1
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408069. [PMID: 39535476 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Tianxiang Li
- Department of Ultrasound, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical. Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Bingxin Gong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sichen Wang
- School of Life Science and Technology, Computational Biology Research Center, Harbin Institute of Technology, Harbin, 150001, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
2
|
Horasan A, Güneş A. Advancing Prostate Cancer Diagnosis: A Deep Learning Approach for Enhanced Detection in MRI Images. Diagnostics (Basel) 2024; 14:1871. [PMID: 39272656 PMCID: PMC11393904 DOI: 10.3390/diagnostics14171871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/04/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Prostate cancer remains a leading cause of mortality among men globally, necessitating advancements in diagnostic methodologies to improve detection and treatment outcomes. Magnetic Resonance Imaging has emerged as a crucial technique for the detection of prostate cancer, with current research focusing on the integration of deep learning frameworks to refine this diagnostic process. This study employs a comprehensive approach using multiple deep learning models, including a three-dimensional (3D) Convolutional Neural Network, a Residual Network, and an Inception Network to enhance the accuracy and robustness of prostate cancer detection. By leveraging the complementary strengths of these models through an ensemble method and soft voting technique, the study aims to achieve superior diagnostic performance. The proposed methodology demonstrates state-of-the-art results, with the ensemble model achieving an overall accuracy of 91.3%, a sensitivity of 90.2%, a specificity of 92.1%, a precision of 89.8%, and an F1 score of 90.0% when applied to MRI images from the SPIE-AAPM-NCI PROSTATEx dataset. Evaluation of the models involved meticulous pre-processing, data augmentation, and the use of advanced deep-learning architectures to analyze the whole MRI slices and volumes. The findings highlight the potential of using an ensemble approach to significantly improve prostate cancer diagnostics, offering a robust and precise tool for clinical applications.
Collapse
Affiliation(s)
- Alparslan Horasan
- Computer Engineering Department, Istanbul Aydin University, 34150 Istanbul, Turkey
| | - Ali Güneş
- Computer Engineering Department, Istanbul Aydin University, 34150 Istanbul, Turkey
| |
Collapse
|
3
|
Nardone V, Marmorino F, Germani MM, Cichowska-Cwalińska N, Menditti VS, Gallo P, Studiale V, Taravella A, Landi M, Reginelli A, Cappabianca S, Girnyi S, Cwalinski T, Boccardi V, Goyal A, Skokowski J, Oviedo RJ, Abou-Mrad A, Marano L. The Role of Artificial Intelligence on Tumor Boards: Perspectives from Surgeons, Medical Oncologists and Radiation Oncologists. Curr Oncol 2024; 31:4984-5007. [PMID: 39329997 PMCID: PMC11431448 DOI: 10.3390/curroncol31090369] [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: 07/29/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
The integration of multidisciplinary tumor boards (MTBs) is fundamental in delivering state-of-the-art cancer treatment, facilitating collaborative diagnosis and management by a diverse team of specialists. Despite the clear benefits in personalized patient care and improved outcomes, the increasing burden on MTBs due to rising cancer incidence and financial constraints necessitates innovative solutions. The advent of artificial intelligence (AI) in the medical field offers a promising avenue to support clinical decision-making. This review explores the perspectives of clinicians dedicated to the care of cancer patients-surgeons, medical oncologists, and radiation oncologists-on the application of AI within MTBs. Additionally, it examines the role of AI across various clinical specialties involved in cancer diagnosis and treatment. By analyzing both the potential and the challenges, this study underscores how AI can enhance multidisciplinary discussions and optimize treatment plans. The findings highlight the transformative role that AI may play in refining oncology care and sustaining the efficacy of MTBs amidst growing clinical demands.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80131 Naples, Italy
| | - Federica Marmorino
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Marco Maria Germani
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | | | | | - Paolo Gallo
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80131 Naples, Italy
| | - Vittorio Studiale
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Ada Taravella
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Matteo Landi
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80131 Naples, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80131 Naples, Italy
| | - Sergii Girnyi
- Department of General Surgery and Surgical Oncology, "Saint Wojciech" Hospital, "Nicolaus Copernicus" Health Center, 80-462 Gdańsk, Poland
| | - Tomasz Cwalinski
- Department of General Surgery and Surgical Oncology, "Saint Wojciech" Hospital, "Nicolaus Copernicus" Health Center, 80-462 Gdańsk, Poland
| | - Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy
| | - Aman Goyal
- Adesh Institute of Medical Sciences and Research, Bathinda 151109, Punjab, India
| | - Jaroslaw Skokowski
- Department of General Surgery and Surgical Oncology, "Saint Wojciech" Hospital, "Nicolaus Copernicus" Health Center, 80-462 Gdańsk, Poland
- Department of Medicine, Academy of Applied Medical and Social Sciences-AMiSNS: Akademia Medycznych I Spolecznych Nauk Stosowanych, 82-300 Elbląg, Poland
| | - Rodolfo J Oviedo
- Nacogdoches Medical Center, Nacogdoches, TX 75965, USA
- Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX 77021, USA
- College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Adel Abou-Mrad
- Centre Hospitalier Universitaire d'Orléans, 45100 Orléans, France
| | - Luigi Marano
- Department of General Surgery and Surgical Oncology, "Saint Wojciech" Hospital, "Nicolaus Copernicus" Health Center, 80-462 Gdańsk, Poland
- Department of Medicine, Academy of Applied Medical and Social Sciences-AMiSNS: Akademia Medycznych I Spolecznych Nauk Stosowanych, 82-300 Elbląg, Poland
| |
Collapse
|
4
|
Zhang Y, Hu Y, Zhao S, Xu S. Validation of the 2018 FIGO staging system for stage IIIC cervical cancer by determining the metabolic and radiomic heterogeneity of primary tumors based on 18F-FDG PET/CT. Abdom Radiol (NY) 2024; 49:2027-2039. [PMID: 38526594 DOI: 10.1007/s00261-024-04226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 03/26/2024]
Abstract
PURPOSE This study aimed to validate the 2018 FIGO staging system of cervical cancer (CC) by determining the metabolic and radiomic heterogeneity of primary tumors between stage IIIC1 and IIIC2. METHODS 168 patients with squamous cell CC underwent pre-treatment fluorine-18 fluorodeoxyglucose positron emission computed tomography (18F-FDG PET/CT) and were randomly allocated to training and testing cohorts with a 7:3 ratio. Radiomics features were extracted from the primary tumors based on CT and PET data. Ten metabolic parameters of the primary tumors were also assessed. After feature selection, three logistic regression radiomics models, involving (1) 2 CT features, (2) 3 PET features, and (3) 2 CT features + 3 PET features, respectively, and one random forest model were established. Finally, area under the curve (AUC) values and calibration curves were used to evaluate the 4 models. RESULTS The IIIC1 and IIIC2 groups did not differ significantly in age, weight, height, or the 10 major metabolic parameters (P > 0.05). The AUCs of the 4 models were 0.577, 0.639, 0.763, and 0.506, respectively, in the training cohort, and 0.789, 0.699, 0.761, and 0.538, respectively, in the testing cohort. The model fit of the logistic regression model based on CT + PET data was good in both the training and testing cohorts. CONCLUSION Our study offers additional diagnostic options for PALN metastasis, which could impact treatment decisions. Our results indirectly support the conclusions of previous studies recommending that primary tumors should be considered during IIIC staging.
Collapse
Affiliation(s)
- Yun Zhang
- Department of PET/CT Center, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yuxiao Hu
- Department of PET/CT Center, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| | - Shuang Zhao
- Department of PET/CT Center, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Shanshan Xu
- Department of PET/CT Center, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Swain IX, Kresak AM. Iron Supplementation Increases Tumor Burden and Alters Protein Expression in a Mouse Model of Human Intestinal Cancer. Nutrients 2024; 16:1316. [PMID: 38732562 PMCID: PMC11085868 DOI: 10.3390/nu16091316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Iron supplements are widely consumed. However, excess iron may accelerate intestinal tumorigenesis. To determine the effect of excess iron on intestinal tumor burden and protein expression changes between tumor and normal tissues, ApcMin/+ mice were fed control (adequate) and excess iron (45 and 450 mg iron/kg diet, respectively; n = 9/group) for 10 wk. Tumor burden was measured, and two-dimensional fluorescence difference gel electrophoresis was used to identify differentially expressed proteins in tumor and normal intestinal tissues. There was a significant increase (78.3%; p ≤ 0.05) in intestinal tumor burden (mm2/cm) with excess iron at wk 10. Of 980 analyzed protein spots, 69 differentially expressed (p ≤ 0.05) protein isoforms were identified, representing 55 genes. Of the isoforms, 56 differed (p ≤ 0.05) between tumor vs. normal tissues from the adequate iron group and 23 differed (p ≤ 0.05) between tumors from the adequate vs. excess iron. Differentially expressed proteins include those involved in cell integrity and adaptive response to reactive oxygen species (including, by gene ID: ANPEP, DPP7, ITGB1, PSMA1 HSPA5). Biochemical pathway analysis found that iron supplementation modulated four highly significant (p ≤ 0.05) functional networks. These findings enhance our understanding of interplay between dietary iron and intestinal tumorigenesis and may help develop more specific dietary guidelines regarding trace element intake.
Collapse
Affiliation(s)
- Ian X. Swain
- Department of Pathology, School of Medicine, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA;
| | | |
Collapse
|
6
|
Huang W, Xiong W, Tang L, Chen C, Yuan Q, Zhang C, Zhou K, Sun Z, Zhang T, Han Z, Feng H, Liang X, Zhong Y, Deng H, Yu L, Xu Y, Wang W, Shen L, Li G, Jiang Y. Non-invasive CT imaging biomarker to predict immunotherapy response in gastric cancer: a multicenter study. J Immunother Cancer 2023; 11:e007807. [PMID: 38179695 PMCID: PMC10668251 DOI: 10.1136/jitc-2023-007807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Despite remarkable benefits have been provided by immune checkpoint inhibitors in gastric cancer (GC), predictions of treatment response and prognosis remain unsatisfactory, making identifying biomarkers desirable. The aim of this study was to develop and validate a CT imaging biomarker to predict the immunotherapy response in patients with GC and investigate the associated immune infiltration patterns. METHODS This retrospective study included 294 GC patients who received anti-PD-1/PD-L1 immunotherapy from three independent medical centers between January 2017 and April 2022. A radiomics score (RS) was developed from the intratumoral and peritumoral features on pretreatment CT images to predict immunotherapy-related progression-free survival (irPFS). The performance of the RS was evaluated by the area under the time-dependent receiver operating characteristic curve (AUC). Multivariable Cox regression analysis was performed to construct predictive nomogram of irPFS. The C-index was used to determine the performance of the nomogram. Bulk RNA sequencing of tumors from 42 patients in The Cancer Genome Atlas was used to investigate the RS-associated immune infiltration patterns. RESULTS Overall, 89 of 294 patients (median age, 57 years (IQR 48-66 years); 171 males) had an objective response to immunotherapy. The RS included 13 CT features that yielded AUCs of 12-month irPFS of 0.787, 0.810 and 0.785 in the training, internal validation, and external validation 1 cohorts, respectively, and an AUC of 24-month irPFS of 0.805 in the external validation 2 cohort. Patients with low RS had longer irPFS in each cohort (p<0.05). Multivariable Cox regression analyses showed RS is an independent prognostic factor of irPFS. The nomogram that integrated the RS and clinical characteristics showed improved performance in predicting irPFS, with C-index of 0.687-0.778 in the training and validation cohorts. The CT imaging biomarker was associated with M1 macrophage infiltration. CONCLUSION The findings of this prognostic study suggest that the non-invasive CT imaging biomarker can effectively predict immunotherapy outcomes in patients with GC and is associated with innate immune signaling, which can serve as a potential tool for individual treatment decisions.
Collapse
Affiliation(s)
- Weicai Huang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Wenjun Xiong
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Cheng Zhang
- Department of Gastrointestinal Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Kangneng Zhou
- University of Science and Technology, Beijing, China
| | - Zepang Sun
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Taojun Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Zhen Han
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Hao Feng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Xiaokun Liang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yonghong Zhong
- Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haijun Deng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Lequan Yu
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong, China
| | - Yikai Xu
- Department of Medical Imaging Center, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Yuming Jiang
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
7
|
Zhang G, Zhang Z, Pei Y, Hu W, Xue Y, Ning R, Guo X, Sun Y, Zhang Q. Biological and clinical significance of radiomics features obtained from magnetic resonance imaging preceding pre-carbon ion radiotherapy in prostate cancer based on radiometabolomics. Front Endocrinol (Lausanne) 2023; 14:1272806. [PMID: 38027108 PMCID: PMC10644841 DOI: 10.3389/fendo.2023.1272806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction We aimed to investigate the feasibility of metabolomics to explain the underlying biological implications of radiomics features obtained from magnetic resonance imaging (MRI) preceding carbon ion radiotherapy (CIRT) in patients with prostate cancer and to further explore the clinical significance of radiomics features on the prognosis of patients, based on their biochemical recurrence (BCR) status. Methods Metabolomic results obtained using high-performance liquid chromatography coupled with tandem mass spectrometry of urine samples, combined with pre-RT radiomic features extracted from MRI images, were evaluated to investigate their biological significance. Receiver operating characteristic (ROC) curve analysis was subsequently conducted to examine the correlation between these biological implications and clinical BCR status. Statistical and metabolic pathway analyses were performed using MetaboAnalyst and R software. Results Correlation analysis revealed that methionine alteration extent was significantly related to four radiomic features (Contrast, Difference Variance, Small Dependence High Gray Level Emphasis, and Mean Absolute Deviation), which were significantly correlated with BCR status. The area under the curve (AUC) for BCR prediction of these four radiomic features ranged from 0.704 to 0.769, suggesting that the higher the value of these four radiomic features, the greater the decrease in methionine levels after CIRT and the lower the probability of BCR. Pre-CIRT MRI radiomic features were associated with CIRT-suppressed metabolites. Discussion These radiomic features can be used to predict the alteration in the amplitude of methionine after CIRT and the BCR status, which may contribute to the optimization of the CIRT strategy and deepen the understanding of PCa.
Collapse
Affiliation(s)
- Guangyuan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Yulei Pei
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Wei Hu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Yushan Xue
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Renli Ning
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Xiaomao Guo
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Yun Sun
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Qing Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| |
Collapse
|
8
|
Orsini A, Diquigiovanni C, Bonora E. Omics Technologies Improving Breast Cancer Research and Diagnostics. Int J Mol Sci 2023; 24:12690. [PMID: 37628869 PMCID: PMC10454385 DOI: 10.3390/ijms241612690] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer (BC) has yielded approximately 2.26 million new cases and has caused nearly 685,000 deaths worldwide in the last two years, making it the most common diagnosed cancer type in the world. BC is an intricate ecosystem formed by both the tumor microenvironment and malignant cells, and its heterogeneity impacts the response to treatment. Biomedical research has entered the era of massive omics data thanks to the high-throughput sequencing revolution, quick progress and widespread adoption. These technologies-liquid biopsy, transcriptomics, epigenomics, proteomics, metabolomics, pharmaco-omics and artificial intelligence imaging-could help researchers and clinicians to better understand the formation and evolution of BC. This review focuses on the findings of recent multi-omics-based research that has been applied to BC research, with an introduction to every omics technique and their applications for the different BC phenotypes, biomarkers, target therapies, diagnosis, treatment and prognosis, to provide a comprehensive overview of the possibilities of BC research.
Collapse
Affiliation(s)
| | - Chiara Diquigiovanni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40131 Bologna, Italy; (A.O.); (E.B.)
| | | |
Collapse
|
9
|
Chaddad A, Tan G, Liang X, Hassan L, Rathore S, Desrosiers C, Katib Y, Niazi T. Advancements in MRI-Based Radiomics and Artificial Intelligence for Prostate Cancer: A Comprehensive Review and Future Prospects. Cancers (Basel) 2023; 15:3839. [PMID: 37568655 PMCID: PMC10416937 DOI: 10.3390/cancers15153839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The use of multiparametric magnetic resonance imaging (mpMRI) has become a common technique used in guiding biopsy and developing treatment plans for prostate lesions. While this technique is effective, non-invasive methods such as radiomics have gained popularity for extracting imaging features to develop predictive models for clinical tasks. The aim is to minimize invasive processes for improved management of prostate cancer (PCa). This study reviews recent research progress in MRI-based radiomics for PCa, including the radiomics pipeline and potential factors affecting personalized diagnosis. The integration of artificial intelligence (AI) with medical imaging is also discussed, in line with the development trend of radiogenomics and multi-omics. The survey highlights the need for more data from multiple institutions to avoid bias and generalize the predictive model. The AI-based radiomics model is considered a promising clinical tool with good prospects for application.
Collapse
Affiliation(s)
- Ahmad Chaddad
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
- The Laboratory for Imagery, Vision and Artificial Intelligence, École de Technologie Supérieure (ETS), Montreal, QC H3C 1K3, Canada
| | - Guina Tan
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
| | - Xiaojuan Liang
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
| | - Lama Hassan
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
| | | | - Christian Desrosiers
- The Laboratory for Imagery, Vision and Artificial Intelligence, École de Technologie Supérieure (ETS), Montreal, QC H3C 1K3, Canada
| | - Yousef Katib
- Department of Radiology, Taibah University, Al Madinah 42361, Saudi Arabia
| | - Tamim Niazi
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
10
|
Dinis Fernandes C, Schaap A, Kant J, van Houdt P, Wijkstra H, Bekers E, Linder S, Bergman AM, van der Heide U, Mischi M, Zwart W, Eduati F, Turco S. Radiogenomics Analysis Linking Multiparametric MRI and Transcriptomics in Prostate Cancer. Cancers (Basel) 2023; 15:3074. [PMID: 37370685 DOI: 10.3390/cancers15123074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Prostate cancer (PCa) is a highly prevalent cancer type with a heterogeneous prognosis. An accurate assessment of tumor aggressiveness can pave the way for tailored treatment strategies, potentially leading to better outcomes. While tumor aggressiveness is typically assessed based on invasive methods (e.g., biopsy), radiogenomics, combining diagnostic imaging with genomic information can help uncover aggressive (imaging) phenotypes, which in turn can provide non-invasive advice on individualized treatment regimens. In this study, we carried out a parallel analysis on both imaging and transcriptomics data in order to identify features associated with clinically significant PCa (defined as an ISUP grade ≥ 3), subsequently evaluating the correlation between them. Textural imaging features were extracted from multi-parametric MRI sequences (T2W, DWI, and DCE) and combined with DCE-derived parametric pharmacokinetic maps obtained using magnetic resonance dispersion imaging (MRDI). A transcriptomic analysis was performed to derive functional features on transcription factors (TFs), and pathway activity from RNA sequencing data, here referred to as transcriptomic features. For both the imaging and transcriptomic features, different machine learning models were separately trained and optimized to classify tumors in either clinically insignificant or significant PCa. These models were validated in an independent cohort and model performance was used to isolate a subset of relevant imaging and transcriptomic features to be further investigated. A final set of 31 imaging features was correlated to 33 transcriptomic features obtained on the same tumors. Five significant correlations (p < 0.05) were found, of which, three had moderate strength (|r| ≥ 0.5). The strongest significant correlations were seen between a perfusion-based imaging feature-MRDI A median-and the activities of the TFs STAT6 (-0.64) and TFAP2A (-0.50). A higher-order T2W textural feature was also significantly correlated to the activity of the TF STAT6 (-0.58). STAT6 plays an important role in controlling cell proliferation and migration. Loss of the AP2alpha protein expression, quantified by TFAP2A, has been strongly associated with aggressiveness and progression in PCa. According to our findings, a combination of texture features extracted from T2W and DCE, as well as perfusion-based pharmacokinetic features, can be considered for the prediction of clinically significant PCa, with the pharmacokinetic MRDI A feature being the most correlated with the underlying transcriptomic information. These results highlight a link between quantitative imaging features and the underlying transcriptomic landscape of prostate tumors.
Collapse
Affiliation(s)
- Catarina Dinis Fernandes
- Electrical Engineering Department, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Annekoos Schaap
- Electrical Engineering Department, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Joan Kant
- Biomedical Engineering-Computational Biology Department, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Petra van Houdt
- Department of Radiation Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Hessel Wijkstra
- Electrical Engineering Department, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Department of Urology, Amsterdam University Medical Centers, 1100 DD Amsterdam, The Netherlands
| | - Elise Bekers
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Simon Linder
- Division of Oncogenomics, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Andries M Bergman
- Division of Oncogenomics, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Division of Medical Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Uulke van der Heide
- Department of Radiation Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Massimo Mischi
- Electrical Engineering Department, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Wilbert Zwart
- Biomedical Engineering-Computational Biology Department, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
- Division of Oncogenomics, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Federica Eduati
- Biomedical Engineering-Computational Biology Department, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Simona Turco
- Electrical Engineering Department, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
11
|
Fischer S, Spath N, Hamed M. Data-Driven Radiogenomic Approach for Deciphering Molecular Mechanisms Underlying Imaging Phenotypes in Lung Adenocarcinoma: A Pilot Study. Int J Mol Sci 2023; 24:4947. [PMID: 36902378 PMCID: PMC10003564 DOI: 10.3390/ijms24054947] [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: 12/23/2022] [Revised: 02/13/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
The heterogeneity of lung tumor nodules is reflected in their phenotypic characteristics in radiological images. The radiogenomics field employs quantitative image features combined with transcriptome expression levels to understand tumor heterogeneity molecularly. Due to the different data acquisition techniques for imaging traits and genomic data, establishing meaningful connections poses a challenge. We analyzed 86 image features describing tumor characteristics (such as shape and texture) with the underlying transcriptome and post-transcriptome profiles of 22 lung cancer patients (median age 67.5 years, from 42 to 80 years) to unravel the molecular mechanisms behind tumor phenotypes. As a result, we were able to construct a radiogenomic association map (RAM) linking tumor morphology, shape, texture, and size with gene and miRNA signatures, as well as biological correlates of GO terms and pathways. These indicated possible dependencies between gene and miRNA expression and the evaluated image phenotypes. In particular, the gene ontology processes "regulation of signaling" and "cellular response to organic substance" were shown to be reflected in CT image phenotypes, exhibiting a distinct radiomic signature. Moreover, the gene regulatory networks involving the TFs TAL1, EZH2, and TGFBR2 could reflect how the texture of lung tumors is potentially formed. The combined visualization of transcriptomic and image features suggests that radiogenomic approaches could identify potential image biomarkers for underlying genetic variation, allowing a broader view of the heterogeneity of the tumors. Finally, the proposed methodology could also be adapted to other cancer types to expand our knowledge of the mechanistic interpretability of tumor phenotypes.
Collapse
Affiliation(s)
- Sarah Fischer
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Ernst-Heydemannstr. 8, 18057 Rostock, Germany
- Department of Systems Biology and Bioinformatics, University of Rostock, Ulmenstr. 69, 18057 Rostock, Germany
| | - Nicolas Spath
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Ernst-Heydemannstr. 8, 18057 Rostock, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Arnold-Hellerstr. 3, 24105 Kiel, Germany
| | - Mohamed Hamed
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Ernst-Heydemannstr. 8, 18057 Rostock, Germany
| |
Collapse
|
12
|
Jia Y, Quan S, Ren J, Wu H, Liu A, Gao Y, Hao F, Yang Z, Zhang T, Hu H. MRI radiomics predicts progression-free survival in prostate cancer. Front Oncol 2022; 12:974257. [PMID: 36110963 PMCID: PMC9468743 DOI: 10.3389/fonc.2022.974257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/02/2022] [Indexed: 01/31/2023] Open
Abstract
Objective To assess the predictive value of magnetic resonance imaging (MRI) radiomics for progression-free survival (PFS) in patients with prostate cancer (PCa). Methods 191 patients with prostate cancer confirmed by puncture biopsy or surgical pathology were included in this retrospective study, including 133 in the training group and 58 in the validation group. All patients underwent T2WI and DWI serial scans. Three radiomics models were constructed using univariate logistic regression and Gradient Boosting Decision Tree(GBDT) for feature screening, followed by Cox risk regression to construct a mixed model combining radiomics features and clinicopathological risk factors and to draw a nomogram. The performance of the models was evaluated by receiver operating characteristic curve (ROC), calibration curve and decision curve analysis. The Kaplan-Meier method was applied for survival analysis. Results Compared with the radiomics model, the hybrid model consisting of a combination of radiomics features and clinical data performed the best in predicting PFS in PCa patients, with AUCs of 0.926 and 0.917 in the training and validation groups, respectively. Decision curve analysis showed that the radiomics nomogram had good clinical application and the calibration curve proved to have good stability. Survival curves showed that PFS was shorter in the high-risk group than in the low-risk group. Conclusion The hybrid model constructed from radiomics and clinical data showed excellent performance in predicting PFS in prostate cancer patients. The nomogram provides a non-invasive diagnostic tool for risk stratification of clinical patients.
Collapse
Affiliation(s)
- Yushan Jia
- Affiliated Hospital, Inner Mongolia Medical University, Hohhot, China
| | - Shuai Quan
- Department of Pharmaceuticals Diagnosis, GE Healthcare (China), Shanghai, China
| | - Jialiang Ren
- Department of Pharmaceuticals Diagnosis, GE Healthcare (China), Shanghai, China
| | - Hui Wu
- Department of Radiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China,*Correspondence: Hui Wu, ; Aishi Liu,
| | - Aishi Liu
- Department of Radiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China,*Correspondence: Hui Wu, ; Aishi Liu,
| | - Yang Gao
- Department of Radiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Fene Hao
- Department of Radiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhenxing Yang
- Department of Radiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Tong Zhang
- Affiliated Hospital, Inner Mongolia Medical University, Hohhot, China
| | - He Hu
- Affiliated Hospital, Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
13
|
Radiogenomic analysis reveals tumor heterogeneity of triple-negative breast cancer. Cell Rep Med 2022; 3:100694. [PMID: 35858585 PMCID: PMC9381418 DOI: 10.1016/j.xcrm.2022.100694] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/04/2022]
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with an adverse prognosis and significant tumor heterogeneity. Here, we extract quantitative radiomic features from contrast-enhanced magnetic resonance images to construct a breast cancer radiomic dataset (n = 860) and a TNBC radiogenomic dataset (n = 202). We develop and validate radiomic signatures that can fairly differentiate TNBC from other breast cancer subtypes and distinguish molecular subtypes within TNBC. A radiomic feature that captures peritumoral heterogeneity is determined to be a prognostic factor for recurrence-free survival (p = 0.01) and overall survival (p = 0.004) in TNBC. Combined with the established matching TNBC transcriptomic and metabolomic data, we demonstrate that peritumoral heterogeneity is associated with immune suppression and upregulated fatty acid synthesis in tumor samples. Collectively, this multi-omic dataset serves as a useful public resource to promote precise subtyping of TNBC and helps to understand the biological significance of radiomics. A radiomic signature identifies TNBC from other subtypes of breast cancer Radiomic features are predictive of TNBC molecular subtypes A radiomic feature reflecting peritumoral heterogeneity indicates TNBC prognosis Peritumoral heterogeneity correlates with metabolic and immune abnormalities in TNBC
Collapse
|
14
|
Cho HH, Kim CK, Park H. Overview of radiomics in prostate imaging and future directions. Br J Radiol 2022; 95:20210539. [PMID: 34797688 PMCID: PMC8978251 DOI: 10.1259/bjr.20210539] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent advancements in imaging technology and analysis methods have led to an analytic framework known as radiomics. This framework extracts comprehensive high-dimensional features from imaging data and performs data mining to build analytical models for improved decision-support. Its features include many categories spanning texture and shape; thus, it can provide abundant information for precision medicine. Many studies of prostate radiomics have shown promising results in the assessment of pathological features, prediction of treatment response, and stratification of risk groups. Herein, we aimed to provide a general overview of radiomics procedures, discuss technical issues, explain various clinical applications, and suggest future research directions, especially for prostate imaging.
Collapse
Affiliation(s)
- Hwan-Ho Cho
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea.,Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Chan Kyo Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyunjin Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea.,School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
15
|
Prostate Cancer Radiogenomics-From Imaging to Molecular Characterization. Int J Mol Sci 2021; 22:ijms22189971. [PMID: 34576134 PMCID: PMC8465891 DOI: 10.3390/ijms22189971] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/06/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022] Open
Abstract
Radiomics and genomics represent two of the most promising fields of cancer research, designed to improve the risk stratification and disease management of patients with prostate cancer (PCa). Radiomics involves a conversion of imaging derivate quantitative features using manual or automated algorithms, enhancing existing data through mathematical analysis. This could increase the clinical value in PCa management. To extract features from imaging methods such as magnetic resonance imaging (MRI), the empiric nature of the analysis using machine learning and artificial intelligence could help make the best clinical decisions. Genomics information can be explained or decoded by radiomics. The development of methodologies can create more-efficient predictive models and can better characterize the molecular features of PCa. Additionally, the identification of new imaging biomarkers can overcome the known heterogeneity of PCa, by non-invasive radiological assessment of the whole specific organ. In the future, the validation of recent findings, in large, randomized cohorts of PCa patients, can establish the role of radiogenomics. Briefly, we aimed to review the current literature of highly quantitative and qualitative results from well-designed studies for the diagnoses, treatment, and follow-up of prostate cancer, based on radiomics, genomics and radiogenomics research.
Collapse
|
16
|
Target Heterogeneity in Oncology: The Best Predictor for Differential Response to Radioligand Therapy in Neuroendocrine Tumors and Prostate Cancer. Cancers (Basel) 2021; 13:cancers13143607. [PMID: 34298822 PMCID: PMC8304541 DOI: 10.3390/cancers13143607] [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] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary In the era of precision medicine, novel targets have emerged on the surface of cancer cells, which have been exploited for the purpose of radioligand therapy. However, there have been variations in the way these receptors are expressed, especially in prostate cancers and neuroendocrine tumors. This variable expression of receptors across the grades of cancers led to the concept of ‘target heterogeneity’, which has not just impacted therapeutic decisions but also their outcomes. Radiopharmaceuticals targeting receptors need to be used when there are specific indicators—either clinical, radiological, or at molecular level—warranting their use. In addition, response to these radioligands can be assessed using different techniques, whereby we can prognosticate further outcomes. We shall also discuss, in this review, the conventional as well as novel approaches of detecting heterogeneity in prostate cancers and neuroendocrine tumors. Abstract Tumor or target heterogeneity (TH) implies presence of variable cellular populations having different genomic characteristics within the same tumor, or in different tumor sites of the same patient. The challenge is to identify this heterogeneity, as it has emerged as the most common cause of ‘treatment resistance’, to current therapeutic agents. We have focused our discussion on ‘Prostate Cancer’ and ‘Neuroendocrine Tumors’, and looked at the established methods for demonstrating heterogeneity, each with its advantages and drawbacks. Also, the available theranostic radiotracers targeting PSMA and somatostatin receptors combined with targeted systemic agents, have been described. Lu-177 labeled PSMA and DOTATATE are the ‘standard of care’ radionuclide therapeutic tracers for management of progressive treatment-resistant prostate cancer and NET. These approved therapies have shown reasonable benefit in treatment outcome, with improvement in quality of life parameters. Various biomarkers and predictors of response to radionuclide therapies targeting TH which are currently available and those which can be explored have been elaborated in details. Imaging-based features using artificial intelligence (AI) need to be developed to further predict the presence of TH. Also, novel theranostic tools binding to newer targets on surface of cancer cell should be explored to overcome the treatment resistance to current treatment regimens.
Collapse
|
17
|
Thenault R, Gasmi A, Khene ZE, Bensalah K, Mathieu R. Radiogenomics in prostate cancer evaluation. Curr Opin Urol 2021; 31:424-429. [PMID: 34009176 DOI: 10.1097/mou.0000000000000902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Radiogenomics, fusion between radiomics and genomics, represents a new field of research to improve cancer comprehension and evaluation. In this review, we give an overview of radiogenomics and its most recent and relevant applications in prostate cancer management. RECENT FINDINGS Literature about radiogenomics in prostate cancer emerged last 5 years but remains scarce. Radiogenomics in prostate cancer mainly rely on MRI-based features. Several imaging biomarkers, mostly based on the identification of radiomic features from deep learning studies, have been studied for the prediction of genomic profiles, such as PTEN Decipher Oncotype DX or Prolaris expression. However, despite promising results, several limitations still preclude any integration of radiogenomics in daily practice. SUMMARY In the future, the emergence of artificial intelligence in urology, with an increasing use of radiomics and genomics data, may enable radiogenomics to assume a growing role in the evaluation of prostate cancer, with a noninvasive and personal approach in the field of personalized medicine. Further efforts are necessary for integration of this promising approach in prostate cancer decision-making.
Collapse
Affiliation(s)
- Ronan Thenault
- Department of Urology, Service d'urologie, Rennes University Hospital, Hôpital Pontchaillou
| | - Anis Gasmi
- Department of Urology, Service d'urologie, Rennes University Hospital, Hôpital Pontchaillou
| | - Zine-Edine Khene
- Department of Urology, Service d'urologie, Rennes University Hospital, Hôpital Pontchaillou
| | - Karim Bensalah
- Department of Urology, Service d'urologie, Rennes University Hospital, Hôpital Pontchaillou
| | - Romain Mathieu
- Department of Urology, Service d'urologie, Rennes University Hospital, Hôpital Pontchaillou
- IRSET, Rennes, France
| |
Collapse
|
18
|
Lorkowski J, Kolaszyńska O, Pokorski M. Artificial Intelligence and Precision Medicine: A Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1375:1-11. [PMID: 34138457 DOI: 10.1007/5584_2021_652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This article aims to present how the advanced solutions of artificial intelligence and precision medicine work together to refine medical management. Multi-omics seems the most suitable approach for biological analysis of data on precision medicine and artificial intelligence. We searched PubMed and Google Scholar databases to collect pertinent articles appearing up to 5 March 2021. Genetics, oncology, radiology, and the recent coronavirus disease (COVID-19) pandemic were chosen as representative fields addressing the cross-compliance of artificial intelligence (AI) and precision medicine based on the highest number of articles, topicality, and interconnectedness of the issue. Overall, we identified and perused 1572 articles. AI is a breakthrough that takes part in shaping the Fourth Industrial Revolution in medicine and health care, changing the long-time accepted diagnostic and treatment regimens and approaches. AI-based link prediction models may be outstandingly helpful in the literature search for drug repurposing or finding new therapeutical modalities in rapidly erupting wide-scale diseases such as the recent COVID-19.
Collapse
Affiliation(s)
- Jacek Lorkowski
- Department of Orthopedics, Traumatology and Sports Medicine, Central Clinical Hospital of the Ministry of Internal Affairs and Administration, Warsaw, Poland. .,Faculty of Health Sciences, Medical University of Mazovia, Warsaw, Poland.
| | - Oliwia Kolaszyńska
- Department of Cardiology, Independent Public Regional Hospital, Szczecin, Poland
| | - Mieczysław Pokorski
- Institute of Health Sciences, Opole University, Opole, Poland.,Faculty of Health Sciences, The Jan Długosz University in Częstochowa, Częstochowa, Poland
| |
Collapse
|
19
|
Martínez-González LJ, Sánchez-Conde V, González-Cabezuelo JM, Antunez-Rodríguez A, Andrés-León E, Robles-Fernandez I, Lorente JA, Vázquez-Alonso F, Alvarez-Cubero MJ. Identification of MicroRNAs as Viable Aggressiveness Biomarkers for Prostate Cancer. Biomedicines 2021; 9:biomedicines9060646. [PMID: 34198846 PMCID: PMC8227559 DOI: 10.3390/biomedicines9060646] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/22/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
MiRNAs play a relevant role in PC (prostate cancer) by the regulation in the expression of several pathways’ AR (androgen receptor), cellular cycle, apoptosis, MET (mesenchymal epithelium transition), or metastasis. Here, we report the role of several miRNAs’ expression patterns, such as miR-93-5p, miR-23c, miR-210-3p, miR-221-3p, miR-592, miR-141, miR-375, and miR-130b, with relevance in processes like cell proliferation and MET. Using Trizol® extraction protocol and TaqMan™ specific probes for amplification, we performed miRNAs’ analysis of 159 PC fresh tissues and 60 plasmas from peripheral blood samples. We had clinical data from all samples including PSA, Gleason, TNM, and D’Amico risk. Moreover, a bioinformatic analysis in TCGA (The Cancer Genome Atlas) was included to analyze the effect of the most relevant miRNAs according to aggressiveness in an extensive cohort (n = 531). We found that miR-210-3p, miR-23c, miR-592, and miR-93-5p are the most suitable biomarkers for PC aggressiveness and diagnosis, respectively. In fact, according with our results, miR-93-5p seems the most promising non-invasive biomarker for PC. To sum up, miR-210-3p, miR-23c, miR-592, and miR-93-5p miRNAs are suggested to be potential biomarkers for PC risk stratification that could be included in non-invasive strategies such as liquid biopsy in precision medicine for PC management.
Collapse
Affiliation(s)
- Luis Javier Martínez-González
- GENYO. Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, Genomics Unit, PTS Granada-Avenida de la Ilustración, 114-18016 Granada, Spain;
- Correspondence: author: (L.J.M.-G.); (M.J.A.-C.); Tel.: +34-958-715-500 (ext. 108) (L.J.M.-G.); +34-958-248-945 (M.J.A.-C.); Fax: +34-958-637-071 (L.J.M.-G.)
| | - Victor Sánchez-Conde
- Urology Department, Hospital Virgen de las Nieves, 18014 Granada, Spain; (V.S.-C.); (F.V.-A.)
| | | | - Alba Antunez-Rodríguez
- GENYO. Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, Genomics Unit, PTS Granada-Avenida de la Ilustración, 114-18016 Granada, Spain;
| | - Eduardo Andrés-León
- Bioinformatics Unit, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Spanish National Research Council (CSIC), 18016 Granada, Spain;
| | - Inmaculada Robles-Fernandez
- GENYO. Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, Liquid Biopsy and Cancer Interception Group, PTS Granada, 114-18016 Granada, Spain; (I.R.-F.); (J.A.L.)
| | - Jose Antonio Lorente
- GENYO. Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, Liquid Biopsy and Cancer Interception Group, PTS Granada, 114-18016 Granada, Spain; (I.R.-F.); (J.A.L.)
- University of Granada, Legal Medicine and Toxicology Department, Faculty of Medicine, PTS Granada, 18016 Granada, Spain
| | - Fernando Vázquez-Alonso
- Urology Department, Hospital Virgen de las Nieves, 18014 Granada, Spain; (V.S.-C.); (F.V.-A.)
| | - María Jesus Alvarez-Cubero
- GENYO. Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, Liquid Biopsy and Cancer Interception Group, PTS Granada, 114-18016 Granada, Spain; (I.R.-F.); (J.A.L.)
- University of Granada, Department of Biochemistry and Molecular Biology III, Faculty of Medicine, PTS Granada, 18016 Granada, Spain
- Nutrition, Diet and Risk Assessment Group, Bio-Health Research Institute (ibs.GRANADA Instituto de Investigación Biosanitaria), 18014 Granada, Spain
- Correspondence: author: (L.J.M.-G.); (M.J.A.-C.); Tel.: +34-958-715-500 (ext. 108) (L.J.M.-G.); +34-958-248-945 (M.J.A.-C.); Fax: +34-958-637-071 (L.J.M.-G.)
| |
Collapse
|
20
|
Liu L, Yi X, Lu C, Pang Y, Zu X, Chen M, Guan X. Background, applications and challenges of radiogenomics in genitourinary tumor. Am J Cancer Res 2021; 11:1936-1945. [PMID: 34094662 PMCID: PMC8167692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 03/03/2021] [Indexed: 06/12/2023] Open
Abstract
Genitourinary tumors are groups of tumors with high complexity and heterogeneity. For long-term monitoring, biomarkers that can be used in detection, grading and treatment response assessment are needed. With rapid development in imaging technology and cancer genomics, radiogenomics, the combination of "radiology" and "genomics", has emerged as a powerful tool in oncology practice in recent years because imaging can provide some information that genomic test cannot as gene expression and mutation status are usually evaluated on a small portion of the tumor and are usually not powerful enough to reflect tumor heterogeneity. Radiogenomics investigates the correlations between imaging features and gene expression of a disease, especially in oncologic diseases. It aims to detect the disease's mutation status and supplement genomic analysis based on imaging analysis, providing additional findings for diagnosis, treatment decisions, evaluation of treatment response and prognosis prediction of the disease. Recent years have seen an increase in the number of studies investigating the application of radiogenomics in genitourinary tumors. Many studies have shown promising results. However, there still exist limitations and challenges. In this review, we will summarize recent applications of radiogenomics in genitourinary tumors and discuss limitiations, challenges and future directions of radiogenomics.
Collapse
Affiliation(s)
- Longfei Liu
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Xiaoping Yi
- Department of Radiology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Can Lu
- Department of Nephrology, The Second Xiangya Hospital of Central South UniversityChangsha 410000, Hunan, P. R. China
| | - Yingxian Pang
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Minfeng Chen
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Xiao Guan
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| |
Collapse
|
21
|
Magnetic Resonance Imaging Based Radiomic Models of Prostate Cancer: A Narrative Review. Cancers (Basel) 2021; 13:cancers13030552. [PMID: 33535569 PMCID: PMC7867056 DOI: 10.3390/cancers13030552] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary The increasing interest in implementing artificial intelligence in radiomic models has occurred alongside advancement in the tools used for computer-aided diagnosis. Such tools typically apply both statistical and machine learning methodologies to assess the various modalities used in medical image analysis. Specific to prostate cancer, the radiomics pipeline has multiple facets that are amenable to improvement. This review discusses the steps of a magnetic resonance imaging based radiomics pipeline. Present successes, existing opportunities for refinement, and the most pertinent pending steps leading to clinical validation are highlighted. Abstract The management of prostate cancer (PCa) is dependent on biomarkers of biological aggression. This includes an invasive biopsy to facilitate a histopathological assessment of the tumor’s grade. This review explores the technical processes of applying magnetic resonance imaging based radiomic models to the evaluation of PCa. By exploring how a deep radiomics approach further optimizes the prediction of a PCa’s grade group, it will be clear how this integration of artificial intelligence mitigates existing major technological challenges faced by a traditional radiomic model: image acquisition, small data sets, image processing, labeling/segmentation, informative features, predicting molecular features and incorporating predictive models. Other potential impacts of artificial intelligence on the personalized treatment of PCa will also be discussed. The role of deep radiomics analysis-a deep texture analysis, which extracts features from convolutional neural networks layers, will be highlighted. Existing clinical work and upcoming clinical trials will be reviewed, directing investigators to pertinent future directions in the field. For future progress to result in clinical translation, the field will likely require multi-institutional collaboration in producing prospectively populated and expertly labeled imaging libraries.
Collapse
|
22
|
Liu L, Yi X, Lu C, Qi L, Zhang Y, Li M, Xiao Q, Wang C, Zhang L, Pang Y, Wang Y, Guan X. Applications of radiomics in genitourinary tumors. Am J Cancer Res 2020; 10:2293-2308. [PMID: 32905456 PMCID: PMC7471369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 07/02/2020] [Indexed: 06/11/2023] Open
Abstract
Genitourinary tumors are heterogeneous groups of tumors with high morbidity and mortality rates. Confronted with existing problems in the management of genitourinary tumors, a personalized imaging method called radiomics shows great potential in areas including detection, grading, and treatment response assessment. Radiomics is characterized by extraction of quantitative imaging features which are not visible to the naked eye from conventional imaging modalities such as computed tomography (CT), magnetic resonance imaging (MRI) and positron emission tomography-computed tomography (PET-CT), followed by data analysis and model building. It outperforms other invasive methods in terms of non-invasiveness, low cost and high efficiency. Recently, a number of studies have evaluated the application of radiomics in patients with genitourinary tumors with promising data. The combination of radiomics and clinical/laboratory factors provides added value in many studies. Despite this, there are limitations and challenges to be overcome before a more extensive clinical application in the future. In this article, we will introduce the concept, significance and workflow of radiomics, review their current applications in patients with genitourinary tumors and discuss limitations and future directions of radiomics. It would help multidisciplinary team involved in the treatment of patients with genitourinary tumors to achieve a better understanding of the results of radiomics study toward a personalized medicine.
Collapse
Affiliation(s)
- Longfei Liu
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Xiaoping Yi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Department of Radiology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Can Lu
- Department of Nephrology, The Second Xiangya Hospital of Central South UniversityChangsha 410000, Hunan, P. R. China
| | - Lin Qi
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Youming Zhang
- Department of Radiology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Minghao Li
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Qiao Xiao
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Cikui Wang
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Liang Zhang
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Yingxian Pang
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Yong Wang
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| | - Xiao Guan
- Department of Urology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
| |
Collapse
|