1
|
Dahl O, Myklebust MP. A study of microRNAs as new prognostic biomarkers in anal cancer patients. Acta Oncol 2024; 63:456-465. [PMID: 38899393 PMCID: PMC11332526 DOI: 10.2340/1651-226x.2024.27976] [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: 11/22/2023] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND MicroRNA (MiR) influences the growth of cancer by regulation of mRNA for 50-60% of all genes. We present as per our knowledge the first global analysis of microRNA expression in anal cancer patients and their prognostic impact. METHODS Twenty-nine patients with T1-4 N0-3 M0 anal cancer treated with curative intent from September 2003 to April 2011 were included in the study. RNA was extracted from fresh frozen tissue and sequenced using NGS. Differentially expressed microRNAs were identified using the R-package DEseq2 and the endpoints were time to progression (TTP) and cancer specific survival (CSS). RESULTS Five microRNAs were significantly associated with 5-year progression free survival (PFS): Low expression of two microRNAs was associated with higher PFS, miR-1246 (100% vs. 55.6%, p = 0.008), and miR-135b-5p (92.9% vs. 59.3%, p = 0.041). On the other hand, high expressions of three microRNAs were associated with higher PFS, miR-148a-3p (93.3% vs. 53.6%, p = 0.025), miR-99a-5p (92.9% vs. 57.1%, p = 0.016), and let-7c-3p (92.9% vs. 57.1%, p = 0.016). Corresponding findings were documented for CSS. INTERPRETATION Our study identified five microRNAs as prognostic markers in anal cancer. MiR-1246 and microRNA-135b-5p were oncoMiRs (miRs with oncogene effects), while miR-148a-3p, miR- 99a-5p, and let-7c-3p acted as tumour suppressors in anal cancer patients.
Collapse
Affiliation(s)
- Olav Dahl
- Department of Oncology, Haukeland University Hospital, Bergen, Norway; University of Bergen, Bergen Norway.
| | | |
Collapse
|
2
|
Thavanesan N, Vigneswaran G, Bodala I, Underwood TJ. The Oesophageal Cancer Multidisciplinary Team: Can Machine Learning Assist Decision-Making? J Gastrointest Surg 2023; 27:807-822. [PMID: 36689150 PMCID: PMC10073064 DOI: 10.1007/s11605-022-05575-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/10/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND The complexity of the upper gastrointestinal (UGI) multidisciplinary team (MDT) is continually growing, leading to rising clinician workload, time pressures, and demands. This increases heterogeneity or 'noise' within decision-making for patients with oesophageal cancer (OC) and may lead to inconsistent treatment decisions. In recent decades, the application of artificial intelligence (AI) and more specifically the branch of machine learning (ML) has led to a paradigm shift in the perceived utility of statistical modelling within healthcare. Within oesophageal cancer (OC) care, ML techniques have already been applied with early success to the analyses of histological samples and radiology imaging; however, it has not yet been applied to the MDT itself where such models are likely to benefit from incorporating information-rich, diverse datasets to increase predictive model accuracy. METHODS This review discusses the current role the MDT plays in modern UGI cancer care as well as the utilisation of ML techniques to date using histological and radiological data to predict treatment response, prognostication, nodal disease evaluation, and even resectability within OC. RESULTS The review finds that an emerging body of evidence is growing in support of ML tools within multiple domains relevant to decision-making within OC including automated histological analysis and radiomics. However, to date, no specific application has been directed to the MDT itself which routinely assimilates this information. CONCLUSIONS The authors feel the UGI MDT offers an information-rich, diverse array of data from which ML offers the potential to standardise, automate, and produce more consistent, data-driven MDT decisions.
Collapse
Affiliation(s)
- Navamayooran Thavanesan
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, University Hospitals Southampton, Southampton, UK.
| | - Ganesh Vigneswaran
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, University Hospitals Southampton, Southampton, UK
| | - Indu Bodala
- School of Electronics and Computer Science, University of Southampton, Southampton, UK
| | - Timothy J Underwood
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, University Hospitals Southampton, Southampton, UK
| |
Collapse
|
3
|
Wang W, Ye S, Yang J, Huang Z, Lin L, Zhu Y, Chen D. Effect of microRNA-1246 Derived from Exosomes on Apoptosis of Astroglioma Cells. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Glioma is a common cancer in the central system. Exosomes play a key role in malignancies. This study mainly investigates the effect and mechanism of microRNA-1246 from self-derived exosomes on the apoptotic activities of astroglioma cells. Samples of malignant glioma were collected
to measure microRNA-1246 expression. The glioma cells were cultured and their secreted exosomes were collected. Cells were randomized into NC group, miRNA-1246-mimic group and miRNA-1246-inhibitor group followed by analysis of invasion capability, expression of miR-1246 and CAMD1 gene, and
AMD1 and apoptosis-related proteins expression by Western-blot as well as the relationship between miRNA-1246 and CAMD1. Under electron microscope, exosomes exhibited round shapes with a diameter of 50–290 nm and a positive expression of CD9 and CD63. miRNA-1246 was upregulated in exosomes
from astroglioma patients. miRNA-1246 downregulated CADMI and apoptosis-related protein Bcl-2, but upregulated Caspase-3 and pro-apoptosis proteins in glioma cells. Moreover, miRNA-1246 facilitates astroglioma cells invasion while restraining apoptotic activities. CADM1 was confirmed to be
a target of miRNA-1246. In conclusion, miR-1246 is highly expressed in exosomes that originated from astroglioma cells and suppressed the apoptosis of glioma cells via targeting CAMD1 genes.
Collapse
Affiliation(s)
- Wei Wang
- Department of Emergency, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Sunzhi Ye
- Department of Emergency, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jiajia Yang
- Department of Neurology, The First Hospital Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Zhaofeng Huang
- Department of Emergency, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Luyang Lin
- Department of Emergency, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yingying Zhu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Daqing Chen
- Department of Emergency, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
4
|
Kline A, Wang H, Li Y, Dennis S, Hutch M, Xu Z, Wang F, Cheng F, Luo Y. Multimodal machine learning in precision health: A scoping review. NPJ Digit Med 2022; 5:171. [PMID: 36344814 PMCID: PMC9640667 DOI: 10.1038/s41746-022-00712-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
Machine learning is frequently being leveraged to tackle problems in the health sector including utilization for clinical decision-support. Its use has historically been focused on single modal data. Attempts to improve prediction and mimic the multimodal nature of clinical expert decision-making has been met in the biomedical field of machine learning by fusing disparate data. This review was conducted to summarize the current studies in this field and identify topics ripe for future research. We conducted this review in accordance with the PRISMA extension for Scoping Reviews to characterize multi-modal data fusion in health. Search strings were established and used in databases: PubMed, Google Scholar, and IEEEXplore from 2011 to 2021. A final set of 128 articles were included in the analysis. The most common health areas utilizing multi-modal methods were neurology and oncology. Early fusion was the most common data merging strategy. Notably, there was an improvement in predictive performance when using data fusion. Lacking from the papers were clear clinical deployment strategies, FDA-approval, and analysis of how using multimodal approaches from diverse sub-populations may improve biases and healthcare disparities. These findings provide a summary on multimodal data fusion as applied to health diagnosis/prognosis problems. Few papers compared the outputs of a multimodal approach with a unimodal prediction. However, those that did achieved an average increase of 6.4% in predictive accuracy. Multi-modal machine learning, while more robust in its estimations over unimodal methods, has drawbacks in its scalability and the time-consuming nature of information concatenation.
Collapse
Affiliation(s)
- Adrienne Kline
- Department of Preventive Medicine, Northwestern University, Chicago, 60201, IL, USA
| | - Hanyin Wang
- Department of Preventive Medicine, Northwestern University, Chicago, 60201, IL, USA
| | - Yikuan Li
- Department of Preventive Medicine, Northwestern University, Chicago, 60201, IL, USA
| | - Saya Dennis
- Department of Preventive Medicine, Northwestern University, Chicago, 60201, IL, USA
| | - Meghan Hutch
- Department of Preventive Medicine, Northwestern University, Chicago, 60201, IL, USA
| | - Zhenxing Xu
- Department of Population Health Sciences, Cornell University, New York, 10065, NY, USA
| | - Fei Wang
- Department of Population Health Sciences, Cornell University, New York, 10065, NY, USA
| | - Feixiong Cheng
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, 44195, OH, USA
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University, Chicago, 60201, IL, USA.
| |
Collapse
|
5
|
Ghafouri-Fard S, Khoshbakht T, Hussen BM, Taheri M, Samadian M. A Review on the Role of miR-1246 in the Pathoetiology of Different Cancers. Front Mol Biosci 2022; 8:771835. [PMID: 35047553 PMCID: PMC8762223 DOI: 10.3389/fmolb.2021.771835] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/22/2021] [Indexed: 01/22/2023] Open
Abstract
miR-1246 is a microRNA firstly recognized through application of a high throughput sequencing technique in human embryonic stem cells. Subsequent studies have shown the role of this microRNA in the carcinogenesis. miR-1246 has been found to exert oncogenic roles in colorectal, breast, renal, oral, laryngeal, pancreatic and ovarian cancers as well as melanoma and glioma. In lung, cervical and liver cancers, studies have reported contradictory results regarding the role of miR-1246. miR-1246 has been reported to regulate activity of RAF/MEK/ERK, GSK3β, Wnt/β-catenin, JAK/STAT, PI3K/AKT, THBS2/MMP and NOTCH2 pathways. In addition to affecting cell cycle progression and proliferation, miR-1246 can influence stemness and resistance of cancer cells to therapeutics. In the current review, we describe the summary of in vitro and in vivo studies about the influence of miR-1246 in carcinogenesis in addition to studies that measured expression levels of miR-1246 in clinical samples.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayyebeh Khoshbakht
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Mohammad Samadian
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Aalami AH, Abdeahad H, Shoghi A, Mesgari M, Amirabadi A, Sahebkar A. Brain Tumors and Circulating microRNAs: A Systematic Review and Diagnostic Meta-Analysis. Expert Rev Mol Diagn 2021; 22:201-211. [PMID: 34906021 DOI: 10.1080/14737159.2022.2019016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE Brain tumors (BT) are among the most prevalent cancers in recent years. Various studies have examined the diagnostic role of microRNAs in different diseases; however, their diagnostic role in BT has not been comprehensively investigated. Therefore, this meta-analysis was performed to assess microRNAs in the blood of patients with BTs accurately. METHODS Twenty-six eligible studies were included for analysis. The pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), area under curve (AUC), Q*index, summary receiver-operating characteristic (SROC) were assessed using the Meta-Disc V.1.4 and Comprehensive Meta-Analysis V.3.3 software. The Egger's test was used to evaluate publication bias in this study. RESULTS The diagnostic accuracy of microRNA was high in identifying BT based on the pooled sensitivity 0.82 (95% CI: 0.816 - 0.84), specificity 0.82 (95% CI: 0.817 - 0.84), PLR 5.101 (95% CI: 3.99 - 6.51), NLR 0.187 (95% CI: 0.149 - 0.236), DOR 34.07 (95% CI: 22.56 - 51.43) as well as AUC (0.92), and Q*-index (0.86). Subgroup analyses was also performed for sample types (serum/plasma), reference genes (RNU6, miR-39, and miR-24), and region to determine the diagnostic power of microRNAs in the diagnosis of BT using pooled sensitivity, specificity, PLR, NLR, AUC, and DOR. CONCLUSION This meta-analysis proved that circulating microRNAs were the potential markers for BT and could potentially be used as non-invasive early detection biomarkers.
Collapse
Affiliation(s)
- Amir Hossein Aalami
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Hossein Abdeahad
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Ali Shoghi
- Neurosurgery Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Mesgari
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Amir Amirabadi
- Department of Internal Medicine, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran.,Solid Tumors Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,School of Pharmacy, University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Hoshino I, Yokota H, Iwatate Y, Mori Y, Kuwayama N, Ishige F, Itami M, Uno T, Nakamura Y, Tatsumi Y, Shimozato O, Nagase H. Prediction of the differences in tumor mutation burden between primary and metastatic lesions by radiogenomics. Cancer Sci 2021; 113:229-239. [PMID: 34689378 PMCID: PMC8748253 DOI: 10.1111/cas.15173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor mutational burden (TMB) is gaining attention as a biomarker for responses to immune checkpoint inhibitors in cancer patients. In this study, we evaluated the status of TMB in primary and liver metastatic lesions in patients with colorectal cancer (CRC). In addition, the status of TMB in primary and liver metastatic lesions was inferred by radiogenomics on the basis of computed tomography (CT) images. The study population included 24 CRC patients with liver metastases. DNA was extracted from primary and liver metastatic lesions obtained from the patients and TMB values were evaluated by next‐generation sequencing. The TMB value was considered high when it equaled to or exceeded 10/100 Mb. Radiogenomic analysis of TMB was performed by machine learning using CT images and the construction of prediction models. In 7 out of 24 patients (29.2%), the TMB status differed between the primary and liver metastatic lesions. Radiogenomic analysis was performed to predict whether TMB status was high or low. The maximum values for the area under the receiver operating characteristic curve were 0.732 and 0.812 for primary CRC and CRC with liver metastasis, respectively. The sensitivity, specificity, and accuracy of the constructed models for TMB status discordance were 0.857, 0.600, and 0.682, respectively. Our results suggested that accurate inference of the TMB status is possible using radiogenomics. Therefore, radiogenomics could facilitate the diagnosis, treatment, and prognosis of patients with CRC in the clinical setting.
Collapse
Affiliation(s)
- Isamu Hoshino
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Hajime Yokota
- Department of Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yosuke Iwatate
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, Chiba, Japan
| | - Yasukuni Mori
- Faculty of Engineering, Graduate School of Engineering, Chiba University, Chiba, Japan
| | - Naoki Kuwayama
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Fumitaka Ishige
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, Chiba, Japan
| | - Makiko Itami
- Division of Clinical Pathology, Chiba Cancer Center, Chiba, Japan
| | - Takashi Uno
- Department of Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yuki Nakamura
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba Cancer Center, Chiba, Japan
| | - Yasutoshi Tatsumi
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba Cancer Center, Chiba, Japan
| | - Osamu Shimozato
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba Cancer Center, Chiba, Japan
| | - Hiroki Nagase
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba Cancer Center, Chiba, Japan
| |
Collapse
|
8
|
Brancato V, Garbino N, Mannelli L, Aiello M, Salvatore M, Franzese M, Cavaliere C. Impact of radiogenomics in esophageal cancer on clinical outcomes: A pilot study. World J Gastroenterol 2021; 27:6110-6127. [PMID: 34629823 PMCID: PMC8476334 DOI: 10.3748/wjg.v27.i36.6110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/16/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Esophageal cancer (ESCA) is the sixth most common malignancy in the world, and its incidence is rapidly increasing. Recently, several microRNAs (miRNAs) and messenger RNA (mRNA) targets were evaluated as potential biomarkers and regulators of epigenetic mechanisms involved in early diagnosis. In addition, computed tomography (CT) radiomic studies on ESCA improved the early stage identification and the prediction of response to treatment. Radiogenomics provides clinically useful prognostic predictions by linking molecular characteristics such as gene mutations and gene expression patterns of malignant tumors with medical images and could provide more opportunities in the management of patients with ESCA. AIM To explore the combination of CT radiomic features and molecular targets associated with clinical outcomes for characterization of ESCA patients. METHODS Of 15 patients with diagnosed ESCA were included in this study and their CT imaging and transcriptomic data were extracted from The Cancer Imaging Archive and gene expression data from The Cancer Genome Atlas, respectively. Cancer stage, history of significant alcohol consumption and body mass index (BMI) were considered as clinical outcomes. Radiomic analysis was performed on CT images acquired after injection of contrast medium. In total, 1302 radiomics features were extracted from three-dimensional regions of interest by using PyRadiomics. Feature selection was performed using a correlation filter based on Spearman's correlation (ρ) and Wilcoxon-rank sum test respect to clinical outcomes. Radiogenomic analysis involved ρ analysis between radiomic features associated with clinical outcomes and transcriptomic signatures consisting of eight N6-methyladenosine RNA methylation regulators and five up-regulated miRNA. The significance level was set at P < 0.05. RESULTS Of 25, five and 29 radiomic features survived after feature selection, considering stage, alcohol history and BMI as clinical outcomes, respectively. Radiogenomic analysis with stage as clinical outcome revealed that six of the eight mRNA regulators and two of the five up-regulated miRNA were significantly correlated with ten and three of the 25 selected radiomic features, respectively (-0.61 < ρ < -0.60 and 0.53 < ρ < 0.69, P < 0.05). Assuming alcohol history as clinical outcome, no correlation was found between the five selected radiomic features and mRNA regulators, while a significant correlation was found between one radiomic feature and three up-regulated miRNAs (ρ = -0.56, ρ = -0.64 and ρ = 0.61, P < 0.05). Radiogenomic analysis with BMI as clinical outcome revealed that four mRNA regulators and one up-regulated miRNA were significantly correlated with 10 and two radiomic features, respectively (-0.67 < ρ < -0.54 and 0.53 < ρ < 0.71, P < 0.05). CONCLUSION Our study revealed interesting relationships between the expression of eight N6-methyladenosine RNA regulators, as well as five up-regulated miRNAs, and CT radiomic features associated with clinical outcomes of ESCA patients.
Collapse
|
9
|
Ouyang ML, Wang YR, Deng QS, Zhu YF, Zhao ZH, Wang L, Wang LX, Tang K. Development and Validation of a 18F-FDG PET-Based Radiomic Model for Evaluating Hypermetabolic Mediastinal-Hilar Lymph Nodes in Non-Small-Cell Lung Cancer. Front Oncol 2021; 11:710909. [PMID: 34568038 PMCID: PMC8457532 DOI: 10.3389/fonc.2021.710909] [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: 05/17/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022] Open
Abstract
Background Accurate evaluation of lymph node (LN) status is critical for determining the treatment options in patients with non-small cell lung cancer (NSCLC). This study aimed to develop and validate a 18F-FDG PET-based radiomic model for the identification of metastatic LNs from the hypermetabolic mediastinal–hilar LNs in NSCLC. Methods We retrospectively reviewed 259 patients with hypermetabolic LNs who underwent pretreatment 18F-FDG PET/CT and were pathologically confirmed as NSCLC from two centers. Two hundred twenty-eight LNs were allocated to a training cohort (LN = 159) and an internal validation cohort (LN = 69) from one center (7:3 ratio), and 60 LNs were enrolled to an external validation cohort from the other. Radiomic features were extracted from LNs of PET images. A PET radiomics signature was constructed by multivariable logistic regression after using the least absolute shrinkage and selection operator (LASSO) method with 10-fold cross-validation. The PET radiomics signature (model 1) and independent predictors from CT image features and clinical data (model 2) were incorporated into a combined model (model 3). A nomogram was plotted for the complex model, and the performance of the nomogram was assessed by its discrimination, calibration, and clinical usefulness. Results The area under the curve (AUC) values of model 1 were 0.820, 0.785, and 0.808 in the training, internal, and external validation cohorts, respectively, showing good diagnostic efficacy for lymph node metastasis (LNM). Furthermore, model 2 was able to discriminate metastatic LNs in the training (AUC 0.780), internal (AUC 0.794), and external validation cohorts (AUC 0.802), respectively. Model 3 showed optimal diagnostic performance among the three cohorts, with an AUC of 0.874, 0.845, and 0.841, respectively. The nomogram based on the model 3 showed good discrimination and calibration. Conclusions Our study revealed that PET radiomics signature, especially when integrated with CT imaging features, showed the ability to identify true and false positives of mediastinal–hilar LNM detected by PET/CT in patients with NSCLC, which would help clinicians to make individual treatment decisions.
Collapse
Affiliation(s)
- Ming-Li Ouyang
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi-Ran Wang
- Department of Medical Engineering, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing-Shan Deng
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ye-Fei Zhu
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Zhen-Hua Zhao
- Department of Radiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, China
| | - Ling Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liang-Xing Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kun Tang
- Department of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Wang Z, Gao D, Wang S, Lin H, Wang Y, Xu W. Exosomal microRNA-1246 from human umbilical cord mesenchymal stem cells potentiates myocardial angiogenesis in chronic heart failure. Cell Biol Int 2021; 45:2211-2225. [PMID: 34270841 DOI: 10.1002/cbin.11664] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/08/2021] [Accepted: 07/03/2021] [Indexed: 12/19/2022]
Abstract
microRNAs (miRNAs) are of importance to chronic heart failure (CHF). However, the relevance of the exosomal miRNAs produced during CHF remains unknown. Our purpose here was to examine the relevance of exosomal microRNA-1246 (miR-1246) released from human umbilical cord mesenchymal stem cell (hucMSC) during CHF and the mechanism of action. Cardiac function, myocardial infarction area, apoptosis, and angiogenesis were all evaluated in a CHF rat model following treatment with hucMSC-derived exosomes (hucMSC-Exos). H9C2 and human umbilical vascular endothelial cells (HUVECs) were subjected to oxygen and glucose deprivation and exosome treatment to quantify the cell proliferation and apoptosis in H9C2 cells and the tube formation capacity of the HUVECs. A dual-luciferase activity reporter assay was conducted to validate the interaction between miR-1246 and serine protease 23 (PRSS23). HucMSCs treatment led to a reduction in H9C2 apoptosis and an increase in HUVEC angiogenesis, which were mitigated when hucMSCs were treated with a miR-1246 inhibitor. We also confirmed that PRSS23 is a putative target of miR-1246 and that miR-1246 attenuated hypoxia-induced myocardial tissue damage by targeting PRSS23 and inhibiting the activation of the Snail/alpha-smooth muscle actin signaling. Our findings suggest that exosomal miR-1246 from hucMSCs protects the heart from failure by targeting PRSS23.
Collapse
Affiliation(s)
- Zicheng Wang
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Da Gao
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Shengjie Wang
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Haiyan Lin
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Yanwei Wang
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Weifeng Xu
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
11
|
Hoshino I, Yokota H. Radiogenomics of gastroenterological cancer: The dawn of personalized medicine with artificial intelligence-based image analysis. Ann Gastroenterol Surg 2021; 5:427-435. [PMID: 34337291 PMCID: PMC8316732 DOI: 10.1002/ags3.12437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/29/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Radiogenomics is a new field of medical science that integrates two omics, radiomics and genomics, and may bring a major paradigm shift in traditional personalized medicine strategies that require tumor tissue samples. In addition, the acquisition of the data does not require special imaging equipment or special imaging conditions, and it is possible to use image information from computed tomography, magnetic resonance imaging, positron emission tomography-computed tomography in clinical practice, so the versatility and cost-effectiveness of radiogenomics are expected. So far, the field of radiogenomics has developed, especially in the fields of brain tumors and breast cancer, but recently, reports of radiogenomic research on gastroenterological cancer are increasing. This review provides an overview of radiogenomic research methods and summarizes the current radiogenomic research in gastroenterological cancer. In addition, the application of artificial intelligence is considered to be indispensable for the integrated analysis of enormous omics information in the future, and the future direction of this research, including the latest technologies, will be discussed.
Collapse
Affiliation(s)
- Isamu Hoshino
- Division of Gastroenterological SurgeryChiba Cancer CenterChibaJapan
| | - Hajime Yokota
- Department of Diagnostic Radiology and Radiation OncologyGraduate School of MedicineChiba UniversityChibaJapan
| |
Collapse
|
12
|
Lu L, Sun SH, Yang H, E L, Guo P, Schwartz LH, Zhao B. Radiomics Prediction of EGFR Status in Lung Cancer-Our Experience in Using Multiple Feature Extractors and The Cancer Imaging Archive Data. ACTA ACUST UNITED AC 2021; 6:223-230. [PMID: 32548300 PMCID: PMC7289249 DOI: 10.18383/j.tom.2020.00017] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We investigated the performance of multiple radiomics feature extractors/software on predicting epidermal growth factor receptor mutation status in 228 patients with non–small cell lung cancer from publicly available data sets in The Cancer Imaging Archive. The imaging and clinical data were split into training (n = 105) and validation cohorts (n = 123). Two of the most cited open-source feature extractors, IBEX (1563 features) and Pyradiomics (1319 features), and our in-house software, Columbia Image Feature Extractor (CIFE) (1160 features), were used to extract radiomics features. Univariate and multivariate analyses were performed sequentially to predict EGFR mutation status using each individual feature extractor. Our univariate analysis integrated an unsupervised clustering method to identify nonredundant and informative candidate features for the creation of prediction models by multivariate analyses. In training, unsupervised clustering-based univariate analysis identified 5, 6, and 4 features from IBEX, Pyradiomics, and CIFE as candidate features, respectively. Multivariate prediction models using these features from IBEX, Pyradiomics, and CIFE yielded similar areas under the receiver operating characteristic curve of 0.68, 0.67, and 0.69. However, in validation, areas under the receiver operating characteristic curve of multivariate prediction models from IBEX, Pyradiomics, and CIFE decreased to 0.54, 0.56 and 0.64, respectively. Different feature extractors select different radiomics features, which leads to prediction models with varying performance. However, correlation between those selected features from different extractors may indicate these features measure similar imaging phenotypes associated with similar biological characteristics. Overall, attention should be paid to the generalizability of individual radiomics features and radiomics prediction models.
Collapse
Affiliation(s)
- Lin Lu
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY; and
| | - Shawn H Sun
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY; and
| | - Hao Yang
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY; and
| | - Linning E
- Department of Radiology, Shanxi DAYI Hospital, Taiyuan, Shanxi, China
| | - Pingzhen Guo
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY; and
| | - Lawrence H Schwartz
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY; and
| | - Binsheng Zhao
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY; and
| |
Collapse
|
13
|
Xie CY, Pang CL, Chan B, Wong EYY, Dou Q, Vardhanabhuti V. Machine Learning and Radiomics Applications in Esophageal Cancers Using Non-Invasive Imaging Methods-A Critical Review of Literature. Cancers (Basel) 2021; 13:2469. [PMID: 34069367 PMCID: PMC8158761 DOI: 10.3390/cancers13102469] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 11/16/2022] Open
Abstract
Esophageal cancer (EC) is of public health significance as one of the leading causes of cancer death worldwide. Accurate staging, treatment planning and prognostication in EC patients are of vital importance. Recent advances in machine learning (ML) techniques demonstrate their potential to provide novel quantitative imaging markers in medical imaging. Radiomics approaches that could quantify medical images into high-dimensional data have been shown to improve the imaging-based classification system in characterizing the heterogeneity of primary tumors and lymph nodes in EC patients. In this review, we aim to provide a comprehensive summary of the evidence of the most recent developments in ML application in imaging pertinent to EC patient care. According to the published results, ML models evaluating treatment response and lymph node metastasis achieve reliable predictions, ranging from acceptable to outstanding in their validation groups. Patients stratified by ML models in different risk groups have a significant or borderline significant difference in survival outcomes. Prospective large multi-center studies are suggested to improve the generalizability of ML techniques with standardized imaging protocols and harmonization between different centers.
Collapse
Affiliation(s)
- Chen-Yi Xie
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Chun-Lap Pang
- Department of Radiology, The Christies’ Hospital, Manchester M20 4BX, UK;
- Division of Dentistry, School of Medical Sciences, University of Manchester, Manchester M15 6FH, UK
| | - Benjamin Chan
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (B.C.); (E.Y.-Y.W.)
| | - Emily Yuen-Yuen Wong
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (B.C.); (E.Y.-Y.W.)
| | - Qi Dou
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China;
| | - Varut Vardhanabhuti
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| |
Collapse
|
14
|
Wei L, Owen D, Rosen B, Guo X, Cuneo K, Lawrence TS, Ten Haken R, El Naqa I. A deep survival interpretable radiomics model of hepatocellular carcinoma patients. Phys Med 2021; 82:295-305. [PMID: 33714190 PMCID: PMC8035300 DOI: 10.1016/j.ejmp.2021.02.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 02/13/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
This work aims to identify a new radiomics signature using imaging phenotypes and clinical variables for risk prediction of overall survival (OS) in hepatocellular carcinoma (HCC) patients treated with stereotactic body radiation therapy (SBRT). 167 patients were retrospectively analyzed with repeated nested cross-validation to mitigate overfitting issues. 56 radiomic features were extracted from pre-treatment contrast-enhanced (CE) CT images. 37 clinical factors were obtained from patients' electronic records. Variational autoencoders (VAE) based survival models were designed for radiomics and clinical features and a convolutional neural network (CNN) survival model was used for the CECT. Finally, radiomics, clinical and raw image deep learning network (DNN) models were combined to predict the risk probability for OS. The final models yielded c-indices of 0.579 (95%CI: 0.544-0.621), 0.629 (95%CI: 0.601-0.643), 0.581 (95%CI: 0.553-0.613) and 0.650 (95%CI: 0.635-0.683) for radiomics, clinical, image input and combined models on nested cross validation scheme, respectively. Integrated gradients method was used to interpret the trained models. Our interpretability analysis of the DNN showed that the top ranked features were clinical liver function and liver exclusive of tumor radiomics features, which suggests a prominent role of side effects and toxicities in liver outside the tumor region in determining the survival rate of these patients. In summary, novel deep radiomic analysis provides improved performance for risk assessment of HCC prognosis compared with Cox survival models and may facilitate stratification of HCC patients and personalization of their treatment strategies. Liver function was found to contribute most to the OS for these HCC patients and radiomics can aid in their management.
Collapse
Affiliation(s)
- Lise Wei
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA.
| | - Dawn Owen
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Benjamin Rosen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Xinzhou Guo
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Kyle Cuneo
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Randall Ten Haken
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Issam El Naqa
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
15
|
Liu Z, Wu K, Wu B, Tang X, Yuan H, Pang H, Huang Y, Zhu X, Luo H, Qi Y. Imaging genomics for accurate diagnosis and treatment of tumors: A cutting edge overview. Biomed Pharmacother 2020; 135:111173. [PMID: 33383370 DOI: 10.1016/j.biopha.2020.111173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Imaging genomics refers to the establishment of the connection between invasive gene expression features and non-invasive imaging features. Tumor imaging genomics can not only understand the macroscopic phenotype of tumor, but also can deeply analyze the cellular and molecular characteristics of tumor tissue. In recent years, tumor imaging genomics has been a key in the field of medicine. The incidence of cancer in China has increased significantly, which is the main reason of disease death of urban residents. With the rapid development of imaging medicine, depending on imaging genomics, many experts have made remarkable achievements in tumor screening and diagnosis, prognosis evaluation, new treatment targets and understanding of tumor biological mechanism. This review analyzes the relationship between tumor radiology and gene expression, which provides a favorable direction for clinical staging, prognosis evaluation and accurate treatment of tumors.
Collapse
Affiliation(s)
- Zhen Liu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Kefeng Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Binhua Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiaoning Tang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Huiqing Yuan
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Hao Pang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Yongmei Huang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiao Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Yi Qi
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| |
Collapse
|
16
|
Abdi E, Latifi-Navid S, Abdi F, Taherian-Esfahani Z. Emerging circulating MiRNAs and LncRNAs in upper gastrointestinal cancers. Expert Rev Mol Diagn 2020; 20:1121-1138. [DOI: 10.1080/14737159.2020.1842199] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Esmat Abdi
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Saeid Latifi-Navid
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Fatemeh Abdi
- Department of Engineering Sciences, Faculty of Advanced Technologies, University of Mohaghegh Ardabili, Namin, Iran
| | - Zahra Taherian-Esfahani
- Medical Genetics Laboratory, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Huang S, Wei YK, Kaliamurthi S, Cao Y, Nangraj AS, Sui X, Chu D, Wang H, Wei DQ, Peslherbe GH, Selvaraj G, Shi J. Circulating miR-1246 Targeting UBE2C, TNNI3, TRAIP, UCHL1 Genes and Key Pathways as a Potential Biomarker for Lung Adenocarcinoma: Integrated Biological Network Analysis. J Pers Med 2020; 10:162. [PMID: 33050659 PMCID: PMC7712139 DOI: 10.3390/jpm10040162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
Analysis of circulating miRNAs (cmiRNAs) before surgical operation (BSO) and after the surgical operation (ASO) has been informative for lung adenocarcinoma (LUAD) diagnosis, progression, and outcomes of treatment. Thus, we performed a biological network analysis to identify the potential target genes (PTGs) of the overexpressed cmiRNA signatures from LUAD samples that had undergone surgical therapy. Differential expression (DE) analysis of microarray datasets, including cmiRNAs (GSE137140) and cmRNAs (GSE69732), was conducted using the Limma package. cmiR-1246 was predicted as a significantly upregulated cmiRNA of LUAD samples BSO and ASO. Then, 9802 miR-1246 target genes (TGs) were predicted using 12 TG prediction platforms (MiRWalk, miRDB, and TargetScan). Briefly, 425 highly expressed overlapping miRNA-1246 TGs were observed between the prediction platform and the cmiRNA dataset. ClueGO predicted cell projection morphogenesis, chemosensory behavior, and glycosaminoglycan binding, and the PI3K-Akt signaling pathways were enriched metabolic interactions regulating miRNA-1245 overlapping TGs in LUAD. Using 425 overlapping miR-1246 TGs, a protein-protein interaction network was constructed. Then, 12 PTGs of three different Walktrap modules were identified; among them, ubiquitin-conjugating enzyme E2C (UBE2C), troponin T1(TNNT1), T-cell receptor alpha locus interacting protein (TRAIP), and ubiquitin c-terminal hydrolase L1(UCHL1) were positively correlated with miR-1246, and the high expression of these genes was associated with better overall survival of LUAD. We conclude that PTGs of cmiRNA-1246 and key pathways, namely, ubiquitin-mediated proteolysis, glycosaminoglycan binding, the DNA metabolic process, and the PI3K-Akt-mTOR signaling pathway, the neurotrophin and cardiomyopathy signaling pathway, and the MAPK signaling pathway provide new insights on a noninvasive prognostic biomarker for LUAD.
Collapse
Affiliation(s)
- Siyuan Huang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou 450052, China; (S.H.); (X.S.)
| | - Yong-Kai Wei
- College of Science, Henan University of Technology, Zhengzhou 450001, China;
| | - Satyavani Kaliamurthi
- Centre for Research in Molecular Modeling and Department of Chemistry and Biochemistry, Concordia University, 7141 Sherbrooke Street West, Montréal, QC H4B 1R6, Canada; (S.K.); (D.-Q.W.); (G.H.P.); (G.S.)
- Center of Interdisciplinary Science-Computational Life Sciences, College of Biological Engineering, Henan University of Technology, No.100, Lianhua Street, Hi-Tech Development Zone, Zhengzhou 450001, China
| | - Yanghui Cao
- Department of General Surgery, Henan Tumor Hospital, No.127 Dongming Road, Zhengzhou 450008, China;
| | - Asma Sindhoo Nangraj
- The State Key Laboratory of Microbial Metabolism, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Xin Sui
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou 450052, China; (S.H.); (X.S.)
| | - Dan Chu
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou 450052, China; (D.C.); (H.W.)
| | - Huan Wang
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou 450052, China; (D.C.); (H.W.)
| | - Dong-Qing Wei
- Centre for Research in Molecular Modeling and Department of Chemistry and Biochemistry, Concordia University, 7141 Sherbrooke Street West, Montréal, QC H4B 1R6, Canada; (S.K.); (D.-Q.W.); (G.H.P.); (G.S.)
- Center of Interdisciplinary Science-Computational Life Sciences, College of Biological Engineering, Henan University of Technology, No.100, Lianhua Street, Hi-Tech Development Zone, Zhengzhou 450001, China
- The State Key Laboratory of Microbial Metabolism, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Gilles H. Peslherbe
- Centre for Research in Molecular Modeling and Department of Chemistry and Biochemistry, Concordia University, 7141 Sherbrooke Street West, Montréal, QC H4B 1R6, Canada; (S.K.); (D.-Q.W.); (G.H.P.); (G.S.)
| | - Gurudeeban Selvaraj
- Centre for Research in Molecular Modeling and Department of Chemistry and Biochemistry, Concordia University, 7141 Sherbrooke Street West, Montréal, QC H4B 1R6, Canada; (S.K.); (D.-Q.W.); (G.H.P.); (G.S.)
- Center of Interdisciplinary Science-Computational Life Sciences, College of Biological Engineering, Henan University of Technology, No.100, Lianhua Street, Hi-Tech Development Zone, Zhengzhou 450001, China
| | - Jiang Shi
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou 450052, China; (D.C.); (H.W.)
| |
Collapse
|
18
|
Iwatate Y, Hoshino I, Yokota H, Ishige F, Itami M, Mori Y, Chiba S, Arimitsu H, Yanagibashi H, Nagase H, Takayama W. Radiogenomics for predicting p53 status, PD-L1 expression, and prognosis with machine learning in pancreatic cancer. Br J Cancer 2020; 123:1253-1261. [PMID: 32690867 PMCID: PMC7555500 DOI: 10.1038/s41416-020-0997-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 06/21/2020] [Accepted: 06/29/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Radiogenomics is an emerging field that integrates "Radiomics" and "Genomics". In the current study, we aimed to predict the genetic information of pancreatic tumours in a simple, inexpensive, and non-invasive manner, using cancer imaging analysis and radiogenomics. We focused on p53 mutations, which are highly implicated in pancreatic ductal adenocarcinoma (PDAC), and PD-L1, a biomarker for immune checkpoint inhibitor-based therapies. METHODS Overall, 107 patients diagnosed with PDAC were retrospectively examined. The relationship between p53 mutations as well as PD-L1 abnormal expression and clinicopathological factors was investigated using immunohistochemistry. Imaging features (IFs) were extracted from CT scans and were used to create prediction models of p53 and PD-L1 status. RESULTS We found that p53 and PD-L1 are significant independent prognostic factors (P = 0.008, 0.013, respectively). The area under the curve for p53 and PD-L1 predictive models was 0.795 and 0.683, respectively. Radiogenomics-predicted p53 mutations were significantly associated with poor prognosis (P = 0.015), whereas the predicted abnormal expression of PD-L1 was not significant (P = 0.096). CONCLUSIONS Radiogenomics could predict p53 mutations and in turn the prognosis of PDAC patients. Hence, prediction of genetic information using radiogenomic analysis may aid in the development of precision medicine.
Collapse
Affiliation(s)
- Yosuke Iwatate
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| | - Isamu Hoshino
- Division of Gastroenterological Surgery, Chiba Cancer Center, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan.
| | - Hajime Yokota
- Department of Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba, 260-8670, Japan
| | - Fumitaka Ishige
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| | - Makiko Itami
- Division of Clinical Pathology, Chiba Cancer Center, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| | - Yasukuni Mori
- Graduate School of Engineering, Faculty of Engineering, Chiba University, Yayoi-cho 1-33, Inage-ku, Chiba, 263-8522, Japan
| | - Satoshi Chiba
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| | - Hidehito Arimitsu
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| | - Hiroo Yanagibashi
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| | - Hiroki Nagase
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba Cancer Center, 666-2 Nitonacho, Chuo-ku, Chiba, 260-8717, Japan
| | - Wataru Takayama
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, 666-2 Nitona-cho, Chuo-ku, Chiba, 260-8717, Japan
| |
Collapse
|
19
|
Wang X, Chai Z, Pan G, Hao Y, Li B, Ye T, Li Y, Long F, Xia L, Liu M. ExoBCD: a comprehensive database for exosomal biomarker discovery in breast cancer. Brief Bioinform 2020; 22:5860692. [PMID: 32591816 DOI: 10.1093/bib/bbaa088] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/08/2020] [Accepted: 04/26/2020] [Indexed: 12/24/2022] Open
Abstract
Effective and safe implementation of precision oncology for breast cancer is a vital strategy to improve patient outcomes, which relies on the application of reliable biomarkers. As 'liquid biopsy' and novel resource for biomarkers, exosomes provide a promising avenue for the diagnosis and treatment of breast cancer. Although several exosome-related databases have been developed, there is still lacking of an integrated database for exosome-based biomarker discovery. To this end, a comprehensive database ExoBCD (https://exobcd.liumwei.org) was constructed with the combination of robust analysis of four high-throughput datasets, transcriptome validation of 1191 TCGA cases and manual mining of 950 studies. In ExoBCD, approximately 20 900 annotation entries were integrated from 25 external sources and 306 exosomal molecules (49 potential biomarkers and 257 biologically interesting molecules). The latter could be divided into 3 molecule types, including 121 mRNAs, 172 miRNAs and 13 lncRNAs. Thus, the well-linked information about molecular characters, experimental biology, gene expression patterns, overall survival, functional evidence, tumour stage and clinical use were fully integrated. As a data-driven and literature-based paradigm proposed of biomarker discovery, this study also demonstrated the corroborative analysis and identified 36 promising molecules, as well as the most promising prognostic biomarkers, IGF1R and FRS2. Taken together, ExoBCD is the first well-corroborated knowledge base for exosomal studies of breast cancer. It not only lays a foundation for subsequent studies but also strengthens the studies of probing molecular mechanisms, discovering biomarkers and developing meaningful clinical use.
Collapse
Affiliation(s)
- Xuanyi Wang
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Zixuan Chai
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Guizhi Pan
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Youjin Hao
- College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Ting Ye
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yinghong Li
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Fei Long
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Lixin Xia
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Mingwei Liu
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|