1
|
Qayyum M, Nayab S, Siddique I, Ghallab A. Analysis of time-fractional cancer-tumor immunotherapy model using modified He-Laplace algorithm. Sci Rep 2025; 15:8929. [PMID: 40087323 PMCID: PMC11909156 DOI: 10.1038/s41598-024-82170-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/03/2024] [Indexed: 03/17/2025] Open
Abstract
Cancer encompasses various diseases characterized by the uncontrolled growth of abnormal cells, which can invade healthy tissues and spread throughout the body, making it the second leading cause of death worldwide. This study presents a fractional cancer treatment model with immunotherapy to enhance understanding of cancer's mathematical framework and behavior. The model comprises fractional differential equations analyzed using the Caputo-fractional derivative, aiming to control cancer growth while considering cell population metrics. A framework integrating various homotopies and Laplace transforms is developed to explore cancer's complexities. Simultaneous solution profiles for effector immune cells and tumor cells illustrate their mutual influence. The model examines parameters such as the death rate of immune cells, natural tumor growth rate, rate of immune cells killing fractional tumor cells and numerous others graphically for clarity. The fractional parameter β is visually represented through 2D, 3D, and contour plots. This comprehensive analysis validates the proposed approach, suggesting its applicability to other complex cancer treatment models for better decision-making in cancer treatment.
Collapse
Affiliation(s)
- Mubashir Qayyum
- Department of Sciences and Humanities, National University of Computer and Emerging Sciences, Lahore, Pakistan
| | - Sidra Nayab
- Department of Sciences and Humanities, National University of Computer and Emerging Sciences, Lahore, Pakistan
| | - Imran Siddique
- Department of Mathematics, University of Sargodha, Sargodha, 40100, Pakistan
- Mathematics in Applied Sciences and Engineering Research Group, Scientific Research Center, Al-Ayen University, Nasiriyah, 64001, Iraq
| | - Abdullatif Ghallab
- Department of Computer Science, University of Science and Technology, P.O. Box: 13064, Sana'a, Yemen.
| |
Collapse
|
2
|
McMahon AN, Reis IM, Takita C, Wright JL, Hu JJ. Metabolomic Profiling of Disease Progression Following Radiotherapy for Breast Cancer. Cancers (Basel) 2025; 17:891. [PMID: 40075737 PMCID: PMC11899340 DOI: 10.3390/cancers17050891] [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: 01/09/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND This study aims to explore metabolic biomarkers and pathways in breast cancer prognosis. METHODS We performed a global post-radiotherapy (RT) urinary metabolomic analysis of 120 breast cancer patients: 60 progression-free (PF) patients as the reference and 60 with progressive disease (PD: recurrence, second primary, metastasis, or death). UPLC-MS/MS (Metabolon Inc.) identified 1742 biochemicals (1258 known and 484 unknown structures). Following normalization to osmolality, log transformation, and imputation of missing values, a Welch's two-sample t-test was used to identify biochemicals and metabolic pathways that differed between PF and PD groups. Data analysis and visualization were performed with MetaboAnalyst. RESULTS Metabolic biomarkers and pathways that significantly differed between the PD and PF groups were the following: amino acid metabolism, including phenylalanine, tyrosine, and tryptophan biosynthesis (impact value (IV) = 1.00; p = 0.0007); histidine metabolism (IV = 0.60; p < 0.0001); and arginine and proline metabolism (IV = 0.70; p = 0.0035). Metabolites of carbohydrate metabolism, including glucose (p = 0.0197), sedoheptulose (p = 0.0115), and carboxymethyl lysine (p = 0.0098), were elevated in patients with PD. Gamma-glutamyl amino acids, myo-inositol, and oxidative stress biomarkers, including 7-Hydroxyindole Sulfate and sulfate, were elevated in patients who died (p ≤ 0.05). CONCLUSIONS Amino acid metabolism emerged as a key pathway in breast cancer progression, while carbohydrate and oxidative stress metabolites also showed potential utility as biomarkers for breast cancer progression. These findings demonstrate applications of metabolomics in identifying metabolic biomarkers and pathways as potential targets for predicting breast cancer progression.
Collapse
Affiliation(s)
- Alexandra N. McMahon
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.N.M.); (I.M.R.)
| | - Isildinha M. Reis
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.N.M.); (I.M.R.)
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Cristiane Takita
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jean L. Wright
- Department of Radiation Oncology, University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA;
| | - Jennifer J. Hu
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.N.M.); (I.M.R.)
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| |
Collapse
|
3
|
Sharafi Monfared M, Nazmi S, Parhizkar F, Jafari D. Soluble B7 and TNF family in colorectal cancer: Serum level, prognostic and treatment value. Hum Immunol 2025; 86:111232. [PMID: 39793378 DOI: 10.1016/j.humimm.2025.111232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Soluble immune checkpoints (sIC) are crucial factors in the immune system. They regulate immune responses by transforming intercellular signals via binding to their membrane-bound receptor or ligand. Moreover, soluble ICs are vital in immune regulation, cancer development, and prognosis. They can be identified and measured in various tumor microenvironments. Recently, sICs have become increasingly important in clinically assessing malignancies like colorectal cancer (CRC) patients. This review explores the evolving role of the soluble B7 family and soluble tumor necrosis factor (TNF) superfamily members in predicting disease progression, treatment response, and overall patient outcomes in CRC. We comprehensively analyze the diagnostic and prognostic potential of soluble immune checkpoints in CRC. Understanding the role of these soluble immune checkpoints in CRC management and their potential as targets for precision medicine approaches can be critical for improving outcomes for patients with colorectal cancer.
Collapse
Affiliation(s)
- Mohanna Sharafi Monfared
- Student's Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran; School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Sina Nazmi
- Student's Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran; School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Forough Parhizkar
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Davood Jafari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
4
|
Liu C, Chen H, Ma Y, Zhang L, Chen L, Huang J, Zhao Z, Jiang H, Kong J. Clinical metabolomics in type 2 diabetes mellitus: from pathogenesis to biomarkers. Front Endocrinol (Lausanne) 2025; 16:1501305. [PMID: 40070584 PMCID: PMC11893406 DOI: 10.3389/fendo.2025.1501305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025] Open
Abstract
As a multidimensional metabolic disorder, the disability and death rate of type 2 diabetes mellitus (T2DM) has increased over time. T2DM covers a wide range of pathological manifestations ranging from hyperglycemia to multi-organ failure, and it has the potential to evolve into acute complications, including ketosis and chronic complications such as peripheral neuropathy, retinopathy, and nephropathy. T2DM mainly occurs in microvascular and large vessels and thus it is restricted for the clinician to diagnose and prescribe. However, the pathological mechanism and clinical diagnosis are inadequate. High-throughput metabolomics, characterized by non-invasive diagnostic techniques to identify potential biomarkers and distinct stages of T2DM, has been increasingly recognized as a vigorous tool with latent capacity for clinical translation. The pathological stratification of T2DM can significantly reduce disability and mortality rates. By tracing the metabolome and associated pathways from impaired fasting blood glucose or impaired glucose tolerance to severe organ failure, the chief contributions of large, independent population-based cohorts are summarized herein. These results facilitate understanding the pathophysiology and mechanism and supports research in accurate diagnosis, risk prediction, curative effect, distinct stages, and prognosis judgment of T2DM.
Collapse
Affiliation(s)
- Chuanxin Liu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Hetao Chen
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yujin Ma
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Lei Zhang
- Department of Integrative Medicine, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Lulu Chen
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Department of Clinical Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jiarui Huang
- Department of Critical Care Medicine, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Zizhe Zhao
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Hongwei Jiang
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jiao Kong
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Lu G, Su Z, Yu X, He Y, Sha T, Yan K, Guo H, Tao Y, Liao L, Zhang Y, Lu G, Gong W. Differentiating Pulmonary Nodule Malignancy Using Exhaled Volatile Organic Compounds: A Prospective Observational Study. Cancer Med 2025; 14:e70545. [PMID: 39777868 PMCID: PMC11706237 DOI: 10.1002/cam4.70545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Advances in imaging technology have enhanced the detection of pulmonary nodules. However, determining malignancy often requires invasive procedures or repeated radiation exposure, underscoring the need for safer, noninvasive diagnostic alternatives. Analyzing exhaled volatile organic compounds (VOCs) shows promise, yet its effectiveness in assessing the malignancy of pulmonary nodules remains underexplored. METHODS Employing a prospective study design from June 2023 to January 2024 at the Affiliated Hospital of Yangzhou University, we assessed the malignancy of pulmonary nodules using the Mayo Clinic model and collected exhaled breath samples alongside lifestyle and health examination data. We applied five machine learning (ML) algorithms to develop predictive models which were evaluated using area under the curve (AUC), sensitivity, specificity, and other relevant metrics. RESULTS A total of 267 participants were enrolled, including 210 with low-risk and 57 with moderate-risk pulmonary nodules. Univariate analysis identified 11 exhaled VOCs associated with nodule malignancy, alongside two lifestyle factors (smoke index and sites of tobacco smoke inhalation) and one clinical metric (nodule diameter) as independent predictors for moderate-risk nodules. The logistic regression model integrating lifestyle and health data achieved an AUC of 0.91 (95% CI: 0.8611-0.9658), while the random forest model incorporating exhaled VOCs achieved an AUC of 0.99 (95% CI: 0.974-1.00). Calibration curves indicated strong concordance between predicted and observed risks. Decision curve analysis confirmed the net benefit of these models over traditional methods. A nomogram was developed to aid clinicians in assessing nodule malignancy based on VOCs, lifestyle, and health data. CONCLUSIONS The integration of ML algorithms with exhaled biomarkers and clinical data provides a robust framework for noninvasive assessment of pulmonary nodules. These models offer a safer alternative to traditional methods and may enhance early detection and management of pulmonary nodules. Further validation through larger, multicenter studies is necessary to establish their generalizability. TRIAL REGISTRATION Number ChiCTR2400081283.
Collapse
Affiliation(s)
- Guangyu Lu
- Department of Health Management CenterAffiliated Hospital of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
- School of Public HealthMedical College of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Zhixia Su
- School of Public HealthMedical College of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Xiaoping Yu
- Department of Health Management CenterAffiliated Hospital of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Yuhang He
- School of NursingMedical College of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Taining Sha
- School of Public HealthMedical College of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Kai Yan
- School of Public HealthMedical College of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Hong Guo
- Department of Thoracic SurgeryAffiliated Hospital of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Yujian Tao
- Department of Respiratory and Critical Care MedicineAffiliated Hospital of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Liting Liao
- Department of Basic MedicineMedical College of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Yanyan Zhang
- Testing Center of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Guotao Lu
- Yangzhou Key Laboratory of Pancreatic DiseaseInstitute of Digestive Diseases, Affiliated Hospital of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
- Pancreatic Center, Department of GastroenterologyAffiliated Hospital of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| | - Weijuan Gong
- Department of Health Management CenterAffiliated Hospital of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
- Department of Basic MedicineMedical College of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
- Yangzhou Key Laboratory of Pancreatic DiseaseInstitute of Digestive Diseases, Affiliated Hospital of Yangzhou University, Yangzhou UniversityYangzhouJiangsuChina
| |
Collapse
|
6
|
Sumon MSI, Malluhi M, Anan N, AbuHaweeleh MN, Krzyslak H, Vranic S, Chowdhury MEH, Pedersen S. Integrative Stacking Machine Learning Model for Small Cell Lung Cancer Prediction Using Metabolomics Profiling. Cancers (Basel) 2024; 16:4225. [PMID: 39766124 PMCID: PMC11727543 DOI: 10.3390/cancers16244225] [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: 11/02/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 01/15/2025] Open
Abstract
Background: Small cell lung cancer (SCLC) is an extremely aggressive form of lung cancer, characterized by rapid progression and poor survival rates. Despite the importance of early diagnosis, the current diagnostic techniques are invasive and restricted. Methods: This study presents a novel stacking-based ensemble machine learning approach for classifying small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) using metabolomics data. The analysis included 191 SCLC cases, 173 NSCLC cases, and 97 healthy controls. Feature selection techniques identified significant metabolites, with positive ions proving more relevant. Results: For multi-class classification (control, SCLC, NSCLC), the stacking ensemble achieved 85.03% accuracy and 92.47 AUC using Support Vector Machine (SVM). Binary classification (SCLC vs. NSCLC) further improved performance, with ExtraTreesClassifier reaching 88.19% accuracy and 92.65 AUC. SHapley Additive exPlanations (SHAP) analysis revealed key metabolites like benzoic acid, DL-lactate, and L-arginine as significant predictors. Conclusions: The stacking ensemble approach effectively leverages multiple classifiers to enhance overall predictive performance. The proposed model effectively captures the complementary strengths of different classifiers, enhancing the detection of SCLC and NSCLC. This work accentuates the potential of combining metabolomics with advanced machine learning for non-invasive early lung cancer subtype detection, offering an alternative to conventional biopsy methods.
Collapse
Affiliation(s)
| | - Marwan Malluhi
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (M.M.); (M.N.A.); (S.V.)
| | - Noushin Anan
- Department of Electrical Engineering, Qatar University, Doha 2713, Qatar; (M.S.I.S.); (N.A.)
| | | | - Hubert Krzyslak
- Department of Clinical Biochemistry, Aalborg University Hospital, 9000 Aalborg, Denmark;
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (M.M.); (M.N.A.); (S.V.)
| | | | - Shona Pedersen
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (M.M.); (M.N.A.); (S.V.)
| |
Collapse
|
7
|
Xu X, Zeng C, Qing B, He Y, Song G, Wang J, Yu S, Zhang T, Wei Q, Liu L, Wen H, Hu J, Zhang W, Li Y, Chen Y, Xia Z. Development of a urine-based metabolomics approach for multi-cancer screening and tumor origin prediction. Front Immunol 2024; 15:1449103. [PMID: 39735533 PMCID: PMC11671364 DOI: 10.3389/fimmu.2024.1449103] [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: 06/14/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Background Cancer remains a leading cause of mortality worldwide. A non-invasive screening solution was required for early diagnosis of cancer. Multi-cancer early detection (MCED) tests have been considered to address the challenge by simultaneously identifying multiple types of cancer within a single test using minimally invasive blood samples. However, a multi-cancer screening strategy utilizing urine-based metabolomics has not yet been developed. Methods We enrolled 911 cancer patients with 548 lung cancer (LC), 177 with gastric cancer (GC), and 186 with colorectal cancer (CRC), alongside 563 individuals with non-cancerous benign diseases and 229 healthy controls (HC) and investigated the metabolic profiles of urine samples. Participants were randomly allocated to discovery and validation cohorts. The discovery cohort was used for identifying multi-cancer and tissue-specific signatures to build the cancer screening and tumor origin prediction models, while the validation cohort was employed for assessing the performance of these models. Results We identified and annotated a total of 360 metabolites from the urine samples. Using the LASSO regression algorithm, 18 metabolites were characterized as urinary metabolic biomarkers and exhibited excellent discriminative performance between cancer patients and HC with AUC of 0.96 in the validation cohort. In comparison with the performance of traditional tumor markers CEA, the screening model performed higher sensitivity across the cancer stages, with a particularly increase in sensitivity among early-stage cancer patients. Moreover, the screening model also exhibited in high classification of cancers from non-cancerous group, comprising with HC and benign disease participants. Furthermore, two non-overlapping metabolic panels were selected to differentiate LC from Non-LC and GC from CRC with the AUC values of 0.87 and 0.83 in validation cohorts, respectively. Additionally, the model accurately predicted the origin of three lethal cancers: lung, gastric, and colorectal, with an overall accuracy of 0.75. The AUC values for LC, GC, and CRC were 0.88, 0.88, and 0.80, respectively. Discussion Our study demonstrates the potential of urine-based metabolomics for multi-cancer early detection. The approach offers non-invasive cancer screening, promising widespread implementation in population-based programs for early detection and improved outcomes. Further validation and expansion are needed for broader clinical applicability.
Collapse
Affiliation(s)
- Xinping Xu
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chunyan Zeng
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bei Qing
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yun He
- Metanotitia Inc., Shenzhen, China
| | - Guodong Song
- The Second Hospital of Tianjin Medical University, Tianjin, China
| | | | - Shuqi Yu
- Metanotitia Inc., Shenzhen, China
| | | | | | - Li Liu
- Metanotitia Inc., Shenzhen, China
| | - He Wen
- Metanotitia Inc., Shenzhen, China
| | | | - Wei Zhang
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan Li
- Metanotitia Inc., Shenzhen, China
| | - Youxiang Chen
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenkun Xia
- The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
8
|
Labory J, Njomgue-Fotso E, Bottini S. Benchmarking feature selection and feature extraction methods to improve the performances of machine-learning algorithms for patient classification using metabolomics biomedical data. Comput Struct Biotechnol J 2024; 23:1274-1287. [PMID: 38560281 PMCID: PMC10979063 DOI: 10.1016/j.csbj.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Objective Classification tasks are an open challenge in the field of biomedicine. While several machine-learning techniques exist to accomplish this objective, several peculiarities associated with biomedical data, especially when it comes to omics measurements, prevent their use or good performance achievements. Omics approaches aim to understand a complex biological system through systematic analysis of its content at the molecular level. On the other hand, omics data are heterogeneous, sparse and affected by the classical "curse of dimensionality" problem, i.e. having much fewer observation, samples (n) than omics features (p). Furthermore, a major problem with multi-omics data is the imbalance either at the class or feature level. The objective of this work is to study whether feature extraction and/or feature selection techniques can improve the performances of classification machine-learning algorithms on omics measurements. Methods Among all omics, metabolomics has emerged as a powerful tool in cancer research, facilitating a deeper understanding of the complex metabolic landscape associated with tumorigenesis and tumor progression. Thus, we selected three publicly available metabolomics datasets, and we applied several feature extraction techniques both linear and non-linear, coupled or not with feature selection methods, and evaluated the performances regarding patient classification in the different configurations for the three datasets. Results We provide general workflow and guidelines on when to use those techniques depending on the characteristics of the data available. To further test the extension of our approach to other omics data, we have included a transcriptomics and a proteomics data. Overall, for all datasets, we showed that applying supervised feature selection improves the performances of feature extraction methods for classification purposes. Scripts used to perform all analyses are available at: https://github.com/Plant-Net/Metabolomic_project/.
Collapse
Affiliation(s)
- Justine Labory
- Université Côte d′Azur, Center of Modeling Simulation and Interactions, Nice, France
- INRAE, Université Côte d′Azur, CNRS, Institut Sophia Agrobiotech, Sophia-Antipolis, France
- Université Côte d′Azur, Inserm U1081, CNRS UMR 7284, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | | | - Silvia Bottini
- Université Côte d′Azur, Center of Modeling Simulation and Interactions, Nice, France
- INRAE, Université Côte d′Azur, CNRS, Institut Sophia Agrobiotech, Sophia-Antipolis, France
| |
Collapse
|
9
|
Beyoğlu D, Popov YV, Idle JR. Metabolomic Hallmarks of Obesity and Metabolic Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2024; 25:12809. [PMID: 39684520 DOI: 10.3390/ijms252312809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
From a detailed review of 90 experimental and clinical metabolomic investigations of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD), we have developed metabolomic hallmarks for both obesity and MASLD. Obesity studies were conducted in mice, rats, and humans, with consensus biomarker groups in plasma/serum being essential and nonessential amino acids, energy metabolites, gut microbiota metabolites, acylcarnitines and lysophosphatidylcholines (LPC), which formed the basis of the six metabolomic hallmarks of obesity. Additionally, mice and rats shared elevated cholesterol, humans and rats shared elevated fatty acids, and humans and mice shared elevated VLDL/LDL, bile acids and phosphatidylcholines (PC). MASLD metabolomic studies had been performed in mice, rats, hamsters, cows, geese, blunt snout breams, zebrafish, and humans, with the biomarker groups in agreement between experimental and clinical investigations being energy metabolites, essential and nonessential amino acids, fatty acids, and bile acids, which lay the foundation of the five metabolomic hallmarks of MASLD. Furthermore, the experimental group had higher LPC/PC and cholesteryl esters, and the clinical group had elevated acylcarnitines, lysophosphatidylethanolamines/phosphatidylethanolamines (LPE/PE), triglycerides/diglycerides, and gut microbiota metabolites. These metabolomic hallmarks aid in the understanding of the metabolic role played by obesity in MASLD development, inform mechanistic studies into underlying disease pathogenesis, and are critical for new metabolite-inspired therapies.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| | - Yury V Popov
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jeffrey R Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| |
Collapse
|
10
|
Bhardwaj JK, Siwach A, Sachdeva SN. Metabolomics and cellular altered pathways in cancer biology: A review. J Biochem Mol Toxicol 2024; 38:e23807. [PMID: 39148273 DOI: 10.1002/jbt.23807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
Cancer is a deadly disease that affects a cell's metabolism and surrounding tissues. Understanding the fundamental mechanisms of metabolic alterations in cancer cells would assist in developing cancer treatment targets and approaches. From this perspective, metabolomics is a great analytical tool to clarify the mechanisms of cancer therapy as well as a useful tool to investigate cancer from a distinct viewpoint. It is a powerful emerging technology that detects up to thousands of molecules in tissues and biofluids. Like other "-omics" technologies, metabolomics involves the comprehensive investigation of micromolecule metabolites and can reveal important details about the cancer state that is otherwise not apparent. Recent developments in metabolomics technologies have made it possible to investigate cancer metabolism in greater depth and comprehend how cancer cells utilize metabolic pathways to make the amino acids, nucleotides, and lipids required for tumorigenesis. These new technologies have made it possible to learn more about cancer metabolism. Here, we review the cellular and systemic effects of cancer and cancer treatments on metabolism. The current study provides an overview of metabolomics, emphasizing the current technologies and their use in clinical and translational research settings.
Collapse
Affiliation(s)
- Jitender Kumar Bhardwaj
- Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra, Haryana, India
| | - Anshu Siwach
- Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra, Haryana, India
| | - Som Nath Sachdeva
- Department of Civil Engineering, National Institute of Technology, Kurukshetra and Kurukshetra University, Kurukshetra, Haryana, India
| |
Collapse
|
11
|
Ding Z, Chen J, Li B, Ji X. Inflammatory factors and risk of lung adenocarcinoma: a Mendelian randomization study mediated by blood metabolites. Front Endocrinol (Lausanne) 2024; 15:1446863. [PMID: 39257908 PMCID: PMC11384989 DOI: 10.3389/fendo.2024.1446863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/29/2024] [Indexed: 09/12/2024] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most common type of lung cancer, and its pathogenesis remains not fully elucidated. Inflammation and metabolic dysregulation are considered to play crucial roles in LUAD development, but their causal relationships and specific mechanisms remain unclear. Methods This study employed a two-sample Mendelian randomization (MR) approach to systematically evaluate the causal associations between 91 circulating inflammatory factors, 1,400 serum metabolites, and LUAD. We utilized LUAD genome-wide association studies (GWAS) data from the FinnGen biobank and GWAS data of metabolites and inflammatory factors from the GWAS catalog to conduct two-sample MR analyses. For the identified key metabolites, we further used mediator MR to investigate their mediating effects in the influence of IL-17A on LUAD and explored potential mechanisms through protein-protein interaction and functional enrichment analyses. Results The MR analyses revealed that IL-17A (OR 0.78, 95%CI 0.62-0.99) was negatively associated with LUAD, while 71 metabolites were significantly associated with LUAD. Among them, ferulic acid 4-sulfate may play a crucial mediating role in the suppression of LUAD by IL-17A (OR 0.87, 95%CI 0.78-0.97). IL-17A may exert its anti-LUAD effects through extensive interactions with genes related to ferulic acid 4-sulfate metabolism (such as SULT1A1, CYP1A1, etc.), inhibiting oxidative stress and inflammatory responses, as well as downstream tumor-related pathways of ferulic acid 4-sulfate (such as MAPK, NF-κB, etc.). Conclusion This study discovered causal associations between IL-17A, multiple serum metabolites, and LUAD occurrence, revealing the key role of inflammatory and metabolic dysregulation in LUAD pathogenesis. Our findings provide new evidence-based medical support for specific inflammatory factors and metabolites as early predictive and risk assessment biomarkers for LUAD, offering important clues for subsequent mechanistic studies and precision medicine applications.
Collapse
Affiliation(s)
- Zheng Ding
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Liaoning, Shenyang, China
| | - Juan Chen
- Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Liaoning, Shenyang, China
| | - Bohan Li
- Department of Urinary Surgery, The First Affiliated Hospital of China Medical University, Liaoning, Shenyang, China
| | - Xinyu Ji
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Liaoning, Shenyang, China
| |
Collapse
|
12
|
Dalal B, Tada T, Patel DP, Pine SR, Khan M, Oike T, Kanke Y, Parker AL, Haznadar M, Toulabi L, Krausz KW, Robles AI, Bowman ED, Gonzalez FJ, Harris CC. Urinary Metabolite Diagnostic and Prognostic Liquid Biopsy Biomarkers of Lung Cancer in Nonsmokers and Tobacco Smokers. Clin Cancer Res 2024; 30:3592-3602. [PMID: 38837903 PMCID: PMC11325153 DOI: 10.1158/1078-0432.ccr-24-0637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/23/2024] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
PURPOSE Nonsmokers account for 10% to 13% of all lung cancer cases in the United States. Etiology is attributed to multiple risk factors including exposure to secondhand smoking, asbestos, environmental pollution, and radon, but these exposures are not within the current eligibility criteria for early lung cancer screening by low-dose CT (LDCT). EXPERIMENTAL DESIGN Urine samples were collected from two independent cohorts comprising 846 participants (exploratory cohort) and 505 participants (validation cohort). The cancer urinary biomarkers, creatine riboside (CR) and N-acetylneuraminic acid (NANA), were analyzed and quantified using liquid chromatography-mass spectrometry to determine if nonsmoker cases can be distinguished from sex and age-matched controls in comparison with tobacco smoker cases and controls, potentially leading to more precise eligibility criteria for LDCT screening. RESULTS Urinary levels of CR and NANA were significantly higher and comparable in nonsmokers and tobacco smoker cases than population controls in both cohorts. Receiver operating characteristic analysis for combined CR and NANA levels in nonsmokers of the exploratory cohort resulted in better predictive performance with the AUC of 0.94, whereas the validation cohort nonsmokers had an AUC of 0.80. Kaplan-Meier survival curves showed that high levels of CR and NANA were associated with increased cancer-specific death in nonsmokers as well as tobacco smoker cases in both cohorts. CONCLUSIONS Measuring CR and NANA in urine liquid biopsies could identify nonsmokers at high risk for lung cancer as candidates for LDCT screening and warrant prospective studies of these biomarkers.
Collapse
Affiliation(s)
- Bhavik Dalal
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Takeshi Tada
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Daxesh P Patel
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Sharon R Pine
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mohammed Khan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Takahiro Oike
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Yasuyuki Kanke
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Amelia L Parker
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Majda Haznadar
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Leila Toulabi
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - Elise D Bowman
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
13
|
Shi S, Luo D, Yang Y, Wang X. Integrative Omics Analysis Reveals Metabolic Features of Ground-Glass Opacity-Associated Lung Cancer. J Cancer 2024; 15:1848-1862. [PMID: 38434969 PMCID: PMC10905408 DOI: 10.7150/jca.92437] [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: 11/21/2023] [Accepted: 01/19/2024] [Indexed: 03/05/2024] Open
Abstract
Background: Ground-glass opacity (GGO)-associated cancers are increasingly prevalent, exhibiting unique clinical and molecular features that suggest the need for a distinct treatment strategy. However, the metabolic characteristics and vulnerabilities of GGO-associated lung cancers remain unexplored. Methods: We conducted metabolomic and transcriptomic analyses on 40 pairs of GGO-associated lung cancer tissues and adjacent normal tissues. By integrating data from TCGA database and single-cell RNA sequencing, we aimed to identify aberrant metabolic pathways, establish a metabolite-associated gene signature, and pinpoint key metabolic genes. The physiological effect of key genes was detected in vitro and vivo assays. Results: We identified a 30-gene metabolite-associated signature and discovered aberrant metabolic pathways for GGO-associated lung cancer at both metabolic and transcriptional levels. Patients with this signature displayed specific prognostic and molecular features. Cox regression analysis, based on the Cancer Genome Atlas Program (TCGA) data, further narrowed down the metabolite-related gene signature, resulting in a 5-gene signature. Confirmed by single-cell RNA sequencing (GSE203360), the 5-gene signature was mainly expressed in cancer cells of GGO tissue. Real-time quantitative PCR (RT-qPCR) further validated the differential expression of these genes between GGO and adjacent normal tissue obtained from pulmonary surgery. Finally, our integrative analysis unveiled aberrant histidine metabolism at both the multi-omics and single-cell levels. Moreover, we identified MAOB as a key metabolic gene, demonstrating its ability to suppress cell proliferation, migration, and invasion in LUAD cell lines, both in vitro and in vivo. Conclusions: We identified a specific metabolite-associated gene signature and identified aberrant histidine metabolism in GGO-associated lung cancer from multiple perspectives. Notably, MAOB, a crucial component in histidine metabolism, demonstrated a significant inhibitory effect on the proliferation and metastasis of LUAD, indicating its potential significance in pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Shuai Shi
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan Province, China
| | - Dayuan Luo
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan Province, China
| | - Yanyi Yang
- Heath Management Center, Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan Province, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan Province, China
| |
Collapse
|
14
|
Bhalla S, Yi S, Gerber DE. Emerging Strategies in Lung Cancer Screening: Blood and Beyond. Clin Chem 2024; 70:60-67. [PMID: 38175587 PMCID: PMC11161198 DOI: 10.1093/clinchem/hvad137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/02/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Although low dose computed tomography (LDCT)-based lung cancer screening (LCS) can decrease lung cancer-related mortality among high-risk individuals, it remains an imperfect and substantially underutilized process. LDCT-based LCS may result in false-positive findings, which can lead to invasive procedures and potential morbidity. Conversely, current guidelines may fail to capture at-risk individuals, particularly those from under-represented minority populations. To address these limitations, numerous biomarkers have emerged to complement LDCT and improve early lung cancer detection. CONTENT This review focuses primarily on blood-based biomarkers, including protein, microRNAs, circulating DNA, and methylated DNA panels, in current clinical development for LCS. We also examine other emerging biomarkers-utilizing airway epithelia, exhaled breath, sputum, and urine-under investigation. We highlight challenges and limitations of biomarker testing, as well as recent strategies to integrate molecular strategies with imaging technologies. SUMMARY Multiple biomarkers are under active investigation for LCS, either to improve risk-stratification after nodule detection or to optimize risk-based patient selection for LDCT-based screening. Results from ongoing and future clinical trials will elucidate the clinical utility of biomarkers in the LCS paradigm.
Collapse
Affiliation(s)
- Sheena Bhalla
- Department of Internal Medicine (Division of Hematology-Oncology), UT Southwestern Medical Center, Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, United States
| | - Sofia Yi
- School of Medicine, UT Southwestern Medical Center, Dallas, TX, United States
| | - David E Gerber
- Department of Internal Medicine (Division of Hematology-Oncology), UT Southwestern Medical Center, Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, United States
- Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
15
|
Liang S, Cao X, Wang Y, Leng P, Wen X, Xie G, Luo H, Yu R. Metabolomics Analysis and Diagnosis of Lung Cancer: Insights from Diverse Sample Types. Int J Med Sci 2024; 21:234-252. [PMID: 38169594 PMCID: PMC10758149 DOI: 10.7150/ijms.85704] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/14/2023] [Indexed: 01/05/2024] Open
Abstract
Lung cancer is a highly fatal disease that poses a significant global health burden. The absence of characteristic clinical symptoms frequently results in the diagnosis of most patients at advanced stages of lung cancer. Although low-dose computed tomography (LDCT) screening has become increasingly prevalent in clinical practice, its high rate of false positives continues to present a significant challenge. In addition to LDCT screening, tumor biomarker detection represents a critical approach for early diagnosis of lung cancer; unfortunately, no tumor marker with optimal sensitivity and specificity is currently available. Metabolomics has recently emerged as a promising field for developing novel tumor biomarkers. In this paper, we introduce metabolic pathways, instrument platforms, and a wide variety of sample types for lung cancer metabolomics. Specifically, we explore the strengths, limitations, and distinguishing features of various sample types employed in lung cancer metabolomics research. Additionally, we present the latest advances in lung cancer metabolomics research that utilize diverse sample types. We summarize and enumerate research studies that have investigated lung cancer metabolomics using different metabolomic sample types. Finally, we provide a perspective on the future of metabolomics research in lung cancer. Our discussion of the potential of metabolomics in developing new tumor biomarkers may inspire further study and innovation in this dynamic field.
Collapse
Affiliation(s)
- Simin Liang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiujun Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yingshuang Wang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ping Leng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiaoxia Wen
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Guojing Xie
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Huaichao Luo
- Department of Clinical Laboratory, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China (UESTC), Chengdu, China
| | - Rong Yu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
16
|
Ghosh C, Hu J, Kebebew E. Advances in translational research of the rare cancer type adrenocortical carcinoma. Nat Rev Cancer 2023; 23:805-824. [PMID: 37857840 DOI: 10.1038/s41568-023-00623-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2023] [Indexed: 10/21/2023]
Abstract
Adrenocortical carcinoma is a rare malignancy with an annual worldwide incidence of 1-2 cases per 1 million and a 5-year survival rate of <60%. Although adrenocortical carcinoma is rare, such rare cancers account for approximately one third of patients diagnosed with cancer annually. In the past decade, there have been considerable advances in understanding the molecular basis of adrenocortical carcinoma. The genetic events associated with adrenocortical carcinoma in adults are distinct from those of paediatric cases, which are often associated with germline or somatic TP53 mutations and have a better prognosis. In adult primary adrenocortical carcinoma, the main somatic genetic alterations occur in genes that encode proteins involved in the WNT-β-catenin pathway, cell cycle and p53 apoptosis pathway, chromatin remodelling and telomere maintenance pathway, cAMP-protein kinase A (PKA) pathway or DNA transcription and RNA translation pathways. Recently, integrated molecular studies of adrenocortical carcinomas, which have characterized somatic mutations and the methylome as well as gene and microRNA expression profiles, have led to a molecular classification of these tumours that can predict prognosis and have helped to identify new therapeutic targets. In this Review, we summarize these recent translational research advances in adrenocortical carcinoma, which it is hoped could lead to improved patient diagnosis, treatment and outcome.
Collapse
Affiliation(s)
| | - Jiangnan Hu
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Electron Kebebew
- Department of Surgery, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
17
|
Yan Q, He D, Walker DI, Uppal K, Wang X, Orimoloye HT, Jones DP, Ritz BR, Heck JE. The neonatal blood spot metabolome in retinoblastoma. EJC PAEDIATRIC ONCOLOGY 2023; 2:100123. [PMID: 38130370 PMCID: PMC10735245 DOI: 10.1016/j.ejcped.2023.100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Background Retinoblastoma is rare but nevertheless the most common pediatric eye cancer that occurs in children under age 5. High-resolution metabolomics (HRM) is a powerful analytical approach to profile metabolic features and pathways or identify metabolite biomarkers. To date, no studies have used pre-diagnosis blood samples from retinoblastoma cases and compared them to healthy controls to elucidate early perturbations in tumor pathways. Objectives Here, we report on metabolic profiles of neonatal blood comparing cases later in childhood diagnosed with retinoblastoma and controls. Methods We employed untargeted metabolomics analysis using neonatal dried blood spots for 1327 children (474 retinoblastoma cases and 853 healthy controls) born in California from 1983 to 2011. Cases were selected from the California Cancer Registry and controls, frequency matched to cases by birth year, from California birth rolls. We performed high-resolution metabolomics to extract metabolic features, partial least squares discriminant analysis (PLS-DA) and logistic regression to identify features associated with disease, and Mummichog pathway analysis to characterize enriched biological pathways. Results PLS-DA identified 1917 discriminative features associated with retinoblastoma and Mummichog identified 14 retinoblastoma-related enriched pathways including linoleate metabolism, pentose phosphate pathway, pyrimidine metabolism, fructose and mannose metabolism, vitamin A metabolism, as well as fatty acid and lipid metabolism. Interpretation Our findings linked a retinoblastoma diagnosis in early life to newborn blood metabolome perturbations indicating alterations in inflammatory pathways and energy metabolism. Neonatal blood spots may provide a venue for early detection for this or potentially other childhood cancers.
Collapse
Affiliation(s)
- Qi Yan
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Di He
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Douglas I. Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Xuexia Wang
- Department of Mathematics, University of North Texas, Denton, TX, USA
| | - Helen T. Orimoloye
- College of Health and Public Service, University of North Texas, Denton, TX, USA
| | - Dean P. Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA, USA
- Department of Medicine, Emory University, Atlanta, GA, USA
| | - Beate R. Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
- Department of Neurology, UCLA School of Medicine, CA, USA
| | - Julia E. Heck
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
- College of Health and Public Service, University of North Texas, Denton, TX, USA
| |
Collapse
|
18
|
Chen CJ, Lee DY, Yu J, Lin YN, Lin TM. Recent advances in LC-MS-based metabolomics for clinical biomarker discovery. MASS SPECTROMETRY REVIEWS 2023; 42:2349-2378. [PMID: 35645144 DOI: 10.1002/mas.21785] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/14/2021] [Accepted: 11/18/2021] [Indexed: 06/15/2023]
Abstract
The employment of liquid chromatography-mass spectrometry (LC-MS) untargeted and targeted metabolomics has led to the discovery of novel biomarkers and improved the understanding of various disease mechanisms. Numerous strategies have been reported to expand the metabolite coverage in LC-MS-untargeted and targeted metabolomics. To improve the sensitivity of low-abundance or poor-ionized metabolites for reducing the amount of clinical sample, chemical derivatization methods are used to target different functional groups. Proper sample preparation is beneficial for reducing the matrix effect, maintaining the stability of the LC-MS system, and increasing the metabolite coverage. Machine learning has recently been integrated into the workflow of LC-MS metabolomics to accelerate metabolite identification and data-processing automation, and increase the accuracy of disease classification and clinical outcome prediction. Due to the rapidly growing utility of LC-MS metabolomics in discovering disease markers, this review will address the recent advances in the field and offer perspectives on various strategies for expanding metabolite coverage, chemical derivatization, sample preparation, clinical disease markers, and machining learning for disease modeling.
Collapse
Affiliation(s)
- Chao-Jung Chen
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Der-Yen Lee
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Jiaxin Yu
- AI Innovation Center, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Ning Lin
- Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Tsung-Min Lin
- Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
19
|
Almalki AH. Recent Analytical Advances for Decoding Metabolic Reprogramming in Lung Cancer. Metabolites 2023; 13:1037. [PMID: 37887362 PMCID: PMC10609104 DOI: 10.3390/metabo13101037] [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: 08/24/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 10/28/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Metabolic reprogramming is a fundamental trait associated with lung cancer development that fuels tumor proliferation and survival. Monitoring such metabolic pathways and their intermediate metabolites can provide new avenues concerning treatment strategies, and the identification of prognostic biomarkers that could be utilized to monitor drug responses in clinical practice. In this review, recent trends in the analytical techniques used for metabolome mapping of lung cancer are capitalized. These techniques include nuclear magnetic resonance (NMR), gas chromatography-mass spectrometry (GC-MS), liquid chromatography-mass spectrometry (LC-MS), and imaging mass spectrometry (MSI). The advantages and limitations of the application of each technique for monitoring the metabolite class or type are also highlighted. Moreover, their potential applications in the analysis of many biological samples will be evaluated.
Collapse
Affiliation(s)
- Atiah H. Almalki
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
- Addiction and Neuroscience Research Unit, Health Science Campus, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
20
|
Shields PG. Role of untargeted omics biomarkers of exposure and effect for tobacco research. ADDICTION NEUROSCIENCE 2023; 7:100098. [PMID: 37396411 PMCID: PMC10310069 DOI: 10.1016/j.addicn.2023.100098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Tobacco research remains a clear priority to improve individual and population health, and has recently become more complex with emerging combustible and noncombustible tobacco products. The use of omics methods in prevention and cessation studies are intended to identify new biomarkers for risk, compared risks related to other products and never use, and compliance for cessation and reinitation. to assess the relative effects of tobacco products to each other. They are important for the prediction of reinitiation of tobacco use and relapse prevention. In the research setting, both technical and clinical validation is required, which presents a number of complexities in the omics methodologies from biospecimen collection and sample preparation to data collection and analysis. When the results identify differences in omics features, networks or pathways, it is unclear if the results are toxic effects, a healthy response to a toxic exposure or neither. The use of surrogate biospecimens (e.g., urine, blood, sputum or nasal) may or may not reflect target organs such as the lung or bladder. This review describes the approaches for the use of omics in tobacco research and provides examples of prior studies, along with the strengths and limitations of the various methods. To date, there is little consistency in results, likely due to small number of studies, limitations in study size, the variability in the analytic platforms and bioinformatic pipelines, differences in biospecimen collection and/or human subject study design. Given the demonstrated value for the use of omics in clinical medicine, it is anticipated that the use in tobacco research will be similarly productive.
Collapse
Affiliation(s)
- Peter G. Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH
| |
Collapse
|
21
|
Yu CT, Farhat Z, Livinski AA, Loftfield E, Zanetti KA. Characteristics of Cancer Epidemiology Studies That Employ Metabolomics: A Scoping Review. Cancer Epidemiol Biomarkers Prev 2023; 32:1130-1145. [PMID: 37410086 PMCID: PMC10472112 DOI: 10.1158/1055-9965.epi-23-0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/26/2023] [Accepted: 06/28/2023] [Indexed: 07/07/2023] Open
Abstract
An increasing number of cancer epidemiology studies use metabolomics assays. This scoping review characterizes trends in the literature in terms of study design, population characteristics, and metabolomics approaches and identifies opportunities for future growth and improvement. We searched PubMed/MEDLINE, Embase, Scopus, and Web of Science: Core Collection databases and included research articles that used metabolomics to primarily study cancer, contained a minimum of 100 cases in each main analysis stratum, used an epidemiologic study design, and were published in English from 1998 to June 2021. A total of 2,048 articles were screened, of which 314 full texts were further assessed resulting in 77 included articles. The most well-studied cancers were colorectal (19.5%), prostate (19.5%), and breast (19.5%). Most studies used a nested case-control design to estimate associations between individual metabolites and cancer risk and a liquid chromatography-tandem mass spectrometry untargeted or semi-targeted approach to measure metabolites in blood. Studies were geographically diverse, including countries in Asia, Europe, and North America; 27.3% of studies reported on participant race, the majority reporting White participants. Most studies (70.2%) included fewer than 300 cancer cases in their main analysis. This scoping review identified key areas for improvement, including needs for standardized race and ethnicity reporting, more diverse study populations, and larger studies.
Collapse
Affiliation(s)
- Catherine T. Yu
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland
| | - Zeinab Farhat
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Alicia A. Livinski
- National Institutes of Health Library, Office of Research Services, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - Erikka Loftfield
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Krista A. Zanetti
- Office of Nutrition Research, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
22
|
Kamali AN, Bautista JM, Eisenhut M, Hamedifar H. Immune checkpoints and cancer immunotherapies: insights into newly potential receptors and ligands. Ther Adv Vaccines Immunother 2023; 11:25151355231192043. [PMID: 37662491 PMCID: PMC10469281 DOI: 10.1177/25151355231192043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 07/14/2023] [Indexed: 09/05/2023] Open
Abstract
Checkpoint markers and immune checkpoint inhibitors have been increasingly identified and developed as potential immunotherapeutic targets in various human cancers. Despite valuable efforts to discover novel immune checkpoints and their ligands, the precise roles of their therapeutic functions, as well as the broad identification of their counterpart receptors, remain to be addressed. In this context, it has been suggested that various putative checkpoint receptors can be induced upon activation. In the tumor microenvironment, T cells, as crucial immune response against malignant diseases as well as other immune central effector cells, such as natural killer cells, are regulated via co-stimulatory or co-inhibitory signals from immune or tumor cells. Studies have shown that exposure of T cells to tumor antigens upregulates the expression of inhibitory checkpoint receptors, leading to T-cell dysfunction or exhaustion. Although targeting immune checkpoint regulators has shown relative clinical efficacy in some tumor types, most trials in the field of cancer immunotherapies have revealed unsatisfactory results due to de novo or adaptive resistance in cancer patients. To overcome these obstacles, combinational therapies with newly discovered inhibitory molecules or combined blockage of several checkpoints provide a rationale for further research. Moreover, precise identification of their receptors counterparts at crucial checkpoints is likely to promise effective therapies. In this review, we examine the prospects for the application of newly emerging checkpoints, such as T-cell immunoglobulin and mucin domain 3, lymphocyte activation gene-3, T-cell immunoreceptor with Ig and ITIM domains (TIGIT), V-domain Ig suppressor of T-cell activation (VISTA), new B7 family proteins, and B- and T-cell lymphocyte attenuator, in association with immunotherapy of malignancies. In addition, their clinical and biological significance is discussed, including their expression in various human cancers, along with their roles in T-cell-mediated immune responses.
Collapse
Affiliation(s)
- Ali N. Kamali
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Simin Dasht Industrial Area, Karaj, Iran
- CinnaGen Research and Production Co., Alborz 3165933155, Iran
| | - José M. Bautista
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
- Research Institute Hospital 12 de Octubre, Madrid, Spain
| | - Michael Eisenhut
- Department of Pediatrics, Luton and Dunstable University Hospital NHS Foundation Trust, Luton, UK
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| |
Collapse
|
23
|
Yin H, Yuan Y, Xin L, Hang Q, Zhao L, Qin F, Xiong Z. pH-responsive magnetic graphene oxide composite as an adsorbent with high affinity for rapid capture of nucleosides. Mikrochim Acta 2023; 190:365. [PMID: 37612484 DOI: 10.1007/s00604-023-05945-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/06/2023] [Indexed: 08/25/2023]
Abstract
A novel pH-responsive magnetic graphene oxide composite (MGO@PEI-BA) is proposed for the first time as an adsorbent for the rapid capture and detection of nucleosides (cytidine, uridine, guanosine, and adenosine). The morphology, structure, and magnetic properties of the composite were evaluated using various characterization techniques. The results indicated that the composite was successfully fabricated. A series of parameters that affect extraction and elution were optimized through one-factor-at-a-time and Box-Behnken design of response surface methodology (BBD-RSM). The unique layered structures and easily accessible active sites of the composite facilitated molecular transport, resulting in instantaneous equilibrium of nucleosides adsorption within 5 min. Based on this study, a magnetic dispersive micro-solid-phase extraction (MD-μ-SPE) method assisted by the MGO@PEI-BA was developed in combination with UHPLC-UV analysis for the determination of nucleosides in rat urine. Under the optimum conditions, a wide linear range (10-2000 ng mL-1), good linearity (r > 0.99), low detection limits (1-3 ng mL-1), low relative standard deviations (RSDs ≤ 3.9%), and satisfactory recoveries (82.7-96.3%) were achieved. These results demonstrate that the MGO@PEI-BA is an excellent adsorbent for extracting nucleosides from biological samples.
Collapse
Affiliation(s)
- Huawen Yin
- School of Pharmacy, Shenyang Pharmaceutical University, No. 26 Huatuo Rd, High & New Tech Development Zone, 117004, Benxi, Liaoning Province, People's Republic of China
| | - Yue Yuan
- School of Pharmacy, Shenyang Pharmaceutical University, No. 26 Huatuo Rd, High & New Tech Development Zone, 117004, Benxi, Liaoning Province, People's Republic of China
| | - Ling Xin
- School of Pharmacy, Shenyang Pharmaceutical University, No. 26 Huatuo Rd, High & New Tech Development Zone, 117004, Benxi, Liaoning Province, People's Republic of China
| | - Qian Hang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 26 Huatuo Rd, High & New Tech Development Zone, 117004, Benxi, Liaoning Province, People's Republic of China
| | - Longshan Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 26 Huatuo Rd, High & New Tech Development Zone, 117004, Benxi, Liaoning Province, People's Republic of China
| | - Feng Qin
- School of Pharmacy, Shenyang Pharmaceutical University, No. 26 Huatuo Rd, High & New Tech Development Zone, 117004, Benxi, Liaoning Province, People's Republic of China.
| | - Zhili Xiong
- School of Pharmacy, Shenyang Pharmaceutical University, No. 26 Huatuo Rd, High & New Tech Development Zone, 117004, Benxi, Liaoning Province, People's Republic of China.
| |
Collapse
|
24
|
Zhang Y, Yang Z, Tang Y, Guo C, Lin D, Cheng L, Hu X, Zhang K, Li G. Hallmark guided identification and characterization of a novel immune-relevant signature for prognostication of recurrence in stage I-III lung adenocarcinoma. Genes Dis 2023; 10:1657-1674. [PMID: 37397559 PMCID: PMC10311029 DOI: 10.1016/j.gendis.2022.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/07/2022] [Accepted: 07/16/2022] [Indexed: 11/23/2022] Open
Abstract
The high risk of postoperative mortality in lung adenocarcinoma (LUAD) patients is principally driven by cancer recurrence and low response rates to adjuvant treatment. Here, A combined cohort containing 1,026 stage I-III patients was divided into the learning (n = 678) and validation datasets (n = 348). The former was used to establish a 16-mRNA risk signature for recurrence prediction with multiple statistical algorithms, which was verified in the validation set. Univariate and multivariate analyses confirmed it as an independent indicator for both recurrence-free survival (RFS) and overall survival (OS). Distinct molecular characteristics between the two groups including genomic alterations, and hallmark pathways were comprehensively analyzed. Remarkably, the classifier was tightly linked to immune infiltrations, highlighting the critical role of immune surveillance in prolonging survival for LUAD. Moreover, the classifier was a valuable predictor for therapeutic responses in patients, and the low-risk group was more likely to yield clinical benefits from immunotherapy. A transcription factor regulatory protein-protein interaction network (TF-PPI-network) was constructed via weighted gene co-expression network analysis (WGCNA) concerning the hub genes of the signature. The constructed multidimensional nomogram dramatically increased the predictive accuracy. Therefore, our signature provides a forceful basis for individualized LUAD management with promising potential implications.
Collapse
Affiliation(s)
- Yongqiang Zhang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510620, China
| | - Zhao Yang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuqin Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Chengbin Guo
- Center for Biomedicine and Innovations, Faculty of Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Danni Lin
- Center for Biomedicine and Innovations, Faculty of Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Linling Cheng
- Center for Biomedicine and Innovations, Faculty of Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Xun Hu
- Clinical Research Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- Biorepository, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kang Zhang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Center for Biomedicine and Innovations, Faculty of Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Gen Li
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510620, China
| |
Collapse
|
25
|
Oike T, Osu N, Yoshimoto Y, Obinata H, Yoshikawa K, Harris CC, Ohno T. Pilot study of plasma creatine riboside as a potential biomarker for cervical cancer. Heliyon 2023; 9:e16684. [PMID: 37292314 PMCID: PMC10245246 DOI: 10.1016/j.heliyon.2023.e16684] [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: 10/05/2022] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/10/2023] Open
Abstract
This pilot study aimed primarily to evaluate plasma levels of a novel metabolite, creatine riboside, in patients with cervical cancer (discovery and validation cohorts, n = 11 for each) compared with non-cancer subjects (controls, n = 30). We found that the pre-treatment plasma creatine riboside level was significantly higher in the discovery cohort than in controls. The cut-off value determined from the discovery cohort distinguished 90.9% of the patients in the validation cohort from controls. Unbiased principal component analysis of plasma metabolites in high-creatine riboside samples demonstrated enrichment of pathways involved in arginine and creatine metabolism. These data indicate the potential utility of plasma creatine riboside as a biomarker of cervical cancer.
Collapse
Affiliation(s)
- Takahiro Oike
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma, 371-8511, Japan
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Naoto Osu
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yuya Yoshimoto
- Department of Radiation Oncology, School of Medicine, Fukushima Medical University, 1, Hikarigaoka, Fukushima, Fukushima, 960-1295, Japan
| | - Hideru Obinata
- Laboratory for Analytical Instruments, Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Kazuhiro Yoshikawa
- Division of Research Creation and Biobank, Research Creation Support Center, Aichi Medical University, 1-1, Yazako-karimata, Nagakute, Aichi, 480-1103, Japan
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr, Bethesda, MD, 20814, USA
| | - Tatsuya Ohno
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma, 371-8511, Japan
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
26
|
Xu Y, Dong X, Qin C, Wang F, Cao W, Li J, Yu Y, Zhao L, Tan F, Chen W, Li N, He J. Metabolic biomarkers in lung cancer screening and early diagnosis (Review). Oncol Lett 2023; 25:265. [PMID: 37216157 PMCID: PMC10193366 DOI: 10.3892/ol.2023.13851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 03/29/2023] [Indexed: 05/24/2023] Open
Abstract
Late diagnosis is one of the major contributing factors to the high mortality rate of lung cancer, which is now the leading cause of cancer-associated mortality worldwide. At present, low-dose CT (LDCT) screening in the high-risk population, in which lung cancer incidence is higher than that of the low-risk population is the predominant diagnostic strategy. Although this has efficiently reduced lung cancer mortality in large randomized trials, LDCT screening has high false-positive rates, resulting in excessive subsequent follow-up procedures and radiation exposure. Complementation of LDCT examination with biofluid-based biomarkers has been documented to increase efficacy, and this type of preliminary screening can potentially reduce potential radioactive damage to low-risk populations and the burden of hospital resources. Several molecular signatures based on components of the biofluid metabolome that can possibly discriminate patients with lung cancer from healthy individuals have been proposed over the past two decades. In the present review, advancements in currently available technologies in metabolomics were reviewed, with particular focus on their possible application in lung cancer screening and early detection.
Collapse
Affiliation(s)
- Yongjie Xu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Xuesi Dong
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Chao Qin
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Fei Wang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Wei Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Jiang Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Yiwen Yu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Liang Zhao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Ni Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. 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 100021, P.R. China
| |
Collapse
|
27
|
Yang Q, Luo J, Xu H, Huang L, Zhu X, Li H, Yang R, Peng B, Sun D, Zhu Q, Liu F. Metabolomic investigation of urinary extracellular vesicles for early detection and screening of lung cancer. J Nanobiotechnology 2023; 21:153. [PMID: 37189121 DOI: 10.1186/s12951-023-01908-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/24/2023] [Indexed: 05/17/2023] Open
Abstract
Lung cancer is a prevalent cancer type worldwide that often remains asymptomatic in its early stages and is frequently diagnosed at an advanced stage with a poor prognosis due to the lack of effective diagnostic techniques and molecular biomarkers. However, emerging evidence suggests that extracellular vesicles (EVs) may promote lung cancer cell proliferation and metastasis, and modulate the anti-tumor immune response in lung cancer carcinogenesis, making them potential biomarkers for early cancer detection. To investigate the potential of urinary EVs for non-invasive detection and screening of patients at early stages, we studied metabolomic signatures of lung cancer. Specifically, we conducted metabolomic analysis of 102 EV samples and identified metabolome profiles of urinary EVs, including organic acids and derivatives, lipids and lipid-like molecules, organheterocyclic compounds, and benzenoids. Using machine learning with a random forest model, we screened for potential markers of lung cancer and identified a marker panel consisting of Kanzonol Z, Xanthosine, Nervonyl carnitine, and 3,4-Dihydroxybenzaldehyde, which exhibited a diagnostic potency of 96% for the testing cohort (AUC value). Importantly, this marker panel also demonstrated effective prediction for the validation set, with an AUC value of 84%, indicating the reliability of the marker screening process. Our findings suggest that the metabolomic analysis of urinary EVs provides a promising source of non-invasive markers for lung cancer diagnostics. We believe that the EV metabolic signatures could be used to develop clinical applications for the early detection and screening of lung cancer, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Jiaxin Luo
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Hao Xu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Liu Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xinxi Zhu
- Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hengrui Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Rui Yang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bo Peng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Da Sun
- Institute of Life Sciences & Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou, 325035, China
| | - Qingfu Zhu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Fei Liu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
28
|
Jordaens S, Zwaenepoel K, Tjalma W, Deben C, Beyers K, Vankerckhoven V, Pauwels P, Vorsters A. Urine biomarkers in cancer detection: A systematic review of preanalytical parameters and applied methods. Int J Cancer 2023; 152:2186-2205. [PMID: 36647333 DOI: 10.1002/ijc.34434] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/25/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023]
Abstract
The aim of this review was to explore the status of urine sampling as a liquid biopsy for noninvasive cancer research by reviewing used preanalytical parameters and protocols. We searched two main health sciences databases, PubMed and Web of Science. From all eligible publications (2010-2022), information was extracted regarding: (a) study population characteristics, (b) cancer type, (c) urine preanalytics, (d) analyte class, (e) isolation method, (f) detection method, (g) comparator used, (h) biomarker type, (i) conclusion and (j) sensitivity and specificity. The search query identified 7835 records, of which 924 unique publications remained after screening the title, abstract and full text. Our analysis demonstrated that many publications did not report information about the preanalytical parameters of their urine samples, even though several other studies have shown the importance of standardization of sample handling. Interestingly, it was noted that urine is used for many cancer types and not just cancers originating from the urogenital tract. Many different types of relevant analytes have been shown to be found in urine. Additionally, future considerations and recommendations are discussed: (a) the heterogeneous nature of urine, (b) the need for standardized practice protocols and (c) the road toward the clinic. Urine is an emerging liquid biopsy with broad applicability in different analytes and several cancer types. However, standard practice protocols for sample handling and processing would help to elaborate the clinical utility of urine in cancer research, detection and disease monitoring.
Collapse
Affiliation(s)
- Stephanie Jordaens
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium.,Novosanis NV, Wijnegem, Belgium
| | - Karen Zwaenepoel
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium.,Laboratory of Pathological Anatomy, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Wiebren Tjalma
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium.,Multidisciplinary Breast Clinic, Gynecological Oncology Unit, Department of Obstetrics and Gynecology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Christophe Deben
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | | | - Vanessa Vankerckhoven
- Novosanis NV, Wijnegem, Belgium.,Center for Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium.,Laboratory of Pathological Anatomy, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Alex Vorsters
- Center for Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
29
|
Schmidt F, Kohlbrenner D, Malesevic S, Huang A, Klein SD, Puhan MA, Kohler M. Mapping the landscape of lung cancer breath analysis: A scoping review (ELCABA). Lung Cancer 2023; 175:131-140. [PMID: 36529115 DOI: 10.1016/j.lungcan.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/23/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022]
Abstract
Lung cancer is the leading cause of cancer death worldwide due to its late-stage detection. Lung cancer screening, including low-dose computed tomography (low-dose CT), provides an initial clinical solution. Nevertheless, further innovations and refinements would help to alleviate remaining limitations. The non-invasive, gentle, and fast nature of breath analysis (BA) makes this technology highly attractive to supplement low-dose CT for an improved screening algorithm. However, BA has not taken hold in everyday clinical practice. One reason might be the heterogeneity and variety of BA methods. This scoping review is a comprehensive summary of study designs, breath analytical methods, and suggested biomarkers in lung cancer. Furthermore, this synthesis provides a framework with core outcomes for future studies in lung cancer BA. This work supports future research for evidence synthesis, meta-analysis, and translation into clinical routine workflows.
Collapse
Affiliation(s)
- Felix Schmidt
- University of Zurich, Faculty of Medicine, Zurich, Switzerland; University Hospital Zurich, Department of Pulmonology, Zurich, Switzerland.
| | - Dario Kohlbrenner
- University of Zurich, Faculty of Medicine, Zurich, Switzerland; University Hospital Zurich, Department of Pulmonology, Zurich, Switzerland
| | - Stefan Malesevic
- University of Zurich, Faculty of Medicine, Zurich, Switzerland; University Hospital Zurich, Department of Pulmonology, Zurich, Switzerland
| | - Alice Huang
- University Hospital Zurich, Department of Medical Oncology and Hematology, Zurich, Switzerland
| | - Sabine D Klein
- University of Zurich, University Library, Zurich, Switzerland
| | - Milo A Puhan
- University of Zurich, Epidemiology, Biostatistics and Prevention Institute, Zurich, Switzerland
| | - Malcolm Kohler
- University of Zurich, Faculty of Medicine, Zurich, Switzerland; University Hospital Zurich, Department of Pulmonology, Zurich, Switzerland; University of Zurich, Zurich Centre for Integrative Human Physiology, Zurich, Switzerland
| |
Collapse
|
30
|
Matsuta R, Yamamoto H, Tomita M, Saito R. iDMET: network-based approach for integrating differential analysis of cancer metabolomics. BMC Bioinformatics 2022; 23:508. [PMID: 36443658 PMCID: PMC9706903 DOI: 10.1186/s12859-022-05068-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Comprehensive metabolomic analyses have been conducted in various institutes and a large amount of metabolomic data are now publicly available. To help fully exploit such data and facilitate their interpretation, metabolomic data obtained from different facilities and different samples should be integrated and compared. However, large-scale integration of such data for biological discovery is challenging given that they are obtained from various types of sample at different facilities and by different measurement techniques, and the target metabolites and sensitivities to detect them also differ from study to study. RESULTS We developed iDMET, a network-based approach to integrate metabolomic data from different studies based on the differential metabolomic profiles between two groups, instead of the metabolite profiles themselves. As an application, we collected cancer metabolomic data from 27 previously published studies and integrated them using iDMET. A pair of metabolomic changes observed in the same disease from two studies were successfully connected in the network, and a new association between two drugs that may have similar effects on the metabolic reactions was discovered. CONCLUSIONS We believe that iDMET is an efficient tool for integrating heterogeneous metabolomic data and discovering novel relationships between biological phenomena.
Collapse
Affiliation(s)
- Rira Matsuta
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Fujisawa, Kanagawa, 252-8520, Japan
- Human Metabolome Technologies, Inc., 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | - Hiroyuki Yamamoto
- Human Metabolome Technologies, Inc., 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan.
| | - Masaru Tomita
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Fujisawa, Kanagawa, 252-8520, Japan
| | - Rintaro Saito
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0052, Japan
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Fujisawa, Kanagawa, 252-8520, Japan
| |
Collapse
|
31
|
Saman H, Raza A, Patil K, Uddin S, Crnogorac-Jurcevic T. Non-Invasive Biomarkers for Early Lung Cancer Detection. Cancers (Basel) 2022; 14:5782. [PMID: 36497263 PMCID: PMC9739091 DOI: 10.3390/cancers14235782] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/27/2022] Open
Abstract
Worldwide, lung cancer (LC) is the most common cause of cancer death, and any delay in the detection of new and relapsed disease serves as a major factor for a significant proportion of LC morbidity and mortality. Though invasive methods such as tissue biopsy are considered the gold standard for diagnosis and disease monitoring, they have several limitations. Therefore, there is an urgent need to identify and validate non-invasive biomarkers for the early diagnosis, prognosis, and treatment of lung cancer for improved patient management. Despite recent progress in the identification of non-invasive biomarkers, currently, there is a shortage of reliable and accessible biomarkers demonstrating high sensitivity and specificity for LC detection. In this review, we aim to cover the latest developments in the field, including the utility of biomarkers that are currently used in LC screening and diagnosis. We comment on their limitations and summarise the findings and developmental stages of potential molecular contenders such as microRNAs, circulating tumour DNA, and methylation markers. Furthermore, we summarise research challenges in the development of biomarkers used for screening purposes and the potential clinical applications of newly discovered biomarkers.
Collapse
Affiliation(s)
- Harman Saman
- Hamad Medical Corporation, Doha 3050, Qatar
- Barts Cancer Institute, Queen Mary University of London, London EC1M 5PZ, UK
| | - Afsheen Raza
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha 3050, Qatar
| | - Kalyani Patil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Laboratory of Animal Research Centre, Qatar University, Doha 2731, Qatar
| | | |
Collapse
|
32
|
Parker AL, Bowman E, Zingone A, Ryan BM, Cooper WA, Kohonen-Corish M, Harris CC, Cox TR. Extracellular matrix profiles determine risk and prognosis of the squamous cell carcinoma subtype of non-small cell lung carcinoma. Genome Med 2022; 14:126. [PMID: 36404344 PMCID: PMC9677915 DOI: 10.1186/s13073-022-01127-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/14/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Squamous cell carcinoma (SqCC) is a subtype of non-small cell lung cancer for which patient prognosis remains poor. The extracellular matrix (ECM) is critical in regulating cell behavior; however, its importance in tumor aggressiveness remains to be comprehensively characterized. METHODS Multi-omics data of SqCC human tumor specimens was combined to characterize ECM features associated with initiation and recurrence. Penalized logistic regression was used to define a matrix risk signature for SqCC tumors and its performance across a panel of tumor types and in SqCC premalignant lesions was evaluated. Consensus clustering was used to define prognostic matreotypes for SqCC tumors. Matreotype-specific tumor biology was defined by integration of bulk RNAseq with scRNAseq data, cell type deconvolution, analysis of ligand-receptor interactions and enriched biological pathways, and through cross comparison of matreotype expression profiles with aging and idiopathic pulmonary fibrosis lung profiles. RESULTS This analysis revealed subtype-specific ECM signatures associated with tumor initiation that were predictive of premalignant progression. We identified an ECM-enriched tumor subtype associated with the poorest prognosis. In silico analysis indicates that matrix remodeling programs differentially activate intracellular signaling in tumor and stromal cells to reinforce matrix remodeling associated with resistance and progression. The matrix subtype with the poorest prognosis resembles ECM remodeling in idiopathic pulmonary fibrosis and may represent a field of cancerization associated with elevated cancer risk. CONCLUSIONS Collectively, this analysis defines matrix-driven features of poor prognosis to inform precision medicine prevention and treatment strategies towards improving SqCC patient outcome.
Collapse
Affiliation(s)
- Amelia L. Parker
- grid.415306.50000 0000 9983 6924Matrix and Metastasis Lab, Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, 384 Victoria St, Darlinghurst, NSW 2052 Australia ,grid.1005.40000 0004 4902 0432School of Clinical Medicine, UNSW Sydney, Sydney, 2052 Australia
| | - Elise Bowman
- grid.48336.3a0000 0004 1936 8075Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892 USA
| | - Adriana Zingone
- grid.48336.3a0000 0004 1936 8075Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892 USA
| | - Brid M. Ryan
- grid.48336.3a0000 0004 1936 8075Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892 USA ,Present address: MiNA Therapeutics, London, UK
| | - Wendy A. Cooper
- grid.413249.90000 0004 0385 0051Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050 Australia ,grid.1013.30000 0004 1936 834XSydney Medical School, University of Sydney, Sydney, NSW 2050 Australia ,grid.1029.a0000 0000 9939 5719Discipline of Pathology, School of Medicine, Western Sydney University, Liverpool, NSW 2170 Australia
| | - Maija Kohonen-Corish
- grid.417229.b0000 0000 8945 8472Woolcock Institute of Medical Research, Sydney, NSW 2037 Australia ,grid.1005.40000 0004 4902 0432Microbiome Research Centre, School of Clinical Medicine, UNSW Sydney, Sydney, 2052 Australia ,grid.415306.50000 0000 9983 6924Garvan Institute of Medical Research, Darlinghurst, NSW 2010 Australia
| | - Curtis C. Harris
- grid.48336.3a0000 0004 1936 8075Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892 USA
| | - Thomas R. Cox
- grid.415306.50000 0000 9983 6924Matrix and Metastasis Lab, Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, 384 Victoria St, Darlinghurst, NSW 2052 Australia ,grid.1005.40000 0004 4902 0432School of Clinical Medicine, UNSW Sydney, Sydney, 2052 Australia
| |
Collapse
|
33
|
Kim JO, Balshaw R, Trevena C, Banerji S, Murphy L, Dawe D, Tan L, Srinathan S, Buduhan G, Kidane B, Qing G, Domaratzki M, Aliani M. Data-driven identification of plasma metabolite clusters and metabolites of interest for potential detection of early-stage non-small cell lung cancer cases versus cancer-free controls. Cancer Metab 2022; 10:16. [PMID: 36224630 PMCID: PMC9559833 DOI: 10.1186/s40170-022-00294-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Metabolomics is a potential means for biofluid-based lung cancer detection. We conducted a non-targeted, data-driven assessment of plasma from early-stage non-small cell lung cancer (ES-NSCLC) cases versus cancer-free controls (CFC) to explore and identify the classes of metabolites for further targeted metabolomics biomarker development. METHODS Plasma from 250 ES-NSCLC cases and 250 CFCs underwent ultra-high-performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UHPLC-QTOF-MS) in positive and negative electrospray ionization (ESI) modes. Molecular feature extraction, formula generation, and find-by-ion tools annotated metabolic entities. Analysis was restricted to endogenous metabolites present in ≥ 80% of samples. Unsupervised hierarchical cluster analysis identified clusters of metabolites. The metabolites with the strongest correlation with the principal component of each cluster were included in logistic regression modeling to assess discriminatory performance with and without adjustment for clinical covariates. RESULTS A total of 1900 UHPLC-QTOF-MS assessments identified 1667 and 2032 endogenous metabolites in the ESI-positive and ESI-negative modes, respectively. After data filtration, 676 metabolites remained, and 12 clusters of metabolites were identified from each ESI mode. Multivariable logistic regression using the representative metabolite from each cluster revealed effective classification of cases from controls with overall diagnostic accuracy of 91% (ESI positive) and 94% (ESI negative). Metabolites of interest identified for further targeted analysis include the following: 1b, 3a, 12a-trihydroxy-5b-cholanoic acid, pyridoxamine 5'-phosphate, sphinganine 1-phosphate, gamma-CEHC, 20-carboxy-leukotriene B4, isodesmosine, and 18-hydroxycortisol. CONCLUSIONS Plasma-based metabolomic detection of early-stage NSCLC appears feasible. Further metabolomics studies targeting phospholipid, steroid, and fatty acid metabolism are warranted to further develop noninvasive metabolomics-based detection of early-stage NSCLC.
Collapse
Affiliation(s)
- Julian O Kim
- Section of Radiation Oncology, Department of Radiology, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. .,CancerCare Manitoba Research Institute, Winnipeg, Manitoba, Canada.
| | - Robert Balshaw
- George and Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Connel Trevena
- Department of Computer Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shantanu Banerji
- CancerCare Manitoba Research Institute, Winnipeg, Manitoba, Canada.,Section of Medical Oncology, Department of Internal Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Leigh Murphy
- CancerCare Manitoba Research Institute, Winnipeg, Manitoba, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Dawe
- CancerCare Manitoba Research Institute, Winnipeg, Manitoba, Canada.,Section of Medical Oncology, Department of Internal Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lawrence Tan
- Section of Thoracic Surgery, Department of Surgery, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sadeesh Srinathan
- Section of Thoracic Surgery, Department of Surgery, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gordon Buduhan
- Section of Thoracic Surgery, Department of Surgery, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Biniam Kidane
- Section of Thoracic Surgery, Department of Surgery, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gefei Qing
- Department of Human Pathology, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael Domaratzki
- Department of Computer Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michel Aliani
- Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
34
|
Chardin D, Gille C, Pourcher T, Humbert O, Barlaud M. Learning a confidence score and the latent space of a new supervised autoencoder for diagnosis and prognosis in clinical metabolomic studies. BMC Bioinformatics 2022; 23:361. [PMID: 36050631 PMCID: PMC9434875 DOI: 10.1186/s12859-022-04900-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022] Open
Abstract
Background Presently, there is a wide variety of classification methods and deep neural network approaches in bioinformatics. Deep neural networks have proven their effectiveness for classification tasks, and have outperformed classical methods, but they suffer from a lack of interpretability. Therefore, these innovative methods are not appropriate for decision support systems in healthcare. Indeed, to allow clinicians to make informed and well thought out decisions, the algorithm should provide the main pieces of information used to compute the predicted diagnosis and/or prognosis, as well as a confidence score for this prediction. Methods Herein, we used a new supervised autoencoder (SAE) approach for classification of clinical metabolomic data. This new method has the advantage of providing a confidence score for each prediction thanks to a softmax classifier and a meaningful latent space visualization and to include a new efficient feature selection method, with a structured constraint, which allows for biologically interpretable results. Results Experimental results on three metabolomics datasets of clinical samples illustrate the effectiveness of our SAE and its confidence score. The supervised autoencoder provides an accurate localization of the patients in the latent space, and an efficient confidence score. Experiments show that the SAE outperforms classical methods (PLS-DA, Random Forests, SVM, and neural networks (NN)). Furthermore, the metabolites selected by the SAE were found to be biologically relevant. Conclusion In this paper, we describe a new efficient SAE method to support diagnostic or prognostic evaluation based on metabolomics analyses.
Collapse
Affiliation(s)
- David Chardin
- Transporters in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des sciences du vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Université Côte d'Azur (UCA), Nice, France.,Centre Antoine Lacassagne, Université Côte d'Azur (UCA), Nice, France
| | - Cyprien Gille
- Laboratoire d'Informatique, Signaux et Systèmes de Sophia Antipolis (I3S), Centre de Recherche Scientifique (CNRS), Université Côte d'Azur (UCA), Sophia Antipolis, France
| | - Thierry Pourcher
- Transporters in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des sciences du vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Université Côte d'Azur (UCA), Nice, France
| | - Olivier Humbert
- Transporters in Imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institut des sciences du vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Université Côte d'Azur (UCA), Nice, France.,Centre Antoine Lacassagne, Université Côte d'Azur (UCA), Nice, France
| | - Michel Barlaud
- Laboratoire d'Informatique, Signaux et Systèmes de Sophia Antipolis (I3S), Centre de Recherche Scientifique (CNRS), Université Côte d'Azur (UCA), Sophia Antipolis, France.
| |
Collapse
|
35
|
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: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
36
|
Parker AL, Toulabi L, Oike T, Kanke Y, Patel D, Tada T, Taylor S, Beck JA, Bowman E, Reyzer ML, Butcher D, Kuhn S, Pauly GT, Krausz KW, Gonzalez FJ, Hussain SP, Ambs S, Ryan BM, Wang XW, Harris CC. Creatine riboside is a cancer cell-derived metabolite associated with arginine auxotrophy. J Clin Invest 2022; 132:157410. [PMID: 35838048 PMCID: PMC9282934 DOI: 10.1172/jci157410] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/25/2022] [Indexed: 12/17/2022] Open
Abstract
The metabolic dependencies of cancer cells have substantial potential to be exploited to improve the diagnosis and treatment of cancer. Creatine riboside (CR) is identified as a urinary metabolite associated with risk and prognosis in lung and liver cancer. However, the source of high CR levels in patients with cancer as well as their implications for the treatment of these aggressive cancers remain unclear. By integrating multiomics data on lung and liver cancer, we have shown that CR is a cancer cell–derived metabolite. Global metabolomics and gene expression analysis of human tumors and matched liquid biopsies, together with functional studies, revealed that dysregulation of the mitochondrial urea cycle and a nucleotide imbalance were associated with high CR levels and indicators of a poor prognosis. This metabolic phenotype was associated with reduced immune infiltration and supported rapid cancer cell proliferation that drove aggressive tumor growth. CRhi cancer cells were auxotrophic for arginine, revealing a metabolic vulnerability that may be exploited therapeutically. This highlights the potential of CR not only as a poor-prognosis biomarker but also as a companion biomarker to inform the administration of arginine-targeted therapies in precision medicine strategies to improve survival for patients with cancer.
Collapse
Affiliation(s)
- Amelia L Parker
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Leila Toulabi
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Takahiro Oike
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Yasuyuki Kanke
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Daxeshkumar Patel
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Takeshi Tada
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Sheryse Taylor
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Jessica A Beck
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Elise Bowman
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Michelle L Reyzer
- National Research Resource for Imaging Mass Spectrometry, Vanderbilt University, Nashville, Tennessee, USA
| | - Donna Butcher
- Pathology and Histotechnology Laboratory, Frederick National Laboratory, Frederick, Maryland, USA
| | - Skyler Kuhn
- Center for Cancer Research Collaborative Bioinformatics Resource
| | | | | | | | - S Perwez Hussain
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Bríd M Ryan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA.,Liver Cancer Program, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| |
Collapse
|
37
|
Tzanakis K, Nattkemper TW, Niehaus K, Albaum SP. MetHoS: a platform for large-scale processing, storage and analysis of metabolomics data. BMC Bioinformatics 2022; 23:267. [PMID: 35804309 PMCID: PMC9270834 DOI: 10.1186/s12859-022-04793-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 06/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Modern mass spectrometry has revolutionized the detection and analysis of metabolites but likewise, let the data skyrocket with repositories for metabolomics data filling up with thousands of datasets. While there are many software tools for the analysis of individual experiments with a few to dozens of chromatograms, we see a demand for a contemporary software solution capable of processing and analyzing hundreds or even thousands of experiments in an integrative manner with standardized workflows. RESULTS Here, we introduce MetHoS as an automated web-based software platform for the processing, storage and analysis of great amounts of mass spectrometry-based metabolomics data sets originating from different metabolomics studies. MetHoS is based on Big Data frameworks to enable parallel processing, distributed storage and distributed analysis of even larger data sets across clusters of computers in a highly scalable manner. It has been designed to allow the processing and analysis of any amount of experiments and samples in an integrative manner. In order to demonstrate the capabilities of MetHoS, thousands of experiments were downloaded from the MetaboLights database and used to perform a large-scale processing, storage and statistical analysis in a proof-of-concept study. CONCLUSIONS MetHoS is suitable for large-scale processing, storage and analysis of metabolomics data aiming at untargeted metabolomic analyses. It is freely available at: https://methos.cebitec.uni-bielefeld.de/ . Users interested in analyzing their own data are encouraged to apply for an account.
Collapse
Affiliation(s)
- Konstantinos Tzanakis
- International Research Training Group "Computational Methods for the Analysis of the Diversity and Dynamics of Genomes", Faculty of Technology, Bielefeld University, Bielefeld, Germany.
| | - Tim W Nattkemper
- Biodata Mining Group, Center for Biotechnology (CeBiTec), Faculty of Technology, Bielefeld University, Bielefeld, Germany
| | - Karsten Niehaus
- Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Stefan P Albaum
- Bioinformatics Resource Facility, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| |
Collapse
|
38
|
Characteristics of Normalization Methods in Quantitative Urinary Metabolomics—Implications for Epidemiological Applications and Interpretations. Biomolecules 2022; 12:biom12070903. [PMID: 35883459 PMCID: PMC9313036 DOI: 10.3390/biom12070903] [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: 05/21/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 01/25/2023] Open
Abstract
A systematic comparison is presented for the effects of seven different normalization schemes in quantitative urinary metabolomics. Morning spot urine samples were analyzed with nuclear magnetic resonance (NMR) spectroscopy from a population-based group of 994 individuals. Forty-four metabolites were quantified and the metabolite–metabolite associations and the associations of metabolite concentrations with two representative clinical measures, body mass index and mean arterial pressure, were analyzed. Distinct differences were observed when comparing the effects of normalization for the intra-urine metabolite associations with those for the clinical associations. The metabolite–metabolite associations show quite complex patterns of similarities and dissimilarities between the different normalization methods, while the epidemiological association patterns are consistent, leading to the same overall biological interpretations. The results indicate that, in general, the normalization method appears to have only minor influences on standard epidemiological regression analyses with clinical/physiological measures. Multimetabolite normalization schemes showed consistent results with the customary creatinine reference. Nevertheless, interpretations of intra-urine metabolite associations and nuanced understanding of the epidemiological associations call for comparisons with different normalizations and accounting for the physiology, metabolism and kidney function related to the normalization schemes.
Collapse
|
39
|
Thaiparambil J, Dong J, Grimm SL, Perera D, Ambati CSR, Putluri V, Robertson MJ, Patel TD, Mistretta B, Gunaratne PH, Kim MP, Yustein JT, Putluri N, Coarfa C, El‐Zein R. Integrative metabolomics and transcriptomics analysis reveals novel therapeutic vulnerabilities in lung cancer. Cancer Med 2022; 12:584-596. [PMID: 35676822 PMCID: PMC9844651 DOI: 10.1002/cam4.4933] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/22/2022] [Accepted: 05/04/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) comprises the majority (~85%) of all lung tumors, with lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC) being the most frequently diagnosed histological subtypes. Multi-modal omics profiling has been carried out in NSCLC, but no studies have yet reported a unique metabolite-related gene signature and altered metabolic pathways associated with LUAD and LUSC. METHODS We integrated transcriptomics and metabolomics to analyze 30 human lung tumors and adjacent noncancerous tissues. Differential co-expression was used to identify modules of metabolites that were altered between normal and tumor. RESULTS We identified unique metabolite-related gene signatures specific for LUAD and LUSC and key pathways aberrantly regulated at both transcriptional and metabolic levels. Differential co-expression analysis revealed that loss of coherence between metabolites in tumors is a major characteristic in both LUAD and LUSC. We identified one metabolic onco-module gained in LUAD, characterized by nine metabolites and 57 metabolic genes. Multi-omics integrative analysis revealed a 28 metabolic gene signature associated with poor survival in LUAD, with six metabolite-related genes as individual prognostic markers. CONCLUSIONS We demonstrated the clinical utility of this integrated metabolic gene signature in LUAD by using it to guide repurposing of AZD-6482, a PI3Kβ inhibitor which significantly inhibited three genes from the 28-gene signature. Overall, we have integrated metabolomics and transcriptomics analyses to show that LUAD and LUSC have distinct profiles, inferred gene signatures with prognostic value for patient survival, and identified therapeutic targets and repurposed drugs for potential use in NSCLC treatment.
Collapse
Affiliation(s)
| | - Jianrong Dong
- Center for Precision and Environmental HealthBaylor College of MedicineHoustonTexasUSA,Molecular and Cellular Biology DepartmentBaylor College of MedicineHoustonTexasUSA
| | - Sandra L. Grimm
- Center for Precision and Environmental HealthBaylor College of MedicineHoustonTexasUSA,Dan L Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTexasUSA,Advanced Technology CoresBaylor College of MedicineHoustonTexasUSA
| | - Dimuthu Perera
- Advanced Technology CoresBaylor College of MedicineHoustonTexasUSA
| | | | - Vasanta Putluri
- Advanced Technology CoresBaylor College of MedicineHoustonTexasUSA
| | - Matthew J. Robertson
- Dan L Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTexasUSA,Advanced Technology CoresBaylor College of MedicineHoustonTexasUSA
| | - Tajhal D. Patel
- Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma CenterBaylor College of MedicineHoustonTexasUSA
| | - Brandon Mistretta
- Department of Biology and BiochemistryUniversity of HoustonHoustonTexasUSA
| | - Preethi H. Gunaratne
- Molecular and Cellular Biology DepartmentBaylor College of MedicineHoustonTexasUSA,Department of Biology and BiochemistryUniversity of HoustonHoustonTexasUSA
| | - Min P. Kim
- Houston Methodist Cancer CenterHoustonTexasUSA,Division of Thoracic Surgery, Department of SurgeryHouston Methodist HospitalHoustonTexasUSA
| | - Jason T. Yustein
- Molecular and Cellular Biology DepartmentBaylor College of MedicineHoustonTexasUSA,Dan L Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTexasUSA,Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma CenterBaylor College of MedicineHoustonTexasUSA,Integrative Molecular and Biological Sciences ProgramBaylor College of MedicineHoustonTexasUSA
| | - Nagireddy Putluri
- Molecular and Cellular Biology DepartmentBaylor College of MedicineHoustonTexasUSA,Advanced Technology CoresBaylor College of MedicineHoustonTexasUSA
| | - Cristian Coarfa
- Center for Precision and Environmental HealthBaylor College of MedicineHoustonTexasUSA,Molecular and Cellular Biology DepartmentBaylor College of MedicineHoustonTexasUSA,Dan L Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTexasUSA,Advanced Technology CoresBaylor College of MedicineHoustonTexasUSA
| | | |
Collapse
|
40
|
Yu H, Huan T. MAFFIN: Metabolomics Sample Normalization Using Maximal Density Fold Change with High-Quality Metabolic Features and Corrected Signal Intensities. Bioinformatics 2022; 38:3429-3437. [PMID: 35639662 DOI: 10.1093/bioinformatics/btac355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/18/2022] [Accepted: 05/19/2022] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Post-acquisition sample normalization is a critical step in comparative metabolomics to remove the variation introduced by sample amount or concentration difference. Previously reported approaches are either specific to one sample type or built on strong assumptions on data structure, which are limited to certain levels. This encouraged us to develop MAFFIN, an accurate and robust post-acquisition sample normalization workflow that works universally for metabolomics data collected on mass spectrometry (MS) platforms. RESULTS MAFFIN calculates normalization factors using maximal density fold change (MDFC) computed by a kernel density-based approach. Using both simulated data and 20 metabolomics data sets, we showcased that MDFC outperforms four commonly used normalization methods in terms of reducing the intragroup variation among samples. Two essential steps, overlooked in conventional methods, were also examined and incorporated into MAFFIN. (1) MAFFIN uses multiple orthogonal criteria to select high-quality features for normalization factor calculation, which minimizes the bias caused by abiotic features or metabolites with poor quantitative performance. (2) MAFFIN corrects the MS signal intensities of high-quality features using serial quality control (QC) samples, which guarantees the accuracy of fold change calculations. MAFFIN was applied to a human saliva metabolomics study and led to better data separation in principal component analysis (PCA) and more confirmed significantly altered metabolites. AVAILABILITY AND IMPLEMENTATION The MAFFIN algorithm was implemented in an R package named MAFFIN. Package installation, user instruction, and demo data are available at https://github.com/HuanLab/MAFFIN. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Huaxu Yu
- Department of Chemistry, Faculty of Science, The University of British Columbia, Vancouver Campus, 2036 Main Mall, Vancouver, BC, V6T 1Z1, Canada
| | - Tao Huan
- Department of Chemistry, Faculty of Science, The University of British Columbia, Vancouver Campus, 2036 Main Mall, Vancouver, BC, V6T 1Z1, Canada
| |
Collapse
|
41
|
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: 36] [Impact Index Per Article: 12.0] [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
|
42
|
In-situ growth of boronic acid-decorated metal-organic framework on Fe3O4 nanospheres for specific enrichment of cis-diol containing nucleosides. Anal Chim Acta 2022; 1206:339772. [DOI: 10.1016/j.aca.2022.339772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/09/2022] [Accepted: 03/24/2022] [Indexed: 12/26/2022]
|
43
|
Kozłowska L, Santonen T, Duca RC, Godderis L, Jagiello K, Janasik B, Van Nieuwenhuyse A, Poels K, Puzyn T, Scheepers PTJ, Sijko M, Silva MJ, Sosnowska A, Viegas S, Verdonck J, Wąsowicz W. HBM4EU Chromates Study: Urinary Metabolomics Study of Workers Exposed to Hexavalent Chromium. Metabolites 2022; 12:362. [PMID: 35448548 PMCID: PMC9032989 DOI: 10.3390/metabo12040362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Exposure to hexavalent chromium Cr(VI) may occur in several occupational activities, placing workers in many industries at risk for potential related health outcomes. Untargeted metabolomics was applied to investigate changes in metabolic pathways in response to Cr(VI) exposure. We obtained our data from a study population of 220 male workers with exposure to Cr(VI) and 102 male controls from Belgium, Finland, Poland, Portugal and the Netherlands within the HBM4EU Chromates Study. Urinary metabolite profiles were determined using liquid chromatography mass spectrometry, and differences between post-shift exposed workers and controls were analyzed using principal component analysis. Based on the first two principal components, we observed clustering by industrial chromate application, such as welding, chrome plating, and surface treatment, distinct from controls and not explained by smoking status or alcohol use. The changes in the abundancy of excreted metabolites observed in workers reflect fatty acid and monoamine neurotransmitter metabolism, oxidative modifications of amino acid residues, the excessive formation of abnormal amino acid metabolites and changes in steroid and thyrotropin-releasing hormones. The observed responses could also have resulted from work-related factors other than Cr(VI). Further targeted metabolomics studies are needed to better understand the observed modifications and further explore the suitability of urinary metabolites as early indicators of adverse effects associated with exposure to Cr(VI).
Collapse
Affiliation(s)
- Lucyna Kozłowska
- Laboratory of Human Metabolism Research, Department of Dietetics, Warsaw University of Life Sciences, 02776 Warsaw, Poland;
| | - Tiina Santonen
- Finnish Institute of Occupational Health, 00250 Helsinki, Finland;
| | - Radu Corneliu Duca
- Labotoire National de Santé (LNS), Unit Environmental Hygiene and Human Biological Monitoring, Department of Health Protection, 3555 Dudelange, Luxembourg;
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
| | - Lode Godderis
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
- IDEWE, External Service for Prevention and Protection at Work, 3001 Heverlee, Belgium
| | - Karolina Jagiello
- QSAR Laboratory Ltd., 80172 Gdansk, Poland; (K.J.); (T.P.); (A.S.)
- Laboratory of Environmental Chemoinfomatics, Department of Environmental Chemistry and Radiochemistry, Faculty of Chemistry, University of Gdansk, 80308 Gdansk, Poland
| | - Beata Janasik
- Department of Environmental and Biological Monitoring, Nofer Institute of Occupational Medicine, 91348 Lodz, Poland; (B.J.); (W.W.)
| | - An Van Nieuwenhuyse
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
- Laboratoire National de Santé (LNS), Department of Health Protection, 3555 Dudelange, Luxembourg
| | - Katrien Poels
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
| | - Tomasz Puzyn
- QSAR Laboratory Ltd., 80172 Gdansk, Poland; (K.J.); (T.P.); (A.S.)
- Laboratory of Environmental Chemoinfomatics, Department of Environmental Chemistry and Radiochemistry, Faculty of Chemistry, University of Gdansk, 80308 Gdansk, Poland
| | - Paul T. J. Scheepers
- Radboud Institute for Health Sciences, Radboudumc, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands;
| | - Monika Sijko
- Laboratory of Human Metabolism Research, Department of Dietetics, Warsaw University of Life Sciences, 02776 Warsaw, Poland;
| | - Maria João Silva
- Human Genetics Department, National Institute of Health Dr. Ricardo Jorge (INSA), Toxicogenomics and Human Health (ToxOmics), NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal;
| | - Anita Sosnowska
- QSAR Laboratory Ltd., 80172 Gdansk, Poland; (K.J.); (T.P.); (A.S.)
| | - Susana Viegas
- Public Health Research Centre, NOVA National School of Public Health, Universidade NOVA de Lisbon, 1600-560 Lisbon, Portugal;
- Comprehensive Health Research Center (CHRC), 1169-056 Lisbon, Portugal
| | - Jelle Verdonck
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven (University of Leuven), 3000 Leuven, Belgium; (L.G.); (A.V.N.); (K.P.); (J.V.)
| | - Wojciech Wąsowicz
- Department of Environmental and Biological Monitoring, Nofer Institute of Occupational Medicine, 91348 Lodz, Poland; (B.J.); (W.W.)
| | | | | |
Collapse
|
44
|
Modeling of Tumor Growth with Input from Patient-Specific Metabolomic Data. Ann Biomed Eng 2022; 50:314-329. [PMID: 35083584 PMCID: PMC9743982 DOI: 10.1007/s10439-022-02904-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/01/2022] [Indexed: 12/15/2022]
Abstract
Advances in omic technologies have provided insight into cancer progression and treatment response. However, the nonlinear characteristics of cancer growth present a challenge to bridge from the molecular- to the tissue-scale, as tumor behavior cannot be encapsulated by the sum of the individual molecular details gleaned experimentally. Mathematical modeling and computational simulation have been traditionally employed to facilitate analysis of nonlinear systems. In this study, for the first time tumor metabolomic data are linked via mathematical modeling to the tumor tissue-scale behavior, showing the capability to mechanistically simulate cancer progression personalized to omic information obtainable from patient tumor core biopsy analysis. Generally, a higher degree of metabolic dysregulation has been correlated with more aggressive tumor behavior. Accordingly, key parameters influenced by metabolomic data in this model include tumor proliferation, vascularization, aggressiveness, lactic acid production, monocyte infiltration and macrophage polarization, and drug effect. The model enables evaluating interactions of interest between these parameters which drive tumor growth based on the metabolomic data. The results show that the model can group patients consistently with the clinically observed outcomes of response/non-response to chemotherapy. This modeling approach provides a first step towards evaluation of tumor growth based on tumor-specific metabolomic data.
Collapse
|
45
|
Singh A, Prakash V, Gupta N, Kumar A, Kant R, Kumar D. Serum Metabolic Disturbances in Lung Cancer Investigated through an Elaborative NMR-Based Serum Metabolomics Approach. ACS OMEGA 2022; 7:5510-5520. [PMID: 35187366 PMCID: PMC8851899 DOI: 10.1021/acsomega.1c06941] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/18/2022] [Indexed: 06/01/2023]
Abstract
Detection of metabolic disturbances in lung cancer (LC) has the potential to aid early diagnosis/prognosis and hence improve disease management strategies through reliable grading, staging, and determination of neoadjuvant status in LC. However, a majority of previous metabolomics studies compare the normalized spectral features which not only provide ambiguous information but further limit the clinical translation of this information. Various such issues can be resolved by performing the concentration profiling of various metabolites with respect to formate as an internal reference using commercial software Chenomx. Continuing our efforts in this direction, the serum metabolic profiles were measured on 39 LC patients and 42 normal controls (NCs, comparable in age/sex) using high-field 800 MHz NMR spectroscopy and compared using multivariate statistical analysis tools to identify metabolic disturbances and metabolites of diagnostic potential. Partial least-squares discriminant analysis (PLS-DA) model revealed a distinct separation between LC and NC groups and resulted in excellent discriminatory ability with the area under the receiver-operating characteristic (AUROC) = 0.97 [95% CI = 0.89-1.00]. The metabolic features contributing to the differentiation of LC from NC samples were identified first using variable importance in projection (VIP) score analysis and then checked for their statistical significance (with p-value < 0.05) and diagnostic potential using the ROC curve analysis. The analysis revealed relevant metabolic disturbances associated with LC. Among various circulatory metabolites, six metabolites, including histidine, glutamine, glycine, threonine, alanine, and valine, were found to be of apposite diagnostic potential for clinical implications. These metabolic alterations indicated altered glucose metabolism, aberrant fatty acid synthesis, and augmented utilization of various amino acids including active glutaminolysis in LC.
Collapse
Affiliation(s)
- Anjana Singh
- All
India Institute of Medical Sciences (AIIMS), Rishikesh, Uttarakhand 249201, India
- Pulmonary
& Critical Care Medicine, King George’s
Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Ved Prakash
- Pulmonary
& Critical Care Medicine, King George’s
Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Nikhil Gupta
- Centre
of Biomedical Research (CBMR), SGPGIMS, Lucknow, Uttar Pradesh 226014, India
- Department
of Chemistry, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Ashish Kumar
- Department
of Chemistry, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Ravi Kant
- All
India Institute of Medical Sciences (AIIMS), Rishikesh, Uttarakhand 249201, India
| | - Dinesh Kumar
- Centre
of Biomedical Research (CBMR), SGPGIMS, Lucknow, Uttar Pradesh 226014, India
| |
Collapse
|
46
|
Haince JF, Joubert P, Bach H, Ahmed Bux R, Tappia PS, Ramjiawan B. Metabolomic Fingerprinting for the Detection of Early-Stage Lung Cancer: From the Genome to the Metabolome. Int J Mol Sci 2022; 23:ijms23031215. [PMID: 35163138 PMCID: PMC8835988 DOI: 10.3390/ijms23031215] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
The five-year survival rate of lung cancer patients is very low, mainly because most newly diagnosed patients present with locally advanced or metastatic disease. Therefore, early diagnosis is key to the successful treatment and management of lung cancer. Unfortunately, early detection methods of lung cancer are not ideal. In this brief review, we described early detection methods such as chest X-rays followed by bronchoscopy, sputum analysis followed by cytological analysis, and low-dose computed tomography (LDCT). In addition, we discussed the potential of metabolomic fingerprinting, compared to that of other biomarkers, including molecular targets, as a low-cost, high-throughput blood-based test that is both feasible and affordable for early-stage lung cancer screening of at-risk populations. Accordingly, we proposed a paradigm shift to metabolomics as an alternative to molecular and proteomic-based markers in lung cancer screening, which will enable blood-based routine testing and be accessible to those patients at the highest risk for lung cancer.
Collapse
Affiliation(s)
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Pathology, Laval University, Quebec, QC G1V 4G5, Canada;
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC V6H 3Z6, Canada;
| | - Rashid Ahmed Bux
- BioMark Diagnostics Inc., Richmond, BC V6X 2W8, Canada; (J.-F.H.); (R.A.B.)
| | - Paramjit S. Tappia
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Correspondence: ; Tel.: +1-204-258-1230
| | - Bram Ramjiawan
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
- Department of Pharmacology & Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| |
Collapse
|
47
|
Chardin D, Humbert O, Bailleux C, Burel-Vandenbos F, Rigau V, Pourcher T, Barlaud M. Primal-dual for classification with rejection (PD-CR): a novel method for classification and feature selection-an application in metabolomics studies. BMC Bioinformatics 2021; 22:594. [PMID: 34911437 PMCID: PMC8672607 DOI: 10.1186/s12859-021-04478-w] [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: 01/19/2021] [Accepted: 10/29/2021] [Indexed: 11/25/2022] Open
Abstract
Background Supervised classification methods have been used for many years for feature selection in metabolomics and other omics studies. We developed a novel primal-dual based classification method (PD-CR) that can perform classification with rejection and feature selection on high dimensional datasets. PD-CR projects data onto a low dimension space and performs classification by minimizing an appropriate quadratic cost. It simultaneously optimizes the selected features and the prediction accuracy with a new tailored, constrained primal-dual method. The primal-dual framework is general enough to encompass various robust losses and to allow for convergence analysis. Here, we compare PD-CR to three commonly used methods: partial least squares discriminant analysis (PLS-DA), random forests and support vector machines (SVM). We analyzed two metabolomics datasets: one urinary metabolomics dataset concerning lung cancer patients and healthy controls; and a metabolomics dataset obtained from frozen glial tumor samples with mutated isocitrate dehydrogenase (IDH) or wild-type IDH. Results PD-CR was more accurate than PLS-DA, Random Forests and SVM for classification using the 2 metabolomics datasets. It also selected biologically relevant metabolites. PD-CR has the advantage of providing a confidence score for each prediction, which can be used to perform classification with rejection. This substantially reduces the False Discovery Rate. Conclusion PD-CR is an accurate method for classification of metabolomics datasets which can outperform PLS-DA, Random Forests and SVM while selecting biologically relevant features. Furthermore the confidence score provided with PD-CR can be used to perform classification with rejection and reduce the false discovery rate. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04478-w.
Collapse
Affiliation(s)
- David Chardin
- Transporters in imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institute des sciences du vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Université Côte d'Azur (UCA), Nice, France.,Department of Nuclear Medicine, Centre Antoine Lacassagne, Université Côte d'Azur (UCA), Nice, France
| | - Olivier Humbert
- Transporters in imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institute des sciences du vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Université Côte d'Azur (UCA), Nice, France.,Department of Nuclear Medicine, Centre Antoine Lacassagne, Université Côte d'Azur (UCA), Nice, France
| | - Caroline Bailleux
- Transporters in imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institute des sciences du vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Université Côte d'Azur (UCA), Nice, France.,Department of Oncology, Centre Antoine Lacassagne, Université Côte d'Azur (UCA), Nice, France
| | - Fanny Burel-Vandenbos
- Central Laboratory of Pathology, University Hospital and Institute of Biology Valrose, Inserm U1091 - CNRS UMR7277, University Côte d'Azur, Nice, France
| | - Valerie Rigau
- Department of Pathology and Oncobiology, University Hospital, Montpellier, France.,Institute for Neurosciences of Montpellier, INSERM U1051, Montpellier, France
| | - Thierry Pourcher
- Transporters in imaging and Radiotherapy in Oncology (TIRO), Direction de la Recherche Fondamentale (DRF), Institute des sciences du vivant Fréderic Joliot, Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), Université Côte d'Azur (UCA), Nice, France
| | - Michel Barlaud
- Laboratoire d'Informatique, Signaux et Systèmes de Sophia Antipolis (I3S), Université Côte d'Azur (UCA), Centre de Recherche Scientifique (CNRS), Sophia Antipolis, France.
| |
Collapse
|
48
|
Yuan Y, Zhao Z, Xue L, Wang G, Song H, Pang R, Zhou J, Luo J, Song Y, Yin Y. Identification of diagnostic markers and lipid dysregulation in oesophageal squamous cell carcinoma through lipidomic analysis and machine learning. Br J Cancer 2021; 125:351-357. [PMID: 33953345 PMCID: PMC8329198 DOI: 10.1038/s41416-021-01395-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/24/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Oesophageal cancer (EC) ranks high in both morbidity and mortality. A non-invasive and high-sensitivity diagnostic approach is necessary to improve the prognosis of EC patients. METHODS A total of 525 serum samples were subjected to lipidomic analysis. We combined serum lipidomics and machine-learning algorithms to select important metabolite features for the detection of oesophageal squamous cell carcinoma (ESCC), the major subtype of EC in developing countries. A diagnostic model using a panel of selected features was developed and evaluated. Integrative analyses of tissue transcriptome and serum lipidome were conducted to reveal the underlying mechanism of lipid dysregulation. RESULTS Our optimised diagnostic model with a panel of 12 lipid biomarkers together with age and gender reaches a sensitivity of 90.7%, 91.3% and 90.7% and an area under receiver-operating characteristic curve of 0.958, 0.966 and 0.818 in detecting ESCC for the training cohort, validation cohort and independent validation cohort, respectively. Integrative analysis revealed matched variation trend of genes encoding key enzymes in lipid metabolism. CONCLUSIONS We have identified a panel of 12 lipid biomarkers for diagnostic modelling and potential mechanisms of lipid dysregulation in the serum of ESCC patients. This is a reliable, rapid and non-invasive tumour-diagnostic approach for clinical application.
Collapse
Affiliation(s)
- Yuyao Yuan
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| | - Zitong Zhao
- grid.506261.60000 0001 0706 7839State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- grid.506261.60000 0001 0706 7839Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangxi Wang
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| | - Huajie Song
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| | - Ruifang Pang
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China ,grid.440601.70000 0004 1798 0578Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Juntuo Zhou
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| | - Jianyuan Luo
- grid.11135.370000 0001 2256 9319Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yongmei Song
- grid.506261.60000 0001 0706 7839State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Yin
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China ,grid.440601.70000 0004 1798 0578Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
49
|
Igarashi K, Ota S, Kaneko M, Hirayama A, Enomoto M, Katumata K, Sugimoto M, Soga T. High-throughput screening of salivary polyamine markers for discrimination of colorectal cancer by multisegment injection capillary electrophoresis tandem mass spectrometry. J Chromatogr A 2021; 1652:462355. [PMID: 34233246 DOI: 10.1016/j.chroma.2021.462355] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/03/2021] [Accepted: 06/15/2021] [Indexed: 01/05/2023]
Abstract
Polyamine metabolites provide pathophysiological information on disease or therapeutic efficacy, yet rapid screening methods for these biomarkers are lacking. Here, we developed high-throughput polyamine metabolite profiling based on multisegment injection capillary electrophoresis triple quadrupole tandem mass spectrometry (MSI-CE-MS/MS), which allows sequential 40-sample injection followed by electrophoretic separation and specific mass detection. To achieve consecutive analysis of polyamine samples, 1 M formic acid was used as the background electrolyte (BGE). The BGE spacer volume had an apparent effect on peak resolution among samples, and 20 nL was selected as the optimal volume. The use of polyamine isotopomers as the internal standard enabled the correction of matrix effects in MS detection. This method is sensitive, selective and quantitative, and its utility was demonstrated by screening polyamines in 359 salivary samples within 360 min, resulting in discrimination of colorectal cancer patients from noncancer controls.
Collapse
Affiliation(s)
- Kaori Igarashi
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka 997-0052, Japan.
| | - Sana Ota
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka 997-0052, Japan.
| | - Miku Kaneko
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka 997-0052, Japan.
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka 997-0052, Japan.
| | - Masanobu Enomoto
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1, Nishijinjuku, Shinjuku, Tokyo 160-0023, Japan.
| | - Kenji Katumata
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1, Nishijinjuku, Shinjuku, Tokyo 160-0023, Japan.
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka 997-0052, Japan; Research and Development Center for Minimally Invasive Therapies, Medical Research Institute, Tokyo Medical University, 6-1-1, Sinjuku, Tokyo 160-0022, Japan.
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka 997-0052, Japan; Faculty of Environmental Information Studies, Keio University, 5322 Endo, Fujisawa 252-0882, Japan.
| |
Collapse
|
50
|
Metabolic Changes in Early-Stage Non-Small Cell Lung Cancer Patients after Surgical Resection. Cancers (Basel) 2021; 13:cancers13123012. [PMID: 34208545 PMCID: PMC8234274 DOI: 10.3390/cancers13123012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Considerable progress in the treatment of non–small cell lung cancer (NSCLC) has been made possible by large-scale technologies that scan the gene expression in tumor cells. While gene expression is informative, it is the changes to cellular metabolism that directly affect the initiation and the progression of the disease. Altered metabolic processes in cancer include how the tumor cells handle fat, proteins, and sugar, produce energy, divide (grow), or migrate. We have used nuclear magnetic resonance and mass spectrometry to survey and document the metabolic changes in blood and urine samples collected from NSCLC patients before and after their lung tumors were surgically removed. We found several molecular compounds that changed in abundance in the blood or urine after surgery, many of which are related to cancer cell metabolism. Further documentation of these changes in large patient populations will lead to non-invasive ways to screen, diagnose, or monitor disease progression in lung cancer patients. Abstract Metabolic alterations in malignant cells play a vital role in tumor initiation, proliferation, and metastasis. Biofluids from patients with non–small cell lung cancer (NSCLC) harbor metabolic biomarkers with potential clinical applications. In this study, we assessed the changes in the metabolic profile of patients with early-stage NSCLC using mass spectrometry and nuclear magnetic resonance spectroscopy before and after surgical resection. A single cohort of 35 patients provided a total of 29 and 32 pairs of urine and serum samples, respectively, pre-and post-surgery. We identified a profile of 48 metabolites that were significantly different pre- and post-surgery: 17 in urine and 31 in serum. A higher proportion of metabolites were upregulated than downregulated post-surgery (p < 0.01); however, the median fold change (FC) was higher for downregulated than upregulated metabolites (p < 0.05). Purines/pyrimidines and proteins had a larger dysregulation than other classes of metabolites (p < 0.05 for each class). Several of the dysregulated metabolites have been previously associated with cancer, including leucyl proline, asymmetric dimethylarginine, isopentenyladenine, fumaric acid (all downregulated post-surgery), as well as N6-methyladenosine and several deoxycholic acid moieties, which were upregulated post-surgery. This study establishes metabolomic analysis of biofluids as a path to non-invasive diagnostics, screening, and monitoring in NSCLC.
Collapse
|