1
|
Naghavi AO, Bryant JM, Kim Y, Weygand J, Redler G, Sim AJ, Miller J, Coucoules K, Michael LT, Gloria WE, Yang G, Rosenberg SA, Ahmed K, Bui MM, Henderson-Jackson EB, Lee A, Lee CD, Gonzalez RJ, Feygelman V, Eschrich SA, Scott JG, Torres-Roca J, Latifi K, Parikh N, Costello J. Habitat escalated adaptive therapy (HEAT): a phase 2 trial utilizing radiomic habitat-directed and genomic-adjusted radiation dose (GARD) optimization for high-grade soft tissue sarcoma. BMC Cancer 2024; 24:437. [PMID: 38594603 PMCID: PMC11003059 DOI: 10.1186/s12885-024-12151-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 03/20/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Soft tissue sarcomas (STS), have significant inter- and intra-tumoral heterogeneity, with poor response to standard neoadjuvant radiotherapy (RT). Achieving a favorable pathologic response (FPR ≥ 95%) from RT is associated with improved patient outcome. Genomic adjusted radiation dose (GARD), a radiation-specific metric that quantifies the expected RT treatment effect as a function of tumor dose and genomics, proposed that STS is significantly underdosed. STS have significant radiomic heterogeneity, where radiomic habitats can delineate regions of intra-tumoral hypoxia and radioresistance. We designed a novel clinical trial, Habitat Escalated Adaptive Therapy (HEAT), utilizing radiomic habitats to identify areas of radioresistance within the tumor and targeting them with GARD-optimized doses, to improve FPR in high-grade STS. METHODS Phase 2 non-randomized single-arm clinical trial includes non-metastatic, resectable high-grade STS patients. Pre-treatment multiparametric MRIs (mpMRI) delineate three distinct intra-tumoral habitats based on apparent diffusion coefficient (ADC) and dynamic contrast enhanced (DCE) sequences. GARD estimates that simultaneous integrated boost (SIB) doses of 70 and 60 Gy in 25 fractions to the highest and intermediate radioresistant habitats, while the remaining volume receives standard 50 Gy, would lead to a > 3 fold FPR increase to 24%. Pre-treatment CT guided biopsies of each habitat along with clip placement will be performed for pathologic evaluation, future genomic studies, and response assessment. An mpMRI taken between weeks two and three of treatment will be used for biological plan adaptation to account for tumor response, in addition to an mpMRI after the completion of radiotherapy in addition to pathologic response, toxicity, radiomic response, disease control, and survival will be evaluated as secondary endpoints. Furthermore, liquid biopsy will be performed with mpMRI for future ancillary studies. DISCUSSION This is the first clinical trial to test a novel genomic-based RT dose optimization (GARD) and to utilize radiomic habitats to identify and target radioresistance regions, as a strategy to improve the outcome of RT-treated STS patients. Its success could usher in a new phase in radiation oncology, integrating genomic and radiomic insights into clinical practice and trial designs, and may reveal new radiomic and genomic biomarkers, refining personalized treatment strategies for STS. TRIAL REGISTRATION NCT05301283. TRIAL STATUS The trial started recruitment on March 17, 2022.
Collapse
Affiliation(s)
- Arash O Naghavi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - J M Bryant
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Youngchul Kim
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Joseph Weygand
- Department of Radiation Oncology and Applied Sciences, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Gage Redler
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Austin J Sim
- Department of Radiation Oncology, James Cancer Hospital, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Justin Miller
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kaitlyn Coucoules
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Lauren Taylor Michael
- Clinical Trials Office, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Warren E Gloria
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - George Yang
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Stephen A Rosenberg
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kamran Ahmed
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Marilyn M Bui
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Andrew Lee
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Caitlin D Lee
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ricardo J Gonzalez
- Department of Sarcoma, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Vladimir Feygelman
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Steven A Eschrich
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jacob G Scott
- Translational Hematology and Oncology Research, Radiation Oncology Department, Cleveland Clinic, Cleveland, OH, USA
| | - Javier Torres-Roca
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kujtim Latifi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Nainesh Parikh
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - James Costello
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
2
|
Griffith JF, Yip SWY, van der Heijden RA, Valenzuela RF, Yeung DKW. Perfusion Imaging of the Musculoskeletal System. Magn Reson Imaging Clin N Am 2024; 32:181-206. [PMID: 38007280 DOI: 10.1016/j.mric.2023.07.004] [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: 11/27/2023]
Abstract
Perfusion imaging is the aspect of functional imaging, which is most applicable to the musculoskeletal system. In this review, the anatomy and physiology of bone perfusion is briefly outlined as are the methods of acquiring perfusion data on MR imaging. The current clinical indications of perfusion related to the assessment of soft tissue and bone tumors, synovitis, osteoarthritis, avascular necrosis, Keinbock's disease, diabetic foot, osteochondritis dissecans, and Paget's disease of bone are reviewed. Challenges and opportunities related to perfusion imaging of the musculoskeletal system are also briefly addressed.
Collapse
Affiliation(s)
- James F Griffith
- Department of Imaging and Interventional Radiology, Prince of Wales Hospital, The Chinese University of Hong Kong.
| | - Stefanie W Y Yip
- Department of Imaging and Interventional Radiology, Prince of Wales Hospital, The Chinese University of Hong Kong
| | - Rianne A van der Heijden
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Radiology, University of Wisconsin-Madison, Madison, WI, USA
| | - Raul F Valenzuela
- Department of Musculoskeletal Imaging, The University of Texas, MD Anderson Cancer Center, USA
| | - David K W Yeung
- Department of Imaging and Interventional Radiology, Prince of Wales Hospital, The Chinese University of Hong Kong
| |
Collapse
|
3
|
Patkulkar P, Subbalakshmi AR, Jolly MK, Sinharay S. Mapping Spatiotemporal Heterogeneity in Tumor Profiles by Integrating High-Throughput Imaging and Omics Analysis. ACS OMEGA 2023; 8:6126-6138. [PMID: 36844580 PMCID: PMC9948167 DOI: 10.1021/acsomega.2c06659] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/05/2023] [Indexed: 05/14/2023]
Abstract
Intratumoral heterogeneity associates with more aggressive disease progression and worse patient outcomes. Understanding the reasons enabling the emergence of such heterogeneity remains incomplete, which restricts our ability to manage it from a therapeutic perspective. Technological advancements such as high-throughput molecular imaging, single-cell omics, and spatial transcriptomics allow recording of patterns of spatiotemporal heterogeneity in a longitudinal manner, thus offering insights into the multiscale dynamics of its evolution. Here, we review the latest technological trends and biological insights from molecular diagnostics as well as spatial transcriptomics, both of which have witnessed burgeoning growth in the recent past in terms of mapping heterogeneity within tumor cell types as well as the stromal constitution. We also discuss ongoing challenges, indicating possible ways to integrate insights across these methods to have a systems-level spatiotemporal map of heterogeneity in each tumor and a more systematic investigation of the implications of heterogeneity for patient outcomes.
Collapse
|
4
|
Sherminie LPG, Jayatilake ML. Fractal Dimension Analysis of Pixel Dynamic Contrast Enhanced-Magnetic Resonance Imaging Pharmacokinetic Parameters for Discrimination of Benign and Malignant Breast Lesions. JCO Clin Cancer Inform 2023; 7:e2200101. [PMID: 36745858 DOI: 10.1200/cci.22.00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
PURPOSE Breast cancer is the most frequent cancer in women worldwide. However, its diagnosis mostly depends on visual examination of radiologic images, leading to an overdiagnosis with substantial costs. Therefore, a quantitative approach such as dynamic contrast enhanced (DCE)-magnetic resonance imaging (MRI) through pharmacokinetic (PK) modeling is required for reliable analysis. As PK parameters lack information on parameter heterogeneity, texture-based analysis is required to quantify PK parameter heterogeneity. Therefore, this study focused on determining the usefulness of fractal dimension (FD) as a potential imaging biomarker of tumor heterogeneity for discriminating benign and malignant breast lesions. METHODS Parametric maps for PK parameters, extravasation rate of contrast agent from blood plasma to extravascular extracellular space (Ktrans) and volume fraction of extravascular extracellular space (ve), were generated for the regions of interest (ROIs) under the standard model using 18 lesions. Then, tumor ROI and pixel DCE-MRI time-course data were analyzed to extract pixel values of Ktrans and ve. For each ROI, FD values of Ktrans and ve were computed using the blanket method. RESULTS The FD values of Ktrans for benign and malignant lesions varied from 2.96 to 3.49 and from 2.37 to 3.16, respectively, whereas FD values of ve for benign and malignant lesions varied from 3.01 to 5.15 and 2.42 to 3.44, respectively. There were significant differences in FD values derived from Ktrans parametric maps (P = .0053) and ve parametric maps (P = .0271) between benign and malignant lesions according to the statistical analysis. CONCLUSION Incorporating texture heterogeneity changes in breast lesions captured by FD with quantitative DCE-MRI parameters generated under the standard model is a potential marker for prediction of malignant lesions.
Collapse
Affiliation(s)
- Lahanda Purage G Sherminie
- Department of Nuclear Science, Faculty of Science, University of Colombo, Colombo, Sri Lanka.,Department of Radiography/Radiotherapy, Faculty of Allied Health Sciences, University of Peradeniya, Peradeniya, Sri Lanka
| | - Mohan L Jayatilake
- Department of Radiography/Radiotherapy, Faculty of Allied Health Sciences, University of Peradeniya, Peradeniya, Sri Lanka
| |
Collapse
|
5
|
Assessment of Early Response to Lung Cancer Chemotherapy by Semiquantitative Analysis of Dynamic Contrast-Enhanced MRI. DISEASE MARKERS 2022; 2022:2669281. [PMID: 35915736 PMCID: PMC9338849 DOI: 10.1155/2022/2669281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/30/2022] [Accepted: 07/04/2022] [Indexed: 11/28/2022]
Abstract
Objective To evaluate the early chemotherapy response in patients with lung cancer using semiquantitative analysis of dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI). Methods Twenty-two patients with lung cancer treated with chemotherapy were subjected to DCE-MRI at two time points: before starting treatment and after one week of therapy. The image data were collected by DCE-MRI, and the semiquantitative parameters including positive enhancement integral (PEI), signal enhancement ratio (SER), maximum slope of increase (MSI), and time to peak (TTP) were calculated. After chemotherapy, the parameters and relevant variations between the responders and nonresponders were compared with Mann–Whitney U tests. Student's t-test for paired samples was used to evaluate the temporal changes between pre- and posttreatment images. Results The patients were categorized as 13 responders and 9 nonresponders based on the tumor response evaluation. After chemotherapy, the PEI, SER, and MSI were significantly increased in responders compared with the pretreatment values (P < 0.05), while no obvious decrease in TTP was observed (P > 0.05). However, 9 nonresponders showed no significant changes in PEI, SER, MSI, and TTP values, as compared with those of pretreatment (P > 0.05). Moreover, the increase of PEI was more dramatically in responders than in nonresponders (P < 0.05), but no significantly differences were observed in SER, MSI, and TTP (P > 0.05). Conclusion Semiquantitative analysis of DCE-MRI could provide a reliable noninvasive method for assessing early chemotherapy response in lung cancer patients.
Collapse
|
6
|
Zabel WJ, Allam N, Foltz WD, Flueraru C, Taylor E, Vitkin IA. Bridging the macro to micro resolution gap with angiographic optical coherence tomography and dynamic contrast enhanced MRI. Sci Rep 2022; 12:3159. [PMID: 35210476 PMCID: PMC8873467 DOI: 10.1038/s41598-022-07000-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/09/2022] [Indexed: 11/25/2022] Open
Abstract
Dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) is emerging as a valuable tool for non-invasive volumetric monitoring of the tumor vascular status and its therapeutic response. However, clinical utility of DCE-MRI is challenged by uncertainty in its ability to quantify the tumor microvasculature (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\mu \mathrm{m}$$\end{document}μm scale) given its relatively poor spatial resolution (mm scale at best). To address this challenge, we directly compared DCE-MRI parameter maps with co-registered micron-scale-resolution speckle variance optical coherence tomography (svOCT) microvascular images in a window chamber tumor mouse model. Both semi and fully quantitative (Toft’s model) DCE-MRI metrics were tested for correlation with microvascular svOCT biomarkers. svOCT’s derived vascular volume fraction (VVF) and the mean distance to nearest vessel (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\overline{\mathrm{DNV} }$$\end{document}DNV¯) metrics were correlated with DCE-MRI vascular biomarkers such as time to peak contrast enhancement (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$r=-0.81$$\end{document}r=-0.81 and \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$0.83$$\end{document}0.83 respectively, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P<0.0001$$\end{document}P<0.0001 for both), the area under the gadolinium-time concentration curve (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$r=0.50$$\end{document}r=0.50 and \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$-0.48$$\end{document}-0.48 respectively, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P<0.0001$$\end{document}P<0.0001 for both) and \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${k}_{trans}$$\end{document}ktrans (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$r=0.64$$\end{document}r=0.64 and \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$-0.61$$\end{document}-0.61 respectively, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P<0.0001$$\end{document}P<0.0001 for both). Several other correlated micro–macro vascular metric pairs were also noted. The microvascular insights afforded by svOCT may help improve the clinical utility of DCE-MRI for tissue functional status assessment and therapeutic response monitoring applications.
Collapse
Affiliation(s)
- W Jeffrey Zabel
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| | - Nader Allam
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Warren D Foltz
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Costel Flueraru
- National Research Council Canada, Information Communication Technology, Ottawa, Canada
| | - Edward Taylor
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - I Alex Vitkin
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
7
|
Anan N, Zainon R, Tamal M. A review on advances in 18F-FDG PET/CT radiomics standardisation and application in lung disease management. Insights Imaging 2022; 13:22. [PMID: 35124733 PMCID: PMC8817778 DOI: 10.1186/s13244-021-01153-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Radiomics analysis quantifies the interpolation of multiple and invisible molecular features present in diagnostic and therapeutic images. Implementation of 18-fluorine-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) radiomics captures various disorders in non-invasive and high-throughput manner. 18F-FDG PET/CT accurately identifies the metabolic and anatomical changes during cancer progression. Therefore, the application of 18F-FDG PET/CT in the field of oncology is well established. Clinical application of 18F-FDG PET/CT radiomics in lung infection and inflammation is also an emerging field. Combination of bioinformatics approaches or textual analysis allows radiomics to extract additional information to predict cell biology at the micro-level. However, radiomics texture analysis is affected by several factors associated with image acquisition and processing. At present, researchers are working on mitigating these interrupters and developing standardised workflow for texture biomarker establishment. This review article focuses on the application of 18F-FDG PET/CT in detecting lung diseases specifically on cancer, infection and inflammation. An overview of different approaches and challenges encountered on standardisation of 18F-FDG PET/CT technique has also been highlighted. The review article provides insights about radiomics standardisation and application of 18F-FDG PET/CT in lung disease management.
Collapse
|
8
|
Kashyap A, Rapsomaniki MA, Barros V, Fomitcheva-Khartchenko A, Martinelli AL, Rodriguez AF, Gabrani M, Rosen-Zvi M, Kaigala G. Quantification of tumor heterogeneity: from data acquisition to metric generation. Trends Biotechnol 2021; 40:647-676. [PMID: 34972597 DOI: 10.1016/j.tibtech.2021.11.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 01/18/2023]
Abstract
Tumors are unique and complex ecosystems, in which heterogeneous cell subpopulations with variable molecular profiles, aggressiveness, and proliferation potential coexist and interact. Understanding how heterogeneity influences tumor progression has important clinical implications for improving diagnosis, prognosis, and treatment response prediction. Several recent innovations in data acquisition methods and computational metrics have enabled the quantification of spatiotemporal heterogeneity across different scales of tumor organization. Here, we summarize the most promising efforts from a common experimental and computational perspective, discussing their advantages, shortcomings, and challenges. With personalized medicine entering a new era of unprecedented opportunities, our vision is that of future workflows integrating across modalities, scales, and dimensions to capture intricate aspects of the tumor ecosystem and to open new avenues for improved patient care.
Collapse
Affiliation(s)
- Aditya Kashyap
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland
| | | | - Vesna Barros
- Department of Healthcare Informatics, IBM Research, IBM R&D Labs, University of Haifa Campus, Mount Carmel, Haifa, 3498825, Israel; The Hebrew University, The Edmond J. Safra Campus - Givat Ram, Jerusalem, 9190401, Israel
| | - Anna Fomitcheva-Khartchenko
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland; Eidgenössische Technische Hochschule (ETH-Zurich), Vladimir-Prelog-Weg 1-5/10, 8099 Zurich, Switzerland
| | | | | | - Maria Gabrani
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland
| | - Michal Rosen-Zvi
- Department of Healthcare Informatics, IBM Research, IBM R&D Labs, University of Haifa Campus, Mount Carmel, Haifa, 3498825, Israel; The Hebrew University, The Edmond J. Safra Campus - Givat Ram, Jerusalem, 9190401, Israel
| | - Govind Kaigala
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland.
| |
Collapse
|
9
|
Girot C, Volk A, Walczak C, Lassau N, Pitre-Champagnat S. New method for quantification of intratumoral heterogeneity: a feasibility study on K trans maps from preclinical DCE-MRI. MAGMA (NEW YORK, N.Y.) 2021; 34:845-857. [PMID: 34091826 DOI: 10.1007/s10334-021-00930-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/31/2022]
Abstract
OBJECT To develop new imaging biomarkers of therapeutic efficacy through the quantification of intratumoral microvascular heterogeneity. MATERIALS AND METHODS The described method was a combination of non-supervised clustering and extraction of intratumoral complexity features (ICF): number of non-connected objects, volume fraction. It was applied to a set of 3D DCE-MRI Ktrans maps acquired previously on tumor bearing mice prior to and on day 4 of anti-angiogenic treatment. Evolutions of ICF were compared to conventional summary statistics (CSS) and to heterogeneity related whole tumor texture features (TF) on treated (n = 9) and control (n = 6) mice. RESULTS Computed optimal number of clusters per tumor was 4. Several intratumoral features extracted from the clusters were able to monitor a therapy effect. Whereas no feature significantly changed for the control group, 6 features significantly changed for the treated group (4 ICF, 2 CSS). Among these, 5 also significantly differentiated the two groups (3 ICF, 2 CSS). TF failed in demonstrating differences within and between the two groups. DISCUSSION ICF are potential imaging biomarkers for anti-angiogenic therapy assessment. The presented method may be expected to have advantages with respect to texture analysis-based methods regarding interpretability of results and setup of standardized image analysis protocols.
Collapse
Affiliation(s)
- Charly Girot
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France.
| | - Andreas Volk
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Christine Walczak
- Institut Curie, Inserm, Université Paris-Saclay, CNRS, 91405, Orsay, France
| | - Nathalie Lassau
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France.,Département de Radiologie, Gustave Roussy, 94805, Villejuif, France
| | - Stéphanie Pitre-Champagnat
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| |
Collapse
|
10
|
Advanced Imaging and Computational Techniques for the Diagnostic and Prognostic Assessment of Malignant Gliomas. Cancer J 2021; 27:344-352. [PMID: 34570448 DOI: 10.1097/ppo.0000000000000545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ABSTRACT Advanced imaging techniques provide a powerful tool to assess the intratumoral and intertumoral heterogeneity of gliomas. Advances in the molecular understanding of glioma subgroups may allow improved diagnostic assessment combining imaging and molecular tumor features, with enhanced prognostic utility and implications for patient treatment. In this article, a comprehensive overview of the physiologic basis for conventional and advanced imaging techniques is presented, and clinical applications before and after treatment are discussed. An introduction to the principles of radiomics and the advanced integration of imaging, clinical outcomes, and genomic data highlights the future potential for this field of research to better stratify and select patients for standard as well as investigational therapies.
Collapse
|
11
|
Primary Gastro-Intestinal Lymphoma and Gastro-Intestinal Adenocarcinoma: An Initial Study of CT Texture Analysis as Quantitative Biomarkers for Differentiation. Life (Basel) 2021; 11:life11030264. [PMID: 33806817 PMCID: PMC8005065 DOI: 10.3390/life11030264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND To explore the potential role of computed tomography (CT) texture analysis and an imaging biomarker in differentiating primary gastro-intestinal lymphoma (PGIL) from gastro-intestinal adenocarcinoma (GIAC). METHODS A total of 131 patients with surgical pathologically PGIL and GIAC were enrolled in this study. Histogram parameters of arterial and venous phases extracted from contrast enhanced modified discrete cosine transform (MDCT) images were compared between PGIL and GIAC by Mann-Whitney U tests. The optimal parameters for differentiating these two groups were obtained through receiver operating characteristic (ROC) curves and the area under the curve (AUC) was calculated. RESULTS Compared with GIAC, in arterial phase, PGIL had statistically higher 5th, 10th percentiles (p = 0.003 and 0.011) and statistically lower entropy (p = 0.001). In the venous phase, PGIL had statistically lower mean, median, 75th, 90th, 95th percentiles, and entropy (p = 0.036, 0.029, 0.007, 0.001 and 0.001, respectively). For differentiating PGIL from GIAC, V-median + A-5th percentile was an optimal parameter for combined diagnosis (AUC = 0.746, p < 0.0001), and the corresponding sensitivity and specificity were 81.7 and 64.8%, respectively. CONCLUSION CT texture analysis could be useful for differential diagnosis of PGIL and GIAC.
Collapse
|
12
|
Tomaszewski MR, Gillies RJ. The Biological Meaning of Radiomic Features. Radiology 2021; 298:505-516. [PMID: 33399513 PMCID: PMC7924519 DOI: 10.1148/radiol.2021202553] [Citation(s) in RCA: 306] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/30/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
An earlier incorrect version appeared online. This article was corrected on February 10, 2021.
Collapse
Affiliation(s)
- Michal R. Tomaszewski
- From the Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612
| | - Robert J. Gillies
- From the Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612
| |
Collapse
|
13
|
Kalisvaart GM, Bloem JL, Bovée JVMG, van de Sande MAJ, Gelderblom H, van der Hage JA, Hartgrink HH, Krol ADG, de Geus-Oei LF, Grootjans W. Personalising sarcoma care using quantitative multimodality imaging for response assessment. Clin Radiol 2021; 76:313.e1-313.e13. [PMID: 33483087 DOI: 10.1016/j.crad.2020.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/17/2020] [Indexed: 01/18/2023]
Abstract
Over the last decades, technological developments in the field of radiology have resulted in a widespread use of imaging for personalising medicine in oncology, including patients with a sarcoma. New scanner hardware, imaging protocols, image reconstruction algorithms, radiotracers, and contrast media, enabled the assessment of the physical and biological properties of tumours associated with response to treatment. In this context, medical imaging has the potential to select sarcoma patients who do not benefit from (neo-)adjuvant treatment and facilitate treatment adaptation. Due to the biological heterogeneity in sarcomas, the challenge at hand is to acquire a practicable set of imaging features for specific sarcoma subtypes, allowing response assessment. This review provides a comprehensive overview of available clinical data on imaging-based response monitoring in sarcoma patients and future research directions. Eventually, it is expected that imaging-based response monitoring will help to achieve successful modification of (neo)adjuvant treatments and improve clinical care for these patients.
Collapse
Affiliation(s)
- G M Kalisvaart
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands.
| | - J L Bloem
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - J V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - M A J van de Sande
- Department of Orthopaedics, Leiden University Medical Center, Leiden, the Netherlands
| | - H Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - J A van der Hage
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - H H Hartgrink
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - A D G Krol
- Department of Radiation Oncology. Leiden University Medical Center, Leiden, the Netherlands
| | - L F de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands
| | - W Grootjans
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
14
|
Daye D, Tabari A, Kim H, Chang K, Kamran SC, Hong TS, Kalpathy-Cramer J, Gee MS. Quantitative tumor heterogeneity MRI profiling improves machine learning-based prognostication in patients with metastatic colon cancer. Eur Radiol 2021; 31:5759-5767. [PMID: 33454799 DOI: 10.1007/s00330-020-07673-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/28/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Intra-tumor heterogeneity has been previously shown to be an independent predictor of patient survival. The goal of this study is to assess the role of quantitative MRI-based measures of intra-tumor heterogeneity as predictors of survival in patients with metastatic colorectal cancer. METHODS In this IRB-approved retrospective study, we identified 55 patients with stage 4 colon cancer with known hepatic metastasis on MRI. Ninety-four metastatic hepatic lesions were identified on post-contrast images and manually volumetrically segmented. A heterogeneity phenotype vector was extracted from each lesion. Univariate regression analysis was used to assess the contribution of 110 extracted features to survival prediction. A random forest-based machine learning technique was applied to the feature vector and to the standard prognostic clinical and pathologic variables. The dataset was divided into a training and test set at a ratio of 4:1. ROC analysis and confusion matrix analysis were used to assess classification performance. RESULTS Mean survival time was 39 ± 3.9 months for the study population. A total of 22 texture features were associated with patient survival (p < 0.05). The trained random forest machine learning model that included standard clinical and pathological prognostic variables resulted in an area under the ROC curve of 0.83. A model that adds imaging-based heterogeneity features to the clinical and pathological variables resulted in improved model performance for survival prediction with an AUC of 0.94. CONCLUSIONS MRI-based texture features are associated with patient outcomes and improve the performance of standard clinical and pathological variables for predicting patient survival in metastatic colorectal cancer. KEY POINTS • MRI-based tumor heterogeneity texture features are associated with patient survival outcomes. • MRI-based tumor texture features complement standard clinical and pathological variables for prognosis prediction in metastatic colorectal cancer. • Agglomerative hierarchical clustering shows that patient survival outcomes are associated with different MRI tumor profiles.
Collapse
Affiliation(s)
- Dania Daye
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB #290, Boston, MA, 02114, USA.
| | - Azadeh Tabari
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB #290, Boston, MA, 02114, USA
| | - Hyunji Kim
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB #290, Boston, MA, 02114, USA.,Massachusetts Institute of Technology, Boston, MA, USA
| | - Ken Chang
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB #290, Boston, MA, 02114, USA
| | - Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jayashree Kalpathy-Cramer
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB #290, Boston, MA, 02114, USA
| | - Michael S Gee
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB #290, Boston, MA, 02114, USA
| |
Collapse
|
15
|
Hu N, Yin S, Li Q, He H, Zhong L, Gong NJ, Guo J, Cai P, Xie C, Liu H, Qiu B. Evaluating Heterogeneity of Primary Lung Tumor Using Clinical Routine Magnetic Resonance Imaging and a Tumor Heterogeneity Index. Front Oncol 2021; 10:591485. [PMID: 33542900 PMCID: PMC7853693 DOI: 10.3389/fonc.2020.591485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/23/2020] [Indexed: 11/20/2022] Open
Abstract
Objective To improve the assessment of primary tumor heterogeneity in magnetic resonance imaging (MRI) of non-small cell lung cancer (NSCLC), we proposed a method using basic measurements from T1- and T2-weighted MRI. Methods One hundred and four NSCLC patients with different T stages were studied. Fifty-two patients were analyzed as training group and another 52 as testing group. The ratios of standard deviation (SD)/mean signal value of primary tumor from T1-weighted (T1WI), T1-enhanced (T1C), T2-weighted (T2WI), and T2 fat suppression (T2fs) images were calculated. In the training group, correlation analyses were performed between the ratios and T stages. Then an ordinal regression model was built to generate the tumor heterogeneous index (THI) for evaluating the heterogeneity of tumor. The model was validated in the testing group. Results There were 11, 32, 40, and 21 patients with T1, T2, T3, and T4 disease, respectively. In the training group, the median SD/mean on T1WI, T1C, T2WI, and T2fs sequences was 0.11, 0.19, 0.16, and 0.15 respectively. The SD/mean on T1C (p=0.003), T2WI (p=0.000), and T2fs sequences (p=0.002) correlated significantly with T stages. Patients with more advanced T stage showed higher SD/mean on T2-weighted, T2fs, and T1C sequences. The median THI in the training group was 2.15. THI correlated with T stage significantly (p=0.000). In the testing group, THI was also significantly related to T stages (p=0.001). Higher THI had relevance to more advanced T stage. Conclusions The proposed ratio measurements and THI based on MRI can serve as functional radiomic markers that correlated with T stages for evaluating heterogeneity of lung tumors.
Collapse
Affiliation(s)
- Nan Hu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiation Oncology, Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| | - ShaoHan Yin
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiwen Li
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiation Oncology, Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| | - Haoqiang He
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Linchang Zhong
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Nan-Jie Gong
- Vector Lab for Intelligent Medical Imaging and Neural Engineering, International Innovation Center of Tsinghua University, Shanghai, China
| | - Jinyu Guo
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiation Oncology, Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| | - Peiqiang Cai
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chuanmiao Xie
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui Liu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiation Oncology, Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| | - Bo Qiu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiation Oncology, Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| |
Collapse
|
16
|
Abstract
Radiomics describes the extraction of multiple features from medical images, including molecular imaging modalities, that with bioinformatic approaches, provide additional clinically relevant information that may be invisible to the human eye. This information may complement standard radiological interpretation with data that may better characterize a disease or that may provide predictive or prognostic information. Progressing from predefined image features, often describing heterogeneity of voxel intensities within a volume of interest, there is increasing use of machine learning to classify disease characteristics and deep learning methods based on artificial neural networks that can learn features without a priori definition and without the need for preprocessing of images. There have been advances in standardization and harmonization of methods to a level that should support multicenter studies. However, in this relatively early phase of research in the field, there are limited aspects that have been adopted into routine practice. Most of the reports in the molecular imaging field describe radiomic approaches in cancer using 18F-fluorodeoxyglucose positron emission tomography (18F-FDG-PET). In this review, we will describe radiomics in molecular imaging and summarize the pertinent literature in lung cancer where reports are most prevalent and mature.
Collapse
Affiliation(s)
- Gary J R Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK; King's College London & Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, UK.
| | - Vicky Goh
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK; Radiology Department, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| |
Collapse
|
17
|
Thomas JV, Abou Elkassem AM, Ganeshan B, Smith AD. MR Imaging Texture Analysis in the Abdomen and Pelvis. Magn Reson Imaging Clin N Am 2020; 28:447-456. [PMID: 32624161 DOI: 10.1016/j.mric.2020.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Add "which is a" before "distribution"? Texture analysis (TA) is a form of radiomics that refers to quantitative measurements of the histogram, distribution and/or relationship of pixel intensities or gray scales within a region of interest on an image. TA can be applied to MR images of the abdomen and pelvis, with the main strength quantitative analysis of pixel intensities and heterogeneity rather than subjective/qualitative analysis. There are multiple limitations of MRTA. Despite these limitations, there is a growing body of literature supporting MRTA. This review discusses application of MRTA to the abdomen and pelvis.
Collapse
Affiliation(s)
- John V Thomas
- Body Imaging Section, Department of Radiology, University of Alabama at Birmingham, N355 Jefferson Tower, 619 19th Street South, Birmingham, AL 35249-6830, USA.
| | - Asser M Abou Elkassem
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL 35249-6830, USA
| | - Balaji Ganeshan
- Institute of Nuclear Medicine, University College of London, 5th Floor, Tower, 235 Euston Road, London NW1 2BU, UK
| | - Andrew D Smith
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL 35249-6830, USA
| |
Collapse
|
18
|
El Khouli RH, Jacobs MA. Use of MRI for Personalized Treatment of More Aggressive Tumors. Radiology 2020; 295:527-528. [PMID: 32233918 PMCID: PMC7263283 DOI: 10.1148/radiol.2020200678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/10/2020] [Accepted: 03/20/2020] [Indexed: 03/16/2025]
Affiliation(s)
- Riham H. El Khouli
- From the Department of Radiology, Nuclear Medicine and Molecular Imaging, University of Kentucky College of Medicine, 800 Rose St, Lexington, KY 40506-9983 (R.H.E.K.); and Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Md (M.A.J.)
| | - Michael A. Jacobs
- From the Department of Radiology, Nuclear Medicine and Molecular Imaging, University of Kentucky College of Medicine, 800 Rose St, Lexington, KY 40506-9983 (R.H.E.K.); and Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Md (M.A.J.)
| |
Collapse
|
19
|
A prospective, multi-centre trial of multi-parametric MRI as a biomarker in anal carcinoma. Radiother Oncol 2020; 144:7-12. [DOI: 10.1016/j.radonc.2019.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 09/12/2019] [Accepted: 10/01/2019] [Indexed: 11/23/2022]
|
20
|
Peled S, Vangel M, Kikinis R, Tempany CM, Fennessy FM, Fedorov A. Selection of Fitting Model and Arterial Input Function for Repeatability in Dynamic Contrast-Enhanced Prostate MRI. Acad Radiol 2019; 26:e241-e251. [PMID: 30467073 DOI: 10.1016/j.acra.2018.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/19/2018] [Accepted: 10/21/2018] [Indexed: 12/18/2022]
Abstract
RATIONALE AND OBJECTIVES Analysis of dynamic contrast-enhanced (DCE) magnetic resonance imaging is notable for the variability of calculated parameters. The purpose of this study was to evaluate the level of measurement variability and error/variability due to modeling in DCE magnetic resonance imaging parameters. MATERIALS AND METHODS Two prostate DCE scans were performed on 11 treatment-naïve patients with suspected or confirmed prostate peripheral zone cancer within an interval of less than two weeks. Tumor-suspicious and normal-appearing regions of interest (ROI) in the prostate peripheral zone were segmented. Different Tofts-Kety based models and different arterial input functions, with and without bolus arrival time (BAT) correction, were used to extract pharmacokinetic parameters. The percent repeatability coefficient (%RC) of fitted model parameters Ktrans, ve, and kep was calculated. Paired t-tests comparing parameters in tumor-suspicious ROIs and in normal-appearing tissue evaluated each parameter's sensitivity to pathology. RESULTS Although goodness-of-fit criteria favored the four-parameter extended Tofts-Kety model with the BAT correction included, the simplest two-parameter Tofts-Kety model overall yielded the best repeatability scores. The best %RC in the tumor-suspicious ROI was 63% for kep, 28% for ve, and 83% for Ktrans . The best p values for discrimination between tissues were p <10-5 for kep and Ktrans, and p = 0.11 for ve. Addition of the BAT correction to the models did not improve repeatability. CONCLUSION The parameter kep, using an arterial input functions directly measured from blood signals, was more repeatable than Ktrans. Both Ktrans and kep values were highly discriminatory between healthy and diseased tissues in all cases. The parameter ve had high repeatability but could not distinguish the two tissue types.
Collapse
|
21
|
Ahmed Z, Levesque IR. Pharmacokinetic modeling of dynamic contrast-enhanced MRI using a reference region and input function tail. Magn Reson Med 2019; 83:286-298. [PMID: 31393033 DOI: 10.1002/mrm.27913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE Quantitative analysis of dynamic contrast-enhanced MRI (DCE-MRI) requires an arterial input function (AIF) which is difficult to measure. We propose the reference region and input function tail (RRIFT) approach which uses a reference tissue and the washout portion of the AIF. METHODS RRIFT was evaluated in simulations with 100 parameter combinations at various temporal resolutions (5-30 s) and noise levels (σ = 0.01-0.05 mM). RRIFT was compared against the extended Tofts model (ETM) in 8 studies from patients with glioblastoma multiforme. Two versions of RRIFT were evaluated: one using measured patient-specific AIF tails, and another assuming a literature-based AIF tail. RESULTS RRIFT estimated the transfer constant K trans and interstitial volume v e with median errors within 20% across all simulations. RRIFT was more accurate and precise than the ETM at temporal resolutions slower than 10 s. The percentage error of K trans had a median and interquartile range of -9 ± 45% with the ETM and -2 ± 17% with RRIFT at a temporal resolution of 30 s under noiseless conditions. RRIFT was in excellent agreement with the ETM in vivo, with concordance correlation coefficients (CCC) of 0.95 for K trans , 0.96 for v e , and 0.73 for the plasma volume v p using a measured AIF tail. With the literature-based AIF tail, the CCC was 0.89 for K trans , 0.93 for v e and 0.78 for v p . CONCLUSIONS Quantitative DCE-MRI analysis using the input function tail and a reference tissue yields absolute kinetic parameters with the RRIFT method. This approach was viable in simulation and in vivo for temporal resolutions as low as 30 s.
Collapse
Affiliation(s)
- Zaki Ahmed
- Medical Physics Unit, McGill University, Montreal, Canada.,Department of Physics, McGill University, Montreal, Canada
| | - Ives R Levesque
- Medical Physics Unit, McGill University, Montreal, Canada.,Department of Physics, McGill University, Montreal, Canada.,Gerald Bronfman Department of Oncology, McGill University, Montreal, Canada.,Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
22
|
Han Z, Li Y, Zhang J, Liu J, Chen C, van Zijl PC, Liu G. Molecular Imaging of Deoxycytidine Kinase Activity Using Deoxycytidine-Enhanced CEST MRI. Cancer Res 2019; 79:2775-2783. [PMID: 30940660 DOI: 10.1158/0008-5472.can-18-3565] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/26/2019] [Accepted: 03/27/2019] [Indexed: 11/16/2022]
Abstract
Deoxycytidine kinase (DCK) is a key enzyme for the activation of a broad spectrum of nucleoside-based chemotherapy drugs (e.g., gemcitabine); low DCK activity is one of the most important causes of cancer drug-resistance. Noninvasive imaging methods that can quantify DCK activity are invaluable for assessing tumor resistance and predicting treatment efficacy. Here we developed a "natural" MRI approach to detect DCK activity using its natural substrate deoxycytidine (dC) as the imaging probe, which can be detected directly by chemical exchange saturation transfer (CEST) MRI without any synthetic labeling. CEST MRI contrast of dC and its phosphorylated form, dCTP, successfully discriminated DCK activity in two mouse leukemia cell lines with different DCK expression. This dC-enhanced CEST MRI in xenograft leukemic cancer mouse models demonstrated that DCK(+) tumors have a distinctive dynamic CEST contrast enhancement and a significantly higher CEST contrast than DCK(-) tumors (AUC0-60 min = 0.47 ± 0.25 and 0.20 ± 0.13, respectively; P = 0.026, paired Student t test, n = 4) at 1 hour after the injection of dC. dC-enhanced CEST contrast also correlated well with tumor responses to gemcitabine treatment. This study demonstrates a novel MR molecular imaging approach for predicting cancer resistance using natural, nonradioactive, nonmetallic, and clinically available agents. This method has great potential for pursuing personalized chemotherapy by stratifying patients with different DCK activity. SIGNIFICANCE: A new molecular MRI method that detects deoxycytidine kinase activity using its natural substrate deoxycytidine has great translational potential for clinical assessment of tumor resistance and prediction of treatment efficacy.
Collapse
Affiliation(s)
- Zheng Han
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Yuguo Li
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| | - Jia Zhang
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Jing Liu
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland.,Radiology College, Guizhou Medical University, Guiyang, Guizhou, P.R. China
| | - Chuheng Chen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Peter C van Zijl
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| | - Guanshu Liu
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland. .,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| |
Collapse
|
23
|
Morss Clyne A, Swaminathan S, Díaz Lantada A. Biofabrication strategies for creating microvascular complexity. Biofabrication 2019; 11:032001. [PMID: 30743247 DOI: 10.1088/1758-5090/ab0621] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Design and fabrication of effective biomimetic vasculatures constitutes a relevant and yet unsolved challenge, lying at the heart of tissue repair and regeneration strategies. Even if cell growth is achieved in 3D tissue scaffolds or advanced implants, tissue viability inevitably requires vascularization, as diffusion can only transport nutrients and eliminate debris within a few hundred microns. This engineered vasculature may need to mimic the intricate branching geometry of native microvasculature, referred to herein as vascular complexity, to efficiently deliver blood and recreate critical interactions between the vascular and perivascular cells as well as parenchymal tissues. This review first describes the importance of vascular complexity in labs- and organs-on-chips, the biomechanical and biochemical signals needed to create and maintain a complex vasculature, and the limitations of current 2D, 2.5D, and 3D culture systems in recreating vascular complexity. We then critically review available strategies for design and biofabrication of complex vasculatures in cell culture platforms, labs- and organs-on-chips, and tissue engineering scaffolds, highlighting their advantages and disadvantages. Finally, challenges and future directions are outlined with the hope of inspiring researchers to create the reliable, efficient and sustainable tools needed for design and biofabrication of complex vasculatures.
Collapse
Affiliation(s)
- Alisa Morss Clyne
- Vascular Kinetics Laboratory, Mechanical Engineering & Mechanics, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, United States of America
| | | | | |
Collapse
|
24
|
Starkov P, Aguilera TA, Golden DI, Shultz DB, Trakul N, Maxim PG, Le QT, Loo BW, Diehn M, Depeursinge A, Rubin DL. The use of texture-based radiomics CT analysis to predict outcomes in early-stage non-small cell lung cancer treated with stereotactic ablative radiotherapy. Br J Radiol 2018; 92:20180228. [PMID: 30457885 DOI: 10.1259/bjr.20180228] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE: Stereotactic ablative radiotherapy (SABR) is being increasingly used as a non-invasive treatment for early-stage non-small cell lung cancer (NSCLC). A non-invasive method to estimate treatment outcomes in these patients would be valuable, especially since access to tissue specimens is often difficult in these cases. METHODS: We developed a method to predict survival following SABR in NSCLC patients using analysis of quantitative image features on pre-treatment CT images. We developed a Cox Lasso model based on two-dimensional Riesz wavelet quantitative texture features on CT scans with the goal of separating patients based on survival. RESULTS: The median log-rank p-value for 1000 cross-validations was 0.030. Our model was able to separate patients based upon predicted survival. When we added tumor size into the model, the p-value lost its significance, demonstrating that tumor size is not a key feature in the model but rather decreases significance likely due to the relatively small number of events in the dataset. Furthermore, running the model using Riesz features extracted either from the solid component of the tumor or from the ground glass opacity (GGO) component of the tumor maintained statistical significance. However, the p-value improved when combining features from the solid and the GGO components, demonstrating that there are important data that can be extracted from the entire tumor. CONCLUSIONS: The model predicting patient survival following SABR in NSCLC may be useful in future studies by enabling prediction of survival-based outcomes using radiomics features in CT images. ADVANCES IN KNOWLEDGE: Quantitative image features from NSCLC nodules on CT images have been found to significantly separate patient populations based on overall survival (p = 0.04). In the long term, a non-invasive method to estimate treatment outcomes in patients undergoing SABR would be valuable, especially since access to tissue specimens is often difficult in these cases.
Collapse
Affiliation(s)
- Pierre Starkov
- 1 Deparment of Signal Processing & Control, Systems, Centre Suisse d'Electronique et de Microtechnique , Neuchâtel , Switzerland
| | - Todd A Aguilera
- 2 Department of Radiation Oncology, UT Southwestern Medical Center , Dallas, TX , USA
| | - Daniel I Golden
- 3 Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University School of Medicine , Stanford, CA , USA
| | - David B Shultz
- 4 Department of Radiation Oncology, Princess Margaret Cancer Centre , Toronto, ON , Canada
| | - Nicholas Trakul
- 5 Department of Radiation Oncology, Stanford Cancer Institute and Stanford University School of Medicine , Stanford, CA , USA
| | - Peter G Maxim
- 5 Department of Radiation Oncology, Stanford Cancer Institute and Stanford University School of Medicine , Stanford, CA , USA
| | - Quynh-Thu Le
- 5 Department of Radiation Oncology, Stanford Cancer Institute and Stanford University School of Medicine , Stanford, CA , USA
| | - Billy W Loo
- 5 Department of Radiation Oncology, Stanford Cancer Institute and Stanford University School of Medicine , Stanford, CA , USA
| | - Maximillan Diehn
- 5 Department of Radiation Oncology, Stanford Cancer Institute and Stanford University School of Medicine , Stanford, CA , USA
| | - Adrien Depeursinge
- 6 Department of Signal Processing & Control, Systems, Biomedical Imaging Group, École Polytechnique Fédérale de Lausanne , Lausanne , Switzerland.,7 Department of Signal Processing & Control, Systems, Institute of Information Systems, University of Applied Sciences Western Switzerland (HES-SO) , Sierre , Switzerland
| | - Daniel L Rubin
- 3 Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University School of Medicine , Stanford, CA , USA
| |
Collapse
|
25
|
Cook GJR, Azad G, Owczarczyk K, Siddique M, Goh V. Challenges and Promises of PET Radiomics. Int J Radiat Oncol Biol Phys 2018; 102:1083-1089. [PMID: 29395627 PMCID: PMC6278749 DOI: 10.1016/j.ijrobp.2017.12.268] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/14/2017] [Indexed: 01/09/2023]
Abstract
PURPOSE Radiomics describes the extraction of multiple, otherwise invisible, features from medical images that, with bioinformatic approaches, can be used to provide additional information that can predict underlying tumor biology and behavior. METHODS AND MATERIALS Radiomic signatures can be used alone or with other patient-specific data to improve tumor phenotyping, treatment response prediction, and prognosis, noninvasively. The data describing 18F-fluorodeoxyglucose positron emission tomography radiomics, often using texture or heterogeneity parameters, are increasing rapidly. RESULTS In relation to radiation therapy practice, early data have reported the use of radiomic approaches to better define tumor volumes and predict radiation toxicity and treatment response. CONCLUSIONS Although at an early stage of development, with many technical challenges remaining and a need for standardization, promise nevertheless exists that PET radiomics will contribute to personalized medicine, especially with the availability of increased computing power and the development of machine-learning approaches for imaging.
Collapse
Affiliation(s)
- Gary J R Cook
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom.
| | - Gurdip Azad
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Kasia Owczarczyk
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Musib Siddique
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| |
Collapse
|
26
|
Contrast-Enhanced 3-T Perfusion MRI With Quantitative Analysis for the Characterization of Musculoskeletal Tumors: Is It Worth the Trouble? AJR Am J Roentgenol 2018; 211:1092-1098. [DOI: 10.2214/ajr.18.19618] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
27
|
Vallières M, Serban M, Benzyane I, Ahmed Z, Xing S, El Naqa I, Levesque IR, Seuntjens J, Freeman CR. Investigating the role of functional imaging in the management of soft-tissue sarcomas of the extremities. PHYSICS & IMAGING IN RADIATION ONCOLOGY 2018; 6:53-60. [PMID: 33458389 PMCID: PMC7807871 DOI: 10.1016/j.phro.2018.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 04/06/2018] [Accepted: 05/08/2018] [Indexed: 11/16/2022]
Abstract
Background and purpose In this work, we validate a texture-based model computed from positron emission tomography (PET) and magnetic resonance imaging (MRI) for the prediction of lung metastases in soft-tissue sarcomas (STS). We explore functional imaging at different treatment time points and evaluate the feasibility of radiotherapy dose painting as a potential treatment strategy for patients with higher metastatic risk. Materials and methods We acquired fluorodeoxyglucose (FDG)-PET, fluoromisonidazole (FMISO)-PET, diffusion weighting (DW)-MRI and dynamic contrast enhanced (DCE)-MRI data for 18 patients with extremity STS before, during, and after pre-operative radiotherapy. We tested the lung metastases prediction model using pre-treatment images. We evaluated the feasibility of dose painting using volumetric arc therapy (VMAT) via treatment re-planning with a prescription of 50 Gy to the planning target volume (PTV50Gy) and boost doses of 60 Gy to the FDG hypermetabolic gross tumour volume (GTV60Gy) and 65 Gy to the low-perfusion DCE-MRI hypoxic GTV contained within the GTV60Gy (GTV65Gy). Results The texture-based model for lung metastases prediction reached an area under the curve (AUC), sensitivity, specificity and accuracy of 0.71, 0.75, 0.85 and 0.82, respectively. Dose painting resulted in adequate coverage and homogeneity in the re-planned treatments: D95% to the PTV50Gy, GTV60Gy and GTV65Gy were 50.0 Gy, 60.3 Gy and 65.4 Gy, respectively. Conclusions Textural biomarkers extracted from FDG-PET and MRI could be useful to identify STS patients that might benefit from dose escalation. The feasibility of treatment planning with double boost levels to intratumoural GTV functional sub-volumes was established.
Collapse
Affiliation(s)
- Martin Vallières
- Medical Physics Unit, McGill University, Cedars Cancer Centre, McGill University Health Centre – Glen Site, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
- Corresponding author.
| | - Monica Serban
- Medical Physics Unit, McGill University, Cedars Cancer Centre, McGill University Health Centre – Glen Site, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
| | - Ibtissam Benzyane
- Medical Physics Unit, McGill University, Cedars Cancer Centre, McGill University Health Centre – Glen Site, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
| | - Zaki Ahmed
- Medical Physics Unit, McGill University, Cedars Cancer Centre, McGill University Health Centre – Glen Site, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
| | - Shu Xing
- Medical Physics Unit, McGill University, Cedars Cancer Centre, McGill University Health Centre – Glen Site, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
| | - Issam El Naqa
- Department of Radiation Oncology, Physics Division, University of Michigan, 519 W. Williman St. Argus Bldg, Ann Arbor, MI 48103-4943, USA
| | - Ives R. Levesque
- Medical Physics Unit, McGill University, Cedars Cancer Centre, McGill University Health Centre – Glen Site, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
- Research Institute of the McGill University Health Centre, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
| | - Jan Seuntjens
- Medical Physics Unit, McGill University, Cedars Cancer Centre, McGill University Health Centre – Glen Site, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
- Research Institute of the McGill University Health Centre, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
| | - Carolyn R. Freeman
- Research Institute of the McGill University Health Centre, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
- Department of Radiation Oncology, Cedars Cancer Centre, McGill University Health Centre – Glen Site, 1001 boulevard Décarie, Montréal, QC H4A 3J1, Canada
| |
Collapse
|
28
|
Jethanandani A, Lin TA, Volpe S, Elhalawani H, Mohamed ASR, Yang P, Fuller CD. Exploring Applications of Radiomics in Magnetic Resonance Imaging of Head and Neck Cancer: A Systematic Review. Front Oncol 2018; 8:131. [PMID: 29868465 PMCID: PMC5960677 DOI: 10.3389/fonc.2018.00131] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/10/2018] [Indexed: 01/07/2023] Open
Abstract
Background Radiomics has been widely investigated for non-invasive acquisition of quantitative textural information from anatomic structures. While the vast majority of radiomic analysis is performed on images obtained from computed tomography, magnetic resonance imaging (MRI)-based radiomics has generated increased attention. In head and neck cancer (HNC), however, attempts to perform consistent investigations are sparse, and it is unclear whether the resulting textural features can be reproduced. To address this unmet need, we systematically reviewed the quality of existing MRI radiomics research in HNC. Methods Literature search was conducted in accordance with guidelines established by Preferred Reporting Items for Systematic Reviews and Meta-Analyses. Electronic databases were examined from January 1990 through November 2017 for common radiomic keywords. Eligible completed studies were then scored using a standardized checklist that we developed from Enhancing the Quality and Transparency of Health Research guidelines for reporting machine-learning predictive model specifications and results in biomedical research, defined by Luo et al. (1). Descriptive statistics of checklist scores were populated, and a subgroup analysis of methodology items alone was conducted in comparison to overall scores. Results Sixteen completed studies and four ongoing trials were selected for inclusion. Of the completed studies, the nasopharynx was the most common site of study (37.5%). MRI modalities varied with only four of the completed studies (25%) extracting radiomic features from a single sequence. Study sample sizes ranged between 13 and 118 patients (median of 40), and final radiomic signatures ranged from 2 to 279 features. Analyzed endpoints included either segmentation or histopathological classification parameters (44%) or prognostic and predictive biomarkers (56%). Liu et al. (2) addressed the highest number of our checklist items (total score: 48), and a subgroup analysis of methodology checklist items alone did not demonstrate any difference in scoring trends between studies [Spearman’s ρ = 0.94 (p < 0.0001)]. Conclusion Although MRI radiomic applications demonstrate predictive potential in analyzing diverse HNC outcomes, methodological variances preclude accurate and collective interpretation of data.
Collapse
Affiliation(s)
- Amit Jethanandani
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Timothy A Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Baylor College of Medicine, Houston, TX, United States
| | - Stefania Volpe
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Hesham Elhalawani
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Abdallah S R Mohamed
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, University of Alexandria, Alexandria, Egypt.,Graduate School of Biomedical Sciences, The University of Texas Health Science Center, Houston, TX, United States
| | - Pei Yang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Hunan Cancer Hospital, Department of Head and Neck Radiation Oncology, Changsha, China
| | - Clifton D Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Graduate School of Biomedical Sciences, The University of Texas Health Science Center, Houston, TX, United States
| |
Collapse
|
29
|
Cai WL, Hong GB. Quantitative image analysis for evaluation of tumor response in clinical oncology. Chronic Dis Transl Med 2018; 4:18-28. [PMID: 29756120 PMCID: PMC5938243 DOI: 10.1016/j.cdtm.2018.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Indexed: 12/13/2022] Open
Abstract
The objective, accurate, and standardized evaluation of tumor response to treatment is an indispensable procedure in clinical oncology. Compared to manual measurement, computer-assisted linear measurement can significantly improve the accuracy and reproducibility of tumor burden quantification. For irregular-shaped and infiltrating or diffuse tumors, which are difficult to quantify by linear measurement, computer-assisted volumetric measurement may provide a more objective and sensitive quantification to evaluate tumor response to treatment than linear measurement does. In the evaluation of tumor response to novel oncologic treatments such as targeted therapy, changes in overall tumor size do not necessarily reflect tumor response to therapy due to the presence of internal necrosis or hemorrhages. This leads to a new generation of imaging biomarkers to evaluate tumor response by using texture analysis methods, also called radiomics. Computer-assisted texture analysis technology offers a more comprehensive and in-depth imaging biomarker to evaluate tumor response. The application of computer-assisted quantitative imaging analysis techniques not only reduces the inaccuracy and improves the reliability in tumor burden quantification, but facilitates the development of more comprehensive and intelligent approaches to evaluate treatment response, and hence promotes precision imaging in the evaluation of tumor response in clinical oncology. This article summarizes the state-of-the-art technical developments and clinical applications of quantitative imaging analysis in evaluation of tumor response in clinical oncology.
Collapse
Affiliation(s)
- Wen-Li Cai
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Guo-Bin Hong
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| |
Collapse
|
30
|
Li Y, Qiao Y, Chen H, Bai R, Staedtke V, Han Z, Xu J, Chan KWY, Yadav N, Bulte JWM, Zhou S, van Zijl PCM, Liu G. Characterization of tumor vascular permeability using natural dextrans and CEST MRI. Magn Reson Med 2017; 79:1001-1009. [PMID: 29193288 DOI: 10.1002/mrm.27014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/10/2017] [Accepted: 10/26/2017] [Indexed: 12/30/2022]
Abstract
PURPOSE To investigate the use of natural dextrans as nano-sized chemical exchange saturation transfer (CEST) MRI probes for characterizing size-dependent tumor vascular permeability. METHODS Dextrans of different molecular weight (10, 70, 150, and 2000 kD) were characterized for their CEST contrast. Mice (N = 5) bearing CT26 subcutaneous colon tumors were injected intravenously with 10 kD (D10, 6 nm) and 70 kD (D70, 12 nm) dextran at a dose of 375 mg/kg. The CEST-MRI signal in the tumors was assessed before and approximately 40 min after each injection using a dynamic CEST imaging scheme. RESULTS All dextrans of different molecular weights have a strong CEST signal with an apparent maximum of approximately 0.9 ppm. The detectability and effects of pH and saturation conditions (B1 and Tsat ) were investigated. When applied to CT26 tumors, the injection of D10 could produce a significant "dexCEST" enhancement in the majority of the tumor area, whereas the injection of D70 only resulted in an increase in the tumor periphery. Quantitative analysis revealed the differential permeability of CT26 tumors to different size particles, which was validated by fluorescence imaging and immunohistochemistry. CONCLUSIONS As a first application, we used 10- and 70-kD dextrans to visualize the spatially variable, size-dependent permeability in the tumor, indicating that nano-sized dextrans can be used for characterizing tumor vascular permeability with dexCEST MRI and, potentially, for developing dextran-based theranostic drug delivery systems. Magn Reson Med 79:1001-1009, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Yuguo Li
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yuan Qiao
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hanwei Chen
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Radiology, Panyu Central Hospital, Guangzhou, China
| | - Renyuan Bai
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Verena Staedtke
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zheng Han
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiadi Xu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kannie W Y Chan
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Nirbhay Yadav
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeff W M Bulte
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter C M van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guanshu Liu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
31
|
Cancer Metabolism and Tumor Heterogeneity: Imaging Perspectives Using MR Imaging and Spectroscopy. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:6053879. [PMID: 29114178 PMCID: PMC5654284 DOI: 10.1155/2017/6053879] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 07/31/2017] [Accepted: 08/27/2017] [Indexed: 12/26/2022]
Abstract
Cancer cells reprogram their metabolism to maintain viability via genetic mutations and epigenetic alterations, expressing overall dynamic heterogeneity. The complex relaxation mechanisms of nuclear spins provide unique and convertible tissue contrasts, making magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) pertinent imaging tools in both clinics and research. In this review, we summarized MR methods that visualize tumor characteristics and its metabolic phenotypes on an anatomical, microvascular, microstructural, microenvironmental, and metabolomics scale. The review will progress from the utilities of basic spin-relaxation contrasts in cancer imaging to more advanced imaging methods that measure tumor-distinctive parameters such as perfusion, water diffusion, magnetic susceptibility, oxygenation, acidosis, redox state, and cell death. Analytical methods to assess tumor heterogeneity are also reviewed in brief. Although the clinical utility of tumor heterogeneity from imaging is debatable, the quantification of tumor heterogeneity using functional and metabolic MR images with development of robust analytical methods and improved MR methods may offer more critical roles of tumor heterogeneity data in clinics. MRI/MRS can also provide insightful information on pharmacometabolomics, biomarker discovery, disease diagnosis and prognosis, and treatment response. With these future directions in mind, we anticipate the widespread utilization of these MR-based techniques in studying in vivo cancer biology to better address significant clinical needs.
Collapse
|
32
|
Gourtsoyianni S, Doumou G, Prezzi D, Taylor B, Stirling JJ, Taylor NJ, Siddique M, Cook GJR, Glynne-Jones R, Goh V. Primary Rectal Cancer: Repeatability of Global and Local-Regional MR Imaging Texture Features. Radiology 2017; 284:552-561. [PMID: 28481194 PMCID: PMC6150741 DOI: 10.1148/radiol.2017161375] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Purpose To assess the day-to-day repeatability of global and local-regional magnetic resonance (MR) imaging texture features derived from primary rectal cancer. Materials and Methods After ethical approval and patient informed consent were obtained, two pretreatment T2-weighted axial MR imaging studies performed prospectively with the same imaging unit on 2 consecutive days in 14 patients with rectal cancer (11 men [mean age, 61.7 years], three women [mean age, 70.0 years]) were analyzed to extract (a) global first-order statistical histogram and model-based fractal features reflecting the whole-tumor voxel intensity histogram distribution and repeating patterns, respectively, without spatial information and (b) local-regional second-order and high-order statistical texture features reflecting the intensity and spatial interrelationships between adjacent in-plane or multiplanar voxels or regions, respectively. Repeatability was assessed for 46 texture features, and mean difference, 95% limits of agreement, within-subject coefficient of variation (wCV), and repeatability coefficient (r) were recorded. Results Repeatability was better for global parameters than for most local-regional parameters. In particular, histogram mean, median, and entropy, fractal dimension mean and standard deviation, and second-order entropy, homogeneity, difference entropy, and inverse difference moment demonstrated good repeatability, with narrow limits of agreement and wCVs of 10% or lower. Repeatability was poorest for the following high-order gray-level run-length (GLRL) gray-level zone size matrix (GLZSM) and neighborhood gray-tone difference matrix (NGTDM) parameters: GLRL intensity variability, GLZSM short-zone emphasis, GLZSM intensity nonuniformity, GLZSM intensity variability, GLZSM size zone variability, and NGTDM complexity, demonstrating wider agreement limits and wCVs of 50% or greater. Conclusion MR imaging repeatability is better for global texture parameters than for local-regional texture parameters, indicating that global texture parameters should be sufficiently robust for clinical practice. Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Sofia Gourtsoyianni
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| | - Georgia Doumou
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| | - Davide Prezzi
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| | - Benjamin Taylor
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| | - J. James Stirling
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| | - N. Jane Taylor
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| | - Musib Siddique
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| | - Gary J. R. Cook
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| | - Robert Glynne-Jones
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| | - Vicky Goh
- From the Department of Radiology (S.G., D.P., V.G.) and PET Centre (J.J.S., G.J.R.C.), Guy’s and St Thomas’ Hospitals NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH; Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, England (G.D., D.P., B.T., J.J.S., M.S., G.J.R.C., V.G.); and the Cancer Centre, Mount Vernon Hospital, Northwood, England (N.J.T., R.G.)
| |
Collapse
|
33
|
Sujlana P, Skrok J, Fayad LM. Review of dynamic contrast‐enhanced MRI: Technical aspects and applications in the musculoskeletal system. J Magn Reson Imaging 2017; 47:875-890. [DOI: 10.1002/jmri.25810] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022] Open
Affiliation(s)
- Parvinder Sujlana
- The Russell H. Morgan Department of Radiology and Radiological ScienceBaltimore Maryland USA
| | - Jan Skrok
- The Russell H. Morgan Department of Radiology and Radiological ScienceBaltimore Maryland USA
| | - Laura M. Fayad
- The Russell H. Morgan Department of Radiology and Radiological ScienceBaltimore Maryland USA
| |
Collapse
|
34
|
Valdés Hernández MDC, González-Castro V, Chappell FM, Sakka E, Makin S, Armitage PA, Nailon WH, Wardlaw JM. Application of Texture Analysis to Study Small Vessel Disease and Blood-Brain Barrier Integrity. Front Neurol 2017; 8:327. [PMID: 28769863 PMCID: PMC5515862 DOI: 10.3389/fneur.2017.00327] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/22/2017] [Indexed: 11/13/2022] Open
Abstract
Objectives We evaluate the alternative use of texture analysis for evaluating the role of blood–brain barrier (BBB) in small vessel disease (SVD). Methods We used brain magnetic resonance imaging from 204 stroke patients, acquired before and 20 min after intravenous gadolinium administration. We segmented tissues, white matter hyperintensities (WMH) and applied validated visual scores. We measured textural features in all tissues pre- and post-contrast and used ANCOVA to evaluate the effect of SVD indicators on the pre-/post-contrast change, Kruskal–Wallis for significance between patient groups and linear mixed models for pre-/post-contrast variations in cerebrospinal fluid (CSF) with Fazekas scores. Results Textural “homogeneity” increase in normal tissues with higher presence of SVD indicators was consistently more overt than in abnormal tissues. Textural “homogeneity” increased with age, basal ganglia perivascular spaces scores (p < 0.01) and SVD scores (p < 0.05) and was significantly higher in hypertensive patients (p < 0.002) and lacunar stroke (p = 0.04). Hypertension (74% patients), WMH load (median = 1.5 ± 1.6% of intracranial volume), and age (mean = 65.6 years, SD = 11.3) predicted the pre/post-contrast change in normal white matter, WMH, and index stroke lesion. CSF signal increased with increasing SVD post-contrast. Conclusion A consistent general pattern of increasing textural “homogeneity” with increasing SVD and post-contrast change in CSF with increasing WMH suggest that texture analysis may be useful for the study of BBB integrity.
Collapse
Affiliation(s)
- Maria Del C Valdés Hernández
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Victor González-Castro
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Francesca M Chappell
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Eleni Sakka
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen Makin
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Paul A Armitage
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - William H Nailon
- Department of Oncology Physics, University of Edinburgh, Edinburgh, United Kingdom
| | - Joanna M Wardlaw
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
35
|
Haralick texture features from apparent diffusion coefficient (ADC) MRI images depend on imaging and pre-processing parameters. Sci Rep 2017. [PMID: 28642480 PMCID: PMC5481454 DOI: 10.1038/s41598-017-04151-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In recent years, texture analysis of medical images has become increasingly popular in studies investigating diagnosis, classification and treatment response assessment of cancerous disease. Despite numerous applications in oncology and medical imaging in general, there is no consensus regarding texture analysis workflow, or reporting of parameter settings crucial for replication of results. The aim of this study was to assess how sensitive Haralick texture features of apparent diffusion coefficient (ADC) MR images are to changes in five parameters related to image acquisition and pre-processing: noise, resolution, how the ADC map is constructed, the choice of quantization method, and the number of gray levels in the quantized image. We found that noise, resolution, choice of quantization method and the number of gray levels in the quantized images had a significant influence on most texture features, and that the effect size varied between different features. Different methods for constructing the ADC maps did not have an impact on any texture feature. Based on our results, we recommend using images with similar resolutions and noise levels, using one quantization method, and the same number of gray levels in all quantized images, to make meaningful comparisons of texture feature results between different subjects.
Collapse
|
36
|
Cao J, Xiao L, He B, Zhang G, Dong J, Wu Y, Xie H, Wang G, Lin X. Diagnostic value of combined diffusion-weighted imaging with dynamic contrast enhancement MRI in differentiating malignant from benign bone lesions. Clin Radiol 2017; 72:793.e1-793.e9. [PMID: 28545685 DOI: 10.1016/j.crad.2017.04.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 12/26/2016] [Accepted: 04/18/2017] [Indexed: 11/30/2022]
Abstract
AIM To determine the diagnostic value of combined diffusion-weighted imaging (DWI) with dynamic contrast enhancement magnetic resonance imaging (DCE-MRI) in differentiating malignant from benign bone lesions. MATERIALS AND METHODS DWI and DCE-MRI were performed in 36 patients (14 were benign and 22 were malignant). The mean apparent diffusion coefficient (ADC) values and signal enhanced extent (SEE), slope value, and time-signal intensity curve (TIC) type were recorded by two observers. Between-group comparison was made using the independent sample t-test and receiver-operating characteristic (ROC) analysis. RESULTS There was a significant difference between the mean ADC value of the benign ([1.75±0.50]×10-3 mm2/s) and malignant ([1.11±0.47]×10-3 mm2/s) groups (p=0.001). The threshold ADC value of ≤1.10×10-3 mm2/s resulted in a sensitivity of 77.3%, a specificity of 92.9%, and an accuracy of 85.1%. A type III curve was found in 23 cases (21 malignant and two benign), a type II curve was seen in six cases (one malignant and five benign), and a type I curve in seven cases (all were benign). The SEE and slope values in the benign and malignant groups were 227.96±172.08, 325.60±125.86 (p=0.058); 0.97±0.67%/s, 3.19±3.20%/s (p=0.016), respectively. ROC analysis showed a sensitivity of 95.5%, a specificity of 85.7%, and an accuracy of 90.6% for malignancy, based on a slope cut-off value of >1.46%/s. Combining ADC and slope values resulted in a sensitivity of 100%, a specificity of 85.7%, and an accuracy of 92.9%. CONCLUSIONS Both DWI and DCE-MRI showed promising results for differentiating malignant from benign bone lesions. A combination of DWI and DCE-MRI was the most valuable of the three.
Collapse
Affiliation(s)
- J Cao
- Shandong Medical Imaging Research Institute, Shandong University, No. 44 West Wenhua Road, Jinan, 250012 PR China; Central Hospital of Zibo, No. 54 West Gongqingtuan Road, Zibo, 255020 PR China
| | - L Xiao
- Shandong Medical Imaging Research Institute, Shandong University, No. 44 West Wenhua Road, Jinan, 250012 PR China
| | - B He
- Central Hospital of Zibo, No. 54 West Gongqingtuan Road, Zibo, 255020 PR China
| | - G Zhang
- Central Hospital of Zibo, No. 54 West Gongqingtuan Road, Zibo, 255020 PR China
| | - J Dong
- Shandong Medical Imaging Research Institute, Shandong University, No. 44 West Wenhua Road, Jinan, 250012 PR China
| | - Y Wu
- Shandong Medical Imaging Research Institute, Shandong University, No. 44 West Wenhua Road, Jinan, 250012 PR China
| | - H Xie
- Shandong Medical Imaging Research Institute, Shandong University, No. 44 West Wenhua Road, Jinan, 250012 PR China
| | - G Wang
- Shandong Medical Imaging Research Institute, Shandong University, No. 44 West Wenhua Road, Jinan, 250012 PR China
| | - X Lin
- Shandong Medical Imaging Research Institute, Shandong University, No. 44 West Wenhua Road, Jinan, 250012 PR China.
| |
Collapse
|
37
|
Fite BZ, Kheirolomoom A, Foiret JL, Seo JW, Mahakian LM, Ingham ES, Tam SM, Borowsky AD, Curry FRE, Ferrara KW. Dynamic contrast enhanced MRI detects changes in vascular transport rate constants following treatment with thermally-sensitive liposomal doxorubicin. J Control Release 2017; 256:203-213. [PMID: 28395970 DOI: 10.1016/j.jconrel.2017.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 03/16/2017] [Accepted: 04/05/2017] [Indexed: 01/03/2023]
Abstract
Temperature-sensitive liposomal formulations of chemotherapeutics, such as doxorubicin, can achieve locally high drug concentrations within a tumor and tumor vasculature while maintaining low systemic toxicity. Further, doxorubicin delivery by temperature-sensitive liposomes can reliably cure local cancer in mouse models. Histological sections of treated tumors have detected red blood cell extravasation within tumors treated with temperature-sensitive doxorubicin and ultrasound hyperthermia. We hypothesize that the local release of drug into the tumor vasculature and resulting high drug concentration can alter vascular transport rate constants along with having direct tumoricidal effects. Dynamic contrast enhanced MRI (DCE-MRI) coupled with a pharmacokinetic model can detect and quantify changes in such vascular transport rate constants. Here, we set out to determine whether changes in rate constants resulting from intravascular drug release were detectable by MRI. We found that the accumulation of gadoteridol was enhanced in tumors treated with temperature-sensitive liposomal doxorubicin and ultrasound hyperthermia. While the initial uptake rate of the small molecule tracer was slower (k1=0.0478±0.011s-1 versus 0.116±0.047s-1) in treated compared to untreated tumors, the tracer was retained after treatment due to a larger reduction in the rate of clearance (k2=0.291±0.030s-1 versus 0.747±0.24s-1). While DCE-MRI assesses a combination of blood flow and permeability, ultrasound imaging of microvascular flow rate is sensitive only to changes in vascular flow rate; based on this technique, blood flow was not significantly altered 30min after treatment. In summary, DCE-MRI provides a means to detect changes that are associated with treatment by thermally-activated particles and such changes can be exploited to enhance local delivery.
Collapse
Affiliation(s)
- Brett Z Fite
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Azadeh Kheirolomoom
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Josquin L Foiret
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Jai W Seo
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA 95616, USA.
| | - Fitz-Roy E Curry
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA.
| | - Katherine W Ferrara
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| |
Collapse
|
38
|
Scalco E, Rizzo G. Texture analysis of medical images for radiotherapy applications. Br J Radiol 2017; 90:20160642. [PMID: 27885836 PMCID: PMC5685100 DOI: 10.1259/bjr.20160642] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/27/2016] [Accepted: 11/16/2016] [Indexed: 12/29/2022] Open
Abstract
The high-throughput extraction of quantitative information from medical images, known as radiomics, has grown in interest due to the current necessity to quantitatively characterize tumour heterogeneity. In this context, texture analysis, consisting of a variety of mathematical techniques that can describe the grey-level patterns of an image, plays an important role in assessing the spatial organization of different tissues and organs. For these reasons, the potentiality of texture analysis in the context of radiotherapy has been widely investigated in several studies, especially for the prediction of the treatment response of tumour and normal tissues. Nonetheless, many different factors can affect the robustness, reproducibility and reliability of textural features, thus limiting the impact of this technique. In this review, an overview of the most recent works that have applied texture analysis in the context of radiotherapy is presented, with particular focus on the assessment of tumour and tissue response to radiations. Preliminary, the main factors that have an influence on features estimation are discussed, highlighting the need of more standardized image acquisition and reconstruction protocols and more accurate methods for region of interest identification. Despite all these limitations, texture analysis is increasingly demonstrating its ability to improve the characterization of intratumour heterogeneity and the prediction of clinical outcome, although prospective studies and clinical trials are required to draw a more complete picture of the full potential of this technique.
Collapse
Affiliation(s)
- Elisa Scalco
- Institute of Molecular Bioimaging and Physiology (IBFM), Italian
National Research Council (CNR), Milan, Italy
| | - Giovanna Rizzo
- Institute of Molecular Bioimaging and Physiology (IBFM), Italian
National Research Council (CNR), Milan, Italy
| |
Collapse
|
39
|
Wang C, Subashi E, Yin FF, Chang Z. Dynamic fractal signature dissimilarity analysis for therapeutic response assessment using dynamic contrast-enhanced MRI. Med Phys 2016; 43:1335-47. [PMID: 26936718 DOI: 10.1118/1.4941739] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To develop a dynamic fractal signature dissimilarity (FSD) method as a novel image texture analysis technique for the quantification of tumor heterogeneity information for better therapeutic response assessment with dynamic contrast-enhanced (DCE)-MRI. METHODS A small animal antiangiogenesis drug treatment experiment was used to demonstrate the proposed method. Sixteen LS-174T implanted mice were randomly assigned into treatment and control groups (n = 8/group). All mice received bevacizumab (treatment) or saline (control) three times in two weeks, and one pretreatment and two post-treatment DCE-MRI scans were performed. In the proposed dynamic FSD method, a dynamic FSD curve was generated to characterize the heterogeneity evolution during the contrast agent uptake, and the area under FSD curve (AUCFSD) and the maximum enhancement (MEFSD) were selected as representative parameters. As for comparison, the pharmacokinetic parameter K(trans) map and area under MR intensity enhancement curve AUCMR map were calculated. Besides the tumor's mean value and coefficient of variation, the kurtosis, skewness, and classic Rényi dimensions d1 and d2 of K(trans) and AUCMR maps were evaluated for heterogeneity assessment for comparison. For post-treatment scans, the Mann-Whitney U-test was used to assess the differences of the investigated parameters between treatment/control groups. The support vector machine (SVM) was applied to classify treatment/control groups using the investigated parameters at each post-treatment scan day. RESULTS The tumor mean K(trans) and its heterogeneity measurements d1 and d2 values showed significant differences between treatment/control groups in the second post-treatment scan. In contrast, the relative values (in reference to the pretreatment value) of AUCFSD and MEFSD in both post-treatment scans showed significant differences between treatment/control groups. When using AUCFSD and MEFSD as SVM input for treatment/control classification, the achieved accuracies were 93.8% and 93.8% at first and second post-treatment scan days, respectively. In comparison, the classification accuracies using d1 and d2 of K(trans) map were 87.5% and 100% at first and second post-treatment scan days, respectively. CONCLUSIONS As quantitative metrics of tumor contrast agent uptake heterogeneity, the selected parameters from the dynamic FSD method accurately captured the therapeutic response in the experiment. The potential application of the proposed method is promising, and its addition to the existing DCE-MRI techniques could improve DCE-MRI performance in early assessment of treatment response.
Collapse
Affiliation(s)
- Chunhao Wang
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710
| | - Ergys Subashi
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710
| | - Fang-Fang Yin
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710
| | - Zheng Chang
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
40
|
Ting-Fang Shih T. Angiogenesis in hematological malignancy – Evaluated by dynamic contrast-enhanced MRI. JOURNAL OF CANCER RESEARCH AND PRACTICE 2016. [DOI: 10.1016/j.jcrpr.2016.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
41
|
Jansen JFA, Parra C, Lu Y, Shukla-Dave A. Evaluation of Head and Neck Tumors with Functional MR Imaging. Magn Reson Imaging Clin N Am 2016; 24:123-133. [PMID: 26613878 DOI: 10.1016/j.mric.2015.08.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Head and neck cancer is one of the most common cancers worldwide. MR imaging-based diffusion and perfusion techniques enable the noninvasive assessment of tumor biology and physiology, which supplement information obtained from standard structural scans. Diffusion and perfusion MR imaging techniques provide novel biomarkers that can aid monitoring in pretreatment, during treatment, and posttreatment stages to improve patient selection for therapeutic strategies; provide evidence for change of therapy regime; and evaluate treatment response. This review discusses pertinent aspects of the role of diffusion and perfusion MR imaging and computational analysis methods in studying head and neck cancer.
Collapse
Affiliation(s)
- Jacobus F A Jansen
- Department of Radiology, Maastricht University Medical Center, PO Box 5800, Maastricht 6202 AZ, The Netherlands.
| | - Carlos Parra
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Yonggang Lu
- Department of Radiation Oncology, University of Washington, 4921 Parkview Pl, St Louis, MO 63110, USA
| | - Amita Shukla-Dave
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
42
|
Wittenborn TR, Nielsen T, Thomsen JS, Horsman MR, Nygaard JV. Simulation of heterogeneous molecular delivery in tumours using μCT reconstructions and MRI validation. Microvasc Res 2016; 108:69-74. [PMID: 27569845 DOI: 10.1016/j.mvr.2016.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 08/24/2016] [Accepted: 08/24/2016] [Indexed: 10/21/2022]
Abstract
PRIMARY OBJECTIVE Utilizing the detailed vascular network obtained from micro-computed tomography (μCT) to establish a mathematical model of the temporal molecular distribution within a murine C3H mammary carcinoma. PROCEDURES Female CDF1 mice with a C3H mammary carcinoma on the right rear foot were used in this study. Dynamic information for each tumour was achieved by Dynamic Contrast Enhanced-Magnetic Resonance Imaging (DCE-MRI) on a 16.4 T system. Detailed morphologic information on the tumour vasculature was obtained by ex vivo μCT and compared to CD34 immunohistochemical staining of tissue sections. The reconstructed vascular network served as origin for the diffusion (described by the apparent diffusion coefficient) within the tumour (the restricted volume described by the interstitial volume fraction derived from DCE-MRI). The resulting partial differential equation was solved using Finite-Element and a combined mathematical graph describing molecular distribution within the tumour was obtained. RESULTS The established molecular distribution model predicted a heterogeneous distribution throughout the tumour related to the layout of the vascular network. Central tumour section concentration-time curves estimated from the established molecular distribution model were compared with physical measurements obtained by DCE-MRI of the same tumours and showed excellent correlation. CONCLUSIONS A mathematical model describing temporal molecular distribution based on detailed vascular network structures was established and compared to DCE-MRI. The improved morphological insight will enhance future studies of heterogeneous tumours.
Collapse
Affiliation(s)
- Thomas Rea Wittenborn
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Noerrebrogade 44, Building 5, 8000 Aarhus C, Denmark.
| | - Thomas Nielsen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Noerrebrogade 44, Building 5, 8000 Aarhus C, Denmark; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds vej 14, 8000 Aarhus C, Denmark; Center of Functionally Integrative Neuroscience, Aarhus University Hospital, Nørrebrogade 44, Building 10G, 8000 Aarhus C, Denmark
| | - Jesper Skovhus Thomsen
- Department of Biomedicine - Anatomy, Aarhus University, Wilhelm Meyers Allé 3, 8000 Aarhus C, Denmark
| | - Michael Robert Horsman
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Noerrebrogade 44, Building 5, 8000 Aarhus C, Denmark
| | - Jens Vinge Nygaard
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds vej 14, 8000 Aarhus C, Denmark; Department of Engineering, Aarhus University, Inge Lehmanns Gade 10, 8000 Aarhus C, Denmark
| |
Collapse
|
43
|
KWONG TIFFANYC, HSING MITCHELL, LIN YUTING, THAYER DAVID, UNLU MEHMETBURCIN, SU MINYING, GULSEN GULTEKIN. Differentiation of tumor vasculature heterogeneity levels in small animals based on total hemoglobin concentration using magnetic resonance-guided diffuse optical tomography in vivo. APPLIED OPTICS 2016; 55:5479-87. [PMID: 27463894 PMCID: PMC6839944 DOI: 10.1364/ao.55.005479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Insight into the vasculature of the tumor in small animals has the potential to impact many areas of cancer research. The heterogeneity of the vasculature of a tumor is directly related to tumor stage and disease progression. In this small scale animal study, we investigated the feasibility of differentiating tumors with different levels of vasculature heterogeneity in vivo using a previously developed hybrid magnetic resonance imaging (MRI) and diffuse optical tomography (DOT) system for small animal imaging. Cross-sectional total hemoglobin concentration maps of 10 Fisher rats bearing R3230 breast tumors are reconstructed using multi-wavelength DOT measurements both with and without magnetic resonance (MR) structural a priori information. Simultaneously acquired MR structural images are used to guide and constrain the DOT reconstruction, while dynamic contrast-enhanced MR functional images are used as the gold standard to classify the vasculature of the tumor into two types: high versus low heterogeneity. These preliminary results show that the stand-alone DOT is unable to differentiate tumors with low and high vascular heterogeneity without structural a priori information provided by a high resolution imaging modality. The mean total hemoglobin concentrations comparing the vasculature of the tumors with low and high heterogeneity are significant (p-value 0.02) only when MR structural a priori information is utilized.
Collapse
Affiliation(s)
- TIFFANY C. KWONG
- Tu and Yuen Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, Irvine, California 92697, USA
| | - MITCHELL HSING
- Tu and Yuen Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, Irvine, California 92697, USA
- Department of Electrical and Electronic Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - YUTING LIN
- Tu and Yuen Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, Irvine, California 92697, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02144, USA
| | - DAVID THAYER
- Tu and Yuen Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, Irvine, California 92697, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, USA
| | | | - MIN-YING SU
- Tu and Yuen Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, Irvine, California 92697, USA
| | - GULTEKIN GULSEN
- Tu and Yuen Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, Irvine, California 92697, USA
- Corresponding author:
| |
Collapse
|
44
|
Wang C, Subashi E, Liang X, Yin FF, Chang Z. Evaluation of the effect of transcytolemmal water exchange analysis for therapeutic response assessment using DCE-MRI: a comparison study. Phys Med Biol 2016; 61:4763-80. [PMID: 27272391 DOI: 10.1088/0031-9155/61/13/4763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This study compares the shutter-speed (SS) and the Tofts models as used in assessing therapeutic response in a longitudinal DCE-MRI experiment. Sixteen nu/nu mice with implanted colorectal adenocarcinoma cell line (LS-174T) were randomly assigned into treatment/control groups (n = 8/group) and received bevacizumab/saline twice weekly (Day1/Day4/Day8). All mice were scanned at one pre- (Day0) and two post-treatment (Day2/Day9) time points using a high spatiotemporal resolution DCE-MRI pulse sequence. The CA extravasation rate constant [Formula: see text] from the Tofts/SS model and the mean intracellular water residence time [Formula: see text] from the SS model were analyzed. A biological subvolume (BV) within the tumor was identified based on the [Formula: see text] intensity distribution, and the SS model parameters within the BV ([Formula: see text] and [Formula: see text]) were analyzed. It is found that [Formula: see text] and [Formula: see text] have a similar spatial distribution in the tumor volume. The Bayesian information criterion results show that the SS model was a better fit for all scans. At Day9, the treatment group had significantly higher tumor mean [Formula: see text] (p = 0.021), [Formula: see text] (p = 0.021) and [Formula: see text] (p = 0.045). When BV from transcytolemmal water exchange analysis was adopted, the treatment group had higher mean [Formula: see text] at both Day2 (p = 0.038) and Day9 (p = 0.007). Additionally, at Day9, the treatment group had higher mean [Formula: see text] (p = 0.045) and higher [Formula: see text] spatial heterogeneity indices (Rényi dimensions) d 1 (p = 0.010) and d 2 (p = 0.021). When mean [Formula: see text] and its coefficient of variation (CV) were used to separate treatment/control group samples using supporting vector machine, the accuracy of treatment/control classification was 68.8% at Day2 and 87.5% at Day9; in contrast, the Day2/Day9 accuracy were 62.5%/87.5% using tumor mean [Formula: see text] and its CV and were 50.0%/87.5% using tumor mean [Formula: see text] and its CV, respectively. These results suggest that the SS model parameters outperformed the Tofts model parameters in terms of capturing bevacizumab therapeutic effect in this longitudinal experiment.
Collapse
Affiliation(s)
- Chunhao Wang
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
45
|
Accelerated Brain DCE-MRI Using Iterative Reconstruction With Total Generalized Variation Penalty for Quantitative Pharmacokinetic Analysis: A Feasibility Study. Technol Cancer Res Treat 2016; 16:446-460. [PMID: 27215931 DOI: 10.1177/1533034616649294] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To investigate the feasibility of using undersampled k-space data and an iterative image reconstruction method with total generalized variation penalty in the quantitative pharmacokinetic analysis for clinical brain dynamic contrast-enhanced magnetic resonance imaging. METHODS Eight brain dynamic contrast-enhanced magnetic resonance imaging scans were retrospectively studied. Two k-space sparse sampling strategies were designed to achieve a simulated image acquisition acceleration factor of 4. They are (1) a golden ratio-optimized 32-ray radial sampling profile and (2) a Cartesian-based random sampling profile with spatiotemporal-regularized sampling density constraints. The undersampled data were reconstructed to yield images using the investigated reconstruction technique. In quantitative pharmacokinetic analysis on a voxel-by-voxel basis, the rate constant Ktrans in the extended Tofts model and blood flow FB and blood volume VB from the 2-compartment exchange model were analyzed. Finally, the quantitative pharmacokinetic parameters calculated from the undersampled data were compared with the corresponding calculated values from the fully sampled data. To quantify each parameter's accuracy calculated using the undersampled data, error in volume mean, total relative error, and cross-correlation were calculated. RESULTS The pharmacokinetic parameter maps generated from the undersampled data appeared comparable to the ones generated from the original full sampling data. Within the region of interest, most derived error in volume mean values in the region of interest was about 5% or lower, and the average error in volume mean of all parameter maps generated through either sampling strategy was about 3.54%. The average total relative error value of all parameter maps in region of interest was about 0.115, and the average cross-correlation of all parameter maps in region of interest was about 0.962. All investigated pharmacokinetic parameters had no significant differences between the result from original data and the reduced sampling data. CONCLUSION With sparsely sampled k-space data in simulation of accelerated acquisition by a factor of 4, the investigated dynamic contrast-enhanced magnetic resonance imaging pharmacokinetic parameters can accurately estimate the total generalized variation-based iterative image reconstruction method for reliable clinical application.
Collapse
|
46
|
Jansen JFA, Lu Y, Gupta G, Lee NY, Stambuk HE, Mazaheri Y, Deasy JO, Shukla-Dave A. Texture analysis on parametric maps derived from dynamic contrast-enhanced magnetic resonance imaging in head and neck cancer. World J Radiol 2016; 8:90-97. [PMID: 26834947 PMCID: PMC4731352 DOI: 10.4329/wjr.v8.i1.90] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/24/2015] [Accepted: 11/25/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the merits of texture analysis on parametric maps derived from pharmacokinetic modeling with dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) as imaging biomarkers for the prediction of treatment response in patients with head and neck squamous cell carcinoma (HNSCC).
METHODS: In this retrospective study, 19 HNSCC patients underwent pre- and intra-treatment DCE-MRI scans at a 1.5T MRI scanner. All patients had chemo-radiation treatment. Pharmacokinetic modeling was performed on the acquired DCE-MRI images, generating maps of volume transfer rate (Ktrans) and volume fraction of the extravascular extracellular space (ve). Image texture analysis was then employed on maps of Ktrans and ve, generating two texture measures: Energy (E) and homogeneity.
RESULTS: No significant changes were found for the mean and standard deviation for Ktrans and ve between pre- and intra-treatment (P > 0.09). Texture analysis revealed that the imaging biomarker E of ve was significantly higher in intra-treatment scans, relative to pretreatment scans (P < 0.04).
CONCLUSION: Chemo-radiation treatment in HNSCC significantly reduces the heterogeneity of tumors.
Collapse
|
47
|
Singer AD, Pattany PM, Fayad LM, Tresley J, Subhawong TK. Volumetric segmentation of ADC maps and utility of standard deviation as measure of tumor heterogeneity in soft tissue tumors. Clin Imaging 2015; 40:386-91. [PMID: 27133673 DOI: 10.1016/j.clinimag.2015.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/31/2015] [Accepted: 11/05/2015] [Indexed: 12/26/2022]
Abstract
PURPOSE Determine interobserver concordance of semiautomated three-dimensional volumetric and two-dimensional manual measurements of apparent diffusion coefficient (ADC) values in soft tissue masses (STMs) and explore standard deviation (SD) as a measure of tumor ADC heterogeneity. RESULTS Concordance correlation coefficients for mean ADC increased with more extensive sampling. Agreement on the SD of tumor ADC values was better for large regions of interest and multislice methods. Correlation between mean and SD ADC was low, suggesting that these parameters are relatively independent. CONCLUSION Mean ADC of STMs can be determined by volumetric quantification with high interobserver agreement. STM heterogeneity merits further investigation as a potential imaging biomarker that complements other functional magnetic resonance imaging parameters.
Collapse
Affiliation(s)
- Adam D Singer
- Department of Radiology and Imaging Sciences, Emory University Hospital, Atlanta, GA.
| | | | - Laura M Fayad
- Department of Radiology, Johns Hopkins University, Baltimore, MD
| | | | | |
Collapse
|
48
|
Khalifa F, Soliman A, El-Baz A, Abou El-Ghar M, El-Diasty T, Gimel'farb G, Ouseph R, Dwyer AC. Models and methods for analyzing DCE-MRI: a review. Med Phys 2015; 41:124301. [PMID: 25471985 DOI: 10.1118/1.4898202] [Citation(s) in RCA: 209] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To present a review of most commonly used techniques to analyze dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), discusses their strengths and weaknesses, and outlines recent clinical applications of findings from these approaches. METHODS DCE-MRI allows for noninvasive quantitative analysis of contrast agent (CA) transient in soft tissues. Thus, it is an important and well-established tool to reveal microvasculature and perfusion in various clinical applications. In the last three decades, a host of nonparametric and parametric models and methods have been developed in order to quantify the CA's perfusion into tissue and estimate perfusion-related parameters (indexes) from signal- or concentration-time curves. These indexes are widely used in various clinical applications for the detection, characterization, and therapy monitoring of different diseases. RESULTS Promising theoretical findings and experimental results for the reviewed models and techniques in a variety of clinical applications suggest that DCE-MRI is a clinically relevant imaging modality, which can be used for early diagnosis of different diseases, such as breast and prostate cancer, renal rejection, and liver tumors. CONCLUSIONS Both nonparametric and parametric approaches for DCE-MRI analysis possess the ability to quantify tissue perfusion.
Collapse
Affiliation(s)
- Fahmi Khalifa
- BioImaging Laboratory, Department of Bioengineering, University of Louisville, Louisville, Kentucky 40292 and Electronics and Communication Engineering Department, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed Soliman
- BioImaging Laboratory, Department of Bioengineering, University of Louisville, Louisville, Kentucky 40292
| | - Ayman El-Baz
- BioImaging Laboratory, Department of Bioengineering, University of Louisville, Louisville, Kentucky 40292
| | - Mohamed Abou El-Ghar
- Radiology Department, Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt
| | - Tarek El-Diasty
- Radiology Department, Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt
| | - Georgy Gimel'farb
- Department of Computer Science, University of Auckland, Auckland 1142, New Zealand
| | - Rosemary Ouseph
- Kidney Transplantation-Kidney Disease Center, University of Louisville, Louisville, Kentucky 40202
| | - Amy C Dwyer
- Kidney Transplantation-Kidney Disease Center, University of Louisville, Louisville, Kentucky 40202
| |
Collapse
|
49
|
Fowkes LA, Koh DM, Collins DJ, Jerome NP, MacVicar D, Chua SC, Pearson ADJ. Childhood extracranial neoplasms: the role of imaging in drug development and clinical trials. Pediatr Radiol 2015; 45:1600-15. [PMID: 26045035 DOI: 10.1007/s00247-015-3342-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 02/16/2015] [Accepted: 03/16/2015] [Indexed: 12/25/2022]
Abstract
Cancer is the leading cause of death in children older than 1 year of age and new drugs are necessary to improve outcomes. Imaging is crucial to the drug development process and assessment of therapeutic response. In adults, tumours are often assessed with CT using size criteria. Unfortunately, techniques established in adults are not necessarily applicable in children due to differing pathophysiology, ability to cooperate and increased susceptibility to ionising radiation. MRI, in particular quantitative MRI, has to date not been fully utilised in children with extracranial neoplasms. The specific challenges of imaging in children, the potential for functional imaging techniques to inform upon and their inclusion in clinical trials are discussed.
Collapse
Affiliation(s)
- Lucy A Fowkes
- Department of Radiology, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, Surrey, UK.
| | - Dow-Mu Koh
- Department of Radiology, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, Surrey, UK
| | - David J Collins
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, Surrey, UK
| | - Neil P Jerome
- Cancer Research UK and EPSRC Cancer Imaging Centre, Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, Surrey, UK
| | - David MacVicar
- Department of Radiology, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, Surrey, UK
| | - Sue C Chua
- Nuclear Medicine & PET Department, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, Surrey, UK
| | - Andrew D J Pearson
- Paediatric Drug Development Unit, Children and Young People's Unit, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, Surrey, UK
| |
Collapse
|
50
|
Smitha KA, Gupta AK, Jayasree RS. Fractal analysis: fractal dimension and lacunarity from MR images for differentiating the grades of glioma. Phys Med Biol 2015; 60:6937-47. [DOI: 10.1088/0031-9155/60/17/6937] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|