1
|
Ferro A, Bottosso M, Dieci MV, Scagliori E, Miglietta F, Aldegheri V, Bonanno L, Caumo F, Guarneri V, Griguolo G, Pasello G. Clinical applications of radiomics and deep learning in breast and lung cancer: A narrative literature review on current evidence and future perspectives. Crit Rev Oncol Hematol 2024; 203:104479. [PMID: 39151838 DOI: 10.1016/j.critrevonc.2024.104479] [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: 01/10/2024] [Revised: 07/22/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024] Open
Abstract
Radiomics, analysing quantitative features from medical imaging, has rapidly become an emerging field in translational oncology. Radiomics has been investigated in several neoplastic malignancies as it might allow for a non-invasive tumour characterization and for the identification of predictive and prognostic biomarkers. Over the last few years, evidence has been accumulating regarding potential clinical applications of machine learning in many crucial moments of cancer patients' history. However, the incorporation of radiomics in clinical decision-making process is still limited by low data reproducibility and study variability. Moreover, the need for prospective validations and standardizations is emerging. In this narrative review, we summarize current evidence regarding radiomic applications in high-incidence cancers (breast and lung) for screening, diagnosis, staging, treatment choice, response, and clinical outcome evaluation. We also discuss pro and cons of the radiomic approach, suggesting possible solutions to critical issues which might invalidate radiomics studies and propose future perspectives.
Collapse
Affiliation(s)
- Alessandra Ferro
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Michele Bottosso
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Maria Vittoria Dieci
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy.
| | - Elena Scagliori
- Radiology Unit, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Federica Miglietta
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Vittoria Aldegheri
- Radiology Unit, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Laura Bonanno
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Francesca Caumo
- Unit of Breast Radiology, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy
| | - Valentina Guarneri
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Gaia Griguolo
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| | - Giulia Pasello
- Division of Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, via Gattamelata 64, Padua 35128, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, via Giustiniani 2, Padova 35128, Italy
| |
Collapse
|
2
|
Marfisi D, Giannelli M, Marzi C, Del Meglio J, Barucci A, Masturzo L, Vignali C, Mascalchi M, Traino A, Casolo G, Diciotti S, Tessa C. Test-retest repeatability of myocardial radiomic features from quantitative cardiac magnetic resonance T1 and T2 mapping. Magn Reson Imaging 2024; 113:110217. [PMID: 39067653 DOI: 10.1016/j.mri.2024.110217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/14/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Radiomics of cardiac magnetic resonance (MR) imaging has proved to be potentially useful in the study of various myocardial diseases. Therefore, assessing the repeatability degree in radiomic features measurement is of fundamental importance. The aim of this study was to assess test-retest repeatability of myocardial radiomic features extracted from quantitative T1 and T2 maps. A representative group of 24 subjects (mean age 54 ± 18 years) referred for clinical cardiac MR imaging were enrolled in the study. For each subject, T1 and T2 mapping through MOLLI and T2-prepared TrueFISP acquisition sequences, respectively, were performed at 1.5 T. Then, 98 radiomic features of different classes (shape, first-order, second-order) were extracted from a region of interest encompassing the whole left ventricle myocardium in a short axis slice. The repeatability was assessed performing different and complementary analyses: intraclass correlation coefficient (ICC) and limits of agreement (LOA) (i.e., the interval within which 95% of the percentage differences between two repeated measures are expected to lie). Radiomic features were characterized by a relatively wide range of repeatability degree in terms of both ICC and LOA. Overall, 44.9% and 38.8% of radiomic features showed ICC values > 0.75 for T1 and T2 maps, respectively, while 25.5% and 23.4% of radiomic features showed LOA between ±10%. A subset of radiomic features for T1 (Mean, Median, 10Percentile, 90Percentile, RootMeanSquared, Imc2, RunLengthNonUniformityNormalized, RunPercentage and ShortRunEmphasis) and T2 (MaximumDiameter, RunLengthNonUniformityNormalized, RunPercentage, ShortRunEmphasis) maps presented both ICC > 0.75 and LOA between ±5%. Overall, radiomic features extracted from T1 maps showed better repeatability performance than those extracted from T2 maps, with shape features characterized by better repeatability than first-order and textural features. Moreover, only a limited subset of 9 and 4 radiomic features for T1 and T2 maps, respectively, showed high repeatability degree in terms of both ICC and LOA. These results confirm the importance of assessing test-retest repeatability degree in radiomic feature estimation and might be useful for a more effective/reliable use of myocardial T1 and T2 mapping radiomics in clinical or research studies.
Collapse
Affiliation(s)
- Daniela Marfisi
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", 56126 Pisa, Italy
| | - Marco Giannelli
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", 56126 Pisa, Italy.
| | - Chiara Marzi
- Department of Statistics, Computer Science, Applications "Giuseppe Parenti", University of Florence, 50134 Florence, Italy
| | - Jacopo Del Meglio
- Unit of Cardiology, Azienda USL Toscana Nord Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy
| | - Andrea Barucci
- Institute of Applied Physics "Nello Carrara" (IFAC), Council of National Research (CNR), 50019 Sesto Fiorentino, Italy
| | - Luigi Masturzo
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", 56126 Pisa, Italy
| | - Claudio Vignali
- Unit of Radiology, Azienda USL Toscana Nord Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy
| | - Mario Mascalchi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50121 Florence, Italy; Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy
| | - Antonio Traino
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", 56126 Pisa, Italy
| | - Giancarlo Casolo
- Unit of Cardiology, Azienda USL Toscana Nord Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy
| | - Stefano Diciotti
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, 47522 Cesena, Italy
| | - Carlo Tessa
- Unit of Radiology, Azienda USL Toscana Nord Ovest, Apuane Hospital, 54100 Massa, Italy
| |
Collapse
|
3
|
Sha X, Wang C, Qi S, Yuan X, Zhang H, Yang J. The efficacy of CBCT-based radiomics techniques in differentiating between conventional and unicystic ameloblastoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2024; 138:656-665. [PMID: 39227265 DOI: 10.1016/j.oooo.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE The aim of this study was to develop a cone beam computed tomography (CBCT) radiomics-based model that differentiates between conventional and unicystic ameloblastoma (AB). METHODS In this retrospective study, CBCT images were collected from 100 patients who had ABs that were diagnosed histopathologically as conventional or unicystic AB after surgical treatment. The patients were randomly divided into training (70) and validation (30) cohorts. Radiomics features were extracted from the images, and the optimal features were incorporated into 5 models: Logistic Regression, Support Vector Machine, Linear Discriminant Analysis, Random Forest, and XGBoost for prediction of tumor type. Model performance was evaluated using the area under the curve (AUC) from receiver operating characteristic analysis, sensitivity, specificity, accuracy, calibration curves, and decision curve analysis (DCA). RESULTS The 20 optimal radiomics features were incorporated into the Logistic Regression (LR) model, which exhibited the best overall performance with AUC = 0.936 (95% confidence interval [CI] = 0.877-0.996) for the training cohort and AUC = 0.929 (95% CI = 0.832-1.000) for the validation cohort. The nomogram combined the clinical features and the radiomics signature and resulted in the best predictive performance. CONCLUSIONS The LR model demonstrated the ability of radiomics and the nomogram to distinguish between the 2 types of AB and may have the potential to replace biopsies under noninvasive conditions.
Collapse
Affiliation(s)
- Xiaoyan Sha
- Department of Oral and Maxillofacial Radiology, School of Stomatology, Capital Medical University, Beijing, China
| | - Chao Wang
- Department of Clinical Research, SinoUnion Healthcare Inc., Beijing, China
| | - Senrong Qi
- Department of Oral and Maxillofacial Radiology, School of Stomatology, Capital Medical University, Beijing, China
| | - Xiaohong Yuan
- Department of Oral and Maxillofacial Pathology, School of Stomatology, Capital Medical University, Beijing, China
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Xia S, Zheng Y, Hua Q, Wen J, Luo X, Yan J, Bai B, Dong Y, Zhou J. Super-resolution ultrasound and microvasculomics: a consensus statement. Eur Radiol 2024; 34:7503-7513. [PMID: 38811389 DOI: 10.1007/s00330-024-10796-3] [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: 02/26/2024] [Revised: 02/26/2024] [Accepted: 03/27/2024] [Indexed: 05/31/2024]
Abstract
This is a summary of a consensus statement on the introduction of "Ultrasound microvasculomics" produced by The Chinese Artificial Intelligence Alliance for Thyroid and Breast Ultrasound. The evaluation of microvessels is a very important part for the assessment of diseases. Super-resolution ultrasound (SRUS) microvascular imaging surpasses traditional ultrasound imaging in the morphological and functional analysis of microcirculation. SRUS microvascular imaging relies on contrast microbubbles to gain sensitivity to microvessels and improves the spatial resolution of ultrasound blood flow imaging for a more detailed depiction of vascular structures and hemodynamics. This method has been applied in preclinical animal models and pilot clinical studies, involving areas including neurology, oncology, nephrology, and cardiology. However, the current quantitative parameters of SRUS images are not enough for precise evaluation of microvessels. Therefore, by employing omics methods, more quantification indicators can be obtained, enabling a more precise and personalized assessment of microvascular status. Ultrasound microvasculomics - a high-throughput extraction of image features from SRUS images - is one novel approach that holds great promise but needs further validation in both bench and clinical settings. CLINICAL RELEVANCE STATEMENT: Super-resolution Ultrasound (SRUS) blood flow imaging improves spatial resolution. Ultrasound microvasculomics is possible to acquire high-throughput information of features from SRUS images. It provides more precise and abundant micro-blood flow information in clinical medicine. KEY POINTS: This consensus statement reviews the development and application of super-resolution ultrasound (SRUS). The shortcomings of the current quantification indicators of SRUS and strengths of the omics methodology are addressed. "Ultrasound microvasculomics" is introduced for a high-throughput extraction of image features from SRUS images.
Collapse
Affiliation(s)
- ShuJun Xia
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, 200025, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China
| | - YuHang Zheng
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, 200025, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China
| | - Qing Hua
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, 200025, Shanghai, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China
| | - Jing Wen
- Department of Medical Ultrasound, Affiliated Hospital of Guizhou Medical University, 550001, Guiyang, China
| | - XiaoMao Luo
- Department of Medical Ultrasound, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, 650118, Kunming, China
| | - JiPing Yan
- Department of Ultrasound, Shanxi Provincial People's Hospital, 31th Shuangta Street, 030012, Taiyuan, China
| | - BaoYan Bai
- Department of Ultrasound, Affiliated Hospital of Yan 'an University, 43 North Street, Baota District, 716000, Yan'an, China
| | - YiJie Dong
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, 200025, Shanghai, China.
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China.
| | - JianQiao Zhou
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, 200025, Shanghai, China.
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China.
| |
Collapse
|
5
|
O'Connor JPB, Tessyman V, Little RA, Babur M, Forster D, Latif A, Cheung S, Lipowska-Bhalla G, Higgins GS, Asselin MC, Parker GJM, Williams KJ. Combined Oxygen-Enhanced MRI and Perfusion Imaging Detect Hypoxia Modification from Banoxantrone and Atovaquone and Track Their Differential Mechanisms of Action. CANCER RESEARCH COMMUNICATIONS 2024; 4:2565-2574. [PMID: 39240065 PMCID: PMC11443776 DOI: 10.1158/2767-9764.crc-24-0315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/26/2024] [Accepted: 09/04/2024] [Indexed: 09/07/2024]
Abstract
Oxygen-enhanced MRI (OE-MRI) has shown promise for quantifying and spatially mapping tumor hypoxia, either alone or in combination with perfusion imaging. Previous studies have validated the technique in mouse models and in patients with cancer. Here, we report the first evidence that OE-MRI can track change in tumor oxygenation induced by two drugs designed to modify hypoxia. Mechanism of action of banoxantrone and atovaquone were confirmed using in vitro experiments. Next, in vivo OE-MRI studies were performed in Calu6 and U87 xenograft tumor models, alongside fluorine-18-fluoroazomycin arabinoside PET and immunohistochemistry assays of hypoxia. Neither drug altered tumor size. Banoxantrone reduced OE-MRI hypoxic fraction in Calu6 tumors by 52.5% ± 12.0% (P = 0.008) and in U87 tumors by 29.0% ± 15.8% (P = 0.004) after 3 days treatment. Atovaquone reduced OE-MRI hypoxic fraction in Calu6 tumors by 53.4% ± 15.3% (P = 0.002) after 7 days therapy. PET and immunohistochemistry provided independent validation of the MRI findings. Finally, combined OE-MRI and perfusion imaging showed that hypoxic tissue was converted into necrotic tissue when treated by the hypoxia-activated cytotoxic prodrug banoxantrone, whereas hypoxic tissue became normoxic when treated by atovaquone, an inhibitor of mitochondrial complex III of the electron transport chain. OE-MRI detected and quantified hypoxia reduction induced by two hypoxia-modifying therapies and could distinguish between their differential mechanisms of action. These data support clinical translation of OE-MRI biomarkers in clinical trials of hypoxia-modifying agents to identify patients demonstrating biological response and to optimize treatment timing and scheduling. Significance: For the first time, we show that hypoxic fraction measured by oxygen-enhanced MRI (OE-MRI) detected changes in tumor oxygenation induced by two drugs designed specifically to modify hypoxia. Furthermore, when combined with perfusion imaging, OE-MRI hypoxic volume distinguished the two drug mechanisms of action. This imaging technology has potential to facilitate drug development, enrich clinical trial design, and accelerate clinical translation of novel therapeutics into clinical use.
Collapse
Affiliation(s)
- James P B O'Connor
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Radiology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Victoria Tessyman
- Division of Pharmacy and Optometry, University of Manchester, Manchester, United Kingdom
| | - Ross A Little
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Muhammad Babur
- Division of Pharmacy and Optometry, University of Manchester, Manchester, United Kingdom
| | - Duncan Forster
- Cancer Research UK Manchester Centre, University of Manchester, Manchester, United Kingdom
| | - Ayşe Latif
- Division of Pharmacy and Optometry, University of Manchester, Manchester, United Kingdom
| | - Susan Cheung
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Geoff S Higgins
- CRUK/MRC Oxford Institute for Radiation Oncology and Biology, University of Oxford, Oxford, United Kingdom
| | - Marie-Claude Asselin
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Geoff J M Parker
- Bioxydyn Ltd., Manchester, United Kingdom
- Centre for Medical Image Computing, University College London, London, United Kingdom
| | - Kaye J Williams
- Division of Pharmacy and Optometry, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
6
|
Wang B, Liu J, Xie J, Zhang X, Wang Z, Cao Z, Wen D, Wan Hasan WZ, Harun Ramli HR, Dong X. Systematic review and meta-analysis of the prognostic value of 18F-Fluorodeoxyglucose ( 18F-FDG) positron emission tomography (PET) and/or computed tomography (CT)-based radiomics in head and neck cancer. Clin Radiol 2024; 79:757-772. [PMID: 38944542 DOI: 10.1016/j.crad.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 07/01/2024]
Abstract
AIM Radiomics involves the extraction of quantitative data from medical images to facilitate the diagnosis, prognosis, and staging of tumors. This study provides a comprehensive overview of the efficacy of radiomics in prognostic applications for head and neck cancer (HNC) in recent years. It undertakes a systematic review of prognostic models specific to HNC and conducts a meta-analysis to evaluate their predictive performance. MATERIALS AND METHODS This study adhered rigorously to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for literature searches. The literature databases, including PubMed, Embase, Cochrane, and Scopus were systematically searched individually. The methodological quality of the incorporated studies underwent assessment utilizing the radiomics quality score (RQS) tool. A random-effects meta-analysis employing the Harrell concordance index (C-index) was conducted to evaluate the performance of all radiomics models. RESULTS Among the 388 studies retrieved, 24 studies encompassing a total of 6,978 cases were incorporated into the systematic review. Furthermore, eight studies, focusing on overall survival as an endpoint, were included in the meta-analysis. The meta-analysis revealed that the estimated random effect of the C-index for all studies utilizing radiomics alone was 0.77 (0.71-0.82), with a substantial degree of heterogeneity indicated by an I2 of 80.17%. CONCLUSIONS Based on this review, prognostic modeling utilizing radiomics has demonstrated enhanced efficacy for head and neck cancers; however, there remains room for improvement in this approach. In the future, advancements are warranted in the integration of clinical parameters and multimodal features, balancing multicenter data, as well as in feature screening and model construction within this field.
Collapse
Affiliation(s)
- B Wang
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia; Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | - J Liu
- Department of Nursing, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia; Department of Nursing, Chengde Central Hospital, Chengde city, Hebei Province, China.
| | - J Xie
- Department of Automatic, Tsinghua University, Beijing, China.
| | - X Zhang
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | - Z Wang
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | - Z Cao
- Department of Radiology, The Affiliated Hospital of Chengde Medical University, Chengde City, Hebei Province, China.
| | - D Wen
- Institute of Artificial Intelligence, University of Science and Technology Beijing, Beijing, China.
| | - W Z Wan Hasan
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia.
| | - H R Harun Ramli
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia.
| | - X Dong
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China; Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde City, Hebei, China; Hebei International Research Center of Medical Engineering, Chengde Medical University, Hebei, China.
| |
Collapse
|
7
|
Jia X, Carter BW, Duffton A, Harris E, Hobbs R, Li H. Advancing the Collaboration Between Imaging and Radiation Oncology. Semin Radiat Oncol 2024; 34:402-417. [PMID: 39271275 PMCID: PMC11407744 DOI: 10.1016/j.semradonc.2024.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
The fusion of cutting-edge imaging technologies with radiation therapy (RT) has catalyzed transformative breakthroughs in cancer treatment in recent decades. It is critical for us to review our achievements and preview into the next phase for future synergy between imaging and RT. This paper serves as a review and preview for fostering collaboration between these two domains in the forthcoming decade. Firstly, it delineates ten prospective directions ranging from technological innovations to leveraging imaging data in RT planning, execution, and preclinical research. Secondly, it presents major directions for infrastructure and team development in facilitating interdisciplinary synergy and clinical translation. We envision a future where seamless integration of imaging technologies into RT will not only meet the demands of RT but also unlock novel functionalities, enhancing accuracy, efficiency, safety, and ultimately, the standard of care for patients worldwide.
Collapse
Affiliation(s)
- Xun Jia
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD..
| | - Brett W Carter
- Department of Thoracic Imaging, Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Aileen Duffton
- Beatson West of Scotland Cancer Centre, Glasgow, UK.; Institute of Cancer Science, University of Glasgow, UK
| | - Emma Harris
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Robert Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD
| | - Heng Li
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
8
|
Deasy JO. Data Science Opportunities To Improve Radiotherapy Planning and Clinical Decision Making. Semin Radiat Oncol 2024; 34:379-394. [PMID: 39271273 DOI: 10.1016/j.semradonc.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Radiotherapy aims to achieve a high tumor control probability while minimizing damage to normal tissues. Personalizing radiotherapy treatments for individual patients, therefore, depends on integrating physical treatment planning with predictive models of tumor control and normal tissue complications. Predictive models could be improved using a wide range of rich data sources, including tumor and normal tissue genomics, radiomics, and dosiomics. Deep learning will drive improvements in classifying normal tissue tolerance, predicting intra-treatment tumor changes, tracking accumulated dose distributions, and quantifying the tumor response to radiotherapy based on imaging. Mechanistic patient-specific computer simulations ('digital twins') could also be used to guide adaptive radiotherapy. Overall, we are entering an era where improved modeling methods will allow the use of newly available data sources to better guide radiotherapy treatments.
Collapse
Affiliation(s)
- Joseph O Deasy
- Department of Medical Physics, Attending Physicist, Chief, Service for Predictive Informatics, Chair, Memorial Sloan Kettering Cancer Center, New York, NY..
| |
Collapse
|
9
|
Cao Y, Sutera P, Silva Mendes W, Yousefi B, Hrinivich T, Deek M, Phillips R, Song D, Kiess A, Cem Guler O, Torun N, Reyhan M, Sawant A, Marchionni L, Simone NL, Tran P, Onal C, Ren L. Machine learning predicts conventional imaging metastasis-free survival (MFS) for oligometastatic castration-sensitive prostate cancer (omCSPC) using prostate-specific membrane antigen (PSMA) PET radiomics. Radiother Oncol 2024; 199:110443. [PMID: 39094629 PMCID: PMC11405100 DOI: 10.1016/j.radonc.2024.110443] [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: 05/20/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE This study investigated imaging biomarkers derived from PSMA-PET acquired pre- and post-metastasis-directed therapy (MDT) to predict 2-year metastasis-free survival (MFS), which provides valuable early response assessment to improve patient outcomes. MATERIALS/METHODS An international cohort of 117 oligometastatic castration-sensitive prostate cancer (omCSPC) patients, comprising 34 from John Hopkins Hospital (JHH) and 83 from Baskent University (BU), were treated with stereotactic ablative radiation therapy (SABR) MDT with both pre- and post-MDT PSMA-PET/CT scans acquired. PET radiomic features were analyzed from a CT-PET fusion defined gross tumor volume ((GTV) or zone 1), and a 5 mm expansion ring area outside the GTV (zone 2). A total of 1748 PET radiomic features were extracted from these zones. The six most significant features selected using the Chi2 method, along with five clinical parameters (age, Gleason score, number of total lesions, untreated lesions, and pre-MDT prostate-specific antigen (PSA)) were extracted as inputs to the models. Various machine learning models, including Random Forest, Decision Tree, Support Vector Machine, and Naïve Bayesian, were employed for 2-year MFS prediction and tested using leave-one-out and cross-institution validation. RESULTS Six radiomic features, including Total Energy, Entropy, and Standard Deviation from pre-PSMA-PET zone 1, Total Energy and Contrast from post-PSMA-PET zone 1, and Entropy from pre-PSMA-PET zone 2, along with five clinical parameters were selected for predicting 2-year MFS. In a leave-one-out test with all the patients, random forest achieved an accuracy of 80 % and an AUC of 0.82 in predicting 2-year MFS. In cross-institution validation, the model correctly predicted 2-year MFS events with an accuracy of 75 % and an AUC of 0.77 for patients from JHH, and an accuracy of 78 % and an AUC of 0.80 for BU patients, respectively. CONCLUSION Our study demonstrated the promise of using pre- and post-MDT PSMA-PET-based imaging biomarkers for MFS prediction for omCSPC patients.
Collapse
Affiliation(s)
- Yufeng Cao
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Philip Sutera
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William Silva Mendes
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bardia Yousefi
- Fischell Department of Bioengineering, University of Maryland School of Medicine, College Park, MD, USA
| | - Tom Hrinivich
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Ryan Phillips
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Danny Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ana Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ozan Cem Guler
- Baskent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Radiation Oncology, Adana, Turkey
| | - Nese Torun
- Baskent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Nuclear Medicine, Ankara, Turkey
| | - Mehmet Reyhan
- Baskent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Nuclear Medicine, Ankara, Turkey
| | - Amit Sawant
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Nicole L Simone
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Phuoc Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Cem Onal
- Baskent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Radiation Oncology, Adana, Turkey; Baskent University Faculty of Medicine, Department of Radiation Oncology, Ankara, Turkey.
| | - Lei Ren
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
10
|
Um‐e‐Kalsoom, Wang S, Qu J, Liu L. Innovative optical imaging strategies for monitoring immunotherapy in the tumor microenvironments. Cancer Med 2024; 13:e70155. [PMID: 39387259 PMCID: PMC11465031 DOI: 10.1002/cam4.70155] [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: 01/13/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND The tumor microenvironment (TME) plays a critical role in cancer progression and response to immunotherapy. Immunotherapy targeting the immune system has emerged as a promising treatment modality, but challenges in understanding the TME limit its efficacy. Optical imaging strategies offer noninvasive, real-time insights into the interactions between immune cells and the TME. OBJECTIVE This review assesses the progress of optical imaging technologies in monitoring immunotherapy within the TME and explores their potential applications in clinical trials and personalized cancer treatment. METHODS This is a comprehensive literature review based on the advances in optical imaging modalities including fluorescence imaging (FLI), bioluminescence imaging (BLI), and photoacoustic imaging (PAI). These modalities were analyzed for their capacity to provide high-resolution, real-time imaging of immune cell dynamics, tumor vasculature, and other critical components of the TME. RESULTS Optical imaging techniques have shown significant potential in tracking immune cell infiltration, assessing immune checkpoint inhibitors, and visualizing drug delivery within the TME. Technologies like FLI and BLI are pivotal in tracking immune responses in preclinical models, while PAI provides functional imaging with deeper tissue penetration. The integration of these modalities with immunotherapy holds promise for improving treatment monitoring and outcomes. CONCLUSION Optical imaging is a powerful tool for understanding the complexities of the TME and optimizing immunotherapy. Further advancements in imaging technologies, combined with nanomaterial-based approaches, could pave the way for enhanced diagnostic accuracy and therapeutic efficacy in cancer treatment.
Collapse
Affiliation(s)
- Um‐e‐Kalsoom
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| |
Collapse
|
11
|
Friesen E, Hari K, Sheft M, Thiessen JD, Martin M. Magnetic resonance metrics for identification of cuprizone-induced demyelination in the mouse model of neurodegeneration: a review. MAGMA (NEW YORK, N.Y.) 2024; 37:765-790. [PMID: 38635150 DOI: 10.1007/s10334-024-01160-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/17/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024]
Abstract
Neurodegenerative disorders, including Multiple Sclerosis (MS), are heterogenous disorders which affect the myelin sheath of the central nervous system (CNS). Magnetic Resonance Imaging (MRI) provides a non-invasive method for studying, diagnosing, and monitoring disease progression. As an emerging research area, many studies have attempted to connect MR metrics to underlying pathophysiological presentations of heterogenous neurodegeneration. Most commonly, small animal models are used, including Experimental Autoimmune Encephalomyelitis (EAE), Theiler's Murine Encephalomyelitis (TMEV), and toxin models including cuprizone (CPZ), lysolecithin, and ethidium bromide (EtBr). A contrast and comparison of these models is presented, with focus on the cuprizone model, followed by a review of literature studying neurodegeneration using MRI and the cuprizone model. Conventional MRI methods including T1 Weighted (T1W) and T2 Weighted (T2W) Imaging are mentioned. Quantitative MRI methods which are sensitive to diffusion, magnetization transfer, susceptibility, relaxation, and chemical composition are discussed in relation to studying the CPZ model. Overall, additional studies are needed to improve both the sensitivity and specificity of MRI metrics for underlying pathophysiology of neurodegeneration and the relationships in attempts to clear the clinico-radiological paradox. We therefore propose a multiparametric approach for the investigation of MR metrics for underlying pathophysiology.
Collapse
Affiliation(s)
- Emma Friesen
- Chemistry, University of Winnipeg, Winnipeg, Canada.
| | - Kamya Hari
- Physics, University of Winnipeg, Winnipeg, Canada
- Electronics and Communication Engineering, SSN College of Engineering, Chennai, India
| | - Maxina Sheft
- Physics, University of Winnipeg, Winnipeg, Canada
- Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, USA
| | - Jonathan D Thiessen
- Imaging Program, Lawson Health Research Institute, London, Canada
- Medical Biophysics, Western University, London, Canada
- Medical Imaging, Western University, London, Canada
| | | |
Collapse
|
12
|
Xie SH, Zhang WF, Wu Y, Tang ZL, Yang LT, Xue YJ, Lin JB, Kang MQ. Application of predictive model based on CT radiomics and machine learning in diagnosis for occult locally advanced esophageal squamous cell carcinoma before treatment: A two-center study. Transl Oncol 2024; 47:102050. [PMID: 38981245 PMCID: PMC11292555 DOI: 10.1016/j.tranon.2024.102050] [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: 12/09/2023] [Revised: 02/24/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024] Open
Abstract
PURPOSE Development and validation of a radiomics model for predicting occult locally advanced esophageal squamous cell carcinoma (LA-ESCC) on computed tomography (CT) radiomic features before implementation of treatment. METHODS The study retrospectively collected 574 patients with esophageal squamous cell carcinoma (ESCC) from two medical centers, which were divided into three cohorts for training, internal and external validation. After delineating volume of interest (VOI), radiomics features were extracted and subjected to feature selection using three robust methods. Subsequently, 10 machine learning models were constructed, among which the optimal model was utilized to establish a radiomics signature. Furthermore, a predictive nomogram incorporating both clinical and radiomics signatures was developed. The performance of these models was evaluated through receiver operating characteristic curves, calibration curves, decision curve analysis as well as measures including accuracy, sensitivity, and specificity. RESULTS A total of 19 radiomics features were selected. The multilayer perceptron (MLP), which was found to be optimal, achieved an AUC of 0.919, 0.864 and 0.882 in the training, internal and external validation cohorts, respectively. Similarly, MLP showed good accuracy in distinguish occult LA-ESCC in subgroup of cT1-2N0M0 diagnosed by clinicians with 0.803 and 0.789 in two validation cohorts respectively. By incorporating the radiomics signature with clinical signature, a predictive nomogram demonstrated superior prediction performance with an AUC of 0.877 and accuracy of 0.85 in external validation cohort. CONCLUSION The radiomics and machine learning model can offers improved accuracy in prediction of occult LA-ESCC, providing valuable assistance to clinicians when choosing treatment plans.
Collapse
Affiliation(s)
- Shu-Han Xie
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Wan-Fei Zhang
- Department of Thoracic Surgery, Quanzhou First Hospital, Quanzhou, Fujian, China; Department of Thoracic Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Yue Wu
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; The School of Medical Imaging, Fujian Medical University, Fuzhou, Fujian, China
| | - Zi-Lu Tang
- Department of Thoracic Surgery, Quanzhou First Hospital, Quanzhou, Fujian, China; Department of Thoracic Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian, China
| | - Li-Tao Yang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Department of Thoracic Surgery, Baoji Traditional Chinese Medicine Hospital, Baoji, Shaanxi, China
| | - Yun-Jing Xue
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jiang-Bo Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Ming-Qiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China; Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
13
|
Magnuska ZA, Roy R, Palmowski M, Kohlen M, Winkler BS, Pfeil T, Boor P, Schulz V, Krauss K, Stickeler E, Kiessling F. Combining Radiomics and Autoencoders to Distinguish Benign and Malignant Breast Tumors on US Images. Radiology 2024; 312:e232554. [PMID: 39254446 DOI: 10.1148/radiol.232554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Background US is clinically established for breast imaging, but its diagnostic performance depends on operator experience. Computer-assisted (real-time) image analysis may help in overcoming this limitation. Purpose To develop precise real-time-capable US-based breast tumor categorization by combining classic radiomics and autoencoder-based features from automatically localized lesions. Materials and Methods A total of 1619 B-mode US images of breast tumors were retrospectively analyzed between April 2018 and January 2024. nnU-Net was trained for lesion segmentation. Features were extracted from tumor segments, bounding boxes, and whole images using either classic radiomics, autoencoder, or both. Feature selection was performed to generate radiomics signatures, which were used to train machine learning algorithms for tumor categorization. Models were evaluated using the area under the receiver operating characteristic curve (AUC), sensitivity, and specificity and were statistically compared with histopathologically or follow-up-confirmed diagnosis. Results The model was developed on 1191 (mean age, 61 years ± 14 [SD]) female patients and externally validated on 50 (mean age, 55 years ± 15]). The development data set was divided into two parts: testing and training lesion segmentation (419 and 179 examinations) and lesion categorization (503 and 90 examinations). nnU-Net demonstrated precision and reproducibility in lesion segmentation in test set of data set 1 (median Dice score [DS]: 0.90 [IQR, 0.84-0.93]; P = .01) and data set 2 (median DS: 0.89 [IQR, 0.80-0.92]; P = .001). The best model, trained with 23 mixed features from tumor bounding boxes, achieved an AUC of 0.90 (95% CI: 0.83, 0.97), sensitivity of 81% (46 of 57; 95% CI: 70, 91), and specificity of 87% (39 of 45; 95% CI: 77, 87). No evidence of difference was found between model and human readers (AUC = 0.90 [95% CI: 0.83, 0.97] vs 0.83 [95% CI: 0.76, 0.90]; P = .55 and 0.90 vs 0.82 [95% CI: 0.75, 0.90]; P = .45) in tumor classification or between model and histopathologically or follow-up-confirmed diagnosis (AUC = 0.90 [95% CI: 0.83, 0.97] vs 1.00 [95% CI: 1.00,1.00]; P = .10). Conclusion Precise real-time US-based breast tumor categorization was developed by mixing classic radiomics and autoencoder-based features from tumor bounding boxes. ClinicalTrials.gov identifier: NCT04976257 Published under a CC BY 4.0 license. Supplemental material is available for this article. See also the editorial by Bahl in this issue.
Collapse
Affiliation(s)
- Zuzanna Anna Magnuska
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Rijo Roy
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Moritz Palmowski
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Matthias Kohlen
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Brigitte Sophia Winkler
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Tatjana Pfeil
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Peter Boor
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Volkmar Schulz
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Katja Krauss
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Elmar Stickeler
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| | - Fabian Kiessling
- From the Institute for Experimental Molecular Imaging (Z.A.M., R.R., M.P., V.S., F.K.), Institute of Pathology (P.B.), and Department of Obstetrics and Gynecology (M.K., B.S.W., T.P., K.K., E.S.), University Clinic Aachen, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Physics Institute III B, RWTH Aachen University, Aachen, Germany (V.S.); Comprehensive Diagnostic Center Aachen, Uniklinik RWTH Aachen, Aachen, Germany (P.B., V.S., E.S., F.K.); and Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany (P.B., V.S., F.K.)
| |
Collapse
|
14
|
Capasso E, Casella C, Marisei M, Tortora M, Briganti F, Di Lorenzo P. Imaging biobanks: operational limits, medical-legal and ethical reflections. Front Digit Health 2024; 6:1408619. [PMID: 39268200 PMCID: PMC11391398 DOI: 10.3389/fdgth.2024.1408619] [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: 03/28/2024] [Accepted: 08/05/2024] [Indexed: 09/15/2024] Open
Abstract
The extraordinary growth of health technologies has determined an increasing interest in biobanks that represent a unique wealth for research, experimentation, and validation of new therapies. "Human" biobanks are repositories of various types of human biological samples. Through years the paradigm has shifted from spontaneous collections of biological material all over the world to institutional, organized, and well-structured forms. Imaging biobanks represent a novel field and are defined by European Society of Radiology as: "organized databases of medical images, and associated imaging biomarkers shared among multiple researchers, linked to other biorepositories". Modern radiology and nuclear medicine can provide multiple imaging biomarkers, that express the phenotype related to certain diseases, especially in oncology. Imaging biobanks, not a mere catalogue of bioimages associated to clinical data, involve advanced computer technologies to implement the emergent field of radiomics and radiogenomics. Since Europe hosts most of the biobanks, juridical and ethical framework, with a specific referral to Italy, is analyzed. Linking imaging biobanks to traditional ones appears to be a crucial step that needs to be driven by medical imaging community under clear juridical and ethical guidelines.
Collapse
Affiliation(s)
- Emanuele Capasso
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Claudia Casella
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Mariagrazia Marisei
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Mario Tortora
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Francesco Briganti
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Pierpaolo Di Lorenzo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
15
|
Teng X, Wang Y, Nicol AJ, Ching JCF, Wong EKY, Lam KTC, Zhang J, Lee SWY, Cai J. Enhancing the Clinical Utility of Radiomics: Addressing the Challenges of Repeatability and Reproducibility in CT and MRI. Diagnostics (Basel) 2024; 14:1835. [PMID: 39202322 PMCID: PMC11353986 DOI: 10.3390/diagnostics14161835] [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: 06/26/2024] [Revised: 08/03/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Radiomics, which integrates the comprehensive characterization of imaging phenotypes with machine learning algorithms, is increasingly recognized for its potential in the diagnosis and prognosis of oncological conditions. However, the repeatability and reproducibility of radiomic features are critical challenges that hinder their widespread clinical adoption. This review aims to address the paucity of discussion regarding the factors that influence the reproducibility and repeatability of radiomic features and their subsequent impact on the application of radiomic models. We provide a synthesis of the literature on the repeatability and reproducibility of CT/MR-based radiomic features, examining sources of variation, the number of reproducible features, and the availability of individual feature repeatability indices. We differentiate sources of variation into random effects, which are challenging to control but can be quantified through simulation methods such as perturbation, and biases, which arise from scanner variability and inter-reader differences and can significantly affect the generalizability of radiomic model performance in diverse settings. Four suggestions for repeatability and reproducibility studies are suggested: (1) detailed reporting of variation sources, (2) transparent disclosure of calculation parameters, (3) careful selection of suitable reliability indices, and (4) comprehensive reporting of reliability metrics. This review underscores the importance of random effects in feature selection and harmonizing biases between development and clinical application settings to facilitate the successful translation of radiomic models from research to clinical practice.
Collapse
Affiliation(s)
- Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Yongqiang Wang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Alexander James Nicol
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Jerry Chi Fung Ching
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Edwin Ka Yiu Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Kenneth Tsz Chun Lam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Shara Wee-Yee Lee
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong SAR, China; (X.T.); (Y.W.); (A.J.N.); (J.C.F.C.); (E.K.Y.W.); (K.T.C.L.); (J.Z.)
- Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
16
|
Tian S, Zheng J, Ji X, Zhou F, He Z. Construction of a DNA walker nanomachine aptasensor for simultaneous detection of dual-cancer biomarkers. Analyst 2024. [PMID: 39119745 DOI: 10.1039/d4an00865k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
While it is recognized that early diagnosis of cancer-related biomarkers can become an effective avenue for timely treatment and successfully improve patient survival, it remains challenging to get accurate inspection results. Currently, most reported cancer biomarker sensing methods are focused on the quantitative detection of a single type of biomarker, which makes accurate medical diagnostics difficult. In this work, we constructed a DNA walker nanomachine aptasensor based on gold nanoparticles for the simultaneous sensing of dual cancer biomarkers. The aptamers, labelled with a fluorophore, hybridized with complementary strands on the gold nanoparticle surface, serve as a walking track. Target analytes bind to their specific aptamers, leading to the dissociation of the unstable double-strand spherical nucleic acid. Exonuclease I (Exo I) selectively digested the aptamers bound with the target analytes, then the released targets go back to the next apamers on the gold nanopareticles surface for walking. The use of spherical nucleic acid probes improved the sensitivity of analyte detection. Exo I provided a driving power for target recycling and considerably improved the sensitivity of the aptasensor as well. The DNA walker nanomachine aptasensor was successfully applied for the detection of carcinoembryonic antigen (CEA) in the range of 0.167 to 3.34 ng mL-1, and mucin-1 (MUC-1) in the same range. Moreover, we used the two aptamers to construct the DNA walker nanomachine and achieved the simultaneous detection of CEA and MUC-1, thus having great potential for biomolecular logic gate construction and early disease diagnosis.
Collapse
Affiliation(s)
- Songbai Tian
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Province Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China.
- School of Basic Medical Sciences, Hubei University of Medicine, 442000 Shiyan, China
| | - Jiao Zheng
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Province Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China.
- College of Chemistry and Molecular Sciences, Wuhan University, 430072, P. R. China
| | - Xinghu Ji
- College of Chemistry and Molecular Sciences, Wuhan University, 430072, P. R. China
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Province Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China.
| | - Zhike He
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Province Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China.
- College of Chemistry and Molecular Sciences, Wuhan University, 430072, P. R. China
| |
Collapse
|
17
|
Sridharan N, Salem A, Little RA, Tariq M, Cheung S, Dubec MJ, Faivre-Finn C, Parker GJM, Porta N, O'Connor JPB. Measuring repeatability of dynamic contrast-enhanced MRI biomarkers improves evaluation of biological response to radiotherapy in lung cancer. Eur Radiol 2024:10.1007/s00330-024-10970-7. [PMID: 39122855 DOI: 10.1007/s00330-024-10970-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/09/2024] [Accepted: 07/01/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVES To measure dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) biomarker repeatability in patients with non-small cell lung cancer (NSCLC). To use these statistics to identify which individual target lesions show early biological response. MATERIALS AND METHODS A single-centre, prospective DCE-MRI study was performed between September 2015 and April 2017. Patients with NSCLC were scanned before standard-of-care radiotherapy to evaluate biomarker repeatability and two weeks into therapy to evaluate biological response. Volume transfer constant (Ktrans), extravascular extracellular space volume fraction (ve) and plasma volume fraction (vp) were measured at each timepoint along with tumour volume. Repeatability was assessed using a within-subject coefficient of variation (wCV) and repeatability coefficient (RC). Cohort treatment effects on biomarkers were estimated using mixed-effects models. RC limits of agreement revealed which individual target lesions changed beyond that expected with biomarker daily variation. RESULTS Fourteen patients (mean age, 67 years +/- 12, 8 men) had 22 evaluable lesions (12 primary tumours, 8 nodal metastases, 2 distant metastases). The wCV (in 8/14 patients) was between 9.16% to 17.02% for all biomarkers except for vp, which was 42.44%. Cohort-level changes were significant for Ktrans and ve (p < 0.001) and tumour volume (p = 0.002). Ktrans and tumour volume consistently showed the greatest number of individual lesions showing biological response. In distinction, no individual lesions had a real change in ve despite the cohort-level change. CONCLUSION Identifying individual early biological responders provided additional information to that derived from conventional cohort cohort-level statistics, helping to prioritise which parameters would be best taken forward into future studies. CLINICAL RELEVANCE STATEMENT Dynamic contrast-enhanced magnetic resonance imaging biomarkers Ktrans and tumour volume are repeatable and detect early treatment-induced changes at both cohort and individual lesion levels, supporting their use in further evaluation of radiotherapy and targeted therapeutics. KEY POINTS Few literature studies report quantitative imaging biomarker precision, by measuring repeatability or reproducibility. Several DCE-MRI biomarkers of lung cancer tumour microenvironment were highly repeatable. Repeatability coefficient measurements enabled lesion-specific evaluation of early biological response to therapy, improving conventional assessment.
Collapse
Affiliation(s)
- Nivetha Sridharan
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK.
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK.
| | - Ahmed Salem
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Ross A Little
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Maira Tariq
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Susan Cheung
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Michael J Dubec
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Geoffrey J M Parker
- Bioxydyn Ltd, Manchester, UK
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Nuria Porta
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - James P B O'Connor
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK.
- Division of Cancer Sciences, University of Manchester, Manchester, UK.
- Radiology Department, The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
18
|
Parrella G, Annunziata S, Morelli L, Molinelli S, Magro G, Ciocca M, Riva G, Ciccone LP, Iannalfi A, Paganelli C, Orlandi E, Baroni G. A dosiomics approach to treatment outcome modeling in carbon ion radiotherapy for skull base chordomas. Phys Med 2024; 124:103421. [PMID: 38968695 DOI: 10.1016/j.ejmp.2024.103421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/23/2024] [Accepted: 06/29/2024] [Indexed: 07/07/2024] Open
Abstract
PURPOSE To investigate the role of dosiomics features extracted from physical dose (DPHYS), RBE-weighted dose (DRBE) and dose-averaged Linear Energy Transfer (LETd), to predict the risk of local recurrence (LR) in skull base chordoma (SBC) treated with Carbon Ion Radiotherapy (CIRT). Thus, define and evaluate dosiomics-driven tumor control probability (TCP) models. MATERIALS AND METHODS 54 SBC patients were retrospectively selected for this study. A regularized Cox proportional hazard model (r-Cox) and Survival Support Vector Machine (s-SVM) were tuned within a repeated Cross Validation (CV) and patients were stratified in low/high risk of LR. Models' performance was evaluated through Harrell's concordance statistic (C-index), and survival was represented through Kaplan-Meier (KM) curves. A multivariable logistic regression was fit to the selected feature sets to generate a dosiomics-driven TCP model for each map. These were compared to a reference model built with clinical parameters in terms of f-score and accuracy. RESULTS The LETd maps reached a test C-index of 0.750 and 0.786 with r-Cox and s-SVM, and significantly separated KM curves. DPHYS maps and clinical parameters showed promising CV outcomes with C-index above 0.8, despite a poorer performance on the test set and patients stratification. The LETd-based TCP showed a significatively higher f-score (0.67[0.52-0.70], median[IQR]) compared to the clinical model (0.4[0.32-0.63], p < 0.025), while DPHYS achieved a significatively higher accuracy (DPHYS: 0.73[0.65-0.79], Clinical: 0.6 [0.52-0.72]). CONCLUSION This analysis supports the role of LETd as relevant source of prognostic factors for LR in SBC treated with CIRT. This is reflected in the TCP modeling, where LETd and DPHYS showed an improved performance with respect to clinical models.
Collapse
Affiliation(s)
- Giovanni Parrella
- Politecnico di Milano, Department of Electronics, Information and Bioengineering, Milano, Italy.
| | - Simone Annunziata
- Politecnico di Milano, Department of Electronics, Information and Bioengineering, Milano, Italy
| | - Letizia Morelli
- Politecnico di Milano, Department of Electronics, Information and Bioengineering, Milano, Italy
| | - Silvia Molinelli
- Centro Nazionale di Adroterapia Oncologica, Medical Physics Unit, Pavia, Italy
| | - Giuseppe Magro
- Centro Nazionale di Adroterapia Oncologica, Medical Physics Unit, Pavia, Italy
| | - Mario Ciocca
- Centro Nazionale di Adroterapia Oncologica, Medical Physics Unit, Pavia, Italy
| | - Giulia Riva
- Centro Nazionale di Adroterapia Oncologica, Radiotherapy Unit, Pavia, Italy
| | - Lucia Pia Ciccone
- Centro Nazionale di Adroterapia Oncologica, Radiotherapy Unit, Pavia, Italy
| | - Alberto Iannalfi
- Centro Nazionale di Adroterapia Oncologica, Radiotherapy Unit, Pavia, Italy
| | - Chiara Paganelli
- Politecnico di Milano, Department of Electronics, Information and Bioengineering, Milano, Italy
| | - Ester Orlandi
- Centro Nazionale di Adroterapia Oncologica, Radiation Oncology Clinical Unit, Pavia, Italy; University of Pavia, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, Pavia, Italy
| | - Guido Baroni
- Politecnico di Milano, Department of Electronics, Information and Bioengineering, Milano, Italy
| |
Collapse
|
19
|
Paverd H, Zormpas-Petridis K, Clayton H, Burge S, Crispin-Ortuzar M. Radiology and multi-scale data integration for precision oncology. NPJ Precis Oncol 2024; 8:158. [PMID: 39060351 PMCID: PMC11282284 DOI: 10.1038/s41698-024-00656-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
In this Perspective paper we explore the potential of integrating radiological imaging with other data types, a critical yet underdeveloped area in comparison to the fusion of other multi-omic data. Radiological images provide a comprehensive, three-dimensional view of cancer, capturing features that would be missed by biopsies or other data modalities. This paper explores the complexities and challenges of incorporating medical imaging into data integration models, in the context of precision oncology. We present the different categories of imaging-omics integration and discuss recent progress, highlighting the opportunities that arise from bringing together spatial data on different scales.
Collapse
Affiliation(s)
- Hania Paverd
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | | | - Hannah Clayton
- Department of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Sarah Burge
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Mireia Crispin-Ortuzar
- Department of Oncology, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Sushentsev N, Hamm G, Flint L, Birtles D, Zakirov A, Richings J, Ling S, Tan JY, McLean MA, Ayyappan V, Horvat Menih I, Brodie C, Miller JL, Mills IG, Gnanapragasam VJ, Warren AY, Barry ST, Goodwin RJA, Barrett T, Gallagher FA. Metabolic imaging across scales reveals distinct prostate cancer phenotypes. Nat Commun 2024; 15:5980. [PMID: 39013948 PMCID: PMC11252279 DOI: 10.1038/s41467-024-50362-5] [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: 10/03/2023] [Accepted: 07/07/2024] [Indexed: 07/18/2024] Open
Abstract
Hyperpolarised magnetic resonance imaging (HP-13C-MRI) has shown promise as a clinical tool for detecting and characterising prostate cancer. Here we use a range of spatially resolved histological techniques to identify the biological mechanisms underpinning differential [1-13C]lactate labelling between benign and malignant prostate, as well as in tumours containing cribriform and non-cribriform Gleason pattern 4 disease. Here we show that elevated hyperpolarised [1-13C]lactate signal in prostate cancer compared to the benign prostate is primarily driven by increased tumour epithelial cell density and vascularity, rather than differences in epithelial lactate concentration between tumour and normal. We also demonstrate that some tumours of the cribriform subtype may lack [1-13C]lactate labelling, which is explained by lower epithelial lactate dehydrogenase expression, higher mitochondrial pyruvate carrier density, and increased lipid abundance compared to lactate-rich non-cribriform lesions. These findings highlight the potential of combining spatial metabolic imaging tools across scales to identify clinically significant metabolic phenotypes in prostate cancer.
Collapse
Affiliation(s)
- Nikita Sushentsev
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Gregory Hamm
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Lucy Flint
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Daniel Birtles
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Aleksandr Zakirov
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Jack Richings
- Predictive AI & Data, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Stephanie Ling
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Jennifer Y Tan
- Predictive AI & Data, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Mary A McLean
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Vinay Ayyappan
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ines Horvat Menih
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Cara Brodie
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Jodi L Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Ian G Mills
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Vincent J Gnanapragasam
- Department of Urology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Division of Urology, Department of Surgery, University of Cambridge, Cambridge, UK
- Cambridge Urology Translational Research and Clinical Trials Office, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, UK
| | - Anne Y Warren
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Richard J A Goodwin
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Tristan Barrett
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
21
|
Sakai NS, Bray TJP, Taylor SA. Quantitative Magnetic Resonance Imaging (qMRI) of the Small Bowel in Crohn's Disease: State-of-the-Art and Future Directions. J Magn Reson Imaging 2024. [PMID: 38970359 DOI: 10.1002/jmri.29511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 07/08/2024] Open
Abstract
Crohn's disease (CD) is a chronic inflammatory disease of the gastrointestinal tract in which repeated episodes of acute inflammation may lead to long-term bowel damage. Cross-sectional imaging is used in conjunction with endoscopy to diagnose and monitor disease and detect complications. Magnetic resonance imaging (MRI) has demonstrable utility in evaluating inflammatory activity. However, subjective interpretation of conventional MR sequences is limited in its ability to fully phenotype the underlying histopathological processes in chronic disease. In particular, conventional MRI can be confounded by the presence of mural fibrosis and muscle hypertrophy, which can mask or sometimes mimic inflammation. Quantitative MRI (qMRI) methods provide a means to better differentiate mural inflammation from fibrosis and improve quantification of these processes. qMRI may also provide more objective measures of disease activity and enable better tailoring of treatment. Here, we review quantitative MRI methods for imaging the small bowel in CD and consider the path to their clinical translation. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Naomi S Sakai
- Centre for Medical Imaging, University College London, London, UK
| | - Timothy J P Bray
- Centre for Medical Imaging, University College London, London, UK
| | - Stuart A Taylor
- Centre for Medical Imaging, University College London, London, UK
| |
Collapse
|
22
|
Lei J, Huang Y, Zhao Y, Zhou Z, Mao L, Liu Y. Nanotechnology as a new strategy for the diagnosis and treatment of gliomas. J Cancer 2024; 15:4643-4655. [PMID: 39006067 PMCID: PMC11242339 DOI: 10.7150/jca.96859] [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: 04/01/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
Glioma is the most common malignant tumor of the central nervous system (CNS), and is characterized by high aggressiveness and a high recurrence rate. Currently, the main treatments for gliomas include surgical resection, temozolomide chemotherapy and radiotherapy. However, the prognosis of glioma patients after active standardized treatment is still poor, especially for glioblastoma (GBM); the median survival is still only 14.6 months, and the 5-year survival rate is only 4-5%. The current challenges in glioma treatment include difficulty in complete surgical resection, poor blood‒brain barrier (BBB) drug permeability, therapeutic resistance, and difficulty in tumor-specific targeting. In recent years, the rapid development of nanotechnology has provided new directions for diagnosing and treating gliomas. Nanoparticles (NPs) are characterized by excellent surface tunability, precise targeting, excellent biocompatibility, and high safety. In addition, NPs can be used for gene therapy, photodynamic therapy, and antiangiogenic therapy and can be combined with biomaterials for thermotherapy. In recent decades, breakthroughs in diagnosing and treating gliomas have been made with various functional NPs, and NPs are expected to become a new strategy for glioma diagnosis and treatment. In this paper, we review the main obstacles in the treatment of glioma and discuss the potential and challenges of the latest nanotechnology in the diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Jun Lei
- Department of Neurosurgery, The First People's Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yiyang Huang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yichuan Zhao
- Southwest Medical University, Luzhou 646000, China
| | - Zhi Zhou
- Department of Neurosurgery, The First People's Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China
| | - Lei Mao
- Department of Neurosurgery, The First People's Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China
| | - Yanhui Liu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
23
|
Li H, Gong Q, Luo K. Biomarker-driven molecular imaging probes in radiotherapy. Theranostics 2024; 14:4127-4146. [PMID: 38994026 PMCID: PMC11234278 DOI: 10.7150/thno.97768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
Background: Biomarker-driven molecular imaging has emerged as an integral part of cancer precision radiotherapy. The use of molecular imaging probes, including nanoprobes, have been explored in radiotherapy imaging to precisely and noninvasively monitor spatiotemporal distribution of biomarkers, potentially revealing tumor-killing mechanisms and therapy-induced adverse effects during radiation treatment. Methods: We summarized literature reports from preclinical studies and clinical trials, which cover two main parts: 1) Clinically-investigated and emerging imaging biomarkers associated with radiotherapy, and 2) instrumental roles, functions, and activatable mechanisms of molecular imaging probes in the radiotherapy workflow. In addition, reflection and future perspectives are proposed. Results: Numerous imaging biomarkers have been continuously explored in decades, while few of them have been successfully validated for their correlation with radiotherapeutic outcomes and/or radiation-induced toxicities. Meanwhile, activatable molecular imaging probes towards the emerging biomarkers have exhibited to be promising in animal or small-scale human studies for precision radiotherapy. Conclusion: Biomarker-driven molecular imaging probes are essential for precision radiotherapy. Despite very inspiring preliminary results, validation of imaging biomarkers and rational design strategies of probes await robust and extensive investigations. Especially, the correlation between imaging biomarkers and radiotherapeutic outcomes/toxicities should be established through multi-center collaboration involving a large cohort of patients.
Collapse
Affiliation(s)
- Haonan Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, 699 Jinyuan Xi Road, Jimei District, 361021 Xiamen, Fujian, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
24
|
Hájek M, Flögel U, S Tavares AA, Nichelli L, Kennerley A, Kahn T, Futterer JJ, Firsiori A, Grüll H, Saha N, Couñago F, Aydogan DB, Caligiuri ME, Faber C, Bell LC, Figueiredo P, Vilanova JC, Santini F, Mekle R, Waiczies S. MR beyond diagnostics at the ESMRMB annual meeting: MR theranostics and intervention. MAGMA (NEW YORK, N.Y.) 2024; 37:323-328. [PMID: 38865057 PMCID: PMC11316697 DOI: 10.1007/s10334-024-01176-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Affiliation(s)
- Milan Hájek
- Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Institute for Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Adriana A S Tavares
- Centre for Cardiovascular Sciences and Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Lucia Nichelli
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Paris Brain Institute, ICM, Paris, France
- Department of Neuroradiology, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Aneurin Kennerley
- Department of Sports and Exercise Science, Institute of Sport, Manchester Metropolitan University, Manchester, UK
- Department of Biology, University of York, York, UK
| | - Thomas Kahn
- Department of Diagnostic and Interventional Radiology, University of Leipzig, Leipzig, Germany
| | - Jurgen J Futterer
- Minimally Invasive Image-Guided Intervention Center (MAGIC), Department of Medical Imaging, Radboudumc, Nijmegen, The Netherlands
| | - Aikaterini Firsiori
- Unit of Diagnostic and Interventional Neuroradiology, Diagnostic Department, University Hospitals of Geneva, Geneva, Switzerland
| | - Holger Grüll
- Institute of Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Nandita Saha
- Max-Delbrück-Centrum Für Molekulare Medizin (MDC), Berlin Ultrahigh Field Facility, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Felipe Couñago
- Department of Radiation Oncology, Hospital Universitario San Francisco de Asís, Hospital Universitario Vithas La Milagrosa, GenesisCare, 28010, Madrid, Spain
| | - Dogu Baran Aydogan
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maria Eugenia Caligiuri
- Neuroscience Research Center, Department of Medical and Surgical Sciences, Università Degli Studi "Magna Graecia", Catanzaro, Italy
| | - Cornelius Faber
- Translational Research Imaging Center (TRIC), Clinic of Radiology, University of Münster, Münster, Germany
| | - Laura C Bell
- Early Clinical Development, Genentech Inc., South San Francisco, USA
| | - Patrícia Figueiredo
- Institute for Systems and Robotics, ISR-Lisboa, Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Joan C Vilanova
- Department of Radiology, Clínica Girona, Institute of Diagnostic Imaging (IDI) Girona, University of Girona, 17004, Girona, Spain
| | - Francesco Santini
- Department of Radiology, University Hospital of Basel, Basel, Switzerland
- Basel Muscle MRI, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Ralf Mekle
- Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sonia Waiczies
- Max-Delbrück-Centrum Für Molekulare Medizin (MDC), Berlin Ultrahigh Field Facility, Berlin, Germany.
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation Between the Charité Medical Faculty and the MDC, Berlin, Germany.
| |
Collapse
|
25
|
Keaveney S, Hopkinson G, Markus JE, Priest AN, Scurr E, Hughes J, Robertson S, Doran SJ, Collins DJ, Messiou C, Koh DM, Winfield JM. A scan-specific quality control acquisition for clinical whole-body (WB) MRI protocols. Phys Med Biol 2024; 69:125027. [PMID: 38648786 DOI: 10.1088/1361-6560/ad4195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 04/22/2024] [Indexed: 04/25/2024]
Abstract
Objective.Image quality in whole-body MRI (WB-MRI) may be degraded by faulty radiofrequency (RF) coil elements or mispositioning of the coil arrays. Phantom-based quality control (QC) is used to identify broken RF coil elements but the frequency of these acquisitions is limited by scanner and staff availability. This work aimed to develop a scan-specific QC acquisition and processing pipeline to detect broken RF coil elements, which is sufficiently rapid to be added to the clinical WB-MRI protocol. The purpose of this is to improve the quality of WB-MRI by reducing the number of patient examinations conducted with suboptimal equipment.Approach.A rapid acquisition (14 s additional acquisition time per imaging station) was developed that identifies broken RF coil elements by acquiring images from each individual coil element and using the integral body coil. This acquisition was added to one centre's clinical WB-MRI protocol for one year (892 examinations) to evaluate the effect of this scan-specific QC. To demonstrate applicability in multi-centre imaging trials, the technique was also implemented on scanners from three manufacturers.Main results. Over the course of the study RF coil elements were flagged as potentially broken on five occasions, with the faults confirmed in four of those cases. The method had a precision of 80% and a recall of 100% for detecting faulty RF coil elements. The coil array positioning measurements were consistent across scanners and have been used to define the expected variation in signal.Significance. The technique demonstrated here can identify faulty RF coil elements and positioning errors and is a practical addition to the clinical WB-MRI protocol. This approach was fully implemented on systems from two manufacturers and partially implemented on a third. It has potential to reduce the number of clinical examinations conducted with suboptimal hardware and improve image quality across multi-centre studies.
Collapse
Affiliation(s)
- Sam Keaveney
- MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | | | - Julia E Markus
- Centre for Medical Imaging, University College London, London, United Kingdom
| | - Andrew N Priest
- Department of Imaging, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Erica Scurr
- MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Julie Hughes
- MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Scott Robertson
- MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Simon J Doran
- MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - David J Collins
- MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Christina Messiou
- MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Dow-Mu Koh
- MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Jessica M Winfield
- MRI Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
26
|
Horng H, Scott C, Winham S, Jensen M, Pantalone L, Mankowski W, Kerlikowske K, Vachon CM, Kontos D, Shinohara RT. Multivariate testing and effect size measures for batch effect evaluation in radiomic features. Sci Rep 2024; 14:13923. [PMID: 38886407 PMCID: PMC11183083 DOI: 10.1038/s41598-024-64208-z] [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: 01/28/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
While precision medicine applications of radiomics analysis are promising, differences in image acquisition can cause "batch effects" that reduce reproducibility and affect downstream predictive analyses. Harmonization methods such as ComBat have been developed to correct these effects, but evaluation methods for quantifying batch effects are inconsistent. In this study, we propose the use of the multivariate statistical test PERMANOVA and the Robust Effect Size Index (RESI) to better quantify and characterize batch effects in radiomics data. We evaluate these methods in both simulated and real radiomics features extracted from full-field digital mammography (FFDM) data. PERMANOVA demonstrated higher power than standard univariate statistical testing, and RESI was able to interpretably quantify the effect size of site at extremely large sample sizes. These methods show promise as more powerful and interpretable methods for the detection and quantification of batch effects in radiomics studies.
Collapse
Affiliation(s)
- Hannah Horng
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Statistics in Imaging Endeavor (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | | | | | | | - Lauren Pantalone
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Walter Mankowski
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | | | - Despina Kontos
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Columbia University, New York, NY, 10027, USA
| | - Russell T Shinohara
- Department of Radiology, Center for Biomedical Image Computing and Analysis (CBICA), University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Statistics in Imaging Endeavor (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
27
|
Xu L, Wu Y, Shen X, Zhou L, Lu Y, Teng Z, Du J, Ding M, Han H, Niu T. Exploring the biological basis of CT imaging features in pancreatic neuroendocrine tumors: a two-center study. Phys Med Biol 2024; 69:125013. [PMID: 38810631 DOI: 10.1088/1361-6560/ad51c7] [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: 01/08/2024] [Accepted: 05/29/2024] [Indexed: 05/31/2024]
Abstract
Objective.Medical imaging offered a non-invasive window to visualize tumors, with radiomics transforming these images into quantitative data for tumor phenotyping. However, the intricate web linking imaging features, clinical endpoints, and tumor biology was mostly uncharted. This study aimed to unravel the connections between CT imaging features and clinical characteristics, including tumor histopathological grading, clinical stage, and endocrine symptoms, alongside immunohistochemical markers of tumor cell growth, such as the Ki-67 index and nuclear mitosis rate.Approach.We conducted a retrospective analysis of data from 137 patients with pancreatic neuroendocrine tumors who had undergone contrast-enhanced CT scans across two institutions. Our study focused on three clinical factors: pathological grade, clinical stage, and endocrine symptom status, in addition to two immunohistochemical markers: the Ki-67 index and the rate of nuclear mitosis. We computed both predefined (2D and 3D) and learning-based features (via sparse autoencoder, or SAE) from the scans. To unearth the relationships between imaging features, clinical factors, and immunohistochemical markers, we employed the Spearman rank correlation along with the Benjamini-Hochberg method. Furthermore, we developed and validated radiomics signatures to foresee these clinical factors.Main results.The 3D imaging features showed the strongest relationships with clinical factors and immunohistochemical markers. For the association with pathological grade, the mean absolute value of the correlation coefficient (CC) of 2D, SAE, and 3D features was 0.3318 ± 0.1196, 0.2149 ± 0.0361, and 0.4189 ± 0.0882, respectively. While for the association with Ki-67 index and rate of nuclear mitosis, the 3D features also showed higher correlations, with CC as 0.4053 ± 0.0786 and 0.4061 ± 0.0806. In addition, the 3D feature-based signatures showed optimal performance in clinical factor prediction.Significance.We found relationships between imaging features, clinical factors, and immunohistochemical markers. The 3D features showed higher relationships with clinical factors and immunohistochemical markers.
Collapse
Affiliation(s)
- Lei Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, Guangdong, People's Republic of China
| | - Yan Wu
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaoyong Shen
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Luping Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Yongkai Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Ze Teng
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jichen Du
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, People's Republic of China
| | - Mingchao Ding
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, People's Republic of China
| | - Hongbin Han
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
- Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology, Peking University Third Hospital, Beijing, People's Republic of China
| | - Tianye Niu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, Guangdong, People's Republic of China
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, People's Republic of China
| |
Collapse
|
28
|
Yaghoubi Naei V, Ivanova E, Mullally W, O'Leary CG, Ladwa R, O'Byrne K, Warkiani ME, Kulasinghe A. Characterisation of circulating tumor-associated and immune cells in patients with advanced-stage non-small cell lung cancer. Clin Transl Immunology 2024; 13:e1516. [PMID: 38835954 PMCID: PMC11147668 DOI: 10.1002/cti2.1516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/26/2024] [Accepted: 05/16/2024] [Indexed: 06/06/2024] Open
Abstract
Objectives Globally, non-small cell lung cancer (NSCLC) is the most prevalent form of lung cancer and the leading cause of cancer-related deaths. Tumor-associated circulating cells in NSCLC can have a wide variety of morphological and phenotypic characteristics, including epithelial, immunological or hybrid subtypes. The distinctive characteristics and potential clinical significance of these cells in patients with NSCLC are explored in this study. Methods We utilised a spiral microfluidic device to enrich large cells and cell aggregates from the peripheral blood samples of NSCLC patients. These cells were characterised through high-resolution immunofluorescent imaging and statistical analysis, correlating findings with clinical information from our patient cohort. Results We have identified varied populations of heterotypic circulating tumor cell clusters with differing immune cell composition that included a distinct class of atypical tumor-associated macrophages that exhibits unique morphology and cell size. This subtype's prevalence is positively correlated with the tumor stage, progression and metastasis. Conclusions Our study reveals a heterogeneous landscape of circulating tumor cells and their clusters, underscoring the complexity of NSCLC pathobiology. The identification of a unique subtype of atypical tumor-associatedmacrophages that simultaneously express both tumor and immune markers and whose presence correlates with late disease stages, poor clinical outcomes and metastatic risk infers the potential of these cells as biomarkers for NSCLC staging and prognosis. Future studies should focus on the role of these cells in the tumor microenvironment and their potential as therapeutic targets. Additionally, longitudinal studies tracking these cell types through disease progression could provide further insights into their roles in NSCLC evolution and response to treatment.
Collapse
Affiliation(s)
- Vahid Yaghoubi Naei
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNSWAustralia
- Frazer Institute, Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| | - Ekaterina Ivanova
- Cancer and Ageing Research Program, Centre for Genomics and Personalised HealthQueensland University of TechnologyWoolloongabbaQLDAustralia
| | | | | | - Rahul Ladwa
- Frazer Institute, Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
- The Princess Alexandra HospitalBrisbaneQLDAustralia
| | - Ken O'Byrne
- The Princess Alexandra HospitalBrisbaneQLDAustralia
| | - Majid E Warkiani
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNSWAustralia
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
29
|
Bourke G, Wade RG, van Alfen N. Updates in diagnostic tools for diagnosing nerve injury and compressions. J Hand Surg Eur Vol 2024; 49:668-680. [PMID: 38534079 DOI: 10.1177/17531934241238736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Predicting prognosis after nerve injury and compression can be challenging, even for the experienced clinician. Although thorough clinical assessment can aid diagnosis, we cannot always be precise about long-term functional recovery of either motor or sensory nerves. To evaluate the severity of nerve injury, surgical exploration remains the gold standard, particularly after iatrogenic injury and major nerve injury from trauma, such as brachial plexus injury. Recently, advances in imaging techniques (ultrasound, magnetic resonance imaging [MRI] and MR neurography) along with multimodality assessment, including electrodiagnostic testing, have allowed us to have a better preoperative understanding of nerve continuity and prediction of nerve health and possible recovery. This article outlines the current and potential roles for clinical assessment, exploratory surgery, electrodiagnostic testing ultrasound and MRI in entrapment neuropathies, inflammatory neuritis and trauma. Emphasis is placed on those modalities that are improving in diagnostic accuracy of nerve assessment before any surgical intervention.
Collapse
Affiliation(s)
- Gráinne Bourke
- Leeds Institute for Medical Research, University of Leeds, Leeds, UK
- Department of Plastic and Reconstructive Surgery, Leeds Teaching Hospitals Trust, Leeds, UK
| | - Ryckie G Wade
- Leeds Institute for Medical Research, University of Leeds, Leeds, UK
- Department of Plastic and Reconstructive Surgery, Leeds Teaching Hospitals Trust, Leeds, UK
| | - Nens van Alfen
- Department of Neurology, Clinical Neuromuscular Imaging Group, Donders Centre for Neuroscience, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
30
|
Singh S, Mohajer B, Wells SA, Garg T, Hanneman K, Takahashi T, AlDandan O, McBee MP, Jawahar A. Imaging Genomics and Multiomics: A Guide for Beginners Starting Radiomics-Based Research. Acad Radiol 2024; 31:2281-2291. [PMID: 38286723 DOI: 10.1016/j.acra.2024.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/31/2024]
Abstract
Radiomics uses advanced mathematical analysis of pixel-level information from radiologic images to extract existing information in traditional imaging algorithms. It is intended to find imaging biomarkers related to the genomics of tumors or disease patterns that improve medical care by advanced detection of tumor response patterns in tumors and to assess prognosis. Radiomics expands the paradigm of medical imaging to help with diagnosis, management of diseases and prognostication, leveraging image features by extracting information that can be used as imaging biomarkers to predict prognosis and response to treatment. Radiogenomics is an emerging area in radiomics that investigates the association between imaging characteristics and gene expression profiles. There are an increasing number of research publications using different radiomics approaches without a clear consensus on which method works best. We aim to describe the workflow of radiomics along with a guide of what to expect when starting a radiomics-based research project.
Collapse
Affiliation(s)
- Shiva Singh
- Radiology and Imaging Sciences, National Institutes of Health, Bethesda, Maryland
| | - Bahram Mohajer
- Radiology and Radiological Sciences, Johns Hopkins Medicine, Baltimore, Maryland
| | - Shane A Wells
- Radiology, University of Michigan, Ann Arbor, Michigan
| | - Tushar Garg
- Radiology and Radiological Sciences, Johns Hopkins Medicine, Baltimore, Maryland
| | - Kate Hanneman
- Medical Imaging, University of Toronto, Toronto, ON, Canada
| | | | - Omran AlDandan
- Department of Radiology, Imam Abdulrahman Bin Faisal University, College of Medicine: Dammam, Eastern, Saudi Arabia
| | - Morgan P McBee
- Radiology and Radiological Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Anugayathri Jawahar
- Radiology, Northwestern University-Feinberg School of Medicine, 800, Arkes Pavilion, 676 N St. Clair St, Chicago, IL 60611.
| |
Collapse
|
31
|
Sheppard AJ, Theng EH, Paravastu SS, Wojnowski NM, Farhadi F, Morris MA, Hartley IR, Rachel IG, Roszko KL, Collins MT, Saboury B. Spatial Atlas for Mapping Vascular Microcalcification Using 18F-NaF PET/CT: Application in Hyperphosphatemic Familial Tumoral Calcinosis. Arterioscler Thromb Vasc Biol 2024; 44:1432-1446. [PMID: 38660800 PMCID: PMC11111330 DOI: 10.1161/atvbaha.123.320455] [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: 12/18/2023] [Accepted: 03/28/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Vascular calcification causes significant morbidity and occurs frequently in diseases of calcium/phosphate imbalance. Radiolabeled sodium fluoride positron emission tomography/computed tomography has emerged as a sensitive and specific method for detecting and quantifying active microcalcifications. We developed a novel technique to quantify and map total vasculature microcalcification to a common space, allowing simultaneous assessment of global disease burden and precise tracking of site-specific microcalcifications across time and individuals. METHODS To develop this technique, 4 patients with hyperphosphatemic familial tumoral calcinosis, a monogenic disorder of FGF23 (fibroblast growth factor-23) deficiency with a high prevalence of vascular calcification, underwent radiolabeled sodium fluoride positron emission tomography/computed tomography imaging. One patient received serial imaging 1 year after treatment with an IL-1 (interleukin-1) antagonist. A radiolabeled sodium fluoride-based microcalcification score, as well as calcification volume, was computed at all perpendicular slices, which were then mapped onto a standardized vascular atlas. Segment-wise mCSmean and mCSmax were computed to compare microcalcification score levels at predefined vascular segments within subjects. RESULTS Patients with hyperphosphatemic familial tumoral calcinosis had notable peaks in microcalcification score near the aortic bifurcation and distal femoral arteries, compared with a control subject who had uniform distribution of vascular radiolabeled sodium fluoride uptake. This technique also identified microcalcification in a 17-year-old patient, who had no computed tomography-defined calcification. This technique could not only detect a decrease in microcalcification score throughout the patient treated with an IL-1 antagonist but it also identified anatomic areas that had increased responsiveness while there was no change in computed tomography-defined macrocalcification after treatment. CONCLUSIONS This technique affords the ability to visualize spatial patterns of the active microcalcification process in the peripheral vasculature. Further, this technique affords the ability to track microcalcifications at precise locations not only across time but also across subjects. This technique is readily adaptable to other diseases of vascular calcification and may represent a significant advance in the field of vascular biology.
Collapse
Affiliation(s)
- Aaron J Sheppard
- National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD, 20892
- Louisiana State University Health Shreveport, School of Medicine, Shreveport, LA, 71103
| | - Elizabeth H Theng
- National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD, 20892
- Department of Radiology, Stanford School of Medicine, Stanford, CA, 94304
| | - Sriram S Paravastu
- National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD, 20892
- University of Missouri – Kansas City School of Medicine, Kansas City, MO, 64108
| | - Natalia M Wojnowski
- National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD, 20892
- Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Faraz Farhadi
- Radiology and Imaging Sciences, Clinical Center, NIH, Bethesda, MD, 20892
- Geisel School of Medicine, Dartmouth, Hanover, NH, 03755
- Institute of Nuclear Medicine, Bethesda, MD, USA
| | | | - Iris R Hartley
- National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD, 20892
| | - I Gafni Rachel
- National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD, 20892
| | - Kelly L Roszko
- National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD, 20892
| | - Michael T Collins
- National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD, 20892
| | - Babak Saboury
- Radiology and Imaging Sciences, Clinical Center, NIH, Bethesda, MD, 20892
- Institute of Nuclear Medicine, Bethesda, MD, USA
| |
Collapse
|
32
|
Cohen O, Kargar S, Woo S, Vargas A, Otazo R. DCE-Qnet: Deep Network Quantification of Dynamic Contrast Enhanced (DCE) MRI. ARXIV 2024:arXiv:2405.12360v1. [PMID: 38827459 PMCID: PMC11142325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Introduction Quantification of dynamic contrast-enhanced (DCE)-MRI has the potential to provide valuable clinical information, but robust pharmacokinetic modeling remains a challenge for clinical adoption. Methods A 7-layer neural network called DCE-Qnet was trained on simulated DCE-MRI signals derived from the Extended Tofts model with the Parker arterial input function. Network training incorporated B1 inhomogeneities to estimate perfusion (Ktrans, vp, ve), tissue T1 relaxation, proton density and bolus arrival time (BAT). The accuracy was tested in a digital phantom in comparison to a conventional nonlinear least-squares fitting (NLSQ). In vivo testing was conducted in 10 healthy subjects. Regions of interest in the cervix and uterine myometrium were used to calculate the inter-subject variability. The clinical utility was demonstrated on a cervical cancer patient. Test-retest experiments were used to assess reproducibility of the parameter maps in the tumor. Results The DCE-Qnet reconstruction outperformed NLSQ in the phantom. The coefficient of variation (CV) in the healthy cervix varied between 5-51% depending on the parameter. Parameter values in the tumor agreed with previous studies despite differences in methodology. The CV in the tumor varied between 1-47%. Conclusion The proposed approach provides comprehensive DCE-MRI quantification from a single acquisition. DCE-Qnet eliminates the need for separate T1 scan or BAT processing, leading to a reduction of 10 minutes per scan and more accurate quantification.
Collapse
Affiliation(s)
- Ouri Cohen
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Soudabeh Kargar
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sungmin Woo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alberto Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ricardo Otazo
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
33
|
Eertink JJ, Bahce I, Waterton JC, Huisman MC, Boellaard R, Wunder A, Thiele A, Menke-van der Houven van Oordt CW. The development process of 'fit-for-purpose' imaging biomarkers to characterize the tumor microenvironment. Front Med (Lausanne) 2024; 11:1347267. [PMID: 38818386 PMCID: PMC11138661 DOI: 10.3389/fmed.2024.1347267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/24/2024] [Indexed: 06/01/2024] Open
Abstract
Immune-based treatment approaches are successfully used for the treatment of patients with cancer. While such therapies can be highly effective, many patients fail to benefit. To provide optimal therapy choices and to predict treatment responses, reliable biomarkers for the assessment of immune features in patients with cancer are of significant importance. Biomarkers (BM) that enable a comprehensive and repeatable assessment of the tumor microenvironment (TME), the lymphoid system, and the dynamics induced by drug treatment can fill this gap. Medical imaging, notably positron emission tomography (PET) and magnetic resonance imaging (MRI), providing whole-body imaging BMs, might deliver such BMs. However, those imaging BMs must be well characterized as being 'fit for purpose' for the intended use. This review provides an overview of the key steps involved in the development of 'fit-for-purpose' imaging BMs applicable in drug development, with a specific focus on pharmacodynamic biomarkers for assessing the TME and its modulation by immunotherapy. The importance of the qualification of imaging BMs according to their context of use (COU) as defined by the Food and Drug Administration (FDA) and National Institutes of Health Biomarkers, EndpointS, and other Tools (BEST) glossary is highlighted. We elaborate on how an imaging BM qualification for a specific COU can be achieved.
Collapse
Affiliation(s)
- Jakoba J. Eertink
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Idris Bahce
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
- Department of Pulmonary Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - John C. Waterton
- Centre for Imaging Sciences, University of Manchester, Manchester, United Kingdom
| | - Marc C. Huisman
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ronald Boellaard
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Andreas Wunder
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach and der Riss, Germany
| | - Andrea Thiele
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach and der Riss, Germany
| | - Catharina W. Menke-van der Houven van Oordt
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
34
|
Magbanua MJM, Li W, van ’t Veer LJ. Integrating Imaging and Circulating Tumor DNA Features for Predicting Patient Outcomes. Cancers (Basel) 2024; 16:1879. [PMID: 38791958 PMCID: PMC11120531 DOI: 10.3390/cancers16101879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Biomarkers for evaluating tumor response to therapy and estimating the risk of disease relapse represent tremendous areas of clinical need. To evaluate treatment efficacy, tumor response is routinely assessed using different imaging modalities like positron emission tomography/computed tomography or magnetic resonance imaging. More recently, the development of circulating tumor DNA detection assays has provided a minimally invasive approach to evaluate tumor response and prognosis through a blood test (liquid biopsy). Integrating imaging- and circulating tumor DNA-based biomarkers may lead to improvements in the prediction of patient outcomes. For this mini-review, we searched the scientific literature to find original articles that combined quantitative imaging and circulating tumor DNA biomarkers to build prediction models. Seven studies reported building prognostic models to predict distant recurrence-free, progression-free, or overall survival. Three discussed building models to predict treatment response using tumor volume, pathologic complete response, or objective response as endpoints. The limited number of articles and the modest cohort sizes reported in these studies attest to the infancy of this field of study. Nonetheless, these studies demonstrate the feasibility of developing multivariable response-predictive and prognostic models using regression and machine learning approaches. Larger studies are warranted to facilitate the building of highly accurate response-predictive and prognostic models that are generalizable to other datasets and clinical settings.
Collapse
Affiliation(s)
- Mark Jesus M. Magbanua
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94115, USA;
| | - Wen Li
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94115, USA;
| | - Laura J. van ’t Veer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94115, USA;
| |
Collapse
|
35
|
Shalom ES, Kim H, van der Heijden RA, Ahmed Z, Patel R, Hormuth DA, DiCarlo JC, Yankeelov TE, Sisco NJ, Dortch RD, Stokes AM, Inglese M, Grech-Sollars M, Toschi N, Sahoo P, Singh A, Verma SK, Rathore DK, Kazerouni AS, Partridge SC, LoCastro E, Paudyal R, Wolansky IA, Shukla-Dave A, Schouten P, Gurney-Champion OJ, Jiřík R, Macíček O, Bartoš M, Vitouš J, Das AB, Kim SG, Bokacheva L, Mikheev A, Rusinek H, Berks M, Hubbard Cristinacce PL, Little RA, Cheung S, O'Connor JPB, Parker GJM, Moloney B, LaViolette PS, Bobholz S, Duenweg S, Virostko J, Laue HO, Sung K, Nabavizadeh A, Saligheh Rad H, Hu LS, Sourbron S, Bell LC, Fathi Kazerooni A. The ISMRM Open Science Initiative for Perfusion Imaging (OSIPI): Results from the OSIPI-Dynamic Contrast-Enhanced challenge. Magn Reson Med 2024; 91:1803-1821. [PMID: 38115695 DOI: 10.1002/mrm.29909] [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: 04/14/2023] [Revised: 08/22/2023] [Accepted: 10/16/2023] [Indexed: 12/21/2023]
Abstract
PURPOSE K trans $$ {K}^{\mathrm{trans}} $$ has often been proposed as a quantitative imaging biomarker for diagnosis, prognosis, and treatment response assessment for various tumors. None of the many software tools forK trans $$ {K}^{\mathrm{trans}} $$ quantification are standardized. The ISMRM Open Science Initiative for Perfusion Imaging-Dynamic Contrast-Enhanced (OSIPI-DCE) challenge was designed to benchmark methods to better help the efforts to standardizeK trans $$ {K}^{\mathrm{trans}} $$ measurement. METHODS A framework was created to evaluateK trans $$ {K}^{\mathrm{trans}} $$ values produced by DCE-MRI analysis pipelines to enable benchmarking. The perfusion MRI community was invited to apply their pipelines forK trans $$ {K}^{\mathrm{trans}} $$ quantification in glioblastoma from clinical and synthetic patients. Submissions were required to include the entrants'K trans $$ {K}^{\mathrm{trans}} $$ values, the applied software, and a standard operating procedure. These were evaluated using the proposedOSIP I gold $$ \mathrm{OSIP}{\mathrm{I}}_{\mathrm{gold}} $$ score defined with accuracy, repeatability, and reproducibility components. RESULTS Across the 10 received submissions, theOSIP I gold $$ \mathrm{OSIP}{\mathrm{I}}_{\mathrm{gold}} $$ score ranged from 28% to 78% with a 59% median. The accuracy, repeatability, and reproducibility scores ranged from 0.54 to 0.92, 0.64 to 0.86, and 0.65 to 1.00, respectively (0-1 = lowest-highest). Manual arterial input function selection markedly affected the reproducibility and showed greater variability inK trans $$ {K}^{\mathrm{trans}} $$ analysis than automated methods. Furthermore, provision of a detailed standard operating procedure was critical for higher reproducibility. CONCLUSIONS This study reports results from the OSIPI-DCE challenge and highlights the high inter-software variability withinK trans $$ {K}^{\mathrm{trans}} $$ estimation, providing a framework for ongoing benchmarking against the scores presented. Through this challenge, the participating teams were ranked based on the performance of their software tools in the particular setting of this challenge. In a real-world clinical setting, many of these tools may perform differently with different benchmarking methodology.
Collapse
Affiliation(s)
- Eve S Shalom
- School of Physics and Astronomy, University of Leeds, Leeds, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Harrison Kim
- Department of Radiology, University of Alabama, Birmingham, Alabama, USA
| | - Rianne A van der Heijden
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Zaki Ahmed
- Corewell Health William Beaumont University Hospital, Royal Oak, Michigan, USA
| | - Reyna Patel
- Department of Radiology, Neuroradiology Division, Mayo Clinic, Scottsdale, Arizona, USA
| | - David A Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas, Austin, Texas, USA
| | - Julie C DiCarlo
- Biomedical Imaging Center, Livestrong Cancer Institutes, University of Texas at Austin, Austin, Texas, USA
| | - Thomas E Yankeelov
- Departments of Biomedical Engineering, Diagnostic Medicine, Oncology, Livestrong Cancer Institutes, Oden Institute for Computational Engineering and Sciences, The University of Texas, Austin, Texas, USA
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicholas J Sisco
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Richard D Dortch
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Ashley M Stokes
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Marianna Inglese
- Department of Biomedicine and Prevention, University of Rome, Tor Vergata, Italy
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Matthew Grech-Sollars
- Department of Surgery and Cancer, Imperial College, London, UK
- Department of Computer Science, University College London, London, UK
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome, Tor Vergata, Italy
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, Massachusetts, USA
| | - Prativa Sahoo
- University Medical Center Göttingen, Göttingen, Germany
| | - Anup Singh
- Center for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Sanjay K Verma
- Institute of Bioengineering and Bioimaging, Singapore, Singapore
| | - Divya K Rathore
- Institute of Psychiatry, Psychology & Neuroscience, King's College, London, UK
| | - Anum S Kazerouni
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | | | - Eve LoCastro
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ramesh Paudyal
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ivan A Wolansky
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Amita Shukla-Dave
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Pepijn Schouten
- Department of Radiology and Nuclear Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Oliver J Gurney-Champion
- Department of Radiology and Nuclear Medicine, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Radovan Jiřík
- Czech Academy of Sciences, Institute of Scientific Instruments, Brno, Czech Republic
| | - Ondřej Macíček
- Czech Academy of Sciences, Institute of Scientific Instruments, Brno, Czech Republic
| | - Michal Bartoš
- Czech Academy of Sciences, Institute of Information Theory and Automation, Praha, Czech Republic
| | - Jiří Vitouš
- Czech Academy of Sciences, Institute of Scientific Instruments, Brno, Czech Republic
| | | | - S Gene Kim
- Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - Louisa Bokacheva
- Department of Radiology, Grossman School of Medicine, New York University, New York, New York, USA
| | - Artem Mikheev
- Department of Radiology, Grossman School of Medicine, New York University, New York, New York, USA
| | - Henry Rusinek
- Department of Radiology, Grossman School of Medicine, New York University, New York, New York, USA
| | - Michael Berks
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | | | - Ross A Little
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Susan Cheung
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - James P B O'Connor
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- Department of Radiology, The Christie Hospital NHS Trust, Manchester, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Geoff J M Parker
- Center for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- Bioxydyn Ltd, Manchester, UK
| | - Brendan Moloney
- Advanced Imaging Research Center, Oregon Health & Science Institute, Portland, Oregon, USA
| | - Peter S LaViolette
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Samuel Bobholz
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Savannah Duenweg
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John Virostko
- Department of Diagnostic Medicine, University of Texas, Austin, Texas, USA
| | - Hendrik O Laue
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Kyunghyun Sung
- Department of Radiological Sciences, University of California, Los Angeles, California, USA
| | - Ali Nabavizadeh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Data-Driven Discovery, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hamidreza Saligheh Rad
- Quantitative MR Imaging and Spectroscopy Group, Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
- Center for Computational Imaging & Simulation Technologies in Biomedicine, School of Computing/School of Medicine, University of Leeds, Leeds, UK
| | - Leland S Hu
- Neuroradiology Division, Department of Radiology, Mayo Clinic, Phoenix, Arizona, USA
| | - Steven Sourbron
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Laura C Bell
- Clinical Imaging Group, Genentech, Inc., South San Francisco, California, USA
| | - Anahita Fathi Kazerooni
- Quantitative MR Imaging and Spectroscopy Group, Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, USA
| |
Collapse
|
36
|
Dickie BR, Ahmed Z, Arvidsson J, Bell LC, Buckley DL, Debus C, Fedorov A, Floca R, Gutmann I, van der Heijden RA, van Houdt PJ, Sourbron S, Thrippleton MJ, Quarles C, Kompan IN. A community-endorsed open-source lexicon for contrast agent-based perfusion MRI: A consensus guidelines report from the ISMRM Open Science Initiative for Perfusion Imaging (OSIPI). Magn Reson Med 2024; 91:1761-1773. [PMID: 37831600 PMCID: PMC11337559 DOI: 10.1002/mrm.29840] [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: 05/17/2023] [Revised: 07/25/2023] [Accepted: 08/04/2023] [Indexed: 10/15/2023]
Abstract
This manuscript describes the ISMRM OSIPI (Open Science Initiative for Perfusion Imaging) lexicon for dynamic contrast-enhanced and dynamic susceptibility-contrast MRI. The lexicon was developed by Taskforce 4.2 of OSIPI to provide standardized definitions of commonly used quantities, models, and analysis processes with the aim of reducing reporting variability. The taskforce was established in February 2020 and consists of medical physicists, engineers, clinicians, data and computer scientists, and DICOM (Digital Imaging and Communications in Medicine) standard experts. Members of the taskforce collaborated via a slack channel and quarterly virtual meetings. Members participated by defining lexicon items and reporting formats that were reviewed by at least two other members of the taskforce. Version 1.0.0 of the lexicon was subject to open review from the wider perfusion imaging community between January and March 2022, and endorsed by the Perfusion Study Group of the ISMRM in the summer of 2022. The initial scope of the lexicon was set by the taskforce and defined such that it contained a basic set of quantities, processes, and models to enable users to report an end-to-end analysis pipeline including kinetic model fitting. We also provide guidance on how to easily incorporate lexicon items and definitions into free-text descriptions (e.g., in manuscripts and other documentation) and introduce an XML-based pipeline encoding format to encode analyses using lexicon definitions in standardized and extensible machine-readable code. The lexicon is designed to be open-source and extendable, enabling ongoing expansion of its content. We hope that widespread adoption of lexicon terminology and reporting formats described herein will increase reproducibility within the field.
Collapse
Affiliation(s)
- Ben R. Dickie
- Division of Informatics, Imaging, and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Center, Manchester Academic Health Science Center, The University of Manchester, Manchester, UK
| | - Zaki Ahmed
- Corewell Health William Beaumont University Hospital, Royal Oak, Michigan, USA
| | - Jonathan Arvidsson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Laura C. Bell
- Clinical Imaging Group, Genentech, Inc., South San Francisco, California, USA
| | | | | | - Andrey Fedorov
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ralf Floca
- National Center for Radiation Research in Oncology, Heidelberg Institute for Radiation Oncology, Heidelberg, Germany
| | - Ingomar Gutmann
- Faculty of Physics, Physics of Functional Materials, University of Vienna, Vienna, Austria
| | - Rianne A. van der Heijden
- Department of Radiology & Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Petra J. van Houdt
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Steven Sourbron
- Department of Infection, Immunity, and Cardiovascular Diseases, University of Sheffield, Sheffield, UK
| | - Michael J. Thrippleton
- Edinburgh Imaging and Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Chad Quarles
- Department of Cancer Systems Imaging, UT MD Anderson Cancer Center, Houston, Texas, USA
| | - Ina N. Kompan
- Division of Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
37
|
Su T, Zheng Y, Yang H, Ouyang Z, Fan J, Lin L, Lv F. Nomogram for preoperative differentiation of benign and malignant breast tumors using contrast-enhanced cone-beam breast CT (CE CB-BCT) quantitative imaging and assessment features. LA RADIOLOGIA MEDICA 2024; 129:737-750. [PMID: 38512625 DOI: 10.1007/s11547-024-01803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024]
Abstract
PURPOSE Breast cancer's impact necessitates refined diagnostic approaches. This study develops a nomogram using radiology quantitative features from contrast-enhanced cone-beam breast CT for accurate preoperative classification of benign and malignant breast tumors. MATERIAL AND METHODS A retrospective study enrolled 234 females with breast tumors, split into training and test sets. Contrast-enhanced cone-beam breast CT-images were acquired using Koning Breast CT-1000. Quantitative assessment features were extracted via 3D-slicer software, identifying independent predictors. The nomogram was constructed to preoperative differentiation benign and malignant breast tumors. Calibration curve was used to assess whether the model showed favorable correspondence with pathological confirmation. Decision curve analysis confirmed the model's superiority. RESULTS The study enrolled 234 female patients with a mean age of 50.2 years (SD ± 9.2). The training set had 164 patients (89 benign, 75 malignant), and the test set had 70 patients (29 benign, 41 malignant). The nomogram achieved excellent predictive performance in distinguishing benign and malignant breast lesions with an AUC of 0.940 (95% CI 0.900-0.940) in the training set and 0.970 (95% CI 0.940-0.970) in the test set. CONCLUSION This study illustrates the effectiveness of quantitative radiology features derived from contrast-enhanced cone-beam breast CT in distinguishing between benign and malignant breast tumors. Incorporating these features into a nomogram-based diagnostic model allows for breast tumor diagnoses that are objective and possess good accuracy. The application of these insights could substantially increase reliability and efficacy in the management of breast tumors, offering enhanced diagnostic capability.
Collapse
Affiliation(s)
- Tong Su
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China
| | - Yineng Zheng
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China
| | - Hongyu Yang
- Department of Radiology, Chongqing Changshou District People's Hospital, Chongqing, China
| | - Zubin Ouyang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China
| | - Jun Fan
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China
| | - Lin Lin
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China
| | - Fajin Lv
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China.
| |
Collapse
|
38
|
van Houdt PJ, Ragunathan S, Berks M, Ahmed Z, Kershaw LE, Gurney-Champion OJ, Tadimalla S, Arvidsson J, Sun Y, Kallehauge J, Dickie B, Lévy S, Bell L, Sourbron S, Thrippleton MJ. Contrast-agent-based perfusion MRI code repository and testing framework: ISMRM Open Science Initiative for Perfusion Imaging (OSIPI). Magn Reson Med 2024; 91:1774-1786. [PMID: 37667526 DOI: 10.1002/mrm.29826] [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: 03/24/2023] [Revised: 06/30/2023] [Accepted: 07/25/2023] [Indexed: 09/06/2023]
Abstract
PURPOSE Software has a substantial impact on quantitative perfusion MRI values. The lack of generally accepted implementations, code sharing and transparent testing reduces reproducibility, hindering the use of perfusion MRI in clinical trials. To address these issues, the ISMRM Open Science Initiative for Perfusion Imaging (OSIPI) aimed to establish a community-led, centralized repository for sharing open-source code for processing contrast-based perfusion imaging, incorporating an open-source testing framework. METHODS A repository was established on the OSIPI GitHub website. Python was chosen as the target software language. Calls for code contributions were made to OSIPI members, the ISMRM Perfusion Study Group, and publicly via OSIPI websites. An automated unit-testing framework was implemented to evaluate the output of code contributions, including visual representation of the results. RESULTS The repository hosts 86 implementations of perfusion processing steps contributed by 12 individuals or teams. These cover all core aspects of DCE- and DSC-MRI processing, including multiple implementations of the same functionality. Tests were developed for 52 implementations, covering five analysis steps. For T1 mapping, signal-to-concentration conversion and population AIF functions, different implementations resulted in near-identical output values. For the five pharmacokinetic models tested (Tofts, extended Tofts-Kety, Patlak, two-compartment exchange, and two-compartment uptake), differences in output parameters were observed between contributions. CONCLUSIONS The OSIPI DCE-DSC code repository represents a novel community-led model for code sharing and testing. The repository facilitates the re-use of existing code and the benchmarking of new code, promoting enhanced reproducibility in quantitative perfusion imaging.
Collapse
Affiliation(s)
- Petra J van Houdt
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Michael Berks
- Quantitative Biomedical Imaging Laboratory, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Zaki Ahmed
- Corewell Health William Beaumont University Hospital, Diagnostic Radiology, Royal Oak, USA
| | - Lucy E Kershaw
- Edinburgh Imaging and Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Oliver J Gurney-Champion
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Sirisha Tadimalla
- Institute of Medical Physics, The University of Sydney, Sydney, Australia
| | - Jonathan Arvidsson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Yu Sun
- Institute of Medical Physics, The University of Sydney, Sydney, Australia
| | - Jesper Kallehauge
- Aarhus University Hospital, Danish Centre for Particle Therapy, Aarhus, Denmark
- Aarhus University, Department of Clinical Medicine, Aarhus, Denmark
| | - Ben Dickie
- Division of Informatics, Imaging, and Data Science, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, The University of Manchester, Manchester, UK
| | - Simon Lévy
- MR Research Collaborations, Siemens Healthcare Pty Ltd, Melbourne, Australia
| | - Laura Bell
- Genentech, Inc, Clinical Imaging Group, South San Francisco, USA
| | - Steven Sourbron
- University of Sheffield, Department of Infection, Immunity and Cardiovascular Disease, Sheffield, UK
| | - Michael J Thrippleton
- University of Edinburgh, Edinburgh Imaging and Centre for Clinical Brain Sciences, Edinburgh, UK
| |
Collapse
|
39
|
Waterton JC, O'Connor JPB. OSIPI: A Significant Step Towards Reproducible MR Biomarkers. J Magn Reson Imaging 2024. [PMID: 38676411 DOI: 10.1002/jmri.29414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Affiliation(s)
- John C Waterton
- Bioxydyn Ltd, Manchester, UK
- Division of Informatics Imaging & Data Sciences, University of Manchester, Manchester, UK
| | - James P B O'Connor
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| |
Collapse
|
40
|
Dasgupta A, DiCenzo D, Sannachi L, Gandhi S, Pezo RC, Eisen A, Warner E, Wright FC, Look-Hong N, Sadeghi-Naini A, Curpen B, Kolios MC, Trudeau M, Czarnota GJ. Quantitative ultrasound radiomics guided adaptive neoadjuvant chemotherapy in breast cancer: early results from a randomized feasibility study. Front Oncol 2024; 14:1273437. [PMID: 38706611 PMCID: PMC11066296 DOI: 10.3389/fonc.2024.1273437] [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] [Received: 08/06/2023] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
Background In patients with locally advanced breast cancer (LABC) receiving neoadjuvant chemotherapy (NAC), quantitative ultrasound (QUS) radiomics can predict final responses early within 4 of 16-18 weeks of treatment. The current study was planned to study the feasibility of a QUS-radiomics model-guided adaptive chemotherapy. Methods The phase 2 open-label randomized controlled trial included patients with LABC planned for NAC. Patients were randomly allocated in 1:1 ratio to a standard arm or experimental arm stratified by hormonal receptor status. All patients were planned for standard anthracycline and taxane-based NAC as decided by their medical oncologist. Patients underwent QUS imaging using a clinical ultrasound device before the initiation of NAC and after the 1st and 4th weeks of treatment. A support vector machine-based radiomics model developed from an earlier cohort of patients was used to predict treatment response at the 4th week of NAC. In the standard arm, patients continued to receive planned chemotherapy with the treating oncologists blinded to results. In the experimental arm, the QUS-based prediction was conveyed to the responsible oncologist, and any changes to the planned chemotherapy for predicted non-responders were made by the responsible oncologist. All patients underwent surgery following NAC, and the final response was evaluated based on histopathological examination. Results Between June 2018 and July 2021, 60 patients were accrued in the study arm, with 28 patients in each arm available for final analysis. In patients without a change in chemotherapy regimen (53 of 56 patients total), the QUS-radiomics model at week 4 of NAC that was used demonstrated an accuracy of 97%, respectively, in predicting the final treatment response. Seven patients were predicted to be non-responders (observational arm (n=2), experimental arm (n=5)). Three of 5 non-responders in the experimental arm had chemotherapy regimens adapted with an early initiation of taxane therapy or chemotherapy intensification, or early surgery and ended up as responders on final evaluation. Conclusion The study demonstrates the feasibility of QUS-radiomics adapted guided NAC for patients with breast cancer. The ability of a QUS-based model in the early prediction of treatment response was prospectively validated in the current study. Clinical trial registration clinicaltrials.gov, ID NCT04050228.
Collapse
Affiliation(s)
- Archya Dasgupta
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Daniel DiCenzo
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | - Sonal Gandhi
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Rossana C. Pezo
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Andrea Eisen
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ellen Warner
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Frances C. Wright
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Nicole Look-Hong
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Ali Sadeghi-Naini
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Electrical Engineering and Computer Sciences, Lassonde School of Engineering, York University, Toronto, ON, Canada
| | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | | | - Maureen Trudeau
- Division of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Gregory J. Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
41
|
Abdul-Latif M, Chowdhury A, Tharmalingam H, Taylor NJ, Lakhani A, Padhani A, Hoskin P, Tsang Y. Exploratory study of using Magnetic resonance Prognostic Imaging markers for Radiotherapy In Cervix cancer (EMPIRIC): a prospective cohort study protocol. BMJ Open 2024; 14:e077390. [PMID: 38637128 PMCID: PMC11029356 DOI: 10.1136/bmjopen-2023-077390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/29/2024] [Indexed: 04/20/2024] Open
Abstract
INTRODUCTION Radical chemoradiotherapy represents the gold standard for locally advanced cervical cancer. However, despite significant progress in improving local tumour control, distant relapse continues to impact overall survival. The development of predictive and prognostic biomarkers is consequently important to risk-stratify patients and identify populations at higher risk of poorer treatment response and survival outcomes. Exploratory study of using Magnetic resonance Prognostic Imaging markers for Radiotherapy In Cervix cancer (EMPIRIC) is a prospective exploratory cohort study, which aims to investigate the role of multiparametric functional MRI (fMRI) using diffusion-weighed imaging (DWI), dynamic contrast-enhanced (DCE) and blood oxygen level-dependent imaging (BOLD) MRI to assess treatment response and predict outcomes in patients undergoing radical chemoradiotherapy for cervical cancer. METHODS AND ANALYSIS The study aims to recruit 40 patients across a single-centre over 2 years. Patients undergo multiparametric fMRI (DWI, DCE and BOLD-MRI) at three time points: before, during and at the completion of external beam radiotherapy. Tissue and liquid biopsies are collected at diagnosis and post-treatment to identify potential biomarker correlates against fMRI. The primary outcome is to evaluate sensitivity and specificity of quantitative parameters derived from fMRI as predictors of progression-free survival at 2 years following radical chemoradiotherapy for cervical cancer. The secondary outcome is to investigate the roles of fMRI as predictors of overall survival at 2 years and tumour volume reduction across treatment. Statistical analyses using regression models and survival analyses are employed to evaluate the relationships between the derived parameters, treatment response and clinical outcomes. ETHICS AND DISSEMINATION The EMPIRIC study received ethical approval from the NHS Health Research Authority (HRA) on 14 February 2022 (protocol number RD2021-29). Confidentiality and data protection measures are strictly adhered to throughout the study. The findings of this study will be disseminated through peer-reviewed publications and scientific conferences, aiming to contribute to the growing body of evidence on the use of multiparametric MRI in cervical cancer management. TRIAL REGISTRATION NUMBER NCT05532930.
Collapse
Affiliation(s)
- Mohammed Abdul-Latif
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Clinical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | - Amani Chowdhury
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Clinical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | | | | | | | | | - Peter Hoskin
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Clinical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | - Yatman Tsang
- Clinical Oncology, Mount Vernon Cancer Centre, Northwood, UK
- Radiation Medicine, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Rimola J, Beek KJ, Ordás I, Gecse KB, Cuatrecasas M, Stoker J. Contemporary Imaging Assessment of Strictures and Fibrosis in Crohn Disease, With Focus on Quantitative Biomarkers: From the AJR Special Series on Imaging of Fibrosis. AJR Am J Roentgenol 2024; 222:e2329693. [PMID: 37530400 DOI: 10.2214/ajr.23.29693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Patients with Crohn disease commonly have bowel strictures develop, which exhibit varying degrees of inflammation and fibrosis. Differentiation of the distinct inflammatory and fibrotic components of strictures is key for the optimization of therapeutic management and for the development of antifibrotic drugs. Cross-sectional imaging techniques, including ultrasound, CT, and MRI, allow evaluation of the full thickness of the bowel wall as well as extramural complications and associated mesenteric abnormalities. Although promising data have been reported for a range of novel imaging biomarkers for detection of fibrosis and quantification of the degree of fibrosis, these biomarkers lack sufficient validation and standardization for clinical use. Additional methods, including PET with emerging radiotracers, artificial intelligence, and radiomics, are also under investigation for stricture characterization. In this review, we highlight the clinical relevance of identifying fibrosis in Crohn disease, review the histopathologic aspects of strictures in Crohn disease, summarize the morphologic imaging findings of strictures, and explore contemporary developments in the use of cross-sectional imaging techniques for detecting and characterizing intestinal strictures, with attention given to emerging quantitative biomarkers.
Collapse
Affiliation(s)
- Jordi Rimola
- Radiology Department, IBD Unit, Hospital Clínic de Barcelona, Villarroel 170, Escala 3, Planta 1, Barcelona 08036, Spain
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Kim J Beek
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ingrid Ordás
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Gastroenterology Department, IBD Unit, Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Krisztina B Gecse
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Míriam Cuatrecasas
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Pathology Department, IBD Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Jaap Stoker
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Zhang W, Wang S, Dong Q, Chen W, Wang P, Zhu G, Chen X, Cai Y. Predictive nomogram for lymph node metastasis and survival in gastric cancer using contrast-enhanced computed tomography-based radiomics: a retrospective study. PeerJ 2024; 12:e17111. [PMID: 38525272 PMCID: PMC10960528 DOI: 10.7717/peerj.17111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
Background Lymph node involvement significantly impacts the survival of gastric cancer patients and is a crucial factor in determining the appropriate treatment. This study aimed to evaluate the potential of enhanced computed tomography (CT)-based radiomics in predicting lymph node metastasis (LNM) and survival in patients with gastric cancer before surgery. Methods Retrospective analysis of clinical data from 192 patients diagnosed with gastric carcinoma was conducted. The patients were randomly divided into a training cohort (n = 128) and a validation cohort (n = 64). Radiomic features of CT images were extracted using the Pyradiomics software platform, and distinctive features were further selected using a Lasso Cox regression model. Features significantly associated with LNM were identified through univariate and multivariate analyses and combined with radiomic scores to create a nomogram model for predicting lymph node involvement before surgery. The predictive performance of radiomics features, CT-reported lymph node status, and the nomogram model for LNM were compared in the training and validation cohorts by plotting receiver operating characteristic (ROC) curves. High-risk and low-risk groups were identified in both cohorts based on the cut-off value of 0.582 within the radiomics evaluation scheme, and survival rates were compared. Results Seven radiomic features were identified and selected, and patients were stratified into high-risk and low-risk groups using a 0.582 cut-off radiomics score. Univariate and multivariate analyses revealed that radiomics features, diabetes mellitus, Nutrition Risk Screening (NRS) 2002 score, and CT-reported lymph node status were significant predictors of LNM in patients with gastric cancer. A predictive nomogram model was developed by combining these predictors with the radiomics score, which accurately predicted LNM in gastric cancer patients before surgery and outperformed other models in terms of accuracy and sensitivity. The AUC values for the training and validation cohorts were 0.82 and 0.722, respectively. The high-risk and low-risk groups in both the training and validation cohorts showed significant differences in survival rates. Conclusion The radiomics nomogram, based on contrast-enhanced computed tomography (CECT ), is a promising non-invasive tool for preoperatively predicting LNM in gastric cancer patients and postoperative survival.
Collapse
Affiliation(s)
- Weiteng Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sujun Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiantong Dong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenjing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pengfei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guanbao Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaolei Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiqi Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
44
|
McCabe A, Martin S, Rowe S, Shah J, Morgan PS, Borys D, Panek R. Oxygen-enhanced MRI assessment of tumour hypoxia in head and neck cancer is feasible and well tolerated in the clinical setting. Eur Radiol Exp 2024; 8:27. [PMID: 38443722 PMCID: PMC10914657 DOI: 10.1186/s41747-024-00429-1] [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: 09/01/2023] [Accepted: 01/08/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Tumour hypoxia is a recognised cause of radiotherapy treatment resistance in head and neck squamous cell carcinoma (HNSCC). Current positron emission tomography-based hypoxia imaging techniques are not routinely available in many centres. We investigated if an alternative technique called oxygen-enhanced magnetic resonance imaging (OE-MRI) could be performed in HNSCC. METHODS A volumetric OE-MRI protocol for dynamic T1 relaxation time mapping was implemented on 1.5-T clinical scanners. Participants were scanned breathing room air and during high-flow oxygen administration. Oxygen-induced changes in T1 times (ΔT1) and R2* rates (ΔR2*) were measured in malignant tissue and healthy organs. Unequal variance t-test was used. Patients were surveyed on their experience of the OE-MRI protocol. RESULTS Fifteen patients with HNSCC (median age 59 years, range 38 to 76) and 10 non-HNSCC subjects (median age 46.5 years, range 32 to 62) were scanned; the OE-MRI acquisition took less than 10 min and was well tolerated. Fifteen histologically confirmed primary tumours and 41 malignant nodal masses were identified. Median (range) of ΔT1 times and hypoxic fraction estimates for primary tumours were -3.5% (-7.0 to -0.3%) and 30.7% (6.5 to 78.6%) respectively. Radiotherapy-responsive and radiotherapy-resistant primary tumours had mean estimated hypoxic fractions of 36.8% (95% confidence interval [CI] 17.4 to 56.2%) and 59.0% (95% CI 44.6 to 73.3%), respectively (p = 0.111). CONCLUSIONS We present a well-tolerated implementation of dynamic, volumetric OE-MRI of the head and neck region allowing discernment of differing oxygen responses within biopsy-confirmed HNSCC. TRIAL REGISTRATION ClinicalTrials.gov, NCT04724096 . Registered on 26 January 2021. RELEVANCE STATEMENT MRI of tumour hypoxia in head and neck cancer using routine clinical equipment is feasible and well tolerated and allows estimates of tumour hypoxic fractions in less than ten minutes. KEY POINTS • Oxygen-enhanced MRI (OE-MRI) can estimate tumour hypoxic fractions in ten-minute scanning. • OE-MRI may be incorporable into routine clinical tumour imaging. • OE-MRI has the potential to predict outcomes after radiotherapy treatment.
Collapse
Affiliation(s)
- Alastair McCabe
- Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK.
- Department of Clinical Oncology, Nottingham University Hospitals NHS Trust, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK.
| | - Stewart Martin
- Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Selene Rowe
- Department of Radiology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Jagrit Shah
- Department of Radiology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Paul S Morgan
- Mental Health & Clinical Neurosciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Medical Physics & Clinical Engineering, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Damian Borys
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Rafal Panek
- Mental Health & Clinical Neurosciences Unit, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Medical Physics & Clinical Engineering, Nottingham University Hospitals NHS Trust, Nottingham, UK
| |
Collapse
|
45
|
Wu C, Hormuth DA, Easley T, Pineda F, Karczmar GS, Yankeelov TE. Systematic evaluation of MRI-based characterization of tumor-associated vascular morphology and hemodynamics via a dynamic digital phantom. J Med Imaging (Bellingham) 2024; 11:024002. [PMID: 38463607 PMCID: PMC10921778 DOI: 10.1117/1.jmi.11.2.024002] [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: 06/06/2023] [Revised: 01/26/2024] [Accepted: 02/19/2024] [Indexed: 03/12/2024] Open
Abstract
Purpose Validation of quantitative imaging biomarkers is a challenging task, due to the difficulty in measuring the ground truth of the target biological process. A digital phantom-based framework is established to systematically validate the quantitative characterization of tumor-associated vascular morphology and hemodynamics based on dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Approach A digital phantom is employed to provide a ground-truth vascular system within which 45 synthetic tumors are simulated. Morphological analysis is performed on high-spatial resolution DCE-MRI data (spatial/temporal resolution = 30 to 300 μ m / 60 s ) to determine the accuracy of locating the arterial inputs of tumor-associated vessels (TAVs). Hemodynamic analysis is then performed on the combination of high-spatial resolution and high-temporal resolution (spatial/temporal resolution = 60 to 300 μ m / 1 to 10 s) DCE-MRI data, determining the accuracy of estimating tumor-associated blood pressure, vascular extraction rate, interstitial pressure, and interstitial flow velocity. Results The observed effects of acquisition settings demonstrate that, when optimizing the DCE-MRI protocol for the morphological analysis, increasing the spatial resolution is helpful but not necessary, as the location and arterial input of TAVs can be recovered with high accuracy even with the lowest investigated spatial resolution. When optimizing the DCE-MRI protocol for hemodynamic analysis, increasing the spatial resolution of the images used for vessel segmentation is essential, and the spatial and temporal resolutions of the images used for the kinetic parameter fitting require simultaneous optimization. Conclusion An in silico validation framework was generated to systematically quantify the effects of image acquisition settings on the ability to accurately estimate tumor-associated characteristics.
Collapse
Affiliation(s)
- Chengyue Wu
- University of Texas at Austin, Oden Institute for Computational Engineering and Sciences, Austin, Texas, United States
- MD Anderson Cancer Center, Department of Imaging Physics, Houston, Texas, United States
- MD Anderson Cancer Center, Department of Breast Imaging, Houston, Texas, United States
- MD Anderson Cancer Center, Department of Biostatistics, Houston, Texas, United States
| | - David A. Hormuth
- University of Texas at Austin, Oden Institute for Computational Engineering and Sciences, Austin, Texas, United States
- University of Texas at Austin, Livestrong Cancer Institutes, Austin, Texas, United States
| | - Ty Easley
- Washington University in St. Louis, Department of Biomedical Engineering, St. Louis, Missouri, United States
| | - Federico Pineda
- University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | - Gregory S. Karczmar
- University of Chicago, Department of Radiology, Chicago, Illinois, United States
| | - Thomas E. Yankeelov
- University of Texas at Austin, Oden Institute for Computational Engineering and Sciences, Austin, Texas, United States
- MD Anderson Cancer Center, Department of Imaging Physics, Houston, Texas, United States
- University of Texas at Austin, Livestrong Cancer Institutes, Austin, Texas, United States
- University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States
- University of Texas at Austin, Department of Diagnostic Medicine, Austin, Texas, United States
- University of Texas at Austin, Department of Oncology, Austin, Texas, United States
| |
Collapse
|
46
|
Rastogi A, Brugnara G, Foltyn-Dumitru M, Mahmutoglu MA, Preetha CJ, Kobler E, Pflüger I, Schell M, Deike-Hofmann K, Kessler T, van den Bent MJ, Idbaih A, Platten M, Brandes AA, Nabors B, Stupp R, Bernhardt D, Debus J, Abdollahi A, Gorlia T, Tonn JC, Weller M, Maier-Hein KH, Radbruch A, Wick W, Bendszus M, Meredig H, Kurz FT, Vollmuth P. Deep-learning-based reconstruction of undersampled MRI to reduce scan times: a multicentre, retrospective, cohort study. Lancet Oncol 2024; 25:400-410. [PMID: 38423052 DOI: 10.1016/s1470-2045(23)00641-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 03/02/2024]
Abstract
BACKGROUND The extended acquisition times required for MRI limit its availability in resource-constrained settings. Consequently, accelerating MRI by undersampling k-space data, which is necessary to reconstruct an image, has been a long-standing but important challenge. We aimed to develop a deep convolutional neural network (dCNN) optimisation method for MRI reconstruction and to reduce scan times and evaluate its effect on image quality and accuracy of oncological imaging biomarkers. METHODS In this multicentre, retrospective, cohort study, MRI data from patients with glioblastoma treated at Heidelberg University Hospital (775 patients and 775 examinations) and from the phase 2 CORE trial (260 patients, 1083 examinations, and 58 institutions) and the phase 3 CENTRIC trial (505 patients, 3147 examinations, and 139 institutions) were used to develop, train, and test dCNN for reconstructing MRI from highly undersampled single-coil k-space data with various acceleration rates (R=2, 4, 6, 8, 10, and 15). Independent testing was performed with MRIs from the phase 2/3 EORTC-26101 trial (528 patients with glioblastoma, 1974 examinations, and 32 institutions). The similarity between undersampled dCNN-reconstructed and original MRIs was quantified with various image quality metrics, including structural similarity index measure (SSIM) and the accuracy of undersampled dCNN-reconstructed MRI on downstream radiological assessment of imaging biomarkers in oncology (automated artificial intelligence-based quantification of tumour burden and treatment response) was performed in the EORTC-26101 test dataset. The public NYU Langone Health fastMRI brain test dataset (558 patients and 558 examinations) was used to validate the generalisability and robustness of the dCNN for reconstructing MRIs from available multi-coil (parallel imaging) k-space data. FINDINGS In the EORTC-26101 test dataset, the median SSIM of undersampled dCNN-reconstructed MRI ranged from 0·88 to 0·99 across different acceleration rates, with 0·92 (95% CI 0·92-0·93) for 10-times acceleration (R=10). The 10-times undersampled dCNN-reconstructed MRI yielded excellent agreement with original MRI when assessing volumes of contrast-enhancing tumour (median DICE for spatial agreement of 0·89 [95% CI 0·88 to 0·89]; median volume difference of 0·01 cm3 [95% CI 0·00 to 0·03] equalling 0·21%; p=0·0036 for equivalence) or non-enhancing tumour or oedema (median DICE of 0·94 [95% CI 0·94 to 0·95]; median volume difference of -0·79 cm3 [95% CI -0·87 to -0·72] equalling -1·77%; p=0·023 for equivalence) in the EORTC-26101 test dataset. Automated volumetric tumour response assessment in the EORTC-26101 test dataset yielded an identical median time to progression of 4·27 months (95% CI 4·14 to 4·57) when using 10-times-undersampled dCNN-reconstructed or original MRI (log-rank p=0·80) and agreement in the time to progression in 374 (95·2%) of 393 patients with data. The dCNN generalised well to the fastMRI brain dataset, with significant improvements in the median SSIM when using multi-coil compared with single-coil k-space data (p<0·0001). INTERPRETATION Deep-learning-based reconstruction of undersampled MRI allows for a substantial reduction of scan times, with a 10-times acceleration demonstrating excellent image quality while preserving the accuracy of derived imaging biomarkers for the assessment of oncological treatment response. Our developments are available as open source software and hold considerable promise for increasing the accessibility to MRI, pending further prospective validation. FUNDING Deutsche Forschungsgemeinschaft (German Research Foundation) and an Else Kröner Clinician Scientist Endowed Professorship by the Else Kröner Fresenius Foundation.
Collapse
Affiliation(s)
- Aditya Rastogi
- Division for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Gianluca Brugnara
- Division for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martha Foltyn-Dumitru
- Division for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Mustafa Ahmed Mahmutoglu
- Division for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Chandrakanth J Preetha
- Division for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Erich Kobler
- Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Irada Pflüger
- Division for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marianne Schell
- Division for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Katerina Deike-Hofmann
- Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Tobias Kessler
- Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium within German Cancer Research Center, Heidelberg, Germany
| | | | - Ahmed Idbaih
- Assistance Publique-Hôpitaux de Paris, Service de Neurologie 1, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, University of Heidelberg, Mannheim, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium within German Cancer Research Center, Heidelberg, Germany
| | - Alba A Brandes
- Department of Medical Oncology, Azienda UnitàSanitaria Locale of Bologna, Bologna, Italy
| | - Burt Nabors
- Department of Neurology, Division of Neuro-Oncology, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Roger Stupp
- Lou and Jean Malnati Brain Tumor Institute, Robert H Lurie Comprehensive Cancer Center, Northwestern Medicine and Northwestern University, Chicago, USA; Department of Neurological Surgery, Northwestern Medicine and Northwestern University, Chicago, USA; Department of Neurology, Northwestern Medicine and Northwestern University, Chicago, USA
| | - Denise Bernhardt
- Department of Radiation Oncology, School of Medicine and Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Amir Abdollahi
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Institute of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Heidelberg Ion-Beam Therapy Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Thierry Gorlia
- European Organization for Research and Treatment of Cancer, Brussels, Belgium
| | - Jörg-Christian Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany; German Cancer Consortium within German Center Research Center, partner site Munich, Germany
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Klaus H Maier-Hein
- Medical Image Computing, German Cancer Research Center, Heidelberg, Germany; Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Radbruch
- Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Wolfgang Wick
- Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium within German Cancer Research Center, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hagen Meredig
- Division for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix T Kurz
- Division of Diagnostic and Interventional Neuroradiology, Geneva University Hospitals, Geneva, Switzerland; Department of Radiology, German Cancer Research Center, Heidelberg, Germany
| | - Philipp Vollmuth
- Division for Computational Neuroimaging, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany; Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany; Medical Image Computing, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
47
|
Mitchell-Hay R, Ahearn T, Murray A, Waiter G. Phantom study investigating the repeatability of radiomic features with alteration of image acquisition parameters in magnetic resonance imaging. J Med Imaging Radiat Sci 2024; 55:19-28. [PMID: 37932212 DOI: 10.1016/j.jmir.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Magnetic resonance imaging (MRI) has many different alterable parameters that affect how an image appears. This is relevant in radiomics which produces quantitative features through analysis of medical images. One significant acknowledged limitation of radiomics is repeatability. This phantom study aims to further investigate the repeatability of radiomic features (RaF), within MRI, across a range of different echo (TE) and repetition times (TR). METHODS A phantom was scanned 10 times under identical conditions on a 3T scanner using head coil over 4 months. The TE ranged from 80 to 110 ms while the TR from 3000 to 5000 ms. Radiomics analysis was performed on the same segmented section of the phantom across all TE and TR combinations. Intraclass Correlation Coefficient (ICC) was calculated across the different TE and TR ranges to investigate the repeatability of RaF. RESULTS Of 1596 features calculated, 187 features had ICC >0.9 across the range of TE, while 82 features had an ICC >0.9 across a range of TR. 664 had ICC >0.75 across the range of TEs, with 541 across the range of TR values. There was an overlap of 51 features with ICC >0.9. CONCLUSION Repeatability of RaF in MRI is dependent on imaging parameters and careful consideration of these, in combination with variable selection, is required when applying radiomics to MRI.
Collapse
Affiliation(s)
- Rosalind Mitchell-Hay
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, Scotland; Radiology Department, NHS Grampian, Aberdeen, Scotland.
| | - Trevor Ahearn
- Radiology Department, NHS Grampian, Aberdeen, Scotland
| | - Alison Murray
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, Scotland
| | - Gordon Waiter
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, Scotland
| |
Collapse
|
48
|
Kim M, Naish JH, Needleman SH, Tibiletti M, Taylor Y, O'Connor JPB, Parker GJM. Feasibility of dynamic T 2 *-based oxygen-enhanced lung MRI at 3T. Magn Reson Med 2024; 91:972-986. [PMID: 38013206 PMCID: PMC10952203 DOI: 10.1002/mrm.29914] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE To demonstrate proof-of-concept of a T2 *-sensitized oxygen-enhanced MRI (OE-MRI) method at 3T by assessing signal characteristics, repeatability, and reproducibility of dynamic lung OE-MRI metrics in healthy volunteers. METHODS We performed sequence-specific simulations for protocol optimisation and acquired free-breathing OE-MRI data from 16 healthy subjects using a dual-echo RF-spoiled gradient echo approach at 3T across two institutions. Non-linear registration and tissue density correction were applied. Derived metrics included percent signal enhancement (PSE), ∆R2 * and wash-in time normalized for breathing rate (τ-nBR). Inter-scanner reproducibility and intra-scanner repeatability were evaluated using intra-class correlation coefficient (ICC), repeatability coefficient, reproducibility coefficient, and Bland-Altman analysis. RESULTS Simulations and experimental data show negative contrast upon oxygen inhalation, due to substantial dominance of ∆R2 * at TE > 0.2 ms. Density correction improved signal fluctuations. Density-corrected mean PSE values, aligned with simulations, display TE-dependence, and an anterior-to-posterior PSE reduction trend at TE1 . ∆R2 * maps exhibit spatial heterogeneity in oxygen delivery, featuring anterior-to-posterior R2 * increase. Mean T2 * values across 32 scans were 0.68 and 0.62 ms for pre- and post-O2 inhalation, respectively. Excellent or good agreement emerged from all intra-, inter-scanner and inter-rater variability tests for PSE and ∆R2 *. However, ICC values for τ-nBR demonstrated limited agreement between repeated measures. CONCLUSION Our results demonstrate the feasibility of a T2 *-weighted method utilizing a dual-echo RF-spoiled gradient echo approach, simultaneously capturing PSE, ∆R2 * changes, and oxygen wash-in during free-breathing. The excellent or good repeatability and reproducibility on intra- and inter-scanner PSE and ∆R2 * suggest potential utility in multi-center clinical applications.
Collapse
Affiliation(s)
- Mina Kim
- Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing (CMIC)University College LondonLondonUK
| | - Josephine H. Naish
- Bioxydyn LimitedManchesterUK
- BHF Manchester Centre for Heart and Lung Magnetic Resonance Research (MCMR)Manchester University NHS Foundation TrustManchesterUK
| | - Sarah H. Needleman
- Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing (CMIC)University College LondonLondonUK
| | | | - Yohn Taylor
- Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing (CMIC)University College LondonLondonUK
| | - James P. B. O'Connor
- Division of Cancer SciencesUniversity of ManchesterManchesterUK
- Division of Radiotherapy and ImagingInstitute of Cancer ResearchLondonUK
| | - Geoff J. M. Parker
- Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing (CMIC)University College LondonLondonUK
- Bioxydyn LimitedManchesterUK
| |
Collapse
|
49
|
Tarai S, Lundström E, Sjöholm T, Jönsson H, Korenyushkin A, Ahmad N, Pedersen MA, Molin D, Enblad G, Strand R, Ahlström H, Kullberg J. Improved automated tumor segmentation in whole-body 3D scans using multi-directional 2D projection-based priors. Heliyon 2024; 10:e26414. [PMID: 38390107 PMCID: PMC10882139 DOI: 10.1016/j.heliyon.2024.e26414] [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: 11/14/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
Early cancer detection, guided by whole-body imaging, is important for the overall survival and well-being of the patients. While various computer-assisted systems have been developed to expedite and enhance cancer diagnostics and longitudinal monitoring, the detection and segmentation of tumors, especially from whole-body scans, remain challenging. To address this, we propose a novel end-to-end automated framework that first generates a tumor probability distribution map (TPDM), incorporating prior information about the tumor characteristics (e.g. size, shape, location). Subsequently, the TPDM is integrated with a state-of-the-art 3D segmentation network along with the original PET/CT or PET/MR images. This aims to produce more meaningful tumor segmentation masks compared to using the baseline 3D segmentation network alone. The proposed method was evaluated on three independent cohorts (autoPET, CAR-T, cHL) of images containing different cancer forms, obtained with different imaging modalities, and acquisition parameters and lesions annotated by different experts. The evaluation demonstrated the superiority of our proposed method over the baseline model by significant margins in terms of Dice coefficient, and lesion-wise sensitivity and precision. Many of the extremely small tumor lesions (i.e. the most difficult to segment) were missed by the baseline model but detected by the proposed model without additional false positives, resulting in clinically more relevant assessments. On average, an improvement of 0.0251 (autoPET), 0.144 (CAR-T), and 0.0528 (cHL) in overall Dice was observed. In conclusion, the proposed TPDM-based approach can be integrated with any state-of-the-art 3D UNET with potentially more accurate and robust segmentation results.
Collapse
Affiliation(s)
- Sambit Tarai
- Department of Surgical Sciences, Uppsala University, SE-75185, Uppsala, Sweden
| | - Elin Lundström
- Department of Surgical Sciences, Uppsala University, SE-75185, Uppsala, Sweden
| | - Therese Sjöholm
- Department of Surgical Sciences, Uppsala University, SE-75185, Uppsala, Sweden
| | - Hanna Jönsson
- Department of Surgical Sciences, Uppsala University, SE-75185, Uppsala, Sweden
| | | | - Nouman Ahmad
- Department of Surgical Sciences, Uppsala University, SE-75185, Uppsala, Sweden
| | - Mette A Pedersen
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Daniel Molin
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Robin Strand
- Department of Information Technology, Uppsala University, SE-75237, Uppsala, Sweden
| | - Håkan Ahlström
- Department of Surgical Sciences, Uppsala University, SE-75185, Uppsala, Sweden
- Antaros Medical AB, SE-43153, Mölndal, Sweden
| | - Joel Kullberg
- Department of Surgical Sciences, Uppsala University, SE-75185, Uppsala, Sweden
- Antaros Medical AB, SE-43153, Mölndal, Sweden
| |
Collapse
|
50
|
Lewis D, Li KL, Waqar M, Coope DJ, Pathmanaban ON, King AT, Djoukhadar I, Zhao S, Cootes TF, Jackson A, Zhu X. Low-dose GBCA administration for brain tumour dynamic contrast enhanced MRI: a feasibility study. Sci Rep 2024; 14:4905. [PMID: 38418818 PMCID: PMC10902320 DOI: 10.1038/s41598-024-53871-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
A key limitation of current dynamic contrast enhanced (DCE) MRI techniques is the requirement for full-dose gadolinium-based contrast agent (GBCA) administration. The purpose of this feasibility study was to develop and assess a new low GBCA dose protocol for deriving high-spatial resolution kinetic parameters from brain DCE-MRI. Nineteen patients with intracranial skull base tumours were prospectively imaged at 1.5 T using a single-injection, fixed-volume low GBCA dose, dual temporal resolution interleaved DCE-MRI acquisition. The accuracy of kinetic parameters (ve, Ktrans, vp) derived using this new low GBCA dose technique was evaluated through both Monte-Carlo simulations (mean percent deviation, PD, of measured from true values) and an in vivo study incorporating comparison with a conventional full-dose GBCA protocol and correlation with histopathological data. The mean PD of data from the interleaved high-temporal-high-spatial resolution approach outperformed use of high-spatial, low temporal resolution datasets alone (p < 0.0001, t-test). Kinetic parameters derived using the low-dose interleaved protocol correlated significantly with parameters derived from a full-dose acquisition (p < 0.001) and demonstrated a significant association with tissue markers of microvessel density (p < 0.05). Our results suggest accurate high-spatial resolution kinetic parameter mapping is feasible with significantly reduced GBCA dose.
Collapse
Affiliation(s)
- Daniel Lewis
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester, UK.
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Stott Lane, Salford, Greater Manchester, M6 8HD, UK.
| | - Ka-Loh Li
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Mueez Waqar
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester, UK
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - David J Coope
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester, UK
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Omar N Pathmanaban
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester, UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew T King
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester, UK
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Ibrahim Djoukhadar
- Department of Neuroradiology, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Sha Zhao
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Timothy F Cootes
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Alan Jackson
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Xiaoping Zhu
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|