1
|
Sanchez I, Rahman R. Radiogenomics as an Integrated Approach to Glioblastoma Precision Medicine. Curr Oncol Rep 2024; 26:1213-1222. [PMID: 39009914 PMCID: PMC11480134 DOI: 10.1007/s11912-024-01580-z] [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] [Accepted: 07/05/2024] [Indexed: 07/17/2024]
Abstract
PURPOSE OF REVIEW Isocitrate dehydrogenase wild-type glioblastoma is the most aggressive primary brain tumour in adults. Its infiltrative nature and heterogeneity confer a dismal prognosis, despite multimodal treatment. Precision medicine is increasingly advocated to improve survival rates in glioblastoma management; however, conventional neuroimaging techniques are insufficient in providing the detail required for accurate diagnosis of this complex condition. RECENT FINDINGS Advanced magnetic resonance imaging allows more comprehensive understanding of the tumour microenvironment. Combining diffusion and perfusion magnetic resonance imaging to create a multiparametric scan enhances diagnostic power and can overcome the unreliability of tumour characterisation by standard imaging. Recent progress in deep learning algorithms establishes their remarkable ability in image-recognition tasks. Integrating these with multiparametric scans could transform the diagnosis and monitoring of patients by ensuring that the entire tumour is captured. As a corollary, radiomics has emerged as a powerful approach to offer insights into diagnosis, prognosis, treatment, and tumour response through extraction of information from radiological scans, and transformation of these tumour characteristics into quantitative data. Radiogenomics, which links imaging features with genomic profiles, has exhibited its ability in characterising glioblastoma, and determining therapeutic response, with the potential to revolutionise management of glioblastoma. The integration of deep learning algorithms into radiogenomic models has established an automated, highly reproducible means to predict glioblastoma molecular signatures, further aiding prognosis and targeted therapy. However, challenges including lack of large cohorts, absence of standardised guidelines and the 'black-box' nature of deep learning algorithms, must first be overcome before this workflow can be applied in clinical practice.
Collapse
Affiliation(s)
- Isabella Sanchez
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Ruman Rahman
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
2
|
Meng H, Wang TD, Zhuo LY, Hao JW, Sui LY, Yang W, Zang LL, Cui JJ, Wang JN, Yin XP. Quantitative radiomics analysis of imaging features in adults and children Mycoplasma pneumonia. Front Med (Lausanne) 2024; 11:1409477. [PMID: 38831994 PMCID: PMC11146305 DOI: 10.3389/fmed.2024.1409477] [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: 03/30/2024] [Accepted: 04/30/2024] [Indexed: 06/05/2024] Open
Abstract
Purpose This study aims to explore the value of clinical features, CT imaging signs, and radiomics features in differentiating between adults and children with Mycoplasma pneumonia and seeking quantitative radiomic representations of CT imaging signs. Materials and methods In a retrospective analysis of 981 cases of mycoplasmal pneumonia patients from November 2021 to December 2023, 590 internal data (adults:450, children: 140) randomly divided into a training set and a validation set with an 8:2 ratio and 391 external test data (adults:121; children:270) were included. Using univariate analysis, CT imaging signs and clinical features with significant differences (p < 0.05) were selected. After segmenting the lesion area on the CT image as the region of interest, 1,904 radiomic features were extracted. Then, Pearson correlation analysis (PCC) and the least absolute shrinkage and selection operator (LASSO) were used to select the radiomic features. Based on the selected features, multivariable logistic regression analysis was used to establish the clinical model, CT image model, radiomic model, and combined model. The predictive performance of each model was evaluated using ROC curves, AUC, sensitivity, specificity, accuracy, and precision. The AUC between each model was compared using the Delong test. Importantly, the radiomics features and quantitative and qualitative CT image features were analyzed using Pearson correlation analysis and analysis of variance, respectively. Results For the individual model, the radiomics model, which was built using 45 selected features, achieved the highest AUCs in the training set, validation set, and external test set, which were 0.995 (0.992, 0.998), 0.952 (0.921, 0.978), and 0.969 (0.953, 0.982), respectively. In all models, the combined model achieved the highest AUCs, which were 0.996 (0.993, 0.998), 0.972 (0.942, 0.995), and 0.986 (0.976, 0.993) in the training set, validation set, and test set, respectively. In addition, we selected 11 radiomics features and CT image features with a correlation coefficient r greater than 0.35. Conclusion The combined model has good diagnostic performance for differentiating between adults and children with mycoplasmal pneumonia, and different CT imaging signs are quantitatively represented by radiomics.
Collapse
Affiliation(s)
- Huan Meng
- Clinical Medicine School of Hebei University, Baoding, China
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, Baoding, China
| | - Tian-Da Wang
- Clinical Medicine School of Hebei University, Baoding, China
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, Baoding, China
| | - Li-Yong Zhuo
- Clinical Medicine School of Hebei University, Baoding, China
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, Baoding, China
| | - Jia-Wei Hao
- Clinical Medicine School of Hebei University, Baoding, China
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, Baoding, China
| | - Lian-yu Sui
- Clinical Medicine School of Hebei University, Baoding, China
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, Baoding, China
| | - Wei Yang
- Department of Radiology, Baoding First Central Hospital, Baoding, China
| | - Li-Li Zang
- Department of Radiology, Baoding Children's Hospital, Baoding, China
| | - Jing-Jing Cui
- Department of Research and Development, United Imaging Intelligence (Beijing) Co., Beijing, China
| | - Jia-Ning Wang
- Clinical Medicine School of Hebei University, Baoding, China
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, Baoding, China
| | - Xiao-Ping Yin
- Clinical Medicine School of Hebei University, Baoding, China
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Imaging of Inflammation Related Tumors, Baoding, China
| |
Collapse
|
3
|
Lin H, Liu C, Hu A, Zhang D, Yang H, Mao Y. Understanding the immunosuppressive microenvironment of glioma: mechanistic insights and clinical perspectives. J Hematol Oncol 2024; 17:31. [PMID: 38720342 PMCID: PMC11077829 DOI: 10.1186/s13045-024-01544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Glioblastoma (GBM), the predominant and primary malignant intracranial tumor, poses a formidable challenge due to its immunosuppressive microenvironment, thereby confounding conventional therapeutic interventions. Despite the established treatment regimen comprising surgical intervention, radiotherapy, temozolomide administration, and the exploration of emerging modalities such as immunotherapy and integration of medicine and engineering technology therapy, the efficacy of these approaches remains constrained, resulting in suboptimal prognostic outcomes. In recent years, intensive scrutiny of the inhibitory and immunosuppressive milieu within GBM has underscored the significance of cellular constituents of the GBM microenvironment and their interactions with malignant cells and neurons. Novel immune and targeted therapy strategies have emerged, offering promising avenues for advancing GBM treatment. One pivotal mechanism orchestrating immunosuppression in GBM involves the aggregation of myeloid-derived suppressor cells (MDSCs), glioma-associated macrophage/microglia (GAM), and regulatory T cells (Tregs). Among these, MDSCs, though constituting a minority (4-8%) of CD45+ cells in GBM, play a central component in fostering immune evasion and propelling tumor progression, angiogenesis, invasion, and metastasis. MDSCs deploy intricate immunosuppressive mechanisms that adapt to the dynamic tumor microenvironment (TME). Understanding the interplay between GBM and MDSCs provides a compelling basis for therapeutic interventions. This review seeks to elucidate the immune regulatory mechanisms inherent in the GBM microenvironment, explore existing therapeutic targets, and consolidate recent insights into MDSC induction and their contribution to GBM immunosuppression. Additionally, the review comprehensively surveys ongoing clinical trials and potential treatment strategies, envisioning a future where targeting MDSCs could reshape the immune landscape of GBM. Through the synergistic integration of immunotherapy with other therapeutic modalities, this approach can establish a multidisciplinary, multi-target paradigm, ultimately improving the prognosis and quality of life in patients with GBM.
Collapse
Affiliation(s)
- Hao Lin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Chaxian Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ankang Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Duanwu Zhang
- Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
4
|
Tong L, Shi W, Isgut M, Zhong Y, Lais P, Gloster L, Sun J, Swain A, Giuste F, Wang MD. Integrating Multi-Omics Data With EHR for Precision Medicine Using Advanced Artificial Intelligence. IEEE Rev Biomed Eng 2024; 17:80-97. [PMID: 37824325 DOI: 10.1109/rbme.2023.3324264] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
With the recent advancement of novel biomedical technologies such as high-throughput sequencing and wearable devices, multi-modal biomedical data ranging from multi-omics molecular data to real-time continuous bio-signals are generated at an unprecedented speed and scale every day. For the first time, these multi-modal biomedical data are able to make precision medicine close to a reality. However, due to data volume and the complexity, making good use of these multi-modal biomedical data requires major effort. Researchers and clinicians are actively developing artificial intelligence (AI) approaches for data-driven knowledge discovery and causal inference using a variety of biomedical data modalities. These AI-based approaches have demonstrated promising results in various biomedical and healthcare applications. In this review paper, we summarize the state-of-the-art AI models for integrating multi-omics data and electronic health records (EHRs) for precision medicine. We discuss the challenges and opportunities in integrating multi-omics data with EHRs and future directions. We hope this review can inspire future research and developing in integrating multi-omics data with EHRs for precision medicine.
Collapse
|
5
|
Ghimire P, Kinnersley B, Karami G, Arumugam P, Houlston R, Ashkan K, Modat M, Booth TC. Radiogenomic biomarkers for immunotherapy in glioblastoma: A systematic review of magnetic resonance imaging studies. Neurooncol Adv 2024; 6:vdae055. [PMID: 38680991 PMCID: PMC11046988 DOI: 10.1093/noajnl/vdae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024] Open
Abstract
Background Immunotherapy is an effective "precision medicine" treatment for several cancers. Imaging signatures of the underlying genome (radiogenomics) in glioblastoma patients may serve as preoperative biomarkers of the tumor-host immune apparatus. Validated biomarkers would have the potential to stratify patients during immunotherapy clinical trials, and if trials are beneficial, facilitate personalized neo-adjuvant treatment. The increased use of whole genome sequencing data, and the advances in bioinformatics and machine learning make such developments plausible. We performed a systematic review to determine the extent of development and validation of immune-related radiogenomic biomarkers for glioblastoma. Methods A systematic review was performed following PRISMA guidelines using the PubMed, Medline, and Embase databases. Qualitative analysis was performed by incorporating the QUADAS 2 tool and CLAIM checklist. PROSPERO registered: CRD42022340968. Extracted data were insufficiently homogenous to perform a meta-analysis. Results Nine studies, all retrospective, were included. Biomarkers extracted from magnetic resonance imaging volumes of interest included apparent diffusion coefficient values, relative cerebral blood volume values, and image-derived features. These biomarkers correlated with genomic markers from tumor cells or immune cells or with patient survival. The majority of studies had a high risk of bias and applicability concerns regarding the index test performed. Conclusions Radiogenomic immune biomarkers have the potential to provide early treatment options to patients with glioblastoma. Targeted immunotherapy, stratified by these biomarkers, has the potential to allow individualized neo-adjuvant precision treatment options in clinical trials. However, there are no prospective studies validating these biomarkers, and interpretation is limited due to study bias with little evidence of generalizability.
Collapse
Affiliation(s)
- Prajwal Ghimire
- Department of Neurosurgery, Kings College Hospital NHS Foundation Trust, London, UK
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Ben Kinnersley
- Department of Oncology, University College London, London, UK
| | | | | | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, UK
| | - Keyoumars Ashkan
- Department of Neurosurgery, Kings College Hospital NHS Foundation Trust, London, UK
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| |
Collapse
|
6
|
Kang W, Qiu X, Luo Y, Luo J, Liu Y, Xi J, Li X, Yang Z. Application of radiomics-based multiomics combinations in the tumor microenvironment and cancer prognosis. J Transl Med 2023; 21:598. [PMID: 37674169 PMCID: PMC10481579 DOI: 10.1186/s12967-023-04437-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/12/2023] [Indexed: 09/08/2023] Open
Abstract
The advent of immunotherapy, a groundbreaking advancement in cancer treatment, has given rise to the prominence of the tumor microenvironment (TME) as a critical area of research. The clinical implications of an improved understanding of the TME are significant and far-reaching. Radiomics has been increasingly utilized in the comprehensive assessment of the TME and cancer prognosis. Similarly, the advancement of pathomics, which is based on pathological images, can offer additional insights into the panoramic view and microscopic information of tumors. The combination of pathomics and radiomics has revolutionized the concept of a "digital biopsy". As genomics and transcriptomics continue to evolve, integrating radiomics with genomic and transcriptomic datasets can offer further insights into tumor and microenvironment heterogeneity and establish correlations with biological significance. Therefore, the synergistic analysis of digital image features (radiomics, pathomics) and genetic phenotypes (genomics) can comprehensively decode and characterize the heterogeneity of the TME as well as predict cancer prognosis. This review presents a comprehensive summary of the research on important radiomics biomarkers for predicting the TME, emphasizing the interplay between radiomics, genomics, transcriptomics, and pathomics, as well as the application of multiomics in decoding the TME and predicting cancer prognosis. Finally, we discuss the challenges and opportunities in multiomics research. In conclusion, this review highlights the crucial role of radiomics and multiomics associations in the assessment of the TME and cancer prognosis. The combined analysis of radiomics, pathomics, genomics, and transcriptomics is a promising research direction with substantial research significance and value for comprehensive TME evaluation and cancer prognosis assessment.
Collapse
Affiliation(s)
- Wendi Kang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiang Qiu
- Obstetrics and Gynecology Hospital of, Fudan University, Shanghai, 200011, China
| | - Yingen Luo
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Jianwei Luo
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Yang Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junqing Xi
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Zhengqiang Yang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
7
|
Khalili N, Kazerooni AF, Familiar A, Haldar D, Kraya A, Foster J, Koptyra M, Storm PB, Resnick AC, Nabavizadeh A. Radiomics for characterization of the glioma immune microenvironment. NPJ Precis Oncol 2023; 7:59. [PMID: 37337080 DOI: 10.1038/s41698-023-00413-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/02/2023] [Indexed: 06/21/2023] Open
Abstract
Increasing evidence suggests that besides mutational and molecular alterations, the immune component of the tumor microenvironment also substantially impacts tumor behavior and complicates treatment response, particularly to immunotherapies. Although the standard method for characterizing tumor immune profile is through performing integrated genomic analysis on tissue biopsies, the dynamic change in the immune composition of the tumor microenvironment makes this approach not feasible, especially for brain tumors. Radiomics is a rapidly growing field that uses advanced imaging techniques and computational algorithms to extract numerous quantitative features from medical images. Recent advances in machine learning methods are facilitating biological validation of radiomic signatures and allowing them to "mine" for a variety of significant correlates, including genetic, immunologic, and histologic data. Radiomics has the potential to be used as a non-invasive approach to predict the presence and density of immune cells within the microenvironment, as well as to assess the expression of immune-related genes and pathways. This information can be essential for patient stratification, informing treatment decisions and predicting patients' response to immunotherapies. This is particularly important for tumors with difficult surgical access such as gliomas. In this review, we provide an overview of the glioma microenvironment, describe novel approaches for clustering patients based on their tumor immune profile, and discuss the latest progress on utilization of radiomics for immune profiling of glioma based on current literature.
Collapse
Affiliation(s)
- Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ariana Familiar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Debanjan Haldar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Kraya
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jessica Foster
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mateusz Koptyra
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Phillip B Storm
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Li S, Zhou B. A review of radiomics and genomics applications in cancers: the way towards precision medicine. Radiat Oncol 2022; 17:217. [PMID: 36585716 PMCID: PMC9801589 DOI: 10.1186/s13014-022-02192-2] [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: 09/15/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
The application of radiogenomics in oncology has great prospects in precision medicine. Radiogenomics combines large volumes of radiomic features from medical digital images, genetic data from high-throughput sequencing, and clinical-epidemiological data into mathematical modelling. The amalgamation of radiomics and genomics provides an approach to better study the molecular mechanism of tumour pathogenesis, as well as new evidence-supporting strategies to identify the characteristics of cancer patients, make clinical decisions by predicting prognosis, and improve the development of individualized treatment guidance. In this review, we summarized recent research on radiogenomics applications in solid cancers and presented the challenges impeding the adoption of radiomics in clinical practice. More standard guidelines are required to normalize radiomics into reproducible and convincible analyses and develop it as a mature field.
Collapse
Affiliation(s)
- Simin Li
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| | - Baosen Zhou
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| |
Collapse
|
9
|
Zhang C, Zhou Y, Gao Y, Zhu Z, Zeng X, Liang W, Sun S, Chen X, Wang H. Radiated glioblastoma cell-derived exosomal circ_0012381 induce M2 polarization of microglia to promote the growth of glioblastoma by CCL2/CCR2 axis. J Transl Med 2022; 20:388. [PMID: 36058942 PMCID: PMC9441045 DOI: 10.1186/s12967-022-03607-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
Background Radiotherapy is the primary therapeutic option for glioblastoma. Some studies proved that radiotherapy increased the release of exosomes from cells. The mechanism by which these exosomes modify the phenotype of microglia in the tumor microenvironment to further determine the fate of irradiated glioblastoma cells remains to be elucidated. Methods We erected the co-culture system of glioblastoma cells and microglia. After radiation, we analyzing the immunophenotype of microglia and the proliferation of radiated glioblastoma cells. By whole transcriptome sequencing, we analyzed of circRNAs in exosomes from glioblastoma cells and microglia. We used some methods, which included RT-PCR, dual-luciferase reporter, et al., to identify how circ_0012381 from radiated glioblastoma cell-derived exosomes regulated the immunophenotype of microglia to further affect the proliferation of radiated glioblastoma cells. Results Radiated glioblastoma cell-derived exosomes markedly induced M2 microglia polarization. These M2-polarized microglia promoted the proliferation of irradiated glioblastoma cells. Circ_0012381 expression was increased in the irradiated glioblastoma cells, and circ_0012381 entered the microglia via exosomes. Circ_0012381 induced M2 microglia polarization by sponging with miR-340-5p to increase ARG1 expression. M2-polarized microglia suppressed phagocytosis and promoted the growth of the irradiated glioblastoma cells by CCL2/CCR2 axis. Compared with the effects of radiotherapy alone, the inhibition of exosomes significantly inhibited the growth of irradiated glioblastoma cells in a zebrafish model. Conclusions Our data suggested that the inhibition of exosome secretion might represent a potential therapeutic strategy to increase the efficacy of radiotherapy in patients with glioblastoma. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03607-0.
Collapse
Affiliation(s)
- Chunzhi Zhang
- Department of Radiation Oncology, Tianjin Hospital, Tianjin, 300211, China.
| | - Yuan Zhou
- Tianjin Medical University, Tianjin, 300070, China
| | - Ya Gao
- Department of Pathogenic Biology, Basic Medical College, Tianjin Medical University, Tianjin, 300070, China
| | - Ze Zhu
- Department of Pathogenic Biology, Basic Medical College, Tianjin Medical University, Tianjin, 300070, China
| | - Xianliang Zeng
- Department of Radiation Oncology, Tianjin Hospital, Tianjin, 300211, China
| | - Weizi Liang
- Department of Radiation Oncology, Tianjin Hospital, Tianjin, 300211, China
| | - Songwei Sun
- Department of Radiation Oncology, Tianjin Hospital, Tianjin, 300211, China
| | - Xiuli Chen
- Department of Radiation Oncology, Tianjin Hospital, Tianjin, 300211, China
| | - Hu Wang
- Department of Neuro-Surgery, Tianjin Huanhu Hospital, Tianjin, 300350, China
| |
Collapse
|
10
|
Radiomic Signatures Associated with CD8+ Tumour-Infiltrating Lymphocytes: A Systematic Review and Quality Assessment Study. Cancers (Basel) 2022; 14:cancers14153656. [PMID: 35954318 PMCID: PMC9367613 DOI: 10.3390/cancers14153656] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023] Open
Abstract
The tumour immune microenvironment influences the efficacy of immune checkpoint inhibitors. Within this microenvironment are CD8-expressing tumour-infiltrating lymphocytes (CD8+ TILs), which are an important mediator and marker of anti-tumour response. In practice, the assessment of CD8+ TILs via tissue sampling involves logistical challenges. Radiomics, the high-throughput extraction of features from medical images, may offer a novel and non-invasive alternative. We performed a systematic review of the available literature reporting radiomic signatures associated with CD8+ TILs. We also aimed to evaluate the methodological quality of the identified studies using the Radiomics Quality Score (RQS) tool, and the risk of bias and applicability with the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool. Articles were searched from inception until 31 December 2021, in three electronic databases, and screened against eligibility criteria. Twenty-seven articles were included. A wide variety of cancers have been studied. The reported radiomic signatures were heterogeneous, with very limited reproducibility between studies of the same cancer group. The overall quality of studies was found to be less than desirable (mean RQS = 33.3%), indicating a need for technical maturation. Some potential avenues for further investigation are also discussed.
Collapse
|
11
|
Zhang Y, Zhang K, Jia H, Xia B, Zang C, Liu Y, Qian L, Dong J. IVIM-DWI and MRI-based radiomics in cervical cancer: Prediction of concurrent chemoradiotherapy sensitivity in combination with clinical prognostic factors. Magn Reson Imaging 2022; 91:37-44. [PMID: 35568271 DOI: 10.1016/j.mri.2022.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE To identify the feasibility and value of intravoxel incoherent motion diffusion weighted imaging (IVIM-DWI) and magnetic resonance imaging (MRI)-based radiomics combined with clinical prognostic factors (CPF) in predicting concurrent chemoradiotherapy (CCRT) sensitivity of locally advanced cervical cancer (LACC). METHODS A retrospective analysis of 163 patients (assigned to training or test groups) who underwent conventional MRI and IVIM-DWI before CCRT were divided into sensitive and resistant groups according to their efficacy at 6 months after CCRT. Per-treatment IVIM-DWI parameters (ADC, D, D⁎ and f value), 3D texture features (from axial T2WI) and CPF were measured, analyzed and screened. The prediction model and its nomogram were developed by combining screened parameters and then validated internally and externally. RESULTS Clinical stage, f value, D value, InverseVariance, SizeZoneNonUniformity, and Minimum were selected to construct prediction model. All parameters except D value showed independent diagnostic value in multivariate Logistic regression analysis and composed prediction model, with AUCs of 0.987 and 0.984 for training and test groups, respectively. The calibration curve (Brier score of 0.042, C-index of 0.987), decision curve and clinical impact curve further demonstrated the reliability and clinical value of prediction model. CONCLUSION IVIM-DWI, MRI-based radiomics and CPF showed high clinical value in predicting CCRT sensitivity for LACC with better predictive performance when combined.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Radiation Oncology, Anhui Provincial Hospital Affiliated to Anhui Medical University, 17 Lujiang Road, Hefei, Anhui 230001, China
| | - Kaiyue Zhang
- Department of Radiation Oncology, Anhui Provincial Hospital Affiliated to Anhui Medical University, 17 Lujiang Road, Hefei, Anhui 230001, China
| | - Haodong Jia
- Department of Radiation Oncology, Anhui Provincial Hospital Affiliated to Anhui Medical University, 17 Lujiang Road, Hefei, Anhui 230001, China; Department of Radiology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China
| | - Bairong Xia
- Department of Radiology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China; Department of Radiation Oncology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China
| | - Chunbao Zang
- Department of Radiation Oncology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China
| | - Yunqin Liu
- Department of Radiation Oncology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China
| | - Liting Qian
- Department of Radiation Oncology, Anhui Provincial Hospital Affiliated to Anhui Medical University, 17 Lujiang Road, Hefei, Anhui 230001, China; Department of Radiation Oncology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China.
| | - Jiangning Dong
- Department of Radiology, West Branch of the First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Cancer Hospital, 107 Huanhu East Road, Hefei, Anhui 230031, China.
| |
Collapse
|
12
|
Corr F, Grimm D, Saß B, Pojskić M, Bartsch JW, Carl B, Nimsky C, Bopp MHA. Radiogenomic Predictors of Recurrence in Glioblastoma—A Systematic Review. J Pers Med 2022; 12:jpm12030402. [PMID: 35330402 PMCID: PMC8952807 DOI: 10.3390/jpm12030402] [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: 02/10/2022] [Revised: 02/23/2022] [Accepted: 03/01/2022] [Indexed: 12/10/2022] Open
Abstract
Glioblastoma, as the most aggressive brain tumor, is associated with a poor prognosis and outcome. To optimize prognosis and clinical therapy decisions, there is an urgent need to stratify patients with increased risk for recurrent tumors and low therapeutic success to optimize individual treatment. Radiogenomics establishes a link between radiological and pathological information. This review provides a state-of-the-art picture illustrating the latest developments in the use of radiogenomic markers regarding prognosis and their potential for monitoring recurrence. Databases PubMed, Google Scholar, and Cochrane Library were searched. Inclusion criteria were defined as diagnosis of glioblastoma with histopathological and radiological follow-up. Out of 321 reviewed articles, 43 articles met these inclusion criteria. Included studies were analyzed for the frequency of radiological and molecular tumor markers whereby radiogenomic associations were analyzed. Six main associations were described: radiogenomic prognosis, MGMT status, IDH, EGFR status, molecular subgroups, and tumor location. Prospective studies analyzing prognostic features of glioblastoma together with radiological features are lacking. By reviewing the progress in the development of radiogenomic markers, we provide insights into the potential efficacy of such an approach for clinical routine use eventually enabling early identification of glioblastoma recurrence and therefore supporting a further personalized monitoring and treatment strategy.
Collapse
Affiliation(s)
- Felix Corr
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- EDU Institute of Higher Education, Villa Bighi, Chaplain’s House, KKR 1320 Kalkara, Malta
- Correspondence:
| | - Dustin Grimm
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- EDU Institute of Higher Education, Villa Bighi, Chaplain’s House, KKR 1320 Kalkara, Malta
| | - Benjamin Saß
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
| | - Mirza Pojskić
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
| | - Jörg W. Bartsch
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| | - Barbara Carl
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Department of Neurosurgery, Helios Dr. Horst Schmidt Kliniken, Ludwig-Erhard-Strasse 100, 65199 Wiesbaden, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| | - Miriam H. A. Bopp
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| |
Collapse
|
13
|
Su D, Lu Q, Pan Y, Yu Y, Wang S, Zuo Y, Yang L. Immune-related Gene-based Prognostic Signature for the Risk Stratification Analysis of Breast Cancer. Curr Bioinform 2022. [DOI: 10.2174/1574893616666211005110732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Breast cancer has plagued women for many years and caused many deaths
around the world.
Method:
In this study, based on the weighted correlation network analysis, univariate Cox regression
analysis, and least absolute shrinkage and selection operator, 12 immune-related genes were selected to
construct the risk score for breast cancer patients. The multivariable Cox regression analysis, gene set
enrichment analysis, and nomogram were also conducted in this study.
Results:
Good results were obtained in the survival analysis, enrichment analysis, multivariable Cox regression
analysis and immune-related feature analysis. When the risk score model was applied in 22
breast cancer cohorts, the univariate Cox regression analysis demonstrated that the risk score model was
significantly associated with overall survival in most of the breast cancer cohorts.
Conclusion:
Based on these results, we could conclude that the proposed risk score model may be a
promising method and may improve the treatment stratification of breast cancer patients in the future
work.
Collapse
Affiliation(s)
- Dongqing Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Qianzi Lu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yi Pan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yao Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shiyuan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongchun Zuo
- The State Key Laboratory
of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University,
Hohhot, China
| | - Lei Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
14
|
Identification of MAD2L1 as a Potential Biomarker in Hepatocellular Carcinoma via Comprehensive Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9868022. [PMID: 35132379 PMCID: PMC8817109 DOI: 10.1155/2022/9868022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/19/2021] [Accepted: 01/15/2022] [Indexed: 11/17/2022]
Abstract
Background Hepatocellular carcinoma (HCC) is widely acknowledged as a malignant tumor with rapid progression, high recurrence rate, and poor prognosis. At present, there is a paucity of reliable biomarkers at the clinical level to guide the management of HCC and improve patient outcomes. Our research is aimed at assessing the prognostic value of MAD2L1 in HCC. Methods Four datasets, GSE121248, GSE101685, GSE85598, and GSE62232, were selected from the GEO database to analyze differentially expressed genes (DEGs) between HCC and normal liver tissues. After functional analysis, we constructed a protein-protein interaction network (PPI) for DEGs and identified core genes in this network with high connectivity with other genes. We assessed the relationship between core genes and the pathogenesis and prognosis of HCC. Finally, we explored the gene regulatory signaling mechanisms involved in HCC pathogenesis. Results 145 DEGs were screened from the intersection of the four GEO datasets. MAD2L1 was associated with most genes according to the PPI network and was selected as a candidate gene for further study. Survival analysis suggested that high MAD2L1 expression in HCC correlated with a worse prognosis. In addition, real-time quantitative PCR (RT-qPCR), western blot (WB), and immunohistochemistry (IHC) findings suggested that the expression of MAD2L1 was abnormally increased in HCC tissues and cells compared to paraneoplastic tissues and normal hepatocytes. Conclusion We found that high MAD2L1 expression in HCC was significantly associated with overall patient survival and clinical features. We also explored the potential biological properties of this gene.
Collapse
|
15
|
Aftab K, Aamir FB, Mallick S, Mubarak F, Pope WB, Mikkelsen T, Rock JP, Enam SA. Radiomics for precision medicine in glioblastoma. J Neurooncol 2022; 156:217-231. [PMID: 35020109 DOI: 10.1007/s11060-021-03933-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/20/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Being the most common primary brain tumor, glioblastoma presents as an extremely challenging malignancy to treat with dismal outcomes despite treatment. Varying molecular epidemiology of glioblastoma between patients and intra-tumoral heterogeneity explains the failure of current one-size-fits-all treatment modalities. Radiomics uses machine learning to identify salient features of the tumor on brain imaging and promises patient-specific management in glioblastoma patients. METHODS We performed a comprehensive review of the available literature on studies investigating the role of radiomics and radiogenomics models for the diagnosis, stratification, prognostication as well as treatment planning and monitoring of glioblastoma. RESULTS Classifiers based on a combination of various MRI sequences, genetic information and clinical data can predict non-invasive tumor diagnosis, overall survival and treatment response with reasonable accuracy. However, the use of radiomics for glioblastoma treatment remains in infancy as larger sample sizes, standardized image acquisition and data extraction techniques are needed to develop machine learning models that can be translated effectively into clinical practice. CONCLUSION Radiomics has the potential to transform the scope of glioblastoma management through personalized medicine.
Collapse
Affiliation(s)
- Kiran Aftab
- Section of Neurosurgery, Department of Surgery, Aga Khan University, Karachi, Pakistan
| | | | - Saad Mallick
- Medical College, Aga Khan University, Karachi, Pakistan
| | - Fatima Mubarak
- Department of Radiology, Aga Khan University, Karachi, Pakistan
| | - Whitney B Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Tom Mikkelsen
- Departments of Neurology and Neurosurgery, Henry Ford Hospital, Detroit, MI, USA
| | - Jack P Rock
- Department of Neurosurgery, Henry Ford Health System, Detroit, MI, USA
| | - Syed Ather Enam
- Section of Neurosurgery, Department of Surgery, Aga Khan University, Karachi, Pakistan.
| |
Collapse
|
16
|
Wang D, Zhao J, Zhang R, Yan Q, Zhou L, Han X, Qi Y, Yu D. The value of CT radiomic in differentiating mycoplasma pneumoniae pneumonia from streptococcus pneumoniae pneumonia with similar consolidation in children under 5 years. Front Pediatr 2022; 10:953399. [PMID: 36245722 PMCID: PMC9554402 DOI: 10.3389/fped.2022.953399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/17/2022] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE To investigate the value of CT radiomics in the differentiation of mycoplasma pneumoniae pneumonia (MPP) from streptococcus pneumoniae pneumonia (SPP) with similar CT manifestations in children under 5 years. METHODS A total of 102 children with MPP (n = 52) or SPP (n = 50) with similar consolidation and surrounding halo on CT images in Qilu Hospital and Qilu Children's Hospital between January 2017 and March 2022 were enrolled in the retrospective study. Radiomic features of the both lesions on plain CT images were extracted including the consolidation part of the pneumonia or both consolidation and surrounding halo area which were respectively delineated at region of interest (ROI) areas on the maximum axial image. The training cohort (n = 71) and the validation cohort (n = 31) were established by stratified random sampling at a ratio of 7:3. By means of variance threshold, the effective radiomics features, SelectKBest and least absolute shrinkage and selection operator (LASSO) regression method were employed for feature selection and combined to calculate the radiomics score (Rad-score). Six classifiers, including k-nearest neighbor (KNN), support vector machine (SVM), extreme gradient boosting (XGBoost), random forest (RF), logistic regression (LR), and decision tree (DT) were used to construct the models based on radiomic features. The diagnostic performance of these models and the radiomic nomogram was estimated and compared using the area under the receiver operating characteristic (ROC) curve (AUC), and the decision curve analysis (DCA) was used to evaluate which model achieved the most net benefit. RESULTS RF outperformed other classifiers and was selected as the backbone in the classifier with the consolidation + the surrounding halo was taken as ROI to differentiate MPP from SPP in validation cohort. The AUC value of MPP in validation cohort was 0.822, the sensitivity and specificity were 0.81 and 0.81, respectively. CONCLUSION The RF model has the best classification efficiency in the identification of MPP from SPP in children, and the ROI with both consolidation and surrounding halo is most suitable for the delineation.
Collapse
Affiliation(s)
- Dongdong Wang
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianshe Zhao
- Department of Radiology, Children's Hospital Affiliated to Shandong University, Jinan, China
| | - Ran Zhang
- Huiying Medical Technology (Beijing) Co., Ltd., Beijing, China
| | - Qinghu Yan
- Department of Ultrasound, Shandong Public Health Clinical Center, Jinan, China
| | - Lu Zhou
- Department of Cardiac Surgery ICU, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyu Han
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yafei Qi
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dexin Yu
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
17
|
Lee GA, Lin WL, Kuo DP, Li YT, Chang YW, Chen YC, Huang SW, Hsu JBK, Chen CY. Detection of PD-L1 Expression in Temozolomide-Resistant Glioblastoma by Using PD-L1 Antibodies Conjugated with Lipid‑Coated Superparamagnetic Iron Oxide. Int J Nanomedicine 2021; 16:5233-5246. [PMID: 34366665 PMCID: PMC8336995 DOI: 10.2147/ijn.s310464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Targeted superparamagnetic iron oxide (SPIO) nanoparticles are a promising tool for molecular magnetic resonance imaging (MRI) diagnosis. Lipid-coated SPIO nanoparticles have a nonfouling property that can reduce nonspecific binding to off-target cells and prevent agglomeration, making them suitable contrast agents for molecular MRI diagnosis. PD-L1 is a poor prognostic factor for patients with glioblastoma. Most recurrent glioblastomas are temozolomide resistant. Diagnostic probes targeting PD-L1 could facilitate early diagnosis and be used to predict responses to targeted PD-L1 immunotherapy in patients with primary or recurrent glioblastoma. We conjugated lipid-coated SPIO nanoparticles with PD-L1 antibodies to identify PD-L1 expression in glioblastoma or temozolomide-resistant glioblastoma by using MRI. Methods The synthesized PD-L1 antibody-conjugated SPIO (PDL1-SPIO) nanoparticles were characterized using dynamic light scattering, zeta potential assays, transmission electron microscopy images, Prussian blue assay, in vitro cell affinity assay, and animal MRI analysis. Results PDL1-SPIO exhibited a specific binding capacity to PD-L1 of the mouse glioblastoma cell line (GL261). The presence and quantity of PDL1-SPIO in temozolomide-resistant glioblastoma cells and tumor tissue were confirmed through Prussian blue staining and in vivo T2* map MRI, respectively. Conclusion This is the first study to demonstrate that PDL1-SPIO can specifically target temozolomide-resistant glioblastoma with PD-L1 expression in the brain and can be quantified through MRI analysis, thus making it suitable for the diagnosis of PD-L1 expression in temozolomide-resistant glioblastoma in vivo.
Collapse
Affiliation(s)
- Gilbert Aaron Lee
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wan-Li Lin
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Duen-Pang Kuo
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yi-Tien Li
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, Taiwan.,Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wei Chang
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yung-Chieh Chen
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, Taiwan.,Research Center of Translational Imaging, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shiu-Wen Huang
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Justin Bo-Kai Hsu
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Cheng-Yu Chen
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan.,Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, Taiwan.,Research Center of Translational Imaging, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
18
|
Liu D, Chen J, Hu X, Yang K, Liu Y, Hu G, Ge H, Zhang W, Liu H. Imaging-Genomics in Glioblastoma: Combining Molecular and Imaging Signatures. Front Oncol 2021; 11:699265. [PMID: 34295824 PMCID: PMC8290166 DOI: 10.3389/fonc.2021.699265] [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: 04/23/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Based on artificial intelligence (AI), computer-assisted medical diagnosis can scientifically and efficiently deal with a large quantity of medical imaging data. AI technologies including deep learning have shown remarkable progress across medical image recognition and genome analysis. Imaging-genomics attempts to explore the associations between potential gene expression patterns and specific imaging phenotypes. These associations provide potential cellular pathophysiology information, allowing sampling of the lesion habitat with high spatial resolution. Glioblastoma (GB) poses spatial and temporal heterogeneous characteristics, challenging to current precise diagnosis and treatments for the disease. Imaging-genomics provides a powerful tool for non-invasive global assessment of GB and its response to treatment. Imaging-genomics also has the potential to advance our understanding of underlying cancer biology, gene alterations, and corresponding biological processes. This article reviews the recent progress in the utilization of the imaging-genomics analysis in GB patients, focusing on its implications and prospects in individualized diagnosis and management.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu Chen
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Fourth Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Institute of Brain Sciences, The Affilated Nanjing Brain Hosptial of Nanjing Medical University, Nanjing, China
| | - Xinhua Hu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Institute of Brain Sciences, The Affilated Nanjing Brain Hosptial of Nanjing Medical University, Nanjing, China
| | - Kun Yang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Guanjie Hu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Honglin Ge
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenbin Zhang
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Institute of Brain Sciences, The Affilated Nanjing Brain Hosptial of Nanjing Medical University, Nanjing, China
| | - Hongyi Liu
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Institute of Brain Sciences, The Affilated Nanjing Brain Hosptial of Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Pan J, Lei X, Mao X. Identification of KIF4A as a pan-cancer diagnostic and prognostic biomarker via bioinformatics analysis and validation in osteosarcoma cell lines. PeerJ 2021; 9:e11455. [PMID: 34055488 PMCID: PMC8142929 DOI: 10.7717/peerj.11455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/23/2021] [Indexed: 01/17/2023] Open
Abstract
Background Cancer is a disease of abnormal cell proliferation caused by abnormal expression of cancer-related genes. However, it is still difficult to distinguish benign and malignant lesions in many cases. KIF4A has been reported to be associated with a variety of cancer lesions. We aimed to explore whether KIF4A could be used as a biomarker of pan-cancer diagnostic. Methods We identified twenty-eight cell cycle-related genes that were overexpressed in no less than ten types of cancer. We determined KIF4A mRNA and protein expression in osteosarcoma (OS) cells. Furthermore, to determine the effect of KIF4A in OS, we silenced KIF4A in OS cells and detected cell viability, colony formation, invasion, migration, apoptosis and cell cycle parameters. Results KIF4A exhibited upregulated expression in eleven types of cancer. Cell cycle-related genes are extensively overexpressed in various types of cancers. KIF4A overexpression can serve as a diagnostic and prognostic marker in various cancers. Silencing KIF4A inhibited the viability, colony formation, invasion and migration and induced apoptosis and cell cycle arrest of OS cells. Our findings revealed that high expression of KIF4A could serve as a diagnostic and prognostic marker in OS cancers. Conclusion KIF4A could serve as a pan-cancer diagnostic and prognostic marker. KIF4A could be used as a novel therapeutic target for OS.
Collapse
Affiliation(s)
- Jiankang Pan
- Department of Orthopedics, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaohua Lei
- Department of Hepato-Biliary-Pancreatic Surgery, the First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xinzhan Mao
- Department of Orthopedics, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
20
|
Comprehensive Analysis of Prognostic and Genetic Signatures for General Transcription Factor III (GTF3) in Clinical Colorectal Cancer Patients Using Bioinformatics Approaches. Curr Issues Mol Biol 2021; 43:cimb43010002. [PMID: 33925358 PMCID: PMC8935981 DOI: 10.3390/cimb43010002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) has the fourth-highest incidence of all cancer types, and its incidence has steadily increased in the last decade. The general transcription factor III (GTF3) family, comprising GTF3A, GTF3B, GTF3C1, and GTFC2, were stated to be linked with the expansion of different types of cancers; however, their messenger (m)RNA expressions and prognostic values in colorectal cancer need to be further investigated. To study the transcriptomic expression levels of GTF3 gene members in colorectal cancer in both cancerous tissues and cell lines, we first performed high-throughput screening using the Oncomine, GEPIA, and CCLE databases. We then applied the Prognoscan database to query correlations of their mRNA expressions with the disease-specific survival (DSS), overall survival (OS), and disease-free survival (DFS) status of the colorectal cancer patient. Furthermore, proteomics expressions of GTF3 family members in clinical colorectal cancer specimens were also examined using the Human Protein Atlas. Finally, genomic alterations of GTF3 family gene expressions in colorectal cancer and their signal transduction pathways were studied using cBioPortal, ClueGO, CluePedia, and MetaCore platform. Our findings revealed that GTF3 family members' expressions were significantly correlated with the cell cycle, oxidative stress, WNT/β-catenin signaling, Rho GTPases, and G-protein-coupled receptors (GPCRs). Clinically, high GTF3A and GTF3B expressions were significantly correlated with poor prognoses in colorectal cancer patients. Collectively, our study declares that GTF3A was overexpressed in cancer tissues and cell lines, particularly colorectal cancer, and it could possibly step in as a potential prognostic biomarker.
Collapse
|
21
|
d’Este SH, Nielsen MB, Hansen AE. Visualizing Glioma Infiltration by the Combination of Multimodality Imaging and Artificial Intelligence, a Systematic Review of the Literature. Diagnostics (Basel) 2021; 11:diagnostics11040592. [PMID: 33806195 PMCID: PMC8067218 DOI: 10.3390/diagnostics11040592] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/23/2021] [Indexed: 01/14/2023] Open
Abstract
The aim of this study was to systematically review the literature concerning the integration of multimodality imaging with artificial intelligence methods for visualization of tumor cell infiltration in glioma patients. The review was performed in accordance with the preferred reporting items for systematic reviews and meta-analysis (PRISMA) guidelines. The literature search was conducted in PubMed, Embase, The Cochrane Library and Web of Science and yielded 1304 results. 14 studies were included in the qualitative analysis. The reference standard for tumor infiltration was either histopathology or recurrence on image follow-up. Critical assessment was performed according to the Quality Assessment of Diagnostic Accuracy Studies (QUADAS2). All studies concluded their findings to be of significant value for future clinical practice. Diagnostic test accuracy reached an area under the curve of 0.74–0.91 reported in six studies. There was no consensus with regard to included image modalities, models or training and test strategies. The integration of artificial intelligence with multiparametric imaging shows promise for visualizing tumor cell infiltration in glioma patients. This approach can possibly optimize surgical resection margins and help provide personalized radiotherapy planning.
Collapse
Affiliation(s)
- Sabrina Honoré d’Este
- Department of Diagnostic Radiology, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (M.B.N.); (A.E.H.)
- Correspondence:
| | - Michael Bachmann Nielsen
- Department of Diagnostic Radiology, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (M.B.N.); (A.E.H.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Adam Espe Hansen
- Department of Diagnostic Radiology, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark; (M.B.N.); (A.E.H.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|