1
|
Ghadimi K, Abbas I, Karandish A, Crisman C, Eskandar EN, Kobets AJ. Cognitive Decline in Glioblastoma (GB) Patients with Different Treatment Modalities and Insights on Untreated Cases. Curr Oncol 2025; 32:152. [PMID: 40136356 PMCID: PMC11940939 DOI: 10.3390/curroncol32030152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Cognitive decline is common in patients with Glioblastoma (GB), occurring in both treated and untreated cases. It frequently presents as impairments in memory, attention, language, or other cognitive functions. In addition, these cognitive deficits can affect quality of life, functional independence, and overall survival, and they are associated with psychiatric conditions such as anxiety and depression. METHODS This narrative review evaluates cognitive deficits in GB patients, both with and without treatment. It also explores the impact of tumor features such as size, location, and histology, along with patient characteristics such as age and education, and discusses the effects of standard therapies, such as surgery, chemotherapy, and radiotherapy, on cognitive outcomes. RESULTS Cognitive impairment in GB is influenced by tumor- and patient-specific factors, as well as treatment modalities. Initially, combination therapies such as surgery, radiotherapy, and chemotherapy may improve cognitive domains by reducing tumor burden, relieving cerebral edema, and reducing mass effects, subsequently bringing indirect effects of improved mental health and mood. While certain treatments like radiotherapy and chemotherapy carry risks of delayed neurotoxicity, studies indicate that, on balance, treated patients generally show better preservation or improvement in cognitive function than those who go untreated. However, excessive treatment aggressiveness and cumulative neurotoxic effects may diminish cognitive benefits. CONCLUSION Cognitive function is an independent factor in GB, which could affect survival in GB patients, therefore making routine cognitive assessments essential for prognosis, treatment planning, and rehabilitation. Neuroprotective agents, cognitive rehabilitation, and personalized, multidisciplinary strategies can help optimize both survival and cognitive preservation.
Collapse
|
2
|
Rastogi A, Yalavarthy PK. Greybox: A hybrid algorithm for direct estimation of tracer kinetic parameters from undersampled DCE-MRI data. Med Phys 2024; 51:4838-4858. [PMID: 38214325 DOI: 10.1002/mp.16935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND A variety of deep learning-based and iterative approaches are available to predict Tracer Kinetic (TK) parameters from fully sampled or undersampled dynamic contrast-enhanced (DCE) MRI data. However, both the methods offer distinct benefits and drawbacks. PURPOSE To propose a hybrid algorithm (named as 'Greybox'), using both model- as well as DL-based, for solving a multi-parametric non-linear inverse problem of directly estimating TK parameters from undersampled DCE MRI data, which is invariant to undersampling rate. METHODS The proposed algorithm was inspired by plug-and-play algorithms used for solving linear inverse imaging problems. This technique was tested for its effectiveness in solving the nonlinear ill-posed inverse problem of generating 3D TK parameter maps from four-dimensional (4D; Spatial + Temporal) retrospectively undersampled k-space data. The algorithm learns a deep learning-based prior using UNET to estimate theK trans $\mathbf {K_{trans}}$ andV p $\mathbf {V_{p}}$ parameters based on the Patlak pharmacokinetic model, and this trained prior was utilized to estimate the TK parameter maps using an iterative gradient-based optimization scheme. Unlike the existing DL models, this network is invariant to the undersampling rate of the input data. The proposed method was compared with the total variation-based direct reconstruction technique on brain, breast, and prostate DCE-MRI datasets for various undersampling rates using the Radial Golden Angle (RGA) scheme. For the breast dataset, an indirect estimation using the Fast Composite Splitting algorithm was utilized for comparison. Undersampling rates of 8 × $\times$ , 12 × $\times$ and 20 × $\times$ were used for the experiments, and the results were compared using the PSNR and SSIM as metrics. For the breast dataset of 10 patients, data from four patients were utilized for training (1032 samples), two for validation (752 samples), and the entire volume of four patients for testing. Similarly, for the prostate dataset of 18 patients, 10 patients were utilized for training (720 samples), five for validation (216 samples), and the whole volume of three patients for testing. For the brain dataset of nineteen patients, ten patients were used for training (3152 samples), five for validation (1168 samples), and the whole volume of four patients for testing. Statistical tests were also conducted to assess the significance of the improvement in performance. RESULTS The experiments showed that the proposed Greybox performs significantly better than other direct reconstruction methods. The proposed algorithm improved the estimatedK trans $\mathbf {K_{trans}}$ andV p $\mathbf {V_{p}}$ in terms of the peak signal-to-noise ratio by up to 3 dB compared to other standard reconstruction methods. CONCLUSION The proposed hybrid reconstruction algorithm, Greybox, can provide state-of-the-art performance in solving the nonlinear inverse problem of DCE-MRI. This is also the first of its kind to utilize convolutional neural network-based encodings as part of the plug-and-play priors to improve the performance of the reconstruction algorithm.
Collapse
Affiliation(s)
- Aditya Rastogi
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, India
- University Hospital Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
3
|
Hoffmann E, Masthoff M, Kunz WG, Seidensticker M, Bobe S, Gerwing M, Berdel WE, Schliemann C, Faber C, Wildgruber M. Multiparametric MRI for characterization of the tumour microenvironment. Nat Rev Clin Oncol 2024; 21:428-448. [PMID: 38641651 DOI: 10.1038/s41571-024-00891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/21/2024]
Abstract
Our understanding of tumour biology has evolved over the past decades and cancer is now viewed as a complex ecosystem with interactions between various cellular and non-cellular components within the tumour microenvironment (TME) at multiple scales. However, morphological imaging remains the mainstay of tumour staging and assessment of response to therapy, and the characterization of the TME with non-invasive imaging has not yet entered routine clinical practice. By combining multiple MRI sequences, each providing different but complementary information about the TME, multiparametric MRI (mpMRI) enables non-invasive assessment of molecular and cellular features within the TME, including their spatial and temporal heterogeneity. With an increasing number of advanced MRI techniques bridging the gap between preclinical and clinical applications, mpMRI could ultimately guide the selection of treatment approaches, precisely tailored to each individual patient, tumour and therapeutic modality. In this Review, we describe the evolving role of mpMRI in the non-invasive characterization of the TME, outline its applications for cancer detection, staging and assessment of response to therapy, and discuss considerations and challenges for its use in future medical applications, including personalized integrated diagnostics.
Collapse
Affiliation(s)
- Emily Hoffmann
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Max Masthoff
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Bobe
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Mirjam Gerwing
- Clinic of Radiology, University of Münster, Münster, Germany
| | | | | | - Cornelius Faber
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
4
|
Li J, Lin C, Zhu Y, Shao C, Wang T, Chen B. Colorectal cancer cell membrane biomimetic ferroferric oxide nanomaterials for homologous bio-imaging and chemotherapy application. Med Oncol 2023; 40:322. [PMID: 37801170 DOI: 10.1007/s12032-023-02175-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023]
Abstract
The research of nanomaterials for bio-imaging and theranostic are very active nowadays with unprecedented advantages in nanomedicine. Homologous targeting and bio-imaging greatly improve the ability of targeted drug delivery and enhance active targeting and treatment ability of nanomedicine for the tumor. In this work, lycorine hydrochloride (LH) and magnetic iron oxide nanoparticles coated with a colorectal cancer (CRC) cell membrane (LH-Fe3O4@M) were prepared, for homologous targeting, magnetic resonance imaging (MRI), and chemotherapy. Results showed that the LH-Fe3O4@M and Fe3O4@M intensity at HT29 tumor was significantly higher than that Fe3O4@PEG, proving the superior selectivity of cancer cell membrane-camouflaged nanomedicine for homologous tumors and the MRI effect of darkening contrast enhancement were remarkable at HT29 tumor. The LH-Fe3O4@M exhibited excellent chemotherapy effect in CRC models as well as LH alone and achieved a high tumor ablation rate but no damage to normal tissues and cells. Therefore, our biomimetic system achieved a homologous targeting, bio-imaging, and efficient therapeutic effect of CRC.
Collapse
Affiliation(s)
- Jun Li
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Chenyu Lin
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yuqian Zhu
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Tiegong Wang
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China.
| | - Bingdi Chen
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
5
|
A hybrid hemodynamic knowledge-powered and feature reconstruction-guided scheme for breast cancer segmentation based on DCE-MRI. Med Image Anal 2022; 82:102572. [PMID: 36055051 DOI: 10.1016/j.media.2022.102572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 07/08/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022]
Abstract
Automatically and accurately annotating tumor in dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), which provides a noninvasive in vivo method to evaluate tumor vasculature architectures based on contrast accumulation and washout, is a crucial step in computer-aided breast cancer diagnosis and treatment. However, it remains challenging due to the varying sizes, shapes, appearances and densities of tumors caused by the high heterogeneity of breast cancer, and the high dimensionality and ill-posed artifacts of DCE-MRI. In this paper, we propose a hybrid hemodynamic knowledge-powered and feature reconstruction-guided scheme that integrates pharmacokinetics prior and feature refinement to generate sufficiently adequate features in DCE-MRI for breast cancer segmentation. The pharmacokinetics prior expressed by time intensity curve (TIC) is incorporated into the scheme through objective function called dynamic contrast-enhanced prior (DCP) loss. It contains contrast agent kinetic heterogeneity prior knowledge, which is important to optimize our model parameters. Besides, we design a spatial fusion module (SFM) embedded in the scheme to exploit intra-slices spatial structural correlations, and deploy a spatial-kinetic fusion module (SKFM) to effectively leverage the complementary information extracted from spatial-kinetic space. Furthermore, considering that low spatial resolution often leads to poor image quality in DCE-MRI, we integrate a reconstruction autoencoder into the scheme to refine feature maps in an unsupervised manner. We conduct extensive experiments to validate the proposed method and show that our approach can outperform recent state-of-the-art segmentation methods on breast cancer DCE-MRI dataset. Moreover, to explore the generalization for other segmentation tasks on dynamic imaging, we also extend the proposed method to brain segmentation in DSC-MRI sequence. Our source code will be released on https://github.com/AI-medical-diagnosis-team-of-JNU/DCEDuDoFNet.
Collapse
|
6
|
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: 42] [Impact Index Per Article: 10.5] [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
|
7
|
Berks M, Little RA, Watson Y, Cheung S, Datta A, O'Connor JPB, Scaramuzza D, Parker GJM. A model selection framework to quantify microvascular liver function in gadoxetate-enhanced MRI: Application to healthy liver, diseased tissue, and hepatocellular carcinoma. Magn Reson Med 2021; 86:1829-1844. [PMID: 33973674 DOI: 10.1002/mrm.28798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/04/2021] [Accepted: 03/19/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE We introduce a novel, generalized tracer kinetic model selection framework to quantify microvascular characteristics of liver and tumor tissue in gadoxetate-enhanced dynamic contrast-enhanced MRI (DCE-MRI). METHODS Our framework includes a hierarchy of nested models, from which physiological parameters are derived in 2 regimes, corresponding to the active transport and free diffusion of gadoxetate. We use simulations to show the sensitivity of model selection and parameter estimation to temporal resolution, time-series duration, and noise. We apply the framework in 8 healthy volunteers (time-series duration up to 24 minutes) and 10 patients with hepatocellular carcinoma (6 minutes). RESULTS The active transport regime is preferred in 98.6% of voxels in volunteers, 82.1% of patients' non-tumorous liver, and 32.2% of tumor voxels. Interpatient variations correspond to known co-morbidities. Simulations suggest both datasets have sufficient temporal resolution and signal-to-noise ratio, while patient data would be improved by using a time-series duration of at least 12 minutes. CONCLUSIONS In patient data, gadoxetate exhibits different kinetics: (a) between liver and tumor regions and (b) within regions due to liver disease and/or tumor heterogeneity. Our generalized framework selects a physiological interpretation at each voxel, without preselecting a model for each region or duplicating time-consuming optimizations for models with identical functional forms.
Collapse
Affiliation(s)
- Michael Berks
- Division of Cancer Sciences, Quantitative Biomedical Imaging Laboratory, University of Manchester, Manchester, UK
| | - Ross A Little
- Division of Cancer Sciences, Quantitative Biomedical Imaging Laboratory, University of Manchester, Manchester, UK
| | - Yvonne Watson
- Division of Cancer Sciences, Quantitative Biomedical Imaging Laboratory, University of Manchester, Manchester, UK
| | - Sue Cheung
- Division of Cancer Sciences, Quantitative Biomedical Imaging Laboratory, University of Manchester, Manchester, UK
| | - Anubhav Datta
- Division of Cancer Sciences, Quantitative Biomedical Imaging Laboratory, University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| | - James P B O'Connor
- Division of Cancer Sciences, Quantitative Biomedical Imaging Laboratory, University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | | | - Geoff J M Parker
- Division of Cancer Sciences, Quantitative Biomedical Imaging Laboratory, University of Manchester, Manchester, UK
- Bioxydyn Ltd, Manchester, UK
- Centre for Medical Image Computing, University College London, London, UK
| |
Collapse
|
8
|
Ghalati MK, Nunes A, Ferreira H, Serranho P, Bernardes R. Texture Analysis and its Applications in Biomedical Imaging: A Survey. IEEE Rev Biomed Eng 2021; 15:222-246. [PMID: 34570709 DOI: 10.1109/rbme.2021.3115703] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Texture analysis describes a variety of image analysis techniques that quantify the variation in intensity and pattern. This paper provides an overview of several texture analysis approaches addressing the rationale supporting them, their advantages, drawbacks, and applications. This surveys emphasis is in collecting and categorising over five decades of active research on texture analysis. Brief descriptions of different approaches are presented along with application examples. From a broad range of texture analysis applications, this surveys final focus is on biomedical image analysis. An up-to-date list of biological tissues and organs in which disorders produce texture changes that may be used to spot disease onset and progression is provided. Finally, the role of texture analysis methods as biomarkers of disease is summarised.
Collapse
|
9
|
Wang X, Wang Y, Zhang Z, Zhou M, Zhou X, Zhao H, Xing J, Zhou Y. Rim enhancement on hepatobiliary phase of pre-treatment 3.0 T MRI: A potential marker for early chemotherapy response in colorectal liver metastases treated with XELOX. Eur J Radiol 2021; 143:109887. [PMID: 34454297 DOI: 10.1016/j.ejrad.2021.109887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE To assess the value of the enhanced features on the hepatobiliary phase (HBP) of pre-treatment Gd-EOB-DTPA MRI in evaluating response to chemotherapy in colorectal liver metastases (CRLMs). METHODS We retrospectively studied 65 patients with CRLMs who underwent Gd-EOB-DTPA enhanced MRI before chemotherapy from October 2015 to November 2017. The diagnosis of liver metastasis was established on the basis of imaging findings. Two radiologists evaluated the size, contrast-enhanced (CE) patterns of the maximum lesion on the HBP. According to the different CE patterns, we quantified area signal intensity (SI) by applying SI ratio (such as SIcenter/outer and SIrim/center). All of the above parameters were analyzed in terms of chemotherapy response. RESULTS Rim enhancement on the HBP was more frequent in the responding group of 28 patients (72%) than in the non-responding group of eight patients (31%). Additionally, there was a significant association between chemotherapy response and quantitative parameters: including diameter (P = 0.04), SIcenter/outer (P = 0.047) and SIrim/center (P = 0.012). The HBP CE pattern (P = 0.007) and SIcenter/outer (P = 0.022) were independent factors for chemotherapy response. The areas under the curve (AUCs) of the above-mentioned parameters were significant associated with response to chemotherapy, in which diameter, HBP CE patterns, SIcenter/outer, and SIrim/center were 0.638, 0.706, 0.712, and 0.673, respectively. Moreover, the combination of these parameters obtained the largest AUC of 0.821. CONCLUSION The CE patterns, in particular with rim enhancement, and SI ratio parameters on the HBP are useful indicators for early evaluation of therapeutic response after chemotherapy in patients with CRLMs.
Collapse
Affiliation(s)
- Xinxin Wang
- Department of Radiology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150010, Heilongjiang, China
| | - Yu Wang
- Department of Radiology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150010, Heilongjiang, China
| | - Ziqian Zhang
- Department of Radiology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150010, Heilongjiang, China
| | - Meng Zhou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150010, Heilongjiang, China
| | - Xueyan Zhou
- School of Technology, Harbin University, 109 Zhongxing Street, Harbin 150010, Heilongjiang, China.
| | - Hongxin Zhao
- Department of Radiology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150010, Heilongjiang, China
| | - JiQing Xing
- Harbin Engineering University, Harbin 150001, Heilongjiang Province, China.
| | - Yang Zhou
- Department of Radiology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150010, Heilongjiang, China.
| |
Collapse
|
10
|
Zhu G, Wu Z, Lui S, Hu N, Wu M. Advances in Imaging Modalities and Contrast Agents for the Early Diagnosis of Colorectal Cancer. J Biomed Nanotechnol 2021; 17:558-581. [PMID: 35057884 DOI: 10.1166/jbn.2021.3064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Colorectal cancer is one of the most common gastrointestinal cancers worldwide. The mortality rate of colorectal cancer has declined by more than 20% due to the rapid development of early diagnostic techniques and effective treatment. At present, there are many diagnostic modalities
available for the evaluation of colorectal cancer, such as the carcinoembryonic antigen test, the fecal occult blood test, endoscopy, X-ray barium meal, computed tomography, magnetic resonance imaging, and radionuclide examination. Sensitive and specific imaging modalities have played an increasingly
important role in the diagnosis of colorectal cancer following the rapid development of novel contrast agents. This review discusses the applications and challenges of different imaging techniques and contrast agents applied to detect colorectal cancer, for the purpose of the early diagnosis
and treatment of patients with colorectal cancer.
Collapse
Affiliation(s)
- Guannan Zhu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zijun Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Su Lui
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Na Hu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
11
|
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: 311] [Impact Index Per Article: 77.8] [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
|
12
|
Hartmann L, Bundschuh L, Zsótér N, Essler M, Bundschuh RA. Tumor heterogeneity for differentiation between liver tumors and normal liver tissue in 18F-FDG PET/CT. Nuklearmedizin 2021; 60:25-32. [PMID: 33142334 DOI: 10.1055/a-1270-5568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AIM Malignancies show higher spatial heterogeneity than normal tissue. We investigated, if textural parameters from FDG PET describing the heterogeneity function as tool to differentiate between tumor and normal liver tissue. METHODS FDG PET/CT scans of 80 patients with liver metastases and 80 patients with results negative upper abdominal organs were analyzed. Metastases and normal liver tissue were analyzed drawing up to three VOIs with a diameter of 25 mm in healthy liver tissue of the tumoral affected and results negative liver, whilst up to 3 metastases per patient were delineated. Within these VOIs 30 different textural parameters were calculated as well as SUV. The parameters were compared in terms of intra-patient and inter-patient variability (2-sided t test). ROC analysis was performed to analyze predictive power and cut-off values. RESULTS 28 textural parameters differentiated healthy and pathological tissue (p < 0.05) with high sensitivity and specificity. SUV showed ability to differentiate but with a lower significance. 15 textural parameters as well as SUV showed a significant variation between healthy tissues out of tumour infested and negative livers. Mean intra- and inter-patient variability of metastases were found comparable or lower for 6 of the textural features than the ones of SUV. They also showed good values of mean intra- and inter-patient variability of VOIs drawn in liver tissue of patients with metastases and of results negative ones. CONCLUSION Heterogeneity parameters assessed in FDG PET are promising to classify tissue and differentiate malignant lesions usable for more personalized treatment planning, therapy response evaluation and precise delineation of tumors for target volume determination as part of radiation therapy planning.
Collapse
Affiliation(s)
- Lynn Hartmann
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Bonn, Germany
| | - Lena Bundschuh
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Bonn, Germany
| | | | - Markus Essler
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Bonn, Germany
| | | |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Enhanced Rim on MDCT of Colorectal Liver Metastases: Assessment of Ability to Predict Progression-Free Survival and Response to Bevacizumab-Based Chemotherapy. AJR Am J Roentgenol 2020; 215:1377-1383. [PMID: 32991216 DOI: 10.2214/ajr.19.22280] [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: 02/05/2023]
Abstract
OBJECTIVE. The purpose of this article is to evaluate the enhanced rim on the portal venous phase (PVP) on MDCT as a predictor of 1-year progression-free survival (PFS) and response to bevacizumab-based chemotherapy in patients with colorectal liver metastases (CRLM). MATERIALS AND METHODS. We retrospectively identified 111 patients with primary unresectable CRLM treated with bevacizumab-based chemotherapy at two institutions between 2012 and 2018. Pretreatment contrast-enhanced MDCT images were reviewed and data on clinical characteristics were collected from the electronic medical records. Univariable and multivariable analyses were conducted to assess several imaging features and clinical characteristics as potential predictors of 1-year PFS and objective response rate (ORR). RESULTS. After 1 year of follow-up, liver metastatic tumor progression was detected in 52 patients (46.8%) after bevacizumab-based chemotherapy. A log-rank test showed that enhanced rim on PVP (chi-square test, 5.862; p = 0.015) and the occurrence of liver resection surgery (chi-square test, 7.836; p = 0.005) were significant predictors of 1-year PFS. Multivariable analysis showed that enhanced rim on PVP images was an independent predictor of 1-year PFS (hazard ratio, 0.510; 95% CI, 0.282-0.926; p = 0.027) and ORR (odds ratio, 4.694; p < 0.001). CONCLUSION. The presence of an enhanced rim on PVP MDCT is an independent predictor of survival and response to bevacizumab-based chemotherapy among patients with CRLM.
Collapse
|
16
|
Han Y, Chai F, Wei J, Yue Y, Cheng J, Gu D, Zhang Y, Tong T, Sheng W, Hong N, Ye Y, Wang Y, Tian J. Identification of Predominant Histopathological Growth Patterns of Colorectal Liver Metastasis by Multi-Habitat and Multi-Sequence Based Radiomics Analysis. Front Oncol 2020; 10:1363. [PMID: 32923388 PMCID: PMC7456817 DOI: 10.3389/fonc.2020.01363] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/29/2020] [Indexed: 12/21/2022] Open
Abstract
Purpose: Developing an MRI-based radiomics model to effectively and accurately predict the predominant histopathologic growth patterns (HGPs) of colorectal liver metastases (CRLMs). Materials and Methods: In this study, 182 resected and histopathological proven CRLMs of chemotherapy-naive patients from two institutions, including 123 replacement CRLMs and 59 desmoplastic CRLMs, were retrospectively analyzed. Radiomics analysis was performed on two regions of interest (ROI), the tumor zone and the tumor-liver interface (TLI) zone. Decision tree (DT) algorithm was used for radiomics modeling on each MR sequence, and fused radiomics model was constructed by combining the radiomics signature of each sequence. The clinical and combination models were developed through multivariate logistic regression method. The performance of the developed models was assessed by receiver operating characteristic (ROC) curves with indicators of area under curve (AUC), accuracy, sensitivity, and specificity. A nomogram was constructed to evaluate the discrimination, calibration, and usefulness. Results: The fused radiomicstumor and radiomicsTLI models showed better performance than any single sequence and clinical model. In addition, the radiomicsTLI model exhibited better performance than radiomicstumor model (AUC of 0.912 vs. 0.879) in internal validation cohort. The combination model showed good discrimination, and the AUC of nomogram was 0.971, 0.909, and 0.905 in the training, internal validation, and external validation cohorts, respectively. Conclusion: MRI-based radiomics method has high potential in predicting the predominant HGPs of CRLM. Preoperative non-invasive identification of predominant HGPs could further explore the ability of HGPs as a potential biomarker for clinical treatment strategy, reflecting different biological pathways.
Collapse
Affiliation(s)
- Yuqi Han
- School of Life Science and Technology, Xidian University, Xi'an, China.,Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Molecular Imaging, Beijing, China
| | - Fan Chai
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Jingwei Wei
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Molecular Imaging, Beijing, China
| | - Yali Yue
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Dongsheng Gu
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Molecular Imaging, Beijing, China
| | - Yinli Zhang
- Department of Pathology, Peking University People's Hospital, Beijing, China
| | - Tong Tong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiqi Sheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People' Hospital, Beijing, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Molecular Imaging, Beijing, China.,Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China.,Engineering Research Centre of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| |
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
|
Franklin JM, Irving B, Papiez BW, Kallehauge JF, Wang LM, Goldin RD, Harris AL, Anderson EM, Schnabel JA, Chappell MA, Brady M, Sharma RA, Gleeson FV. Tumour subregion analysis of colorectal liver metastases using semi-automated clustering based on DCE-MRI: Comparison with histological subregions and impact on pharmacokinetic parameter analysis. Eur J Radiol 2020; 126:108934. [PMID: 32217426 DOI: 10.1016/j.ejrad.2020.108934] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/21/2020] [Accepted: 03/01/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE To use a novel segmentation methodology based on dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) to define tumour subregions of liver metastases from colorectal cancer (CRC), to compare these with histology, and to use these to compare extracted pharmacokinetic (PK) parameters between tumour subregions. MATERIALS AND METHODS This ethically-approved prospective study recruited patients with CRC and ≥1 hepatic metastases scheduled for hepatic resection. Patients underwent DCE-MRI pre-metastasectomy. Histological sections of resection specimens were spatially matched to DCE-MRI acquisitions and used to define histological subregions of viable and non-viable tumour. A semi-automated voxel-wise image segmentation algorithm based on the DCE-MRI contrast-uptake curves was used to define imaging subregions of viable and non-viable tumour. Overlap of histologically-defined and imaging subregions was compared using the Dice similarity coefficient (DSC). DCE-MRI PK parameters were compared for the whole tumour and histology-defined and imaging-derived subregions. RESULTS Fourteen patients were included in the analysis. Direct histological comparison with imaging was possible in nine patients. Mean DSC for viable tumour subregions defined by imaging and histology was 0.738 (range 0.540-0.930). There were significant differences between Ktrans and kep for viable and non-viable subregions (p < 0.001) and between whole lesions and viable subregions (p < 0.001). CONCLUSION We demonstrate good concordance of viable tumour segmentation based on pre-operative DCE-MRI with a post-operative histological gold-standard. This can be used to extract viable tumour-specific values from quantitative image analysis, and could improve treatment response assessment in clinical practice.
Collapse
Affiliation(s)
- James M Franklin
- Institute of Medical Imaging and Visualisation, Bournemouth University, UK; Radiology Department, Royal Bournemouth and Christchurch Hospitals NS Foundation Trust, UK.
| | - Benjamin Irving
- Institute of Biomedical Engineering (Department of Engineering Science), University of Oxford, UK
| | - Bartlomiej W Papiez
- Institute of Biomedical Engineering (Department of Engineering Science), University of Oxford, UK
| | - Jesper F Kallehauge
- Institute of Biomedical Engineering (Department of Engineering Science), University of Oxford, UK
| | - Lai Mun Wang
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, UK
| | | | | | - Ewan M Anderson
- Radiology Department, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, UK
| | - Julia A Schnabel
- Institute of Biomedical Engineering (Department of Engineering Science), University of Oxford, UK; School of Biomedical Engineering and Imaging Sciences, King's College London, UK
| | - Michael A Chappell
- Institute of Biomedical Engineering (Department of Engineering Science), University of Oxford, UK
| | | | - Ricky A Sharma
- NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Fergus V Gleeson
- Radiology Department, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, UK
| |
Collapse
|
19
|
Thüring J, Kuhl CK, Barabasch A, Hitpass L, Bode M, Bünting N, Bruners P, Krämer NA. Signal changes in T2-weighted MRI of liver metastases under bevacizumab-A practical imaging biomarker? PLoS One 2020; 15:e0230553. [PMID: 32231380 PMCID: PMC7108712 DOI: 10.1371/journal.pone.0230553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/28/2020] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE The purpose of this study was to investigate signal changes in T2-weighted magnetic resonance imaging of liver metastases under treatment with and without bevacizumab-containing chemotherapy and to compare these signal changes to tumor contrast enhancement. MATERIALS AND METHODS Retrospective analysis of 44 patients, aged 36-84 years, who underwent liver magnetic resonance imaging including T2-weighted and dynamic contrast enhancement sequences. Patients received bevacizumab-containing (n = 22) or conventional cytotoxic chemotherapy (n = 22). Magnetic resonance imaging was obtained at baseline and at three follow-ups (on average 3, 6 and 9 months after initial treatment). Three independent readers rated the T2 signal intensity and the relative contrast enhancement of the metastases on a 5-point scale. RESULTS T2 signal intensity of metastases treated with bevacizumab showed a significant (p<0.001) decrease in T2 signal intensity after initial treatment and exhibit compared to conventionally treated metastases significantly (p<0.001 for each follow-up) hypointense (bevacizumab: 0.70 ± 0.83 before vs. -1.55 ± 0.61, -1.91 ± 0.62, and -1.97 ± 0.52; cytotoxic: 0.73 ± 0.79 before vs. -0.69 ± 0.81, -0.71 ± 0.68, and -0.75 ± 0.65 after 3, 6, and 9 months, respectively). T2 signal intensity was strongly correlated with tumor contrast enhancement (r = 0.71; p<0.001). Intra-observer agreement for T2-signal intensity was substantial (κ = 0.75). The agreement for tumoral contrast enhancement between the readers was considerably lower (κ = 0.39). CONCLUSION Liver metastases exhibit considerably hypointense in T2-weighted imaging after treatment with bevacizumab, in contrast to conventionally treated liver metastases. Therefore, T2-weighted imaging seems to reflect the effect of bevacizumab.
Collapse
Affiliation(s)
- Johannes Thüring
- Department of Diagnostic and Interventional Radiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Christiane Katharina Kuhl
- Department of Diagnostic and Interventional Radiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Alexandra Barabasch
- Department of Diagnostic and Interventional Radiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Lea Hitpass
- Department of Diagnostic and Interventional Radiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Maike Bode
- Department of Diagnostic and Interventional Radiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Nina Bünting
- Department of Diagnostic and Interventional Radiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Philipp Bruners
- Department of Diagnostic and Interventional Radiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Nils Andreas Krämer
- Department of Diagnostic and Interventional Radiology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
20
|
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]
|
21
|
Drisis S, El Adoui M, Flamen P, Benjelloun M, Dewind R, Paesmans M, Ignatiadis M, Bali M, Lemort M. Early prediction of neoadjuvant treatment outcome in locally advanced breast cancer using parametric response mapping and radial heterogeneity from breast MRI. J Magn Reson Imaging 2019; 51:1403-1411. [PMID: 31737963 DOI: 10.1002/jmri.26996] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/25/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Early prediction of nonresponse is essential in order to avoid inefficient treatments. PURPOSE To evaluate if parametrical response mapping (PRM)-derived biomarkers could predict early morphological response (EMR) and pathological complete response (pCR) 24-72 hours after initiation of chemotherapy treatment and whether concentric analysis of nonresponding PRM regions could better predict response. STUDY TYPE This was a retrospective analysis of prospectively acquired cohort, nonrandomized, monocentric, diagnostic study. POPULATION Sixty patients were initially recruited, with 39 women participating in the final cohort. FIELD STRENGTH/SEQUENCE A 1.5T scanner was used for MRI examinations. ASSESSMENT Dynamic contrast-enhanced (DCE)-MR images were acquired at baseline (timepoint 1, TP1), 24-72 hours after the first chemotherapy (TP2), and after the end of anthracycline treatment (TP3). PRM was performed after fusion of T1 subtraction images from TP1 and TP2 using an affine registration algorithm. Pixels with an increase of more than 10% of their value (PRMdce+) were corresponding nonresponding regions of the tumor. Patients with a decrease of maximum diameter (%dDmax) between TP1 and TP3 of more than 30% were defined as EMR responders. pCR patients achieved a residual cancer burden score of 0. STATISTICAL TESTS T-test, receiver operating characteristic (ROC) curves, and logistic regression were used for the analysis. RESULTS PRM showed a statistical difference between pCR response groups (P < 0.01) and AUC of 0.88 for the prediction of non-pCR. Logistic regression analysis demonstrated that PRMdce+ and Grade II were significant (P < 0.01) for non-pCR prediction (AUC = 0.94). Peripheral tumor region demonstrated higher performance for the prediction of non-pCR (AUC = 0.85) than intermediate and central zones; however, statistical comparison showed no significant difference. DATA CONCLUSION PRM could be predictive of non-pCR 24-72 hours after initiation of chemotherapy treatment. Moreover, the peripheral region showed increased AUC for non-pCR prediction and increased signal intensity during treatment for non-pCR tumors, information that could be used for optimal tissue sampling. LEVEL OF EVIDENCE 1 Technical Efficacy Stage: 4 J. Magn. Reson. Imaging 2020;51:1403-1411.
Collapse
Affiliation(s)
| | - Mohammed El Adoui
- Medical Imaging Department, Polytechnic University of Mons, Mons, Belgium
| | - Patrick Flamen
- Nuclear Department, Institute Jules Bordet, Brussels, Belgium
| | | | - Roland Dewind
- Pathology Department, Institute Jules Bordet, Brussels, Belgium
| | - Mariane Paesmans
- Statistics Department, Institute Jules Bordet, Brussels, Belgium
| | | | - Maria Bali
- Radiology Department, Institute Jules Bordet, Brussels, Belgium
| | - Marc Lemort
- Radiology Department, Institute Jules Bordet, Brussels, Belgium
| |
Collapse
|
22
|
Russell MR, Graham C, D'Amato A, Gentry-Maharaj A, Ryan A, Kalsi JK, Whetton AD, Menon U, Jacobs I, Graham RLJ. Diagnosis of epithelial ovarian cancer using a combined protein biomarker panel. Br J Cancer 2019; 121:483-489. [PMID: 31388184 PMCID: PMC6738042 DOI: 10.1038/s41416-019-0544-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 07/04/2019] [Accepted: 07/18/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND An early detection tool for EOC was constructed from analysis of biomarker expression data from serum collected during the UKCTOCS. METHODS This study included 49 EOC cases (19 Type I and 30 Type II) and 31 controls, representing 482 serial samples spanning seven years pre-diagnosis. A logit model was trained by analysis of dysregulation of expression data of four putative biomarkers, (CA125, phosphatidylcholine-sterol acyltransferase, vitamin K-dependent protein Z and C-reactive protein); by scoring the specificity associated with dysregulation from the baseline expression for each individual. RESULTS The model is discriminatory, passes k-fold and leave-one-out cross-validations and was further validated in a Type I EOC set. Samples were analysed as a simulated annual screening programme, the algorithm diagnosed cases with >30% PPV 1-2 years pre-diagnosis. For Type II cases (~80% were HGS) the algorithm classified 64% at 1 year and 28% at 2 years tDx as severe. CONCLUSIONS The panel has the potential to diagnose EOC one-two years earlier than current diagnosis. This analysis provides a tangible worked example demonstrating the potential for development as a screening tool and scrutiny of its properties. Limits on interpretation imposed by the number of samples available are discussed.
Collapse
Affiliation(s)
- Matthew R Russell
- Stoller Biomarker Discovery Centre and Manchester Molecular Pathology Innovation Centre, Division of Cancer Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Ciaren Graham
- School of Biological Sciences, Queens University Belfast, Chlorine Gardens, Belfast, BT9 5DL, UK
| | - Alfonsina D'Amato
- Department of Pharmaceutical Sciences, University of Milan, Milano, Lombardy, Italy
| | - Aleksandra Gentry-Maharaj
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, Faculty of Population Health Sciences, University College London, London, UK
| | - Andy Ryan
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, Faculty of Population Health Sciences, University College London, London, UK
| | - Jatinderpal K Kalsi
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, Faculty of Population Health Sciences, University College London, London, UK
| | - Anthony D Whetton
- Stoller Biomarker Discovery Centre and Manchester Molecular Pathology Innovation Centre, Division of Cancer Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Usha Menon
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, Faculty of Population Health Sciences, University College London, London, UK
| | - Ian Jacobs
- Stoller Biomarker Discovery Centre and Manchester Molecular Pathology Innovation Centre, Division of Cancer Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK.
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, Faculty of Population Health Sciences, University College London, London, UK.
- University of New South Wales, UNSW Australia, Level 1, Chancellery Building, Sydney, NSW, 2052, Australia.
| | - Robert L J Graham
- School of Biological Sciences, Queens University Belfast, Chlorine Gardens, Belfast, BT9 5DL, UK.
| |
Collapse
|
23
|
Abstract
Medullary thyroid carcinoma (MTC), arising from the parafollicular C cells of the thyroid, accounts for 1–2% of thyroid cancers. MTC is frequently aggressive and metastasizes to cervical and mediastinal lymph nodes, lungs, liver, and bones. Although a number of new imaging modalities for directing the management of oncologic patients evolved over the last two decades, the clinical application of these novel techniques is limited in MTC. In this article, we review the biology and molecular aspects of MTC as an important background for the use of current imaging modalities and approaches for this tumor. We discuss the modern and currently available imaging techniques—advanced magnetic resonance imaging (MRI)-based techniques such as whole-body MRI, dynamic contrast-enhanced (DCE) technique, diffusion-weighted imaging (DWI), positron emission tomography/computed tomography (PET/CT) with 18F-FDOPA and 18F-FDG, and integrated positron emission tomography/magnetic resonance (PET/MR) hybrid imaging—for primary as well as metastatic MTC tumor, including its metastatic spread to lymph nodes and the most common sites of distant metastases: lungs, liver, and bones.
Collapse
|
24
|
Thibodeau-Antonacci A, Petitclerc L, Gilbert G, Bilodeau L, Olivié D, Cerny M, Castel H, Turcotte S, Huet C, Perreault P, Soulez G, Chagnon M, Kadoury S, Tang A. Dynamic contrast-enhanced MRI to assess hepatocellular carcinoma response to Transarterial chemoembolization using LI-RADS criteria: A pilot study. Magn Reson Imaging 2019; 62:78-86. [PMID: 31247250 DOI: 10.1016/j.mri.2019.06.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/05/2019] [Accepted: 06/23/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE To identify quantitative dynamic contrast-enhanced (DCE)-MRI perfusion parameters indicating tumor response of hepatocellular carcinoma (HCC) to transarterial chemoembolization (TACE). MATERIALS AND METHODS This prospective pilot study was approved by our institutional review board; written and informed consent was obtained for each participant. Patients underwent DCE-MRI examinations before and after TACE. A variable flip-angle unenhanced 3D mDixon sequence was performed for T1 mapping. A dynamic 4D mDixon sequence was performed after contrast injection for assessing dynamic signal enhancement. Nonparametric analysis was conducted on the time-intensity curves. Parametric analysis was performed on the time-concentration curves using a dual-input single-compartment model. Treatment response according to Liver Reporting and Data System (LI-RADS) v2018 was used as the reference standard. The comparisons within groups (before vs. after treatment) and between groups (nonviable vs. equivocal or viable tumor) were performed using nonparametric bootstrap taking into account the clustering effect of lesions in patients. RESULTS Twenty-eight patients with 52 HCCs (size: 10-104 mm) were evaluated. For nonviable tumors (n = 27), time to peak increased from 62.5 ± 18.2 s before to 83.3 ± 12.8 s after treatment (P< 0.01). For equivocal or viable tumors (n = 25), time to peak and mean transit time significantly increased (from 54.4 ± 24.1 s to 69.5 ± 18.9 s, P < 0.01 and from 14.2 ± 11.8 s to 33.9 ± 36.8 s, P= 0.01, respectively) and the transfer constant from the extracellular and extravascular space to the central vein significantly decreased from 14.8 ± 14.1 to 8.1 ± 9.1 s-1 after treatment (P= 0.01). CONCLUSION This prospective pilot DCE-MRI study showed that time to peak significantly changed after TACE treatment for both groups (nonviable tumors and equivocal or viable tumors). In our cohort, several perfusion parameters may provide an objective marker for differentiation of treatment response after TACE in HCC patients.
Collapse
Affiliation(s)
- Alana Thibodeau-Antonacci
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada; Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Léonie Petitclerc
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada; Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | | | - Laurent Bilodeau
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Damien Olivié
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Milena Cerny
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada; Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Hélène Castel
- Department of Hepatology and Liver transplantation, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Simon Turcotte
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada; Department of Surgery, Hepatopancreatobiliary and Liver Transplantation Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Catherine Huet
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Pierre Perreault
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Gilles Soulez
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Miguel Chagnon
- Department of Mathematics and Statistics, Université de Montréal, QC, Canada
| | - Samuel Kadoury
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada; Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada; École Polytechnique, Montréal, Québec, Canada
| | - An Tang
- Department of Radiology, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada; Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.
| |
Collapse
|
25
|
Texture features of colorectal liver metastases on pretreatment contrast-enhanced CT may predict response and prognosis in patients treated with bevacizumab-containing chemotherapy: a pilot study including comparison with standard chemotherapy. Radiol Med 2019; 124:877-886. [PMID: 31172448 DOI: 10.1007/s11547-019-01046-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023]
|
26
|
Wetz C, Genseke P, Apostolova I, Furth C, Ghazzawi S, Rogasch JMM, Schatka I, Kreissl MC, Hofheinz F, Grosser OS, Amthauer H. The association of intra-therapeutic heterogeneity of somatostatin receptor expression with morphological treatment response in patients undergoing PRRT with [177Lu]-DOTATATE. PLoS One 2019; 14:e0216781. [PMID: 31091247 PMCID: PMC6519899 DOI: 10.1371/journal.pone.0216781] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
Aim Purpose of this study was to evaluate the association of the spatial heterogeneity (asphericity, ASP) in intra-therapeutic SPECT/ CT imaging of somatostatin receptor (SSR) positive metastatic gastroenteropancreatic neuroendocrine neoplasms (GEP-NEN) for morphological treatment response to peptide receptor radionuclide therapy (PRRT). Secondly, we correlated ASP derived form a pre-therapeutic OctreoScan (ASP[In]) and an intra-therapeutic [177Lu]-SPECT/CT (ASP[Lu]). Materials and methods Data from first therapy cycle [177Lu-DOTA0-Tyr3]octreotate ([177Lu]-DOTATATE)-PRRT was retrospectively analyzed in 33 patients (m = 20; w = 13; median age, 72 [46–88] years). The evaluation of response to PRRT was performed according to RECIST 1.1 in responding lesions [RL (SD, PR, CR), n = 104] and non-responding lesions [NRL (PD), n = 27]. The association of SSR tumor heterogeneity with morphological response was evaluated by Kruskal-Wallis test and receiver operating characteristic curve (ROC). The optimal threshold for separation (RL vs. NRL) was calculated using the Youden-index. Relationship between pre- and intra-therapeutic ASP was determined with Spearman’s rank correlation coefficient (ρ) and Bland-Altman plots. Results A total of 131 lesions (liver: n = 59, lymph nodes: n = 48, bone: n = 19, pancreas: n = 5) were analyzed. Lesions with higher ASP values showed a significantly poorer response to PRRT (PD, median: 11.3, IQR: 8.5–15.5; SD, median: 3.4, IQR: 2.1–4.5; PR, median 1.7, IQR: 0.9–2.8; CR, median: 0.5, IQR: 0.0–1.3); Kruskal-Wallis, p<0.001). ROC analyses revealed a significant separation between RL and NRL for ASP after 4 months (AUC 0.85, p<0.001) and after 12 months (AUC 0.94, p<0.001). The optimal threshold for ASP was >5.45% (sensitivity 96% and specificity 82%). The correlation coefficient of pre- and intra-therapeutic ASP revealed ρ = 0.72 (p <0.01). The mean absolute difference between ASP[In] and ASP[Lu] was -0.04 (95% Limits of Agreement, -6.1–6.0). Conclusion Pre- and intra-therapeutic ASP shows a strong correlation and might be an useful tool for therapy monitoring.
Collapse
Affiliation(s)
- Christoph Wetz
- Department of Radiology and Nuclear Medicine; University Hospital Magdeburg A.ö.R., Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Genseke
- Department of Radiology and Nuclear Medicine; University Hospital Magdeburg A.ö.R., Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ivayla Apostolova
- Department of Nuclear Medicine, University Medical Center Hamburg UKE, Hamburg, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sammy Ghazzawi
- Department of Radiology and Nuclear Medicine; University Hospital Magdeburg A.ö.R., Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Julian M. M. Rogasch
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael C. Kreissl
- Department of Radiology and Nuclear Medicine; University Hospital Magdeburg A.ö.R., Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Frank Hofheinz
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, PET Center, Dresden, Germany
| | - Oliver S. Grosser
- Department of Radiology and Nuclear Medicine; University Hospital Magdeburg A.ö.R., Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Holger Amthauer
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
27
|
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
|
28
|
Beaton L, Bandula S, Gaze MN, Sharma RA. How rapid advances in imaging are defining the future of precision radiation oncology. Br J Cancer 2019; 120:779-790. [PMID: 30911090 PMCID: PMC6474267 DOI: 10.1038/s41416-019-0412-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 01/23/2019] [Accepted: 02/05/2019] [Indexed: 12/13/2022] Open
Abstract
Imaging has an essential role in the planning and delivery of radiotherapy. Recent advances in imaging have led to the development of advanced radiotherapy techniques—including image-guided radiotherapy, intensity-modulated radiotherapy, stereotactic body radiotherapy and proton beam therapy. The optimal use of imaging might enable higher doses of radiation to be delivered to the tumour, while sparing normal surrounding tissues. In this article, we review how the integration of existing and novel forms of computed tomography, magnetic resonance imaging and positron emission tomography have transformed tumour delineation in the radiotherapy planning process, and how these advances have the potential to allow a more individualised approach to the cancer therapy. Recent data suggest that imaging biomarkers that assess underlying tumour heterogeneity can identify areas within a tumour that are at higher risk of radio-resistance, and therefore potentially allow for biologically focussed dose escalation. The rapidly evolving concept of adaptive radiotherapy, including artificial intelligence, requires imaging during treatment to be used to modify radiotherapy on a daily basis. These advances have the potential to improve clinical outcomes and reduce radiation-related long-term toxicities. We outline how recent technological advances in both imaging and radiotherapy delivery can be combined to shape the future of precision radiation oncology.
Collapse
Affiliation(s)
- Laura Beaton
- Cancer Institute, University College London, London, UK
| | - Steve Bandula
- Cancer Institute, University College London, London, UK.,NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, London, UK
| | - Mark N Gaze
- NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, London, UK
| | - Ricky A Sharma
- Cancer Institute, University College London, London, UK. .,NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, London, UK.
| |
Collapse
|
29
|
Dynamic contrast-enhanced magnetic resonance imaging in locally advanced rectal cancer: role of perfusion parameters in the assessment of response to treatment. Radiol Med 2018; 124:331-338. [PMID: 30560501 DOI: 10.1007/s11547-018-0978-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 12/05/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE To correlate dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parameters to tumor grading and to assess their reliability in predicting pathological complete response (pCR) before neoadjuvant chemoradiotherapy (CRT) in patients with locally advanced rectal cancer (LARC). MATERIALS AND METHODS Forty patients (24 male; mean age, 67.3 ± 8.1 years) with histologically proven LARC who had undergone 3-Tesla DCE-MRI before (MRI_1) and after CRT (MRI_2) between August 2015 and February 2016 were included in this retrospective study. DCE-MRI parameters at MRI_1 and MRI_2 were extracted by two board certified radiologists in consensus reading with Olea Sphere 2.3 software using the extended Tofts model. Based on DCE-MRI results, patients were divided in complete responders (CR) and non-complete responders (nCR) and the perfusion parameters were correlated to tumor grading and pCR. RESULTS Wash-out and Kep at MRI_1 showed significant correlation with LARC grading (P = 0.004 and 0.01, respectively). Ve showed a significant increase between MRI_1 (0.47 ± 0.27) and MRI_2 (0.63 ± 0.23; P = 0.007). Ktrans measured at MRI_1 was significantly higher in CR (0.66 ± 0.48) compared to nCR (0.53 ± 0.34, P = 0.02). CONCLUSION Wash-out and Kep measured before CRT correlate with LARC grading. Ve changes during CRT, while Ktrans measured before CRT may predict the response to therapy. Therefore, DCE-MRI parameters can predict tumor aggressiveness and CRT efficacy, playing a role as imaging biomarkers in patients with LARC.
Collapse
|
30
|
Granata V, Fusco R, Avallone A, Catalano O, Piccirillo M, Palaia R, Nasti G, Petrillo A, Izzo F. A radiologist's point of view in the presurgical and intraoperative setting of colorectal liver metastases. Future Oncol 2018; 14:2189-2206. [PMID: 30084273 DOI: 10.2217/fon-2018-0080] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multidisciplinary management of patients with metastatic colorectal cancer requires in each phase an adequate choice of the most appropriate imaging modality. The first challenging step is liver lesions detection and characterization, using several imaging modality ultrasound, computed tomography, magnetic resonance and positron emission tomography. The criteria to establish the metastases resectability have been modified. Not only the lesions number and site but also the functional volume remnant after surgery and the quality of the nontumoral liver must be taken into account. Radiologists should identify the liver functional volume remnant and during liver surgical procedures should collaborate with the surgeon to identify all lesions, including those that disappeared after the therapy, using intraoperative ultrasound with or without contrast medium.
Collapse
Affiliation(s)
- Vincenza Granata
- Radiology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Roberta Fusco
- Radiology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Antonio Avallone
- Abdominal Oncology Division, Istitutonazionale Tumori - IRCSS - Fondazione G Pascale, Napoli, Italia
| | - Orlando Catalano
- Radiology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Mauro Piccirillo
- Hepatobiliary Surgical Oncology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Raffaele Palaia
- Hepatobiliary Surgical Oncology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Guglielmo Nasti
- Abdominal Oncology Division, Istitutonazionale Tumori - IRCSS - Fondazione G Pascale, Napoli, Italia
| | - Antonella Petrillo
- Radiology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Francesco Izzo
- Hepatobiliary Surgical Oncology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| |
Collapse
|
31
|
Pellino G, Gallo G, Pallante P, Capasso R, De Stefano A, Maretto I, Malapelle U, Qiu S, Nikolaou S, Barina A, Clerico G, Reginelli A, Giuliani A, Sciaudone G, Kontovounisios C, Brunese L, Trompetto M, Selvaggi F. Noninvasive Biomarkers of Colorectal Cancer: Role in Diagnosis and Personalised Treatment Perspectives. Gastroenterol Res Pract 2018; 2018:2397863. [PMID: 30008744 PMCID: PMC6020538 DOI: 10.1155/2018/2397863] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 04/03/2018] [Accepted: 04/15/2018] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related deaths worldwide. It has been estimated that more than one-third of patients are diagnosed when CRC has already spread to the lymph nodes. One out of five patients is diagnosed with metastatic CRC. The stage of diagnosis influences treatment outcome and survival. Notwithstanding the recent advances in multidisciplinary management and treatment of CRC, patients are still reluctant to undergo screening tests because of the associated invasiveness and discomfort (e.g., colonoscopy with biopsies). Moreover, the serological markers currently used for diagnosis are not reliable and, even if they were useful to detect disease recurrence after treatment, they are not always detected in patients with CRC (e.g., CEA). Recently, translational research in CRC has produced a wide spectrum of potential biomarkers that could be useful for diagnosis, treatment, and follow-up of these patients. The aim of this review is to provide an overview of the newer noninvasive or minimally invasive biomarkers of CRC. Here, we discuss imaging and biomolecular diagnostics ranging from their potential usefulness to obtain early and less-invasive diagnosis to their potential implementation in the development of a bespoke treatment of CRC.
Collapse
Affiliation(s)
- Gianluca Pellino
- Unit of General Surgery, Department of Medical, Surgical, Neurological, Metabolic and Ageing Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, Piazza Miraglia 2, 80138 Naples, Italy
- Colorectal Surgery Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Gaetano Gallo
- Department of Medical and Surgical Sciences, OU of General Surgery, University of Catanzaro, Catanzaro, Italy
- Department of Colorectal Surgery, Clinic S. Rita, Vercelli, Italy
| | - Pierlorenzo Pallante
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), Via S. Pansini 5, Naples, Italy
| | - Raffaella Capasso
- Department of Medicine and Health Sciences, University of Molise, Via Francesco de Sanctis 1, 86100 Campobasso, Italy
| | - Alfonso De Stefano
- Department of Abdominal Oncology, Division of Abdominal Medical Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione G. Pascale, ” IRCCS, Naples, Italy
| | - Isacco Maretto
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Umberto Malapelle
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Shengyang Qiu
- Department of Colorectal Surgery, Royal Marsden Hospital, London, UK
| | - Stella Nikolaou
- Department of Colorectal Surgery, Royal Marsden Hospital, London, UK
| | - Andrea Barina
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Giuseppe Clerico
- Department of Colorectal Surgery, Clinic S. Rita, Vercelli, Italy
| | - Alfonso Reginelli
- Department of Internal and Experimental Medicine, Magrassi-Lanzara, Institute of Radiology, Università degli Studi della Campania “Luigi Vanvitelli”, Piazza Miraglia 2, 80138 Naples, Italy
| | - Antonio Giuliani
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Guido Sciaudone
- Unit of General Surgery, Department of Medical, Surgical, Neurological, Metabolic and Ageing Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, Piazza Miraglia 2, 80138 Naples, Italy
| | - Christos Kontovounisios
- Department of Colorectal Surgery, Royal Marsden Hospital, London, UK
- Department of Surgery and Cancer, Chelsea and Westminster Hospital Campus, Imperial College London, London, UK
| | - Luca Brunese
- Department of Medicine and Health Sciences, University of Molise, Via Francesco de Sanctis 1, 86100 Campobasso, Italy
| | - Mario Trompetto
- Department of Colorectal Surgery, Clinic S. Rita, Vercelli, Italy
| | - Francesco Selvaggi
- Unit of General Surgery, Department of Medical, Surgical, Neurological, Metabolic and Ageing Sciences, Università degli Studi della Campania “Luigi Vanvitelli”, Piazza Miraglia 2, 80138 Naples, Italy
| |
Collapse
|
32
|
Werner RA, Lapa C, Ilhan H, Higuchi T, Buck AK, Lehner S, Bartenstein P, Bengel F, Schatka I, Muegge DO, Papp L, Zsótér N, Große-Ophoff T, Essler M, Bundschuh RA. Survival prediction in patients undergoing radionuclide therapy based on intratumoral somatostatin-receptor heterogeneity. Oncotarget 2018; 8:7039-7049. [PMID: 27705948 PMCID: PMC5351689 DOI: 10.18632/oncotarget.12402] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/26/2016] [Indexed: 11/25/2022] Open
Abstract
The NETTER-1 trial demonstrated significantly improved progression-free survival (PFS) for peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumors (NET) emphasizing the high demand for response prediction in appropriate candidates. In this multicenter study, we aimed to elucidate the prognostic value of tumor heterogeneity as assessed by somatostatin receptor (SSTR)-PET/CT. 141 patients with SSTR-expressing tumors were analyzed obtaining SSTR-PET/CT before PRRT (1-6 cycles, 177Lu somatostatin analog). Using the Interview Fusion Workstation (Mediso), a total of 872 metastases were manually segmented. Conventional PET parameters as well as textural features representing intratumoral heterogeneity were computed. The prognostic ability for PFS and overall survival (OS) were examined. After performing Cox regression, independent parameters were determined by ROC analysis to obtain cut-off values to be used for Kaplan-Meier analysis. Within follow-up (median, 43.1 months), 75 patients showed disease progression (median, 22.2 m) and 54 patients died (median, 27.6 m). Cox analysis identified 8 statistically independent heterogeneity parameters for time-to-progression and time-to-death. Among them, the textural feature Entropy predicted both PFS and OS. Conventional PET parameters failed in response prediction. Imaging-based heterogeneity assessment provides prognostic information in PRRT candidates and outperformed conventional PET parameters. Its implementation in clinical practice can pave the way for individualized patient management.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Sebastian Lehner
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Frank Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - László Papp
- Department of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Tobias Große-Ophoff
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| | - Markus Essler
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| |
Collapse
|
33
|
Abstract
Pancreatic adenocarcinoma is a common malignancy that has a poor prognosis. Imaging is vital in its detection, staging, and management. Although a variety of imaging techniques are available, MDCT is the preferred imaging modality for staging and assessing the resectability of pancreatic adenocarcinoma. MR also has an important adjunct role, and may be used in addition to CT or as a problem-solving tool. A dedicated pancreatic protocol should be acquired as a biphasic technique optimized for the detection of pancreatic adenocarcinoma and to allow accurate local and distant disease staging. Emerging techniques like dual-energy CT and texture analysis of CT and MR images have a great potential in improving lesion detection, characterization, and treatment monitoring.
Collapse
|
34
|
Advanced imaging to predict response to chemotherapy in colorectal liver metastases - a systematic review. HPB (Oxford) 2018; 20:120-127. [PMID: 29196021 DOI: 10.1016/j.hpb.2017.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/24/2017] [Accepted: 10/27/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND The assessment of colorectal liver metastases (CRLM) after treatment with chemotherapy is challenging due to morphological and/or functional change without changes in size. The aim of this review was to assess the value of FDG-PET, FDG-PET-CT, CT and MRI in predicting response to chemotherapy in CRLM. METHODS A systematic review was undertaken based on PRISMA statement. PubMed and Embase were searched up to October 2016 for studies on the accuracy of PET, PET-CT, CT and MRI in predicting RECIST or metabolic response to chemotherapy and/or survival in patients with CRLM. Articles evaluating the assessment of response after chemotherapy were excluded. RESULTS Sixteen studies met the inclusion criteria and were included for further analysis. Study results were available for 6 studies for FDG-PET(-CT), 6 studies for CT and 9 studies for MRI. Generally, features predicting RECIST or metabolic response often predicted shorter survival. The ADC (apparent diffusion coefficient, on MRI) seems to be the most promising predictor of response and survival. In CT-related studies, few attenuation-related parameters and texture features show promising results. In FDG-PET(-CT), findings were ambiguous. CONCLUSION Radiological data on the prediction of response to chemotherapy for CRLM is relatively sparse and heterogeneous. Despite that, a promising parameter might be ADC. Second, there seems to be a seemingly counterintuitive correlation between parameters that predict a good response and also predict poor survival.
Collapse
|
35
|
Reyners AKL, Broekman KE, Glaudemans AWJM, Brouwers AH, Arts HJG, van der Zee AGJ, de Vries EGE, Jalving M. Molecular imaging in ovarian cancer. Ann Oncol 2017; 27 Suppl 1:i23-i29. [PMID: 27141066 DOI: 10.1093/annonc/mdw091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer has a high mortality and novel-targeted treatment strategies have not resulted in breakthroughs for this disease. Insight into the molecular characteristics of ovarian tumors may improve diagnosis and selection of patients for treatment with targeted therapies. A potential way to achieve this is by means of molecular imaging. Generic tumor processes, such as glucose metabolism ((18)F-fluorodeoxyglucose) and DNA synthesis ((18)F-fluorodeoxythymidine), can be visualized non-invasively. More specific targets, such as hormone receptors, growth factor receptors, growth factors and targets of immunotherapy, can also be visualized. Molecular imaging can capture data on intra-patient tumor heterogeneity and is of potential value for individualized, target-guided treatment selection. Early changes in molecular characteristics during therapy may serve as early predictors of response. In this review, we describe the current knowledge on molecular imaging in the diagnosis and as an upfront or early predictive biomarker in patients with ovarian cancer.
Collapse
Affiliation(s)
| | | | | | - A H Brouwers
- Department of Nuclear Medicine and Molecular Imaging
| | - H J G Arts
- Department of Gynecological Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A G J van der Zee
- Department of Gynecological Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
36
|
Ning J, Sun Y, Xie S, Zhang B, Huang F, Koken P, Smink J, Yuan C, Chen H. Simultaneous acquisition sequence for improved hepatic pharmacokinetics quantification accuracy (SAHA) for dynamic contrast-enhanced MRI of liver. Magn Reson Med 2017; 79:2629-2641. [PMID: 28905413 DOI: 10.1002/mrm.26915] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/11/2017] [Accepted: 08/19/2017] [Indexed: 12/13/2022]
Abstract
PURPOSE To propose a simultaneous acquisition sequence for improved hepatic pharmacokinetics quantification accuracy (SAHA) method for liver dynamic contrast-enhanced MRI. METHODS The proposed SAHA simultaneously acquired high temporal-resolution 2D images for vascular input function extraction using Cartesian sampling and 3D large-coverage high spatial-resolution liver dynamic contrast-enhanced images using golden angle stack-of-stars acquisition in an interleaved way. Simulations were conducted to investigate the accuracy of SAHA in pharmacokinetic analysis. A healthy volunteer and three patients with cirrhosis or hepatocellular carcinoma were included in the study to investigate the feasibility of SAHA in vivo. RESULTS Simulation studies showed that SAHA can provide closer results to the true values and lower root mean square error of estimated pharmacokinetic parameters in all of the tested scenarios. The in vivo scans of subjects provided fair image quality of both 2D images for arterial input function and portal venous input function and 3D whole liver images. The in vivo fitting results showed that the perfusion parameters of healthy liver were significantly different from those of cirrhotic liver and HCC. CONCLUSIONS The proposed SAHA can provide improved accuracy in pharmacokinetic modeling and is feasible in human liver dynamic contrast-enhanced MRI, suggesting that SAHA is a potential tool for liver dynamic contrast-enhanced MRI. Magn Reson Med 79:2629-2641, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Jia Ning
- Department of Biomedical Engineering, Center for Biomedical Imaging Research, School of Medicine, Tsinghua University, Beijing, China
| | - Yongliang Sun
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Sheng Xie
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | | | | | | | | | - Chun Yuan
- Department of Biomedical Engineering, Center for Biomedical Imaging Research, School of Medicine, Tsinghua University, Beijing, China.,Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Huijun Chen
- Department of Biomedical Engineering, Center for Biomedical Imaging Research, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
37
|
Sadeghi-Naini A, Sannachi L, Tadayyon H, Tran WT, Slodkowska E, Trudeau M, Gandhi S, Pritchard K, Kolios MC, Czarnota GJ. Chemotherapy-Response Monitoring of Breast Cancer Patients Using Quantitative Ultrasound-Based Intra-Tumour Heterogeneities. Sci Rep 2017; 7:10352. [PMID: 28871171 PMCID: PMC5583340 DOI: 10.1038/s41598-017-09678-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer therapies including chemotherapy aim to induce tumour cell death. Cell death introduces alterations in cell morphology and tissue micro-structures that cause measurable changes in tissue echogenicity. This study investigated the effectiveness of quantitative ultrasound (QUS) parametric imaging to characterize intra-tumour heterogeneity and monitor the pathological response of breast cancer to chemotherapy in a large cohort of patients (n = 100). Results demonstrated that QUS imaging can non-invasively monitor pathological response and outcome of breast cancer patients to chemotherapy early following treatment initiation. Specifically, QUS biomarkers quantifying spatial heterogeneities in size, concentration and spacing of acoustic scatterers could predict treatment responses of patients with cross-validated accuracies of 82 ± 0.7%, 86 ± 0.7% and 85 ± 0.9% and areas under the receiver operating characteristic (ROC) curve of 0.75 ± 0.1, 0.80 ± 0.1 and 0.89 ± 0.1 at 1, 4 and 8 weeks after the start of treatment, respectively. The patients classified as responders and non-responders using QUS biomarkers demonstrated significantly different survivals, in good agreement with clinical and pathological endpoints. The results form a basis for using early predictive information on survival-linked patient response to facilitate adapting standard anti-cancer treatments on an individual patient basis.
Collapse
Affiliation(s)
- Ali Sadeghi-Naini
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Lakshmanan Sannachi
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Hadi Tadayyon
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - William T Tran
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Centre for Health and Social Care Research, Sheffield Hallam University, Sheffield, UK
| | - Elzbieta Slodkowska
- Division of Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Maureen Trudeau
- Division of Medical Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Sonal Gandhi
- Division of Medical Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Kathleen Pritchard
- Division of Medical Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Gregory J Czarnota
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada. .,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
38
|
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
|
39
|
Ueno Y, Forghani B, Forghani R, Dohan A, Zeng XZ, Chamming's F, Arseneau J, Fu L, Gilbert L, Gallix B, Reinhold C. Endometrial Carcinoma: MR Imaging-based Texture Model for Preoperative Risk Stratification-A Preliminary Analysis. Radiology 2017; 284:748-757. [PMID: 28493790 DOI: 10.1148/radiol.2017161950] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Purpose To evaluate the associations among mathematical modeling with the use of magnetic resonance (MR) imaging-based texture features and deep myometrial invasion (DMI), lymphovascular space invasion (LVSI), and histologic high-grade endometrial carcinoma. Materials and Methods Institutional review board approval was obtained for this retrospective study. This study included 137 women with endometrial carcinomas measuring greater than 1 cm in maximal diameter who underwent 1.5-T MR imaging before hysterectomy between January 2011 and December 2015. Texture analysis was performed with commercial research software with manual delineation of a region of interest around the tumor on MR images (T2-weighted, diffusion-weighted, and dynamic contrast material-enhanced images and apparent diffusion coefficient maps). Areas under the receiver operating characteristic curve and diagnostic performance of random forest models determined by using a subset of the most relevant texture features were estimated and compared with those of independent and blinded visual assessments by three subspecialty radiologists. Results A total of 180 texture features were extracted and ultimately limited to 11 features for DMI, 12 for LVSI, and 16 for high-grade tumor for random forest modeling. With random forest models, areas under the receiver operating characteristic curve, sensitivity, specificity, accuracy, positive predictive value, and negative predictive value were estimated at 0.84, 79.3%, 82.3%, 81.0%, 76.7%, and 84.4% for DMI; 0.80, 80.9%, 72.5%, 76.6%, 74.3%, and 79.4% for LVSI; and 0.83, 81.0%, 76.8%, 78.1%, 60.7%, and 90.1% for high-grade tumor, respectively. Sensitivity, specificity, accuracy, positive predictive value, and negative predictive value of visual assessment for DMI were 84.5%, 82.3%, 83.2%, 77.7%, and 87.8% (reader 3). Conclusion The mathematical models that incorporated MR imaging-based texture features were associated with the presence of DMI, LVSI, and high-grade tumor and achieved equivalent accuracy to that of subspecialty radiologists for assessment of DMI in endometrial cancers larger than 1 cm. However, these preliminary results must be interpreted with caution until they are validated with an independent data set, because the small sample size relative to the number of features extracted may have resulted in overfitting of the models. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Yoshiko Ueno
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Behzad Forghani
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Reza Forghani
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Anthony Dohan
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Xing Ziggy Zeng
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Foucauld Chamming's
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Jocelyne Arseneau
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Lili Fu
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Lucy Gilbert
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Benoit Gallix
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| | - Caroline Reinhold
- From the Departments of Diagnostic Radiology (Y.U., R.F., A.D., B.G., C.R.), Obstetrics and Gynecology (X.Z.Z., L.G.), and Pathology (J.A., L.F.), Royal Victoria Hospital, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, Canada H4A 3J1; Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan (Y.U.); Reza Forghani Medical Services, Montreal, Canada (B.F., R.F.); Department of Radiology, Jewish General Hospital, Montreal, Canada (R.F.); Department of Body and Interventional Imaging, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Diderot-Paris 7 and INSERM U965, Paris, France (A.D.); and Department of Radiology, Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (F.C.)
| |
Collapse
|
40
|
Thomassin-Naggara I, Soualhi N, Balvay D, Darai E, Cuenod CA. Quantifying tumor vascular heterogeneity with DCE-MRI in complex adnexal masses: A preliminary study. J Magn Reson Imaging 2017; 46:1776-1785. [DOI: 10.1002/jmri.25707] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/02/2017] [Indexed: 01/08/2023] Open
Affiliation(s)
- Isabelle Thomassin-Naggara
- INSERM, UMR970, Parc HEGP Equipe 2, Imagerie de l'angiogenèse; Paris France
- Sorbonne Universités, UPMC Univ Paris 06, IUC; Paris France
- AP-HP, Hôpital Tenon, Department of Radiology; Paris France
| | - Narimane Soualhi
- INSERM, UMR970, Parc HEGP Equipe 2, Imagerie de l'angiogenèse; Paris France
| | - Daniel Balvay
- INSERM, UMR970, Parc HEGP Equipe 2, Imagerie de l'angiogenèse; Paris France
- Plateforme d'Imagerie du Petit Animal; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine; Paris France
| | - Emile Darai
- AP-HP, Hôpital Tenon, Department of Gynaecology and Obstetrics; Paris France
| | | |
Collapse
|
41
|
Abstract
There is interest in identifying and quantifying tumor heterogeneity at the genomic, tissue pathology and clinical imaging scales, as this may help better understand tumor biology and may yield useful biomarkers for guiding therapy-based decision making. This review focuses on the role and value of using x-ray, CT, MRI and PET based imaging methods that identify, measure and map tumor heterogeneity. In particular we highlight the potential value of these techniques and the key challenges required to validate and qualify these biomarkers for clinical use.
Collapse
Affiliation(s)
- James P B O'Connor
- Institute of Cancer Sciences, University of Manchester, Manchester, UK; Department of Radiology, The Christie Hospital NHS Trust, Manchester, UK.
| |
Collapse
|
42
|
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
|
43
|
Russell MR, D'Amato A, Graham C, Crosbie EJ, Gentry-Maharaj A, Ryan A, Kalsi JK, Fourkala EO, Dive C, Walker M, Whetton AD, Menon U, Jacobs I, Graham RL. Novel risk models for early detection and screening of ovarian cancer. Oncotarget 2017; 8:785-797. [PMID: 27903971 PMCID: PMC5352196 DOI: 10.18632/oncotarget.13648] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/14/2016] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Ovarian cancer (OC) is the most lethal gynaecological cancer. Early detection is required to improve patient survival. Risk estimation models were constructed for Type I (Model I) and Type II (Model II) OC from analysis of Protein Z, Fibronectin, C-reactive protein and CA125 levels in prospectively collected samples from the United Kingdom Collaborative Trial of Ovarian Cancer Screening (UKCTOCS). RESULTS Model I identifies cancers earlier than CA125 alone, with a potential lead time of 3-4 years. Model II detects a number of high grade serous cancers at an earlier stage (Stage I/II) than CA125 alone, with a potential lead time of 2-3 years and assigns high risk to patients that the ROCA Algorithm classified as normal. MATERIALS AND METHODS This nested case control study included 418 individual serum samples serially collected from 49 OC cases and 31 controls up to six years pre-diagnosis. Discriminatory logit models were built combining the ELISA results for candidate proteins with CA125 levels. CONCLUSIONS These models have encouraging sensitivities for detecting pre-clinical ovarian cancer, demonstrating improved sensitivity compared to CA125 alone. In addition we demonstrate how the models improve on ROCA for some cases and outline their potential future use as clinical tools.
Collapse
Affiliation(s)
- Matthew R. Russell
- Stoller Biomarker Discovery Centre and Pathology Node, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Alfonsina D'Amato
- Stoller Biomarker Discovery Centre and Pathology Node, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ciaren Graham
- School of Healthcare Science, Manchester Metropolitan University, UK
| | - Emma J Crosbie
- Gynaecological Oncology Research Group, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Aleksandra Gentry-Maharaj
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Andy Ryan
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Jatinderpal K. Kalsi
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Evangelia-Ourania Fourkala
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Michael Walker
- Stoller Biomarker Discovery Centre and Pathology Node, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Anthony D. Whetton
- Stoller Biomarker Discovery Centre and Pathology Node, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Usha Menon
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Ian Jacobs
- Stoller Biomarker Discovery Centre and Pathology Node, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
- University of New South Wales, Australia
| | - Robert L.J. Graham
- Stoller Biomarker Discovery Centre and Pathology Node, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
44
|
Sala E, Mema E, Himoto Y, Veeraraghavan H, Brenton JD, Snyder A, Weigelt B, Vargas HA. Unravelling tumour heterogeneity using next-generation imaging: radiomics, radiogenomics, and habitat imaging. Clin Radiol 2017; 72:3-10. [PMID: 27742105 PMCID: PMC5503113 DOI: 10.1016/j.crad.2016.09.013] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 12/18/2022]
Abstract
Tumour heterogeneity in cancers has been observed at the histological and genetic levels, and increased levels of intra-tumour genetic heterogeneity have been reported to be associated with adverse clinical outcomes. This review provides an overview of radiomics, radiogenomics, and habitat imaging, and examines the use of these newly emergent fields in assessing tumour heterogeneity and its implications. It reviews the potential value of radiomics and radiogenomics in assisting in the diagnosis of cancer disease and determining cancer aggressiveness. This review discusses how radiogenomic analysis can be further used to guide treatment therapy for individual tumours by predicting drug response and potential therapy resistance and examines its role in developing radiomics as biomarkers of oncological outcomes. Lastly, it provides an overview of the obstacles in these emergent fields today including reproducibility, need for validation, imaging analysis standardisation, data sharing and clinical translatability and offers potential solutions to these challenges towards the realisation of precision oncology.
Collapse
Affiliation(s)
- E Sala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - E Mema
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Radiology, New York Presbyterian/Columbia University Medical Center, 622 W 168th St., New York, NY 10032, USA
| | - Y Himoto
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - H Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - J D Brenton
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - A Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - B Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - H A Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
45
|
Hatt M, Tixier F, Pierce L, Kinahan PE, Le Rest CC, Visvikis D. Characterization of PET/CT images using texture analysis: the past, the present… any future? Eur J Nucl Med Mol Imaging 2017; 44:151-165. [PMID: 27271051 PMCID: PMC5283691 DOI: 10.1007/s00259-016-3427-0] [Citation(s) in RCA: 338] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/18/2016] [Indexed: 02/07/2023]
Abstract
After seminal papers over the period 2009 - 2011, the use of texture analysis of PET/CT images for quantification of intratumour uptake heterogeneity has received increasing attention in the last 4 years. Results are difficult to compare due to the heterogeneity of studies and lack of standardization. There are also numerous challenges to address. In this review we provide critical insights into the recent development of texture analysis for quantifying the heterogeneity in PET/CT images, identify issues and challenges, and offer recommendations for the use of texture analysis in clinical research. Numerous potentially confounding issues have been identified, related to the complex workflow for the calculation of textural features, and the dependency of features on various factors such as acquisition, image reconstruction, preprocessing, functional volume segmentation, and methods of establishing and quantifying correspondences with genomic and clinical metrics of interest. A lack of understanding of what the features may represent in terms of the underlying pathophysiological processes and the variability of technical implementation practices makes comparing results in the literature challenging, if not impossible. Since progress as a field requires pooling results, there is an urgent need for standardization and recommendations/guidelines to enable the field to move forward. We provide a list of correct formulae for usual features and recommendations regarding implementation. Studies on larger cohorts with robust statistical analysis and machine learning approaches are promising directions to evaluate the potential of this approach.
Collapse
Affiliation(s)
- Mathieu Hatt
- INSERM, UMR 1101, LaTIM, University of Brest IBSAM, Brest, France.
| | - Florent Tixier
- Nuclear Medicine, University Hospital, Poitiers, France
- Medical school, EE DACTIM, University of Poitiers, Poitiers, France
| | - Larry Pierce
- Imaging Research Laboratory, University of Washington, Seattle, WA, USA
| | - Paul E Kinahan
- Imaging Research Laboratory, University of Washington, Seattle, WA, USA
| | - Catherine Cheze Le Rest
- Nuclear Medicine, University Hospital, Poitiers, France
- Medical school, EE DACTIM, University of Poitiers, Poitiers, France
| | | |
Collapse
|
46
|
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
|
47
|
DCE-MRI Perfusion and Permeability Parameters as predictors of tumor response to CCRT in Patients with locally advanced NSCLC. Sci Rep 2016; 6:35569. [PMID: 27762331 PMCID: PMC5071875 DOI: 10.1038/srep35569] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 09/21/2016] [Indexed: 02/06/2023] Open
Abstract
In this prospective study, 36 patients with stage III non-small cell lung cancers (NSCLC), who underwent dynamic contrast-enhanced MRI (DCE-MRI) before concurrent chemo-radiotherapy (CCRT) were enrolled. Pharmacokinetic analysis was carried out after non-rigid motion registration. The perfusion parameters [including Blood Flow (BF), Blood Volume (BV), Mean Transit Time (MTT)] and permeability parameters [including endothelial transfer constant (Ktrans), reflux rate (Kep), fractional extravascular extracellular space volume (Ve), fractional plasma volume (Vp)] were calculated, and their relationship with tumor regression was evaluated. The value of these parameters on predicting responders were calculated by receiver operating characteristic (ROC) curve. Multivariate logistic regression analysis was conducted to find the independent variables. Tumor regression rate is negatively correlated with Ve and its standard variation Ve_SD and positively correlated with Ktrans and Kep. Significant differences between responders and non-responders existed in Ktrans, Kep, Ve, Ve_SD, MTT, BV_SD and MTT_SD (P < 0.05). ROC indicated that Ve < 0.24 gave the largest area under curve of 0.865 to predict responders. Multivariate logistic regression analysis also showed Ve was a significant predictor. Baseline perfusion and permeability parameters calculated from DCE-MRI were seen to be a viable tool for predicting the early treatment response after CCRT of NSCLC.
Collapse
|
48
|
Van Cutsem E, Verheul HMW, Flamen P, Rougier P, Beets-Tan R, Glynne-Jones R, Seufferlein T. Imaging in Colorectal Cancer: Progress and Challenges for the Clinicians. Cancers (Basel) 2016; 8:cancers8090081. [PMID: 27589804 PMCID: PMC5040983 DOI: 10.3390/cancers8090081] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/22/2016] [Accepted: 08/24/2016] [Indexed: 01/05/2023] Open
Abstract
The use of imaging in colorectal cancer (CRC) has significantly evolved over the last twenty years, establishing important roles in surveillance, diagnosis, staging, treatment selection and follow up. The range of modalities has broadened with the development of novel tracer and contrast agents, and the fusion of technologies such as positron emission tomography (PET) and computed tomography (CT). Traditionally, the most widely used modality for assessing treatment response in metastasised colon and rectal tumours is CT, combined with use of the RECIST guidelines. However, a growing body of evidence suggests that tumour size does not always adequately correlate with clinical outcomes. Magnetic resonance imaging (MRI) is a more versatile technique and dynamic contrast-enhanced (DCE)-MRI and diffusion-weighted (DW)-MRI may be used to evaluate biological and functional effects of treatment. Integrated fluorodeoxyglucose (FDG)-PET/CT combines metabolic and anatomical imaging to improve sensitivity and specificity of tumour detection, and a number of studies have demonstrated improved diagnostic accuracy of this modality in a variety of tumour types, including CRC. These developments have enabled the progression of treatment strategies in rectal cancer and improved the detection of hepatic metastatic disease, yet are not without their limitations. These include technical, economical and logistical challenges, along with a lack of robust evidence for standardisation and formal guidance. In order to successfully apply these novel imaging techniques and utilise their benefit to provide truly personalised cancer care, advances need to be clinically realised in a routine and robust manner.
Collapse
Affiliation(s)
- Eric Van Cutsem
- Department of Gastroenterology/Digestive Oncology, University Hospitals Gasthuisberg Leuven and KU Leuven, 3000 Leuven, Belgium.
| | - Henk M W Verheul
- Division of Medical Oncology, VU University Medical Centre, 1081 HV Amsterdam, The Netherlands.
| | - Patrik Flamen
- Nuclear Medicine Imaging and Therapy Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium.
| | - Philippe Rougier
- Gastroenterology and Digestive Oncology Department, European Hospital, Georges Pompidou, 75015 Paris, France.
| | - Regina Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
| | - Rob Glynne-Jones
- Department of Medical Oncology, Mount Vernon Centre for Cancer Treatment, HA6 2RN Middlesex, UK.
| | - Thomas Seufferlein
- Clinic of Internal Medicine I, University Hospital Ulm, 89081 Ulm, Germany.
| |
Collapse
|
49
|
Sadeghi-Naini A, Vorauer E, Chin L, Falou O, Tran WT, Wright FC, Gandhi S, Yaffe MJ, Czarnota GJ. Early detection of chemotherapy-refractory patients by monitoring textural alterations in diffuse optical spectroscopic images. Med Phys 2016; 42:6130-46. [PMID: 26520706 DOI: 10.1118/1.4931603] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Changes in textural characteristics of diffuse optical spectroscopic (DOS) functional images, accompanied by alterations in their mean values, are demonstrated here for the first time as early surrogates of ultimate treatment response in locally advanced breast cancer (LABC) patients receiving neoadjuvant chemotherapy (NAC). NAC, as a standard component of treatment for LABC patient, induces measurable heterogeneous changes in tumor metabolism which were evaluated using DOS-based metabolic maps. This study characterizes such inhomogeneous nature of response development, by determining alterations in textural properties of DOS images apparent at early stages of therapy, followed later by gross changes in mean values of these functional metabolic maps. METHODS Twelve LABC patients undergoing NAC were scanned before and at four times after treatment initiation, and tomographic DOS images were reconstructed at each time. Ultimate responses of patients were determined clinically and pathologically, based on a reduction in tumor size and assessment of residual tumor cellularity. The mean-value parameters and textural features were extracted from volumetric DOS images for several functional and metabolic parameters prior to the treatment initiation. Changes in these DOS-based biomarkers were also monitored over the course of treatment. The measured biomarkers were applied to differentiate patient responses noninvasively and compared to clinical and pathologic responses. RESULTS Responding and nonresponding patients demonstrated different changes in DOS-based textural and mean-value parameters during chemotherapy. Whereas none of the biomarkers measured prior the start of therapy demonstrated a significant difference between the two patient populations, statistically significant differences were observed at week one after treatment initiation using the relative change in contrast/homogeneity of seven functional maps (0.001<p<0.049), and mean value of water content in tissue (p=0.010). The cross-validated sensitivity and specificity of these parameters at week one of therapy ranged between 80%-100% and 67%-100%, respectively. Higher levels of statistically significant differences were exhibited at week four after start of treatment, with cross-validated sensitivities and specificities ranging between 80% and 100% for three textural and three mean-value parameters. The combination of the textural and mean-value parameters in a "hybrid" profile could better separate the two patient populations early on during a course of treatment, with cross-validated sensitivities and specificities of up to 100% (p=0.001). CONCLUSIONS The results of this study suggest that alterations in textural characteristics of DOS images, in conjunction with changes in their mean values, can classify noninvasively the ultimate clinical and pathologic response of LABC patients to chemotherapy, as early as one week after start of their treatment. This provides a basis for using DOS imaging as a tool for therapy personalization.
Collapse
Affiliation(s)
- Ali Sadeghi-Naini
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N 3M5, Canada; Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; and Department of Radiation Oncology, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - Eric Vorauer
- Department of Medical Physics, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada and Department of Physics, Ryerson University, Toronto, Ontario M5B 2K3, Canada
| | - Lee Chin
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario M4N 3M5, Canada; Department of Medical Physics, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; and Department of Physics, Ryerson University, Toronto, Ontario M5B 2K3, Canada
| | - Omar Falou
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N 3M5, Canada; Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; and Department of Radiation Oncology, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - William T Tran
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | - Frances C Wright
- Division of General Surgery, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada and Department of Surgery, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - Sonal Gandhi
- Division of Medical Oncology, Sunnybrook Health Sciences Centre, and Faculty of Medicine, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - Martin J Yaffe
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada and Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - Gregory J Czarnota
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N 3M5, Canada; Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; and Department of Radiation Oncology, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| |
Collapse
|
50
|
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
|