1
|
Ahamed MT, Forshed J, Levitsky A, Lehtiö J, Bajalan A, Pernemalm M, Eriksson LE, Andersson B. Multiplex plasma protein assays as a diagnostic tool for lung cancer. Cancer Sci 2024; 115:3439-3454. [PMID: 39080998 PMCID: PMC11447887 DOI: 10.1111/cas.16300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 10/04/2024] Open
Abstract
Lack of the established noninvasive diagnostic biomarkers causes delay in diagnosis of lung cancer (LC). The aim of this study was to explore the association between inflammatory and cancer-associated plasma proteins and LC and thereby discover potential biomarkers. Patients referred for suspected LC and later diagnosed with primary LC, other cancers, or no cancer (NC) were included in this study. Demographic information and plasma samples were collected, and diagnostic information was later retrieved from medical records. Relative quantification of 92 plasma proteins was carried out using the Olink Immuno-Onc-I panel. Association between expression levels of panel of proteins with different diagnoses was assessed using generalized linear model (GLM) with the binomial family and a logit-link function, considering confounder effects of age, gender, smoking, and pulmonary diseases. The analysis showed that the combination of five plasma proteins (CD83, GZMA, GZMB, CD8A, and MMP12) has higher diagnostic performance for primary LC in both early and advanced stages compared with NC. This panel demonstrated lower diagnostic performance for other cancer types. Moreover, inclusion of four proteins (GAL9, PDCD1, CD4, and HO1) to the aforementioned panel significantly increased the diagnostic performance for primary LC in advanced stage as well as for other cancers. Consequently, the collective expression profiles of select plasma proteins, especially when analyzed in conjunction, might have the potential to distinguish individuals with LC from NC. This suggests their utility as predictive biomarkers for identification of LC patients. The synergistic application of these proteins as biomarkers could pave the way for the development of diagnostic tools for early-stage LC detection.
Collapse
Affiliation(s)
- Mohammad Tanvir Ahamed
- Department of Learning, Informatics, Management and Ethics (LIME)Karolinska InstitutetStockholmSweden
| | - Jenny Forshed
- Cancer Proteomics Mass Spectrometry, Department of Oncology‐PathologyKarolinska Institutet, Science for Life LaboratoryStockholmSweden
| | - Adrian Levitsky
- Department of Learning, Informatics, Management and Ethics (LIME)Karolinska InstitutetStockholmSweden
| | - Janne Lehtiö
- Cancer Proteomics Mass Spectrometry, Department of Oncology‐PathologyKarolinska Institutet, Science for Life LaboratoryStockholmSweden
| | - Amanj Bajalan
- Department of Microbiology, Tumor & Cell Biology (MTC)Karolinska InstitutetStockholmSweden
| | - Maria Pernemalm
- Cancer Proteomics Mass Spectrometry, Department of Oncology‐PathologyKarolinska Institutet, Science for Life LaboratoryStockholmSweden
| | - Lars E. Eriksson
- Department of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
- School of Health and Psychological Sciences, CityUniversity of LondonLondonUK
- Medical Unit Infectious DiseasesKarolinska University HospitalHuddingeSweden
| | - Björn Andersson
- Department of Cell and molecular Biology (CMB)Karolinska InstitutetStockholmSweden
| |
Collapse
|
2
|
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
|
3
|
Saputra HA, Jannath KA, Kim KB, Park DS, Shim YB. Conducting polymer composite-based biosensing materials for the diagnosis of lung cancer: A review. Int J Biol Macromol 2023; 252:126149. [PMID: 37582435 DOI: 10.1016/j.ijbiomac.2023.126149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/17/2023]
Abstract
The development of a simple and fast cancer detection method is crucial since early diagnosis is a key factor in increasing survival rates for lung cancer patients. Among several diagnosis methods, the electrochemical sensor is the most promising one due to its outstanding performance, portability, real-time analysis, robustness, amenability, and cost-effectiveness. Conducting polymer (CP) composites have been frequently used to fabricate a robust sensor device, owing to their excellent physical and electrochemical properties as well as biocompatibility with nontoxic effects on the biological system. This review brings up a brief overview of the importance of electrochemical biosensors for the early detection of lung cancer, with a detailed discussion on the design and development of CP composite materials for biosensor applications. The review covers the electrochemical sensing of numerous lung cancer markers employing composite electrodes based on the conducting polyterthiophene, poly(3,4-ethylenedioxythiophene), polyaniline, polypyrrole, molecularly imprinted polymers, and others. In addition, a hybrid of the electrochemical biosensors and other techniques was highlighted. The outlook was also briefly discussed for the development of CP composite-based electrochemical biosensors for POC diagnostic devices.
Collapse
Affiliation(s)
- Heru Agung Saputra
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Republic of Korea
| | - Khatun A Jannath
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Republic of Korea
| | - Kwang Bok Kim
- Digital Health Care R&D Department, Korea Institute of Industrial Technology, Cheonan 31056, Republic of Korea
| | - Deog-Su Park
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Republic of Korea
| | - Yoon-Bo Shim
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
4
|
Xu R, Wang J, Zhu Q, Zou C, Wei Z, Wang H, Ding Z, Meng M, Wei H, Xia S, Wei D, Deng L, Zhang S. Integrated models of blood protein and metabolite enhance the diagnostic accuracy for Non-Small Cell Lung Cancer. Biomark Res 2023; 11:71. [PMID: 37475010 PMCID: PMC10360339 DOI: 10.1186/s40364-023-00497-2] [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: 03/29/2023] [Accepted: 05/05/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND For early screening and diagnosis of non-small cell lung cancer (NSCLC), a robust model based on plasma proteomics and metabolomics is required for accurate and accessible non-invasive detection. Here we aim to combine TMT-LC-MS/MS and machine-learning algorithms to establish models with high specificity and sensitivity, and summarize a generalized model building scheme. METHODS TMT-LC-MS/MS was used to discover the differentially expressed proteins (DEPs) in the plasma of NSCLC patients. Plasma proteomics-guided metabolites were selected for clinical evaluation in 110 NSCLC patients who were going to receive therapies, 108 benign pulmonary diseases (BPD) patients, and 100 healthy controls (HC). The data were randomly split into training set and test set in a ratio of 80:20. Three supervised learning algorithms were applied to the training set for models fitting. The best performance models were evaluated with the test data set. RESULTS Differential plasma proteomics and metabolic pathways analyses revealed that the majority of DEPs in NSCLC were enriched in the pathways of complement and coagulation cascades, cholesterol and bile acids metabolism. Moreover, 10 DEPs, 14 amino acids, 15 bile acids, as well as 6 classic tumor biomarkers in blood were quantified using clinically validated assays. Finally, we obtained a high-performance screening model using logistic regression algorithm with AUC of 0.96, sensitivity of 92%, and specificity of 89%, and a diagnostic model with AUC of 0.871, sensitivity of 86%, and specificity of 78%. In the test set, the screening model achieved accuracy of 90%, sensitivity of 91%, and specificity of 90%, and the diagnostic model achieved accuracy of 82%, sensitivity of 77%, and specificity of 86%. CONCLUSIONS Integrated analysis of DEPs, amino acid, and bile acid features based on plasma proteomics-guided metabolite profiling, together with classical tumor biomarkers, provided a much more accurate detection model for screening and differential diagnosis of NSCLC. In addition, this new mathematical modeling based on plasma proteomics-guided metabolite profiling will be used for evaluation of therapeutic efficacy and long-term recurrence prediction of NSCLC.
Collapse
Affiliation(s)
- Runhao Xu
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Clinical Laboratory, Renji Hospital, Shanghai, 200001, China
| | - Jiongran Wang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qingqing Zhu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430000, China
| | - Chen Zou
- Department of Clinical Laboratory, Children's Hospital of Shanghai, Shanghai, 200040, China
| | - Zehao Wei
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430000, China
| | - Hao Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430000, China
| | - Zian Ding
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, 430000, China
| | - Minjie Meng
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Huimin Wei
- Shanghai Cellsolution Biotech Co.,Ltd, Shanghai, 200444, China
| | - Shijin Xia
- Department of Geriatrics, Huadong Hospital, Shanghai Institute of Geriatrics, Fudan University, Shanghai, 200040, China
| | - Dongqing Wei
- Department of Bioinformatics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Nanyang, 473006, Henan, China
| | - Li Deng
- Shanghai Cellsolution Biotech Co.,Ltd, Shanghai, 200444, China.
| | - Shulin Zhang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Nanyang, 473006, Henan, China.
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
5
|
Sharafeldin M, Rusling JF. Multiplexed electrochemical assays for clinical applications. CURRENT OPINION IN ELECTROCHEMISTRY 2023; 39:101256. [PMID: 37006828 PMCID: PMC10062004 DOI: 10.1016/j.coelec.2023.101256] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Rapid, accurate diagnoses are central to future efficient healthcare to identify diseases at early stages, avoid unnecessary treatment, and improve outcomes. Electrochemical techniques have been applied in many ways to support clinical applications by enabling the analysis of relevant disease biomarkers in user-friendly, sensitive, low-cost assays. Electrochemistry offers a launchpad for multiplexed biomarker assays that offer more accurate and precise diagnostics compared to single biomarker assays. In this short review, we underpin the importance of multiplexed analyses and provide a universal overview of current electrochemical assay strategies for multiple biomarkers. We highlight relevant examples of electrochemical methods that successfully quantify important disease biomarkers. Finally, we offer a future outlook on possible strategies that can be employed to increase throughput, sensitivity, and specificity of multiplexed electrochemical assays.
Collapse
Affiliation(s)
| | - James F. Rusling
- Department of Chemistry, University of Connecticut, Storrs, CT 06269-3060
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269-3136
- Department of Surgery and Neag Cancer Center, Uconn Health, Farmington, CT 06030
- School of Chemistry, National University of Ireland at Galway, Galway, Ireland. H91 TK33
| |
Collapse
|
6
|
Lesur A, Bernardin F, Koncina E, Letellier E, Kruppa G, Schmit PO, Dittmar G. Quantification of 782 Plasma Peptides by Multiplexed Targeted Proteomics. J Proteome Res 2023. [PMID: 37011904 DOI: 10.1021/acs.jproteome.2c00575] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Blood analysis is one of the foundations of clinical diagnostics. In recent years, the analysis of proteins in blood samples by mass spectrometry has taken a jump forward in terms of sensitivity and the number of identified proteins. The recent development of parallel reaction monitoring with parallel accumulation and serial fragmentation (prm-PASEF) combines ion mobility as an additional separation dimension. This increases the proteome coverage while allowing the use of shorter chromatographic gradients. To demonstrate the method's full potential, we used an isotope-labeled synthetic peptide mix of 782 peptides, derived from 579 plasma proteins, spiked into blood plasma samples with a prm-PASEF measurement allowing the quantification of 565 plasma proteins by targeted proteomics. As a less time-consuming alternative to the prm-PASEF method, we describe guided data independent acquisition (dia)-PASEF (g-dia-PASEF) and compare its application to prm-PASEF for measuring blood plasma. To demonstrate both methods' performance in clinical samples, 20 patient plasma samples from a colorectal cancer (CRC) cohort were analyzed. The analysis identified 14 differentially regulated proteins between the CRC patient and control individual plasma samples. This shows the technique's potential for the rapid and unbiased screening of blood proteins, abolishing the need for the preselection of potential biomarker proteins.
Collapse
Affiliation(s)
- Antoine Lesur
- Luxembourg Institute of Health, Strassen L-1445, Luxembourg
| | | | - Eric Koncina
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux L-4367, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux L-4367, Luxembourg
| | - Gary Kruppa
- Bruker Daltonics, Billerica, Massachusetts 01821, United States
| | | | - Gunnar Dittmar
- Luxembourg Institute of Health, Strassen L-1445, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux L-4367, Luxembourg
| |
Collapse
|
7
|
Marmor HN, Zorn JT, Deppen SA, Massion PP, Grogan EL. Biomarkers in Lung Cancer Screening: a Narrative Review. CURRENT CHALLENGES IN THORACIC SURGERY 2023; 5:5. [PMID: 37016707 PMCID: PMC10069480 DOI: 10.21037/ccts-20-171] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Although when used as a lung cancer screening tool low-dose computed tomography (LDCT) has demonstrated a significant reduction in lung cancer related mortality, it is not without pitfalls. The associated high false positive rate, inability to distinguish between benign and malignant nodules, cumulative radiation exposure, and resulting patient anxiety have all demonstrated the need for adjunctive testing in lung cancer screening. Current research focuses on developing liquid biomarkers to complement imaging as non-invasive lung cancer diagnostics. Biomarkers can be useful for both the early detection and diagnosis of disease, thereby decreasing the number of unnecessary radiologic tests performed. Biomarkers can stratify cancer risk to further enrich the screening population and augment existing risk prediction. Finally, biomarkers can be used to distinguish benign from malignant nodules in lung cancer screening. While many biomarkers require further validation studies, several, including autoantibodies and blood protein profiling, are available for clinical use. This paper describes the need for biomarkers as a lung cancer screening tool, both in terms of diagnosis and risk assessment. Additionally, this paper will discuss the goals of biomarker use, describe properties of a good biomarker, and review several of the most promising biomarkers currently being studied including autoantibodies, complement fragments, microRNA, blood proteins, circulating tumor DNA, and DNA methylation. Finally, we will describe future directions in the field of biomarker development.
Collapse
Affiliation(s)
- Hannah N. Marmor
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - J. Tyler Zorn
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Stephen A. Deppen
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Pierre P. Massion
- Vanderbilt Ingram Cancer Center, Nashville, TN; Department of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Eric L. Grogan
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN
- Department of Thoracic Surgery, Tennessee Valley VA Healthcare System, Nashville, TN
| |
Collapse
|
8
|
Huang H, Yang Y, Zhu Y, Chen H, Yang Y, Zhang L, Li W. Blood protein biomarkers in lung cancer. Cancer Lett 2022; 551:215886. [PMID: 35995139 DOI: 10.1016/j.canlet.2022.215886] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022]
Abstract
Lung cancer has consistently ranked first as the cause of cancer-associated mortality. The 5-year survival rate has risen slowly, and the main obstacle to improving the prognosis of patients has been that lung cancer is usually diagnosed at an advanced or incurable stage. Thus, early detection and timely intervention are the most effective ways to reduce lung cancer mortality. Tumor-specific molecules and cellular elements are abundant in circulation, providing real-time information in a noninvasive and cost-effective manner during lung cancer development. These circulating biomarkers are emerging as promising tools for early detection of lung cancer and can be used to supplement computed tomography screening, as well as for prognosis prediction and treatment response monitoring. Serum and plasma are the main sources of circulating biomarkers, and protein biomarkers have been most extensively studied. In this review, we summarize the research progress on three most common types of blood protein biomarkers (tumor-associated antigens, autoantibodies, and exosomal proteins) in lung cancer. This review will potentially guide researchers toward a more comprehensive understanding of candidate lung cancer protein biomarkers in the blood to facilitate their translation to the clinic.
Collapse
Affiliation(s)
- Hong Huang
- Institute of Clinical Pathology, Key Laboratory of Transplantation Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yongfeng Yang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China; Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yihan Zhu
- Institute of Clinical Pathology, Key Laboratory of Transplantation Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Hongyu Chen
- Institute of Clinical Pathology, Key Laboratory of Transplantation Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ying Yang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Li Zhang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China; Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Weimin Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China; Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China; The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, 610041, China.
| |
Collapse
|
9
|
Avian C, Mahali MI, Putro NAS, Prakosa SW, Leu JS. Fx-Net and PureNet: Convolutional Neural Network architecture for discrimination of Chronic Obstructive Pulmonary Disease from smokers and healthy subjects through electronic nose signals. Comput Biol Med 2022; 148:105913. [PMID: 35940164 DOI: 10.1016/j.compbiomed.2022.105913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/28/2022] [Accepted: 07/23/2022] [Indexed: 11/03/2022]
Abstract
As one of the most reliable and significant indicators, Chronic Obstructive Pulmonary Disease (COPD) becomes a robust predictor of lung cancer early detection, the world's leading cause of cancer death. One of the methods is to analyze the Volatile Organic Compounds (VOCs) in exhaled breath using electronic noses (E-noses), which have become emerging tools for analyzing breath because of their potential and promising technology for diagnosing. However, the signal processing of the E-Nose sensor becomes vital in exposing information about the subject condition, which most researchers strive to accomplish. We proposed a Convolutional Neural Network (CNN) architecture to classify COPD in smokers and non-smokers, healthy subjects, and smokers from E-Nose signals to contribute to this field. Two models were constructed following E-Nose signal processing state-of-the-arts. One was by combined feature extraction and classifier, and the second was by CNN, which directly processed the raw signal. In addition, various feature extraction and classifier (Machine Learning and CNN) used in prior research were investigated. Using 3K and 5K Fold cross-validation results demonstrated that our proposed models outperformed in Kernel Principal Component Analysis (KPCA) with Fx-ConvNet and Pure-ConvNet. They all reached maximum F1-Score with zero standard deviation values indicating a consistent result. Further experiments also showed that KPCA contributed to the increasing performance of some classifiers with average F1-Score 0.933 and 0.068 as standard deviation values.
Collapse
Affiliation(s)
- Cries Avian
- Department of Electronic and Computer Engineering, National Taiwan University of Science and Technology, Taiwan
| | - Muhammad Izzuddin Mahali
- Department of Electronic and Computer Engineering, National Taiwan University of Science and Technology, Taiwan; Department of Electronics and Informatics Engineering, Faculty of Engineering, Universitas Negeri Yogyakarta, Indonesia
| | - Nur Achmad Sulistyo Putro
- Department of Electronic and Computer Engineering, National Taiwan University of Science and Technology, Taiwan; Department of Computer Science and Electronics, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Indonesia
| | - Setya Widyawan Prakosa
- Department of Electronic and Computer Engineering, National Taiwan University of Science and Technology, Taiwan
| | - Jenq-Shiou Leu
- Department of Electronic and Computer Engineering, National Taiwan University of Science and Technology, Taiwan.
| |
Collapse
|
10
|
Chavez‐Pineda OG, Rodriguez‐Moncayo R, Cedillo‐Alcantar DF, Guevara‐Pantoja PE, Amador‐Hernandez JU, Garcia‐Cordero JL. Microfluidic systems for the analysis of blood‐derived molecular biomarkers. Electrophoresis 2022; 43:1667-1700. [DOI: 10.1002/elps.202200067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 12/19/2022]
Affiliation(s)
- Oriana G. Chavez‐Pineda
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Roberto Rodriguez‐Moncayo
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Diana F. Cedillo‐Alcantar
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Pablo E. Guevara‐Pantoja
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Josue U. Amador‐Hernandez
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Jose L. Garcia‐Cordero
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
- Roche Institute for Translational Bioengineering (ITB) Roche Pharma Research and Early Development, Roche Innovation Center Basel Basel Switzerland
| |
Collapse
|
11
|
Proteomic Analysis of Lung Cancer Types—A Pilot Study. Cancers (Basel) 2022; 14:cancers14112629. [PMID: 35681609 PMCID: PMC9179298 DOI: 10.3390/cancers14112629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the leading cause of tumor-related mortality, therefore significant effort is directed towards understanding molecular alterations occurring at the origin of the disease to improve current treatment options. The aim of our pilot-scale study was to carry out a detailed proteomic analysis of formalin-fixed paraffin-embedded tissue sections from patients with small cell or non-small cell lung cancer (adenocarcinoma, squamous cell carcinoma, and large cell carcinoma). Tissue surface digestion was performed on relatively small cancerous and tumor-adjacent normal regions and differentially expressed proteins were identified using label-free quantitative mass spectrometry and subsequent statistical analysis. Principal component analysis clearly distinguished cancerous and cancer adjacent normal samples, while the four lung cancer types investigated had distinct molecular profiles and gene set enrichment analysis revealed specific dysregulated biological processes as well. Furthermore, proteins with altered expression unique to a specific lung cancer type were identified and could be the targets of future studies.
Collapse
|
12
|
Brzhozovskiy A, Kononikhin A, Bugrova AE, Kovalev GI, Schmit PO, Kruppa G, Nikolaev EN, Borchers CH. The Parallel Reaction Monitoring-Parallel Accumulation-Serial Fragmentation (prm-PASEF) Approach for Multiplexed Absolute Quantitation of Proteins in Human Plasma. Anal Chem 2022; 94:2016-2022. [PMID: 35040635 DOI: 10.1021/acs.analchem.1c03782] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mass spectrometry (MS)-based quantitative proteomic methods have become some of the major tools for protein biomarker discovery and validation. The recently developed parallel reaction monitoring-parallel accumulation-serial fragmentation (prm-PASEF) approach on a Bruker timsTOF Pro mass spectrometer allows the addition of ion mobility as a new dimension to LC-MS-based proteomics and increases proteome coverage at a reduced analysis time. In this study, a prm-PASEF approach was used for the multiplexed absolute quantitation of proteins in human plasma using isotope-labeled peptide standards for 125 plasma proteins, over a broad (104-106) dynamic range. Optimization of LC and MS parameters, such as accumulation time and collision energy, resulted in improved sensitivity for more than half of the targets (73 out of 125 peptides) by increasing the signal-to-noise ratio by a factor of up to 10. Overall, 41 peptides showed up to a 2-fold increase in sensitivity, 25 peptides showed up to a 5-fold increase in sensitivity, and 7 peptides showed up to a 10-fold increase in sensitivity. Implementation of the prm-PASEF method allowed absolute protein quantitation (down to 1.13 fmol) in human plasma samples. A comparison of the concentration values of plasma proteins determined by MRM on a QTRAP instrument and by prm-PASEF on a timsTOF Pro revealed an excellent correlation (R2 = 0.97) with a slope of close to 1 (0.99), demonstrating that prm-PASEF is well suited for "absolute" quantitative proteomics.
Collapse
Affiliation(s)
- Alexander Brzhozovskiy
- Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Alexey Kononikhin
- Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Anna E Bugrova
- Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.,Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Grigoriy I Kovalev
- Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | | | - Gary Kruppa
- Bruker Daltonics, Inc. Billerica, Massachusetts 018215, United States
| | - Evgeny N Nikolaev
- Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Christoph H Borchers
- Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.,Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec H3T 1E2, Canada.,Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, Quebec H3T 1E2, Canada
| |
Collapse
|
13
|
Han M, Pang B, Zhou C, Li X, Wang Q, Jiang J, Li Y. Liquid biopsy of extracellular vesicle biomarkers for prostate cancer personalized treatment decision. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:3-9. [PMID: 39697872 PMCID: PMC11648516 DOI: 10.20517/evcna.2021.20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 12/20/2024]
Abstract
Liquid biopsy of tumor-derived extracellular vesicles (EVs) has great potential as a biomarker source for prostate cancer (CaP) early diagnosis and predicting the stages of cancer. The contents of EVs play an important role in intercellular communication and have specific expression in blood and urine samples from CaP patients. Powered by high-throughput, next-generation sequencing and proteomic technologies, novel EV biomarkers are easily detected in a non-invasive manner in different stages of CaP patients. These identified potential biomarkers can be further validated with a large sample size, machine learning model, and other different methods to improve the sensitivity and specificity of CaP diagnosis. The EV-based liquid biopsy is a novel and less-invasive alternative to surgical biopsies which would enable clinicians to potentially discover a whole picture of tumor through a simple blood or urine sample. In summary, this approach holds promise for developing personalized medicine to guide treatment decisions precisely for CaP patients.
Collapse
Affiliation(s)
- Meng Han
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo 315600, Zhejiang, China
- Ningbo Clinical Research Center for Urological Disease, Ningbo 315600, Zhejiang, China
| | - Bairen Pang
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Cheng Zhou
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo 315600, Zhejiang, China
- Ningbo Clinical Research Center for Urological Disease, Ningbo 315600, Zhejiang, China
| | - Xin Li
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo 315600, Zhejiang, China
- Ningbo Clinical Research Center for Urological Disease, Ningbo 315600, Zhejiang, China
| | - Qi Wang
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia
| | - Junhui Jiang
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo 315600, Zhejiang, China
- Ningbo Clinical Research Center for Urological Disease, Ningbo 315600, Zhejiang, China
| | - Yong Li
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia
| |
Collapse
|
14
|
Chatterjee G, Ferris B, Momenbeitollahi N, Li H. In-silico selection of cancer blood plasma proteins by integrating genomic and proteomic databases. Proteomics 2021; 22:e2100230. [PMID: 34933412 DOI: 10.1002/pmic.202100230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/15/2021] [Accepted: 12/13/2021] [Indexed: 11/11/2022]
Abstract
Blood protein markers have been studied for the clinical management of cancer. Due to the large number of the proteins existing in blood, it is often necessary to pre-select potential protein markers before experimental studies. However, to date there is a lack of automated method for in-silico selection of cancer blood proteins that integrates the information from both genetic and proteomic studies in a cancer-specific manner. In this work, we synthesized both genomic and proteomic information from several open access databases and established a bioinformatic pipeline for in-silico selection of blood plasma proteins overexpressed in specific type of cancer. We demonstrated the workflow of this pipeline with an example of breast cancer, while the methodology was applicable for other cancer types. With this pipeline we obtained 10 candidate biomarkers for breast cancer. The proposed pipeline provides a useful and convenient tool for in-silico selection of candidate blood protein biomarkers for a variety of cancer research.
Collapse
Affiliation(s)
- Gaurab Chatterjee
- School of Engineering, University of Guelph, Guelph, Ontario, Canada
| | - Bryn Ferris
- School of Engineering, University of Guelph, Guelph, Ontario, Canada
| | | | - Huiyan Li
- School of Engineering, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
15
|
Kulyyassov A, Fresnais M, Longuespée R. Targeted liquid chromatography-tandem mass spectrometry analysis of proteins: Basic principles, applications, and perspectives. Proteomics 2021; 21:e2100153. [PMID: 34591362 DOI: 10.1002/pmic.202100153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/08/2021] [Accepted: 09/24/2021] [Indexed: 12/25/2022]
Abstract
Liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) is now the main analytical method for the identification and quantification of peptides and proteins in biological samples. In modern research, identification of biomarkers and their quantitative comparison between samples are becoming increasingly important for discovery, validation, and monitoring. Such data can be obtained following specific signals after fragmentation of peptides using multiple reaction monitoring (MRM) and parallel reaction monitoring (PRM) methods, with high specificity, accuracy, and reproducibility. In addition, these methods allow measurement of the amount of post-translationally modified forms and isoforms of proteins. This review article describes the basic principles of MRM assays, guidelines for sample preparation, recent advanced MRM-based strategies, applications and illustrative perspectives of MRM/PRM methods in clinical research and molecular biology.
Collapse
Affiliation(s)
| | - Margaux Fresnais
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Rémi Longuespée
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
16
|
Liu L, Chen X, Petinrin OO, Zhang W, Rahaman S, Tang ZR, Wong KC. Machine Learning Protocols in Early Cancer Detection Based on Liquid Biopsy: A Survey. Life (Basel) 2021; 11:638. [PMID: 34209249 PMCID: PMC8308091 DOI: 10.3390/life11070638] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
With the advances of liquid biopsy technology, there is increasing evidence that body fluid such as blood, urine, and saliva could harbor the potential biomarkers associated with tumor origin. Traditional correlation analysis methods are no longer sufficient to capture the high-resolution complex relationships between biomarkers and cancer subtype heterogeneity. To address the challenge, researchers proposed machine learning techniques with liquid biopsy data to explore the essence of tumor origin together. In this survey, we review the machine learning protocols and provide corresponding code demos for the approaches mentioned. We discuss algorithmic principles and frameworks extensively developed to reveal cancer mechanisms and consider the future prospects in biomarker exploration and cancer diagnostics.
Collapse
Affiliation(s)
- Linjing Liu
- Department of Computer Science, City University of Hong Kong, Hong Kong, China; (L.L.); (X.C.); (O.O.P.); (W.Z.); (S.R.); (Z.-R.T.)
| | - Xingjian Chen
- Department of Computer Science, City University of Hong Kong, Hong Kong, China; (L.L.); (X.C.); (O.O.P.); (W.Z.); (S.R.); (Z.-R.T.)
| | - Olutomilayo Olayemi Petinrin
- Department of Computer Science, City University of Hong Kong, Hong Kong, China; (L.L.); (X.C.); (O.O.P.); (W.Z.); (S.R.); (Z.-R.T.)
| | - Weitong Zhang
- Department of Computer Science, City University of Hong Kong, Hong Kong, China; (L.L.); (X.C.); (O.O.P.); (W.Z.); (S.R.); (Z.-R.T.)
| | - Saifur Rahaman
- Department of Computer Science, City University of Hong Kong, Hong Kong, China; (L.L.); (X.C.); (O.O.P.); (W.Z.); (S.R.); (Z.-R.T.)
| | - Zhi-Ri Tang
- Department of Computer Science, City University of Hong Kong, Hong Kong, China; (L.L.); (X.C.); (O.O.P.); (W.Z.); (S.R.); (Z.-R.T.)
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Hong Kong, China; (L.L.); (X.C.); (O.O.P.); (W.Z.); (S.R.); (Z.-R.T.)
- Hong Kong Institute for Data Science, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
17
|
Lesur A, Schmit PO, Bernardin F, Letellier E, Brehmer S, Decker J, Dittmar G. Highly Multiplexed Targeted Proteomics Acquisition on a TIMS-QTOF. Anal Chem 2020; 93:1383-1392. [DOI: 10.1021/acs.analchem.0c03180] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Antoine Lesur
- Quantitative Biology Unit, Luxembourg Institute of Health, 1a Rue Thomas Edison, L-1445 Strassen, Luxembourg
| | | | - François Bernardin
- Quantitative Biology Unit, Luxembourg Institute of Health, 1a Rue Thomas Edison, L-1445 Strassen, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, University of Luxembourg, 6 Avenue du Swing, Campus Belval, L-4367 Belvaux, Luxembourg
| | - Sven Brehmer
- Bruker Daltonik GmbH, Fahrenheitstrasse 4, 28359 Bremen, Germany
| | - Jens Decker
- Bruker Daltonik GmbH, Fahrenheitstrasse 4, 28359 Bremen, Germany
| | - Gunnar Dittmar
- Quantitative Biology Unit, Luxembourg Institute of Health, 1a Rue Thomas Edison, L-1445 Strassen, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, 6 Avenue du Swing, Campus Belval, L-4367 Belvaux, Luxembourg
| |
Collapse
|