when needed. The need for improved sensitivity and specificity of NSCLC screening has led to increased interest in combining clinical and radiological data with molecular data. The development of biomarkers is poised to refine inclusion criteria for LDCT screening programs. Biomarkers may also be useful to better characterize the risk of indeterminate nodules found in the course of screening or to refine prognosis and help in the management of screening detected tumors. The clinical implications of these biomarkers are still being investigated and whether or not biomarkers will be included in further decision-making algorithms in the context of screening and early lung cancer management still needs to be determined. However, it seems clear that there is much room for improvement even in early stage lung cancer disease-free survival (DFS) rates; thus, biomarkers may be the key to refine risk-stratification and treatment of these patients. Clinicians’ capacity to register, integrate, and analyze all the available data in both high risk individuals and early stage NSCLC patients will lead to a better understanding of the disease’s mechanisms, and will have a direct impact in diagnosis, treatment, and follow up of these patients. In this review, we aim to summarize all the available data regarding the role of biomarkers in LDCT screening and early stage NSCLC from a multidisciplinary perspective. We have highlighted clinical implications, the need to combine risk stratification, clinical data, radiomics, molecular information and artificial intelligence in order to improve clinical decision-making, especially regarding early diagnostics and adjuvant therapy. We also discuss current and future perspectives for biomarker implementation in routine clinical practice.
Collapse
Affiliation(s)
- María Rodríguez
- Department of Thoracic Surgery, Clínica Universidad de Navarra, Madrid, Spain
| | - Daniel Ajona
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Luis M Seijo
- Department of Pulmonology, Clínica Universidad de Navarra, Madrid, Spain.,Centro de Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Julián Sanz
- Department of Pathology, Clínica Universidad de Navarra, Madrid, Spain
| | - Karmele Valencia
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Jesús Corral
- Department of Oncology, Clínica Universidad de Navarra, Madrid, Spain
| | - Miguel Mesa-Guzmán
- Department of Thoracic Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rubén Pío
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Alfonso Calvo
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Anatomy and Physiology, Schools of Medicine and Sciences, University of Navarra, Pamplona, Spain
| | - María D Lozano
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Anatomy and Physiology, Schools of Medicine and Sciences, University of Navarra, Pamplona, Spain.,Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier J Zulueta
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pulmonology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Luis M Montuenga
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Anatomy and Physiology, Schools of Medicine and Sciences, University of Navarra, Pamplona, Spain
| |
Collapse
|
35
|
González Maldonado S, Johnson T, Motsch E, Delorme S, Kaaks R. Can autoantibody tests enhance lung cancer screening?-an evaluation of EarlyCDT ®-Lung in context of the German Lung Cancer Screening Intervention Trial (LUSI). Transl Lung Cancer Res 2021; 10:233-242. [PMID: 33569307 PMCID: PMC7867751 DOI: 10.21037/tlcr-20-727] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Tumor-associated autoantibodies are considered promising markers for early lung cancer detection; so far, however, their capacity to detect cancer has been tested mostly in a clinical context, but not in population screening settings. This study evaluates the early detection accuracy, in terms of sensitivity and specificity, of EarlyCDT®-Lung-a test panel of seven tumor-associated autoantibodies optimized for lung cancer detection-using blood samples originally collected as part of the German Lung Cancer Screening Intervention Trial. Methods The EarlyCDT®-Lung test was performed for all participants with lung cancer detected via low-dose computed tomography and with available blood samples taken at detection, and for 180 retrospectively selected cancer-free participants at the end of follow-up: 90 randomly selected from among all cancer-free participants (baseline controls) and 90 randomly selected from among cancer-free participants with suspicious imaging findings (suspicious nodules controls). Sensitivity and specificity of lung cancer detection were estimated in the case group and the two control groups, respectively. Results In the case group, the test panel showed a sensitivity of only 13.0% (95% CI: 4.9-26.3%). Specificity was estimated at 88.9% (95% CI: 80.5-94.5%) in the baseline control group, and 91.1% (95% CI: 83.2-96.1%) among controls presenting CT-detected nodules. Conclusions The test panel showed insufficient sensitivity for detecting lung cancer at an equally early stage as with low-dose computed tomography screening.
Collapse
Affiliation(s)
- Sandra González Maldonado
- Division of Cancer Epidemiology (C020), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research, Heidelberg, Germany
| | - Theron Johnson
- Division of Cancer Epidemiology (C020), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
| | - Erna Motsch
- Division of Cancer Epidemiology (C020), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research, Heidelberg, Germany
| | - Stefan Delorme
- Division of Radiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology (C020), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research, Heidelberg, Germany
| |
Collapse
|
36
|
Autoantibodies against tumor-associated antigens in sputum as biomarkers for lung cancer. Transl Oncol 2020; 14:100991. [PMID: 33333369 PMCID: PMC7736713 DOI: 10.1016/j.tranon.2020.100991] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/28/2022] Open
Abstract
Tumor antigens (TAs) can initiate host immune responses and produce TA-associated autoantibody (TAAbs), potential cancer biomarkers. Sputum is directly generated from the upper and lower airways, and thus can be used as a surrogate sample for the diagnosis of lung cancer based on molecular analysis. To develop sputum TAAb biomarkers for the early detection of lung cancer, the leading cause of cancer death, we probed a protein microarray containing more than 9,000 antigens with sputum supernatants of a discovery set of 30 lung cancer patients and 30 cancer-free smokers. Twenty-eight TAs with higher reactivity in sputum of lung cancer cases vs. controls were identified. The diagnostic significance of TAAbs against the TAs was determined by enzyme-linked immunosorbent assays (ELISAs) in sputum of the discovery set and additional 166 lung cancer patients and 213 cancer-free smokers (validation set). Three sputum TAAbs against DDX6, ENO1, and 14-3-3ζ were developed as a biomarker panel with 81% sensitivity and 83% specificity for diagnosis of lung cancer, regardless of stages, locations, and histological types of lung tumors. This study provides the first evidence that sputum TAAbs could be used as biomarkers for the early detection of lung cancer.
Collapse
|
37
|
Ostrin EJ, Sidransky D, Spira A, Hanash SM. Biomarkers for Lung Cancer Screening and Detection. Cancer Epidemiol Biomarkers Prev 2020; 29:2411-2415. [PMID: 33093160 DOI: 10.1158/1055-9965.epi-20-0865] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/01/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
Lung cancer is the leading worldwide cause of cancer mortality, as it is often detected at an advanced stage. Since 2011, low-dose CT scan-based screening has promised a 20% reduction in lung cancer mortality. However, effectiveness of screening has been limited by eligibility only for a high-risk population of heavy smokers and a large number of false positives generated by CT. Biomarkers have tremendous potential to improve early detection of lung cancer by refining lung cancer risk, stratifying positive CT scans, and categorizing intermediate-risk pulmonary nodules. Three biomarker tests (Early CDT-Lung, Nodify XL2, Percepta) have undergone extensive validation and are available to the clinician. The authors discuss these tests, with their clinical applicability and limitations, current ongoing evaluation, and future directions for biomarkers in lung cancer screening and detection.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- Edwin J Ostrin
- Department of General Internal Medicine and Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - David Sidransky
- Department of Otolaryngology, Johns Hopkins Hospital, Baltimore, Maryland
| | - Avrum Spira
- Department of Medicine, Boston University, Boston, Massachusetts.,The Lung Cancer Initiative, Johnson and Johnson, New Brunswick, New Jersey
| | - Samir M Hanash
- McCombs Institute for the Prevention and Treatment of Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
38
|
Zhang X, Liu M, Zhang X, Wang Y, Dai L. Autoantibodies to tumor-associated antigens in lung cancer diagnosis. Adv Clin Chem 2020; 103:1-45. [PMID: 34229848 DOI: 10.1016/bs.acc.2020.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung cancer (LC) accounts for the majority of cancer-related deaths worldwide. Although screening the high-risk population by low-dose CT (LDCT) has reduced mortality, the cost and high false positivity rate has prevented its general diagnostic use. As such, better and more specific minimally invasive biomarkers are needed in general and for early LC detection, specifically. Autoantibodies produced by humoral immune response to tumor-associated antigens (TAA) are emerging as a promising noninvasive biomarker for LC. Given the low sensitivity of any one single autoantibody, a panel approach could provide a more robust and promising strategy to detect early stage LC. In this review, we summarize the background of TAA autoantibodies (TAAb) and the techniques currently used for identifying TAA, as well as recent findings of LC specific antigens and TAAb. This review provides guidance toward the development of accurate and reliable TAAb as immunodiagnostic biomarkers in the early detection of LC.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, Henan, China
| | - Man Liu
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue Zhang
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China
| | - Yulin Wang
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
39
|
SOX2 and squamous cancers. Semin Cancer Biol 2020; 67:154-167. [PMID: 32905832 DOI: 10.1016/j.semcancer.2020.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 11/10/2019] [Accepted: 05/09/2020] [Indexed: 12/20/2022]
Abstract
SOX2 is a pleiotropic nuclear transcription factor with major roles in stem cell biology and in development. Over the last 10 years SOX2 has also been implicated as a lineage-specific oncogene, notably in squamous carcinomas but also neurological tumours, particularly glioblastoma. Squamous carcinomas (SQCs) comprise a common group of malignancies for which there are no targeted therapeutic interventions. In this article we review the molecular epidemiological and laboratory evidence linking SOX2 with squamous carcinogenesis, explore in detail the multifaceted impact of SOX2 in SQC, describe areas of uncertainty and highlight areas for potential future research.
Collapse
|
40
|
Stem signatures associated antibodies yield early diagnosis and precise prognosis predication of patients with non-small cell lung cancer. J Cancer Res Clin Oncol 2020; 147:223-233. [PMID: 32691153 DOI: 10.1007/s00432-020-03325-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 07/14/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND This study was designed to detect patients with early NSCLC with tentatively using the stem signatures associated autoantibodies (AAbs), and to evaluate its latent values in the early diagnosis and precise prognosis prediction. METHODS The serum concentrations of selective antibodies were quantitated by enzyme-linked immunosorbent assay (ELISA), and a total of 458 cases were enrolled (training set = 401; validation set = 57). TCGA databases were used to analyze the distinct expressions and prognostic values of related genes. The optimal cut-off values were 11.60 U/ml for P53, 4.90 U/ml for MAGEA1, 3.85 U/ml for SOX2, and 7.05U/ml for PGP9.5. RESULTS We found that the stem signatures associated antibodies of MAGEA1, PGP9.5, SOX2, and TP53 exhibited high expressions in NSCLC, negatively correlating with the overall survival (OS) (P < 0.05). In the test groups, the diagnosis sensitivity of P53, PGP9.5, SOX2, and MAGEA1 reached to 21.5%, 39.0%, 50.3%, and 35.0%, respectively, and the specificity reached to 98.7%, 99.4%, 92.2%, and 97.4%. The four candidates' panel gave a sensitivity of 71.8% with a specificity of 89%. In the validation group, the detection of the four antibodies in early diagnosis of NSCLC also exhibited high specificity and sensitivity, further consolidating their potential application. CONCLUSIONS The detection regarding stem signatures associated antibodies could be used as effective tools in early NSCLC diagnosis, but not for localized screening of cancers, and their abnormal expression was in accordance with poorer survival.
Collapse
|
41
|
Liu S, Zhang X, Jiang Q, Liang T. Detection of circulating natural antibodies against CD25, MUC1, and VEGFR1 for early diagnosis of non-small cell lung cancer. FEBS Open Bio 2020; 10:1288-1294. [PMID: 32392378 PMCID: PMC7327917 DOI: 10.1002/2211-5463.12878] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/21/2020] [Accepted: 05/07/2020] [Indexed: 12/30/2022] Open
Abstract
We previously demonstrated that a deficiency of natural antibodies against CD25, Mucin 1 (MUC1), and vascular endothelial growth factor receptor 1 (VEGFR1) could contribute to high risk of non-small cell lung cancer (NSCLC). This study was designed to investigate whether natural IgG antibodies against POU domain class 5 transcription factor 1 (POU5F1), tumor necrosis factor-α (TNF-α), and the combination of CD25, VEGFR1, and MUC1 could play an anti-tumorigenic role against developing NSCLC. An ELISA was developed in-house to examine plasma IgG against peptide antigens derived from POU5F1, TNF-α, and a combination of peptide antigens derived from CD25, MUC1, and VEGFR1 in 211 patients with NSCLC and 200 healthy controls. Mann-Whitney U test demonstrated that plasma IgG levels for the combination of peptide antigens derived from CD25, MUC1, and VEGFR1 were significantly lower in NSCLC patients than control subjects (Z = -12.978, P < 0.001) although plasma levels of IgG antibodies for POU5F1 and TNFα were not significantly changed. The in-house ELISA made with the CD25-MUC1-VEGFR1 combination had a sensitivity of 49.6% against a specificity of 95% to detect early-stage NSCLC. In conclusion, natural antibodies against the combination of CD25, VEGFR1, and MUC1 may be an effective biomarker for early diagnosis of NSCLC.
Collapse
Affiliation(s)
- Siqi Liu
- Second Hospital of Jilin UniversityChangchunChina
| | - Xuan Zhang
- Second Hospital of Jilin UniversityChangchunChina
| | | | | |
Collapse
|
42
|
Gasparri R, Sedda G, Spaggiari L. Biomarkers in Early Diagnosis and Early Stage Lung Cancer: The Clinician's Point of View. J Clin Med 2020; 9:E1790. [PMID: 32526831 PMCID: PMC7355900 DOI: 10.3390/jcm9061790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/19/2022] Open
Abstract
Starting from the work of Ulivi and colleagues, we aim to summarize the research area of biomarkers for early diagnosis and early stage lung cancer.
Collapse
Affiliation(s)
- Roberto Gasparri
- Department of Thoracic Surgery, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy; (G.S.); (L.S.)
| | - Giulia Sedda
- Department of Thoracic Surgery, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy; (G.S.); (L.S.)
| | - Lorenzo Spaggiari
- Department of Thoracic Surgery, IEO, European Institute of Oncology IRCCS, Via Ripamonti, 435, 20141 Milan, Italy; (G.S.); (L.S.)
- Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
43
|
Welberry C, Macdonald I, McElveen J, Parsy-Kowalska C, Allen J, Healey G, Irving W, Murray A, Chapman C. Tumor-associated autoantibodies in combination with alpha-fetoprotein for detection of early stage hepatocellular carcinoma. PLoS One 2020; 15:e0232247. [PMID: 32374744 PMCID: PMC7202612 DOI: 10.1371/journal.pone.0232247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 04/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) continues to be a leading challenge in modern oncology. Early detection via blood-based screening tests has the potential to cause a stage-shift at diagnosis and improve clinical outcomes. Tumor associated autoantibodies (TA-AAbs) have previously shown the ability to distinguish HCC from patients with high-risk liver disease. This research aimed to further show the utility of TA-AAbs as biomarkers of HCC and assess their use in combination with Alpha-fetoprotein (AFP) for detection of HCC across multiple tumor stages. METHODS Levels of circulating G class antibodies to 44 recombinant tumor associated antigens and circulating AFP were measured in the serum of patients with HCC, non-cancerous chronic liver disease (NCCLD) and healthy controls via enzyme-linked immunosorbent assay (ELISA). TA-AAb cut-offs were set at the highest Youden's J statistic at a specificity ≥95.00%. Panels of TA-AAbs were formed using net reclassification improvement. AFP was assessed at a cut-off of 200 ng/ml. RESULTS Sensitivities ranged from 1.01% to 12.24% at specificities of 95.96% to 100.00% for single TA-AAbs. An ELISA test measuring a panel of 10 of these TA-AAbs achieved a combined sensitivity of 36.73% at a specificity of 89.89% when distinguishing HCC from NCCLD controls. At a cut-off of 200 ng/ml, AFP achieved a sensitivity of 31.63% at a specificity of 100.00% in the same cohort. Combination of the TA-AAb panel with AFP significantly increased the sensitivity for stage one (40.00%) and two (55.00%) HCC over the TA-AAb panel or AFP alone. CONCLUSIONS A panel of TA-AAbs in combination with AFP could be clinically relevant as a replacement for measuring levels of AFP alone in surveillance and diagnosis strategies. The increased early stage sensitivity could lead to a stage shift with positive prognostic outcomes.
Collapse
Affiliation(s)
- Christopher Welberry
- Oncimmune ltd, Nottingham, United Kingdom
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- * E-mail: ,
| | | | | | | | - Jared Allen
- Oncimmune ltd, Nottingham, United Kingdom
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | | | - William Irving
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, United Kingdom
| | | | - Caroline Chapman
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Bowel Cancer Screening Program, Nottingham University NHS Trust, Nottingham, United Kingdom
| |
Collapse
|
44
|
Wang W, Zhuang R, Ma H, Fang L, Wang Z, Lv W, Hu J. The diagnostic value of a seven-autoantibody panel and a nomogram with a scoring table for predicting the risk of non-small-cell lung cancer. Cancer Sci 2020; 111:1699-1710. [PMID: 32108977 PMCID: PMC7226194 DOI: 10.1111/cas.14371] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/20/2020] [Accepted: 02/22/2020] [Indexed: 12/17/2022] Open
Abstract
The early detection of non-small-cell lung cancer (NSCLC) remains a common concern. The aim of our study was to validate the diagnostic value of a seven-autoantibody (7-AAB) panel compared with radiological diagnosis for NSCLC. We constructed a nomogram and a scoring table based on the 7-AAB panel's result to predict the risk of NSCLC. We prospectively enrolled 268 patients who presented with radiological lesions and underwent both the 7-AAB panel test and pathological diagnosis by surgical resection. A comparison between the 7-AAB panel and radiological diagnosis was performed. A nomogram and a scoring table based on the 7-AAB panel's result to predict the risk of NSCLC were constructed and internally validated. The 7-AAB panel test had a specificity of 90.2% and a positive predictive value (PPV) of 92.7%, which were significantly higher than those of the radiological diagnosis. The 7-AAB panel also showed a preferable sensitivity in patients with early-stage disease. Seven factors, including the 7-AAB panel results, were integrated into the nomogram. For more convenient application, we formulated a scoring table based on the nomogram. The area under the receiver operating characteristic curve was 0.840 and 0.860 in the training group and validation group, respectively, which was higher than that using the 7-AAB panel or radiological diagnosis alone. This study reveals that our 7-AAB panel has clinical value in the diagnosis of NSCLC. The utility of our nomogram and the scoring table indicated that they have the potential to assist clinicians in avoiding unnecessary treatment or needless follow-up.
Collapse
Affiliation(s)
- Weidong Wang
- Department of Thoracic SurgeryThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Runzhou Zhuang
- Department of Thoracic SurgeryThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Honghai Ma
- Department of Thoracic SurgeryThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Likui Fang
- Department of Thoracic SurgeryThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Zhitian Wang
- Department of Thoracic SurgeryThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Wang Lv
- Department of Thoracic SurgeryThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Jian Hu
- Department of Thoracic SurgeryThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
45
|
Garranzo-Asensio M, Guzmán-Aránguez A, Povedano E, Ruiz-Valdepeñas Montiel V, Poves C, Fernandez-Aceñero MJ, Montero-Calle A, Solís-Fernández G, Fernandez-Diez S, Camps J, Arenas M, Rodríguez-Tomàs E, Joven J, Sanchez-Martinez M, Rodriguez N, Dominguez G, Yáñez-Sedeño P, Pingarrón JM, Campuzano S, Barderas R. Multiplexed monitoring of a novel autoantibody diagnostic signature of colorectal cancer using HaloTag technology-based electrochemical immunosensing platform. Theranostics 2020; 10:3022-3034. [PMID: 32194852 PMCID: PMC7053203 DOI: 10.7150/thno.42507] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 01/02/2020] [Indexed: 12/15/2022] Open
Abstract
Background and Purpose: The humoral immune response in cancer patients can be used for early detection of the disease. Autoantibodies raised against tumor-associated antigens (TAAs) are promising clinical biomarkers for reliable cancer diagnosis, prognosis, and therapy monitoring. In this study, an electrochemical disposable multiplexed immunosensing platform able to integrate difficult- and easy-to-express colorectal cancer (CRC) TAAs is reported for the sensitive determination of eight CRC-specific autoantibodies. Methods: The electrochemical immunosensing approach involves the use of magnetic microcarriers (MBs) as solid supports modified with covalently immobilized HaloTag fusion proteins for the selective capture of specific autoantibodies. After magnetic capture of the modified MBs onto screen-printed carbon working electrodes, the amperometric responses measured using the hydroquinone (HQ)/H2O2 system were related to the levels of autoantibodies in plasma. Results: The biosensing platform was applied to the analysis of autoantibodies against 8 TAAs described for the first time in this work in plasma samples from healthy asymptomatic individuals (n=3), and patients with high-risk of developing CRC (n=3), and from patients already diagnosed with colorectal (n=3), lung (n=2) or breast (n=2) cancer. The developed bioplatform demonstrated an improved discrimination between CRC patients and controls (asymptomatic healthy individuals and breast and lung cancer patients) compared to an ELISA-like luminescence test. Conclusions: The proposed methodology uses a just-in-time produced protein in a simpler protocol, with low sample volume, and involves cost-effective instrumentation, which could be used in a high-throughput manner for reliable population screening to facilitate the detection of early CRC patients at affordable cost.
Collapse
Affiliation(s)
- María Garranzo-Asensio
- Departamento de Bioquímica y Biología Molecular, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037 Madrid, Spain
- UFIEC, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda 28220, Madrid, Spain
| | - Ana Guzmán-Aránguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Eloy Povedano
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Víctor Ruiz-Valdepeñas Montiel
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Carmen Poves
- Gastroenterology Unit, Hospital Universitario Clínico San Carlos, E-28040, Madrid, Spain
| | | | - Ana Montero-Calle
- UFIEC, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda 28220, Madrid, Spain
| | | | | | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
| | - Meritxell Arenas
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
| | - Elisabeth Rodríguez-Tomàs
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
- Department of Radiation Oncology, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d´Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus (Spain)
| | | | - Nuria Rodriguez
- Medical Oncology Department, Hospital Universitario La Paz, E-28046, Madrid, Spain
| | - Gemma Dominguez
- Departamento de Medicina, Facultad de Medicina, Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, E-28029, Madrid, Spain
| | - Paloma Yáñez-Sedeño
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - José Manuel Pingarrón
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Susana Campuzano
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Rodrigo Barderas
- UFIEC, Chronic Disease Programme, Instituto de Salud Carlos III, Majadahonda 28220, Madrid, Spain
| |
Collapse
|
46
|
Screening of tumor-associated antigens based on Oncomine database and evaluation of diagnostic value of autoantibodies in lung cancer. Clin Immunol 2019; 210:108262. [PMID: 31629809 DOI: 10.1016/j.clim.2019.108262] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 09/02/2019] [Accepted: 09/20/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVES The purpose of this study is to discover novel tumor-associated antigens (TAAs) to improve the diagnosis of lung cancer (LC). MATERIALS AND METHODS Oncomine database was used to discover potential TAAs from LC tissues, enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of autoantibodies against TAAs in two independent sets (identification set, n = 368; validation set, n = 1011). RESULTS Analyses of sera from identification set showed that the sensitivity of autoantibodies against five TAAs (HMGB3, ZWINT, GREM1, NUSAP1 and MMP12) reached 57.1%, 42.4%, 38.0%, 36.4% and 20.7%, with area under ROC curve (AUC) of 0.85, 0.75, 0.71, 0.73 and 0.70, respectively. It also validated the diagnostic performances of these autoantibodies with AUC of 0.72, 0.65, 0.61, 0.64 and 0.64, respectively. Autoantibody against HMGB3 exhibited significantly increased frequency in early LC (53.3%) compared to advanced LC (29.3%) (P < .05). The positive rates of autoantibody against HMGB3 and NUSAP1 in serum of LC patients without distant metastasis were significantly higher than that of distant metastatic LC (P < .05). When each of the three protein biomarkers (CEA, CA125 and CYFRA21-1) was combined with anti-HMGB3 autoantibody, the sensitivity of early LC increased to 72.7%, 63.3% and 75.9% from 36.4%, 13.3% and 27.6%, respectively. CONCLUSION Autoantibodies against 5 TAAs (HMGB3, ZWINT, GREM1, NUSAP1 and MMP12) might have favorable diagnostic values in LC detection, and autoantibody against HMGB3 has the potential to serve as a serological biomarker in early-stage LC. The combination of protein biomarkers and anti-HMGB3 might contribute to detection of early-stage LC.
Collapse
|
47
|
Jiang D, Wang Y, Liu M, Si Q, Wang T, Pei L, Wang P, Ye H, Shi J, Wang X, Song C, Wang K, Dai L, Zhang J. A panel of autoantibodies against tumor-associated antigens in the early immunodiagnosis of lung cancer. Immunobiology 2019; 225:151848. [PMID: 31980218 DOI: 10.1016/j.imbio.2019.09.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/07/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Lung cancer (LC) is one of the most common malignant tumors worldwide with low five-year survival rate due to lack of effective diagnosis. This study aims to find an optimal combination of autoantibodies for detecting of early-stage LC. METHODS Nine relatively novel autoantibodies against tumor-associated (TAAs) (PSIP1, TOP2A, ACTR3, RPS6KA5, HMGB3, MMP12, GREM1, ZWINT and NUSAP1) were detected by using ELISA. Diagnostic models were developed by using the training set (n = 644) and further validated in another independent set (n = 248). We also evaluated the diagnostic accuracy of the model to detect benign lung diseases (BLD) from the early-stage lung cancer. RESULTS The areas under the receiver operating characteristic curve (AUC) for the model with three TAAs panel (GREM1, HMGB3 and PSIP1) was 0.711(95% CI 0.674-0.746) in the training set and 0.858 (95% CI 0.808-0.899) in the validation set, which demonstrated a higher diagnostic capability. The AUC of this three TAAs model was 0.833 (95%CI 0.780-0.878) in discriminating LC from BLD. This model could identify early-stage LC patients from normal control (NC) individuals, with AUC of 0.687(95% CI 0.634-0.736) in training set and AUC of 0.920(95% CI 0.860-0.960) in validation set, and the overall AUC for early-stage LC was 0.779(95% CI 0.739-0.816) when the training set and validation set were combined. CONCLUSIONS The model with three TAAs panel would detect LC with higher effectiveness, and might be potential screening method for the early LC.
Collapse
Affiliation(s)
- Di Jiang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Yulin Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Man Liu
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Qiufang Si
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Tingting Wang
- Department of Clinical Laboratory, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 451464, Henan, China
| | - Lu Pei
- Department of Clinical Laboratory, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, 450000, Henan, China
| | - Peng Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Hua Ye
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Jianxiang Shi
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Xiao Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Chunhua Song
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Kaijuan Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China.
| | - Jianying Zhang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, 450001, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhenghzou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
48
|
Heo CK, Hwang HM, Lee HJ, Kwak SS, Yoo JS, Yu DY, Lim KJ, Lee S, Cho EW. Serum anti-EIF3A autoantibody as a potential diagnostic marker for hepatocellular carcinoma. Sci Rep 2019; 9:11059. [PMID: 31363116 PMCID: PMC6667438 DOI: 10.1038/s41598-019-47365-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
Tumor-associated autoantibodies are promising diagnostic biomarkers for early detection of tumors. We have screened a novel tumor-associated autoantibody in hepatocellular carcinoma (HCC) model mice. Its target antigen was identified as eukaryotic translation initiation factor 3 subunit A (EIF3A) by proteomic analysis, and the elevated expression of EIF3A in HCC tissues of tumor model mice as well as human patients was shown. Also, its existence in tumor-derived exosomes was revealed, which seem to be the cause of tumor-associated autoantibody production. To use serum anti-EIF3A autoantibody as biomarker, ELISA detecting anti-EIF3A autoantibody in human serum was performed using autoantibody-specific epitope. For the sensitive detection of serum autoantibodies its specific conformational epitopes were screened from the random cyclic peptide library, and a streptavidin antigen displaying anti-EIF3A autoantibody-specific epitope, XC90p2(-CPVRSGFPC-), was used as capture antigen. It distinguished patients with HCC (n = 102) from healthy controls (n = 0285) with a sensitivity of 79.4% and specificity of 83.5% (AUC = 0.87). Also, by simultaneously detecting with other HCC biomarkers, including alpha-fetoprotein, HCC diagnostic sensitivity improved from 79.4% to 85%. Collectively, we suggest that serum anti-EIF3A autoantibody is a useful biomarker for the diagnosis of HCC and the combinational detection of related biomarkers can enhance the accuracy of the cancer diagnosis.
Collapse
Affiliation(s)
- Chang-Kyu Heo
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea.,College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, South Korea
| | - Hai-Min Hwang
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea.,College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, South Korea
| | - Hye-Jung Lee
- Proteometech Inc., 1101 Wooree Venture Town, 466 Gangseo-ro, Gangseo-gu, Seoul, 03722, South Korea.,Graduate Program for Nanomedical Science, Yonsei University, 50 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea
| | - Sang-Seob Kwak
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea.,Department of Functional Genomics, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Jong-Shin Yoo
- Biomedical Omics Group, Korea Basic Science Institute, 162 YeonGuDanji-Ro, Ochang-eup, Cheongju, Chungbuk, 28119, South Korea
| | - Dae-Yeul Yu
- Disease Model Research Laboratory, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Kook-Jin Lim
- Proteometech Inc., 1101 Wooree Venture Town, 466 Gangseo-ro, Gangseo-gu, Seoul, 03722, South Korea
| | - Soojin Lee
- College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, South Korea.
| | - Eun-Wie Cho
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea. .,Department of Functional Genomics, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea.
| |
Collapse
|
49
|
Zang R, Li Y, Jin R, Wang X, Lei Y, Che Y, Lu Z, Mao S, Huang J, Liu C, Zheng S, Zhou F, Wu Q, Gao S, Sun N, He J. Enhancement of diagnostic performance in lung cancers by combining CEA and CA125 with autoantibodies detection. Oncoimmunology 2019; 8:e1625689. [PMID: 31646071 PMCID: PMC6791432 DOI: 10.1080/2162402x.2019.1625689] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/22/2019] [Accepted: 05/26/2019] [Indexed: 01/14/2023] Open
Abstract
Objectives: Although low-dose computed tomography has been confirmed to have meaningful diagnostic utility, lung cancer is still the leading cause of cancer-related deaths for both genders worldwide. Thus, a novel panel with a stronger diagnostic performance for lung cancer is needed. This study aimed to investigate the efficacy of a new panel in lung cancer diagnosis. Materials and Methods: The serum levels of carcinoembryonic antigen (CEA), cancer antigen 125 (CA125) and seven autoantibodies were measured and statistically analyzed in samples from healthy controls and patients with lung cancer. The 316 candidates enrolled in this study were randomly assigned into two groups for the training and validation of a diagnostic panel. Results: An optimal panel with four biomarkers (CEA, CA125, Annexin A1-Ab, and Alpha enolase-Ab) was established, with an area under the receiver operator characteristic (ROC) curve (AUC) of 0.897, a sensitivity of 86.5%, a specificity of 82.3%, a positive predictive value (PPV) of 88.3%, a negative predictive value (NPV) of 79.7%, and a diagnostic accuracy of 84.8% for the training group. The panel was validated, with an AUC of 0.856 and a sensitivity of 87.5% for the validation group. Furthermore, the new panel performed significantly better in lung cancer screening than did CEA and CA125 in all of the cohorts (p< .05). Conclusion: The diagnostic performance of CEA and CA125 was significantly enhanced through their combination with two autoantibodies (Annexin A1-Ab, and Alpha enolase-Ab). Optimization of the measured autoantibodies is critical for generating a panel to detect lung cancer in patients.
Collapse
Affiliation(s)
- 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, 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, China
| | - Runsen Jin
- 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, China
| | - Xinfeng Wang
- 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, China
| | - Yuanyuan Lei
- 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, China
| | - Yun Che
- 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, China
| | - Zhiliang Lu
- 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, China
| | - Shuangshuang Mao
- 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, China
| | - Jianbing Huang
- 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, China
| | - Chengming Liu
- 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, China
| | - Sufei Zheng
- 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, China
| | - Fang 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, China
| | - Qian Wu
- 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, 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, China
| | - Nan Sun
- 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, China
| | - Jie He
- 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, China
| |
Collapse
|
50
|
Seijo LM, Peled N, Ajona D, Boeri M, Field JK, Sozzi G, Pio R, Zulueta JJ, Spira A, Massion PP, Mazzone PJ, Montuenga LM. Biomarkers in Lung Cancer Screening: Achievements, Promises, and Challenges. J Thorac Oncol 2018; 14:343-357. [PMID: 30529598 DOI: 10.1016/j.jtho.2018.11.023] [Citation(s) in RCA: 302] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/20/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
The present review is an update of the research and development efforts regarding the use of molecular biomarkers in the lung cancer screening setting. The two main unmet clinical needs, namely, the refinement of risk to improve the selection of individuals undergoing screening and the characterization of undetermined nodules found during the computed tomography-based screening process are the object of the biomarkers described in the present review. We first propose some principles to optimize lung cancer biomarker discovery projects. Then, we summarize the discovery and developmental status of currently promising molecular candidates, such as autoantibodies, complement fragments, microRNAs, circulating tumor DNA, DNA methylation, blood protein profiling, or RNA airway or nasal signatures. We also mention other emerging biomarkers or new technologies to follow, such as exhaled breath biomarkers, metabolomics, sputum cell imaging, genetic predisposition studies, and the integration of next-generation sequencing into study of circulating DNA. We also underline the importance of integrating different molecular technologies together with imaging, radiomics, and artificial intelligence. We list a number of completed, ongoing, or planned trials to show the clinical utility of molecular biomarkers. Finally, we comment on future research challenges in the field of biomarkers in the context of lung cancer screening and propose a design of a trial to test the clinical utility of one or several candidate biomarkers.
Collapse
Affiliation(s)
- Luis M Seijo
- Clinica Universidad de Navarra, Madrid, Spain; CIBERES, Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Madrid, Spain
| | - Nir Peled
- Oncology Division, The Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka Medical Center and Ben-Gurion University, Beer-Sheva, Israel
| | - Daniel Ajona
- Solid Tumors Program, Centro de Investigación Médica Aplicada, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Mattia Boeri
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - John K Field
- The Roy Castle Lung Cancer Research Programme, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Gabriella Sozzi
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ruben Pio
- Solid Tumors Program, Centro de Investigación Médica Aplicada, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Javier J Zulueta
- Department of Pulmonology, Clinica Universidad de Navarra, Pamplona, Spain; Visiongate Inc., Phoenix, Arizona
| | - Avrum Spira
- Boston University School of Medicine, Boston, Massachusetts
| | | | | | - Luis M Montuenga
- Solid Tumors Program, Centro de Investigación Médica Aplicada, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.
| |
Collapse
|