1
|
Wang R, Liu Z, Yan J, Hua S, Wang Z, Yan Z, Xie X, Hao J, Zhou D, Zhou J, Zhao W, Zheng Y, Tang M, Zhan W. Identification of the histologic transformation of follicular lymphoma using super-resolution microcirculation imaging. Sci Rep 2025; 15:16533. [PMID: 40360644 PMCID: PMC12075498 DOI: 10.1038/s41598-025-01615-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 05/07/2025] [Indexed: 05/15/2025] Open
Abstract
To investigate the ability of super-resolution microcirculation imaging in identifying high-risk histologic transformation (HT) regions, directing the targeted biopsies, and promising timely chemotherapy escalation and outcome improvement. Initially, a retrospective analysis (January 1, 2018-January 1, 2022) of indolent follicular lymphoma (FL) or aggressive diffuse large B cell lymphoma was conducted to identify imaging-based markers of distinguishing aggressive lymphoma from indolent lymphoma. Subsequently, the prospective research consecutively enrolled histologically confirmed FL patients between February 1, 2022, and May 31, 2024, to validate the diagnostic performance of these investigated indicators in differentiating aggressive transformed FL from FL. Diagnostic performance and the diagnostic consistency associated with time were assessed. A total of 132 participants were enrolled: 52 (age 53 years ± 14 [SD]; 34 males) in the retrospective cohort for development and 80 (age 55 years ± 12 [SD]; 34 males), including 10 initial biopsy-confirmed HT, in the prospective cohort for validation. Super-resolution microcirculation imaging demonstrated excellent sensitivity (100%; 95% CI 69.15-100%) and specificity (97.14%; 95% CI 90.06-99.65%) for HT detection, with minimal risk of missed lesions at initial diagnosis. This technique shows potential in early HT detection, facilitating timely chemotherapy escalation and reducing the need for repeat biopsies.
Collapse
Affiliation(s)
- Ronghui Wang
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Zhenhua Liu
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Jipeng Yan
- Ultrasound Lab for Imaging and Sensing, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Siqi Hua
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Zhiqian Wang
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Zixun Yan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Xue Xie
- Department of Ultrasound, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Xuhui District, Shanghai, 200233, People's Republic of China
| | - Junnian Hao
- Department of Ultrasound, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Xuhui District, Shanghai, 200233, People's Republic of China
| | - Dan Zhou
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Jianqiao Zhou
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Weili Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
| | - Yuanyi Zheng
- Department of Ultrasound, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Xuhui District, Shanghai, 200233, People's Republic of China.
| | - Mengxing Tang
- Ultrasound Lab for Imaging and Sensing, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Weiwei Zhan
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
2
|
Lv M, Hui X, Yang X, Li S, Mao Z, Zhang X, Yang K. Comparison of the diagnostic accuracy of enhanced-CT and double contrast-enhanced ultrasound for preoperative T-staging of gastric cancer: a meta-analysis. Cancer Imaging 2025; 25:48. [PMID: 40181411 PMCID: PMC11966938 DOI: 10.1186/s40644-025-00861-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Accurate preoperative staging of gastric cancer (GC) depends on effective diagnostic methods. Enhanced computed tomography (enhanced-CT) is a widely used and reliable preoperative assessment tool for GC, Double Contrast-Enhanced Ultrasound (DCEUS) can display the structure and layers of the gastric wall more accurately, and has high sensitivity (SE) and specificity (SP). OBJECTIVE The present study aims to conduct a comprehensive meta-analysis comparing the preoperative T-staging accuracy of DCEUS and enhanced-CT. METHODS A systematic literature search was conducted across PubMed, Embase, Web of Science, and Cochrane Library to identify eligible articles from inception to February 19, 2024. The study included both prospective and retrospective studies involving patients with GC who underwent DCEUS or enhanced-CT. This encompassed studies utilizing comparative diagnostic test accuracy (CDTA) with both DCEUS and enhanced-CT, as well as studies employing single diagnostic test accuracy (SDTA) with either DCEUS or enhanced-CT alone. Risk of bias was assessed using the Quality Assessment Of Diagnostic Accuracy Studies-C (QUADAS-C) and Assessment Of Diagnostic Accuracy Studies-2 (QUADAS-2) tool. The quality of evidence for each outcome was assessed using GRADE (Grading of Recommendations Assessment, Development and Evaluation). RESULTS A total of 39 studies involving 6,374 patients were included in this meta-analysis. Among these, 3 studies (319 patients) directly compared dynamic contrast-enhanced ultrasound (DCEUS) and enhanced computed tomography (CT), while 31 studies (5,180 patients) evaluated enhanced CT alone, and 5 studies (875 patients) assessed DCEUS alone. For the direct comparison studies (CDTA), DCEUS demonstrated higher sensitivity (SE) and specificity (SP) for T1-T4 staging compared to enhanced CT, with moderate to low certainty of evidence. Specifically, DCEUS showed superior performance in detecting early-stage (T1) and advanced-stage (T4) tumors. Enhanced CT, while effective, had lower sensitivity across all stages, particularly for T1 tumors. In the single-modality studies (SDTA), DCEUS consistently showed higher sensitivity for T2-T4 staging compared to enhanced CT, with comparable specificity. However, the certainty of evidence for indirect comparisons was very low, highlighting the need for further high-quality comparative studies. Overall, DCEUS appears to be a promising modality for gastric cancer T staging, particularly for early-stage detection, but the limited number of direct comparative studies underscores the need for more robust evidence. CONCLUSION Current evidence indicates that DCEUS significantly outperforms enhanced-CT in terms of SE and diagnostic accuracy for preoperative T-staging of GC, while maintaining comparable SP. However, these findings require further validation through rigorous studies with larger sample sizes and improved methodological quality.
Collapse
Affiliation(s)
- MingYue Lv
- The First Clinical Medical College of Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, China
- Evidence-Based Medicine Centre, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
| | - Xu Hui
- Evidence-Based Medicine Centre, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
- Centre for Evidence-Based Social Science/Center for Health Technology Assessment, School of Public Health, Lanzhou University, Lanzhou, 730000, China
- Gansu Key Laboratory of Evidence-Based Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xin Yang
- The First Clinical Medical College of Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, China
- Evidence-Based Medicine Centre, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China
| | - SuSu Li
- The First Clinical Medical College of Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - ZhiGuo Mao
- The First Clinical Medical College of Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - XinHua Zhang
- Department of Ultrasound Medicine, Gansu University of Chinese Medicine, Gansu Provincial People'S Hospital, No. 204, Donggang West Road, Chengguan District, Lanzhou, 730000, China.
| | - KeHu Yang
- Evidence-Based Medicine Centre, School of Basic Medical Science, Lanzhou University, Lanzhou, 730000, China.
- Centre for Evidence-Based Social Science/Center for Health Technology Assessment, School of Public Health, Lanzhou University, Lanzhou, 730000, China.
- Gansu Key Laboratory of Evidence-Based Medicine, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
3
|
Wang D, Wang Q, Su Q, Wang S, Jian Z, Li J, Ye F, Hou Y, Wan M. Multi-Parametric Retinal Microvascular Functional Perfusion Imaging Based on Dynamic Fundus Fluorescence Angiography. IEEE Trans Biomed Eng 2024; 71:3123-3133. [PMID: 38829760 DOI: 10.1109/tbme.2024.3408636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Retinal microvascular disease has caused serious visual impairment widely in the world, which can be hopefully prevented via early and precision microvascular hemodynamic diagnosis. Due to artifacts from choroidal microvessels and tiny movements, current fundus microvascular imaging techniques including fundus fluorescein angiography (FFA) precisely identify retinal microvascular microstructural damage and abnormal hemodynamic changes difficulty, especially in the early stage. Therefore, this study proposes an FFA-based multi-parametric retinal microvascular functional perfusion imaging (RM-FPI) scheme to assess the microstructural damage and quantify its hemodynamic distribution precisely. Herein, a spatiotemporal filter based on singular value decomposition combined with a lognormal fitting model was used to remove the above artifacts. Dynamic FFAs of patients (n = 7) were collected first. The retinal time fluorescence intensity curves were extracted and the corresponding perfusion parameters were estimated after decomposition filtering and model fitting. Compared with in vivo results without filtering and fitting, the signal-to-clutter ratio of retinal perfusion curves, average contrast, and resolution of RM-FPI were up to 7.32 ± 0.43 dB, 14.34 ± 0.24 dB, and 11.0 ± 2.0 µm, respectively. RM-FPI imaged retinal microvascular distribution and quantified its spatial hemodynamic changes, which further characterized the parabolic distribution of local blood flow within diameters ranging from 9 to 400 µm. Finally, RM-FPI was used to quantify, visualize, and diagnose the retinal hemodynamics of retinal vein occlusion from mild to severe. Therefore, this study provided a scheme for early and precision diagnosis of retinal microvascular disease, which might be beneficial in preventing its development.
Collapse
|
4
|
Shrestha B, Stern NB, Zhou A, Dunn A, Porter T. Current trends in the characterization and monitoring of vascular response to cancer therapy. Cancer Imaging 2024; 24:143. [PMID: 39438891 PMCID: PMC11515715 DOI: 10.1186/s40644-024-00767-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/26/2024] [Indexed: 10/25/2024] Open
Abstract
Tumor vascular physiology is an important determinant of disease progression as well as the therapeutic outcome of cancer treatment. Angiogenesis or the lack of it provides crucial information about the tumor's blood supply and therefore can be used as an index for cancer growth and progression. While standalone anti-angiogenic therapy demonstrated limited therapeutic benefits, its combination with chemotherapeutic agents improved the overall survival of cancer patients. This could be attributed to the effect of vascular normalization, a dynamic process that temporarily reverts abnormal vasculature to the normal phenotype maximizing the delivery and intratumor distribution of chemotherapeutic agents. Longitudinal monitoring of vascular changes following antiangiogenic therapy can indicate an optimal window for drug administration and estimate the potential outcome of treatment. This review primarily focuses on the status of various imaging modalities used for the longitudinal characterization of vascular changes before and after anti-angiogenic therapies and their clinical prospects.
Collapse
Affiliation(s)
- Binita Shrestha
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Noah B Stern
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Annie Zhou
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Dunn
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Tyrone Porter
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
5
|
Mori N, Li L, Matsuda M, Mori Y, Mugikura S. Prospects of perfusion contrast-enhanced ultrasound (CE-US) in diagnosing axillary lymph node metastases in breast cancer: a comparison with lymphatic CE-US. J Med Ultrason (2001) 2024; 51:587-597. [PMID: 38642268 PMCID: PMC11499517 DOI: 10.1007/s10396-024-01444-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/18/2024] [Indexed: 04/22/2024]
Abstract
Accurate diagnosis of lymph node (LN) metastasis is vital for prognosis and treatment in patients with breast cancer. Imaging 1modalities such as ultrasound (US), MRI, CT, and 18F-FDG PET/CT are used for preoperative assessment. While conventional US is commonly recommended due to its resolution and sensitivity, it has limitations such as operator subjectivity and difficulty detecting small metastases. This review shows the microanatomy of axillary LNs to enhance accurate diagnosis and the characteristics of contrast-enhanced US (CE-US), which utilizes intravascular microbubble contrast agents, making it ideal for vascular imaging. A significant focus of this review is on distinguishing between two types of CE-US techniques for axillary LN evaluation: perfusion CE-US and lymphatic CE-US. Perfusion CE-US is used to assess LN metastasis via transvenous contrast agent administration, while lymphatic CE-US is used to identify sentinel LNs and diagnose LN metastasis through percutaneous contrast agent administration. This review also highlights the need for future research to clarify the distinction between studies involving "apparently enlarged LNs" and "clinical node-negative" cases in perfusion CE-US research. Such research standardization is essential to ensure accurate diagnostic performance in various clinical studies. Future studies should aim to standardize CE-US methods for improved LN metastasis diagnosis, not only in breast cancer but also across various malignancies.
Collapse
Affiliation(s)
- Naoko Mori
- Department of Radiology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita, 010-8543, Japan.
| | - Li Li
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Masazumi Matsuda
- Department of Radiology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita, 010-8543, Japan
| | - Yu Mori
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Shunji Mugikura
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
- Division of Image Statistics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| |
Collapse
|
6
|
Weng H, Hu H, Zhao Y, Xu Y, Chen P, Huang P. Clinical diagnostic model for predicting indolent or aggressive lymphoma based on clinical information and ultrasound features of superficial lymph nodes. Eur J Radiol 2024; 181:111738. [PMID: 39293239 DOI: 10.1016/j.ejrad.2024.111738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/17/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
PURPOSE The aim of this study was to develop a diagnostic model for predicting indolent lymphoma or aggressive lymphoma using clinical information and ultrasound characteristics of superficial lymph nodes. METHOD Patients with confirmed pathological lymphoma subtypes who had undergone ultrasound and contrast-enhanced ultrasound examinations were enrolled. Clinical and ultrasound imaging features were retrospectively analysed and compared to the pathological results, which were considered the gold standard for diagnosis. Two diagnostic models were developed: a clinical model (Model-C) using clinical data only, and a combined model (Model-US) integrating ultrasound features into the clinical model. The efficacy of these models in differentiating between indolent and aggressive lymphoma was compared. RESULTS In total, 236 consecutive patients were enrolled, including 78 patients with indolent lymphomas and 158 patients with aggressive lymphomas. Receiver operating characteristic (ROC) curve analysis revealed that the areas under the curves of Model-C and Model-US were 0.78 (95 % confidence interval: 0.72-0.84) and 0.87 (95 % confidence interval: 0.82-0.92), respectively (p < 0.001). Model-US was further evaluated for calibration and is presented as a nomogram. CONCLUSIONS The diagnostic model incorporated clinical and ultrasound characteristics and offered a noninvasive method for assessing lymphoma with good discrimination and calibration.
Collapse
Affiliation(s)
- Huifang Weng
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Huisen Hu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Department of Ultrasound in Medicine, Lanxi People's Hospital, Jinhua, Zhejiang 321100, China
| | - Yanan Zhao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yongyuan Xu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Panpan Chen
- Department of Haematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, China.
| |
Collapse
|
7
|
Ding Y, Peng Y, Zhang J, Pan X, Huang X, Zhang CQ. Diagnostic value of contrast-enhanced ultrasound in the diagnosis of papillary thyroid microcarcinoma: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e37768. [PMID: 38608080 PMCID: PMC11018218 DOI: 10.1097/md.0000000000037768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 03/08/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Using meta-analysis to evaluate the diagnostic value of contrast-enhanced ultrasound (CEUS) in the diagnosis of papillary thyroid microcarcinoma (PTMC). METHODS For this systematic review and meta-analysis, we searched PubMed, Cochrane Library, Web of Science, WanFang Data, VPCS Data, and China National Knowledge Infrastructure electronic databases for diagnostic studies on PTMC by CEUS from January 2013 to November 2022. Data were not available or incomplete such as case reports, nonhuman studies, etc, were excluded. Random-effects meta-analyses were used to evaluate the diagnostic accuracy of CEUS in diagnosing PTMC. The quality of the evidence was assessed with the QUADAS-2 scale. This study is registered on PROSPERO, number CRD42023409417. RESULTS Of 1064 records identified, 33 were eligible. The results showed that the pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio of CEUS in diagnosing PTMC were 0.84 (95% confidence interval [CI] = 0.83-0.86), 0.82 (95% CI = 0.80-0.83), 3.90 (95% CI = 3.23-4.72), 0.21 (95% CI = 0.18-0.25), and 20.01 (95% CI = 14.97-26.74), respectively, and the area under the summary receiver operating characteristic curve was 0.8930 (the Q index was 0.8239). The Deek funnel plot indicated publication bias (P ˂.01). CONCLUSION This meta-analysis provides an overview of diagnostic accuracy of CEUS in diagnosing PTMC which indicates CEUS has a good diagnostic value for PTMC. The limitations of this study are publication bias and strong geographical bias.
Collapse
Affiliation(s)
- Yan Ding
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yulan Peng
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jing Zhang
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xueqin Pan
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xu Huang
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Chun-Quan Zhang
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Porte C, Lisson T, Kohlen M, von Maltzahn F, Dencks S, von Stillfried S, Piepenbrock M, Rix A, Dasgupta A, Koczera P, Boor P, Stickeler E, Schmitz G, Kiessling F. Ultrasound Localization Microscopy for Breast Cancer Imaging in Patients: Protocol Optimization and Comparison with Shear Wave Elastography. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:57-66. [PMID: 37805359 DOI: 10.1016/j.ultrasmedbio.2023.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/25/2023] [Accepted: 09/02/2023] [Indexed: 10/09/2023]
Abstract
OBJECTIVE Ultrasound localization microscopy (ULM) has gained increasing attention in recent years because of its ability to visualize blood vessels at super-resolution. The field of oncology, in particular, could benefit from detailed vascular characterization, for example, for diagnosis and therapy monitoring. This study was aimed at refining ULM for breast cancer patients by optimizing the measurement protocol, identifying translational challenges and combining ULM and shear wave elastography. METHODS We computed ULM images of 11 patients with breast cancer by recording contrast-enhanced ultrasound (CEUS) sequences and post-processing them in an offline pipeline. For CEUS, two different doses and injection speeds of SonoVue were applied. The best injection protocol was determined based on quantitative parameters derived from so-called occurrence maps. In addition, a suitable measurement time window was determined, also considering the occurrence of motion. ULM results were compared with shear wave elastography and histological vessel density. RESULTS At the higher dose and injection speed, the highest number of microbubbles, number of tracks and vessel coverage were achieved, leading to the most detailed representation of tumor vasculature. Even at the highest concentration, no significant overlay of microbubble signals occurred. Motion significantly reduced the number of usable frames, thus limiting the measurement window to 3.5 min. ULM vessel coverage was comparable to the histological vessel fraction and correlated significantly with mean tumor elasticity. CONCLUSION The settings for microbubble injection strongly influence ULM images, thus requiring optimized protocols for different indications. Patient and examiner motion was identified as the main translational challenge for ULM.
Collapse
Affiliation(s)
- Céline Porte
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, Aachen, Germany
| | - Thomas Lisson
- Department of Electrical Engineering and Information Technology, Ruhr University Bochum, Bochum, Germany
| | - Matthias Kohlen
- Department of Gynecology and Obstetrics, University Clinic Aachen, RWTH Aachen University, Aachen, Germany
| | - Finn von Maltzahn
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, Aachen, Germany
| | - Stefanie Dencks
- Department of Electrical Engineering and Information Technology, Ruhr University Bochum, Bochum, Germany
| | - Saskia von Stillfried
- Institute of Pathology, University Clinic Aachen, RWTH Aachen University, Aachen, Germany
| | - Marion Piepenbrock
- Department of Electrical Engineering and Information Technology, Ruhr University Bochum, Bochum, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, Aachen, Germany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, Aachen, Germany
| | - Patrick Koczera
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, University Clinic Aachen, RWTH Aachen University, Aachen, Germany
| | - Elmar Stickeler
- Department of Gynecology and Obstetrics, University Clinic Aachen, RWTH Aachen University, Aachen, Germany
| | - Georg Schmitz
- Department of Electrical Engineering and Information Technology, Ruhr University Bochum, Bochum, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, Aachen, Germany; Fraunhofer Institute for Digital Medicine MEVIS, Aachen, Germany.
| |
Collapse
|
9
|
Hoferer I, Jourdain L, Girot C, Benatsou B, Leguerney I, Cournede PH, Marouf A, Hoarau Y, Lassau N, Pitre-Champagnat S. New calibration setup for quantitative DCE-US imaging protocol: Toward standardization. Med Phys 2023; 50:5541-5552. [PMID: 36939058 DOI: 10.1002/mp.16362] [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: 06/12/2022] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 03/21/2023] Open
Abstract
BACKGROUND The DCE-US (Dynamic Contrast-Enhanced Ultrasonography) imaging protocol predicts the vascular modifications compared with Response Evaluation Criteria in Solid Tumors (RECIST) based mainly on morphological changes. A quantitative biomarker has been validated through the DCE-US multi-centric study for early monitoring of the efficiency of anti-angiogenic cancer treatments. In this context, the question of transposing the use of this biomarker to other types of ultrasound scanners, probes and settings has arisen to maintain the follow-up of patients under anti-angiogenic treatments. As a consequence, radiologists encounter standardization issues between the different generations of ultrasound scanners to perform quantitative imaging protocols. PURPOSE The aim of this study was to develop a new calibration setup to transpose the DCE-US imaging protocol to the new generation of ultrasound scanners using both abdominal and linear probes. METHODS This calibration method has been designed to be easily reproducible and optimized, reducing the time required and cost incurred. It is based on an original set-up that includes using a concentration splitter to measure the variation of the harmonic signal intensity, obtained from the Area Under the time-intensity Curve (AUC) as a function of various contrast-agent concentrations. The splitter provided four different concentrations simultaneously ranging from 12.5% to 100% of the initial concentration of the SonoVue contrast agent (Bracco Imaging S.p.A., Milan, Italy), therefore, measuring four AUCs in a single injection. The plot of the AUC as a function of the four contrast agent concentrations represents the intensity variation of the harmonic signal: the slope being the calibration parameter. The standardization through this method implied that both generations of ultrasound scanners had to have the same slopes to be considered as calibrated. This method was tested on two ultrasound scanners from the same manufacturer (Aplio500, Aplioi900, Canon Medical Systems, Tokyo, Japan). The Aplio500 used the settings defined by the initial multicenter DCE-US study. The Mechanical Index (MI) and the Color Gain (CG) of the Aplioi900 have been adjusted to match those of the Aplio500. The reliability of the new setup was evaluated in terms of measurement repeatability, and reproducibility with the agreement between the measurements obtained once the two ultrasound scanners were calibrated. RESULTS The new setup provided excellent repeatability measurements with a value of 96.8%. Once the two ultrasound scanners have been calibrated for both types of probes, the reproducibility was excellent with the agreement between their respective quantitative measurement was at the lowest 95.4% and at the best 98.8%. The settings of the Aplioi900 (Canon Medical Systems) were adjusted to match those of the Aplio500 (Canon Medical Systems) and these validated settings were for the abdominal probe: MI = 0.13 and CG = 34 dB; and for the linear probe: MI = 0.10 and CG = 38 dB. CONCLUSION This new calibration setup provided reliable measurements and enabled the rapid transfer and the use of the DCE-US imaging protocol on new ultrasound scanners, thus permitting a continuation of the therapeutic evaluation of patients through quantitative imaging.
Collapse
Affiliation(s)
- Isaline Hoferer
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy Cancer Campus, Villejuif, France
- Imaging Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Laurene Jourdain
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy Cancer Campus, Villejuif, France
| | - Charly Girot
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy Cancer Campus, Villejuif, France
| | - Baya Benatsou
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy Cancer Campus, Villejuif, France
- Imaging Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Ingrid Leguerney
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy Cancer Campus, Villejuif, France
- Imaging Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Paul-Henry Cournede
- Université Paris-Saclay, CentraleSupélec, Laboratory of Mathematics and Computer Science (MICS), Gif-Sur-Yvette, France
| | | | - Yannick Hoarau
- Université de Strasbourg, CNRS, ICUBE UMR 7357, Strasbourg, France
| | - Nathalie Lassau
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy Cancer Campus, Villejuif, France
- Imaging Department, Gustave Roussy Cancer Campus, Villejuif, France
| | | |
Collapse
|
10
|
Xiao F, Li JM, Han ZY, Liu FY, Yu J, Xie MX, Zhou P, Liang L, Zhou GM, Che Y, Wang SR, Liu C, Cong ZB, Liang P. Multimodality US versus Thyroid Imaging Reporting and Data System Criteria in Recommending Fine-Needle Aspiration of Thyroid Nodules. Radiology 2023; 307:e221408. [PMID: 37367448 DOI: 10.1148/radiol.221408] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Background Current guidelines recommend the use of conventional US for risk stratification and management of thyroid nodules. However, fine-needle aspiration (FNA) is often recommended in benign nodules. Purpose To compare the diagnostic performance of multimodality US (including conventional US, strain elastography, and contrast-enhanced US [CEUS]) with the American College of Radiology Thyroid Imaging Reporting and Data System (TI-RADS) in the recommendation of FNA for thyroid nodules to reduce unnecessary biopsies. Materials and Methods In this prospective study, 445 consecutive participants with thyroid nodules from nine tertiary referral hospitals were recruited between October 2020 and May 2021. With univariable and multivariable logistic regression, the prediction models incorporating sonographic features, evaluated with interobserver agreement, were constructed and internally validated with bootstrap resampling technique. In addition, discrimination, calibration, and decision curve analysis were performed. Results A total of 434 thyroid nodules confirmed at pathologic analysis (259 malignant thyroid nodules) in 434 participants (mean age, 45 years ± 12 [SD]; 307 female participants) were included. Four multivariable models incorporated participant age, nodule features at US (proportion of cystic components, echogenicity, margin, shape, punctate echogenic foci), elastography features (stiffness), and CEUS features (blood volume). In recommending FNA in thyroid nodules, the highest area under the receiver operating characteristic curve (AUC) was 0.85 (95% CI: 0.81, 0.89) for the multimodality US model, and the lowest AUC was 0.63 (95% CI: 0.59, 0.68) for TI-RADS (P < .001). At the 50% risk threshold, 31% (95% CI: 26, 38) of FNA procedures could be avoided with multimodality US compared with 15% (95% CI: 12, 19) with TI-RADS (P < .001). Conclusion Multimodality US had better performance in recommending FNA to avoid unnecessary biopsies than the TI-RADS. Clinical trial registration no. NCT04574258 © RSNA, 2023 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Fan Xiao
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Jian-Ming Li
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Zhi-Yu Han
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Fang-Yi Liu
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Jie Yu
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Ming-Xing Xie
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Ping Zhou
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Lei Liang
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Gui-Ming Zhou
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Ying Che
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Shu-Rong Wang
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Cun Liu
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Zhi-Bin Cong
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| | - Ping Liang
- From the Department of Interventional Ultrasound, Fifth Medical Center of Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China (F.X., J.M.L., Z.Y.H., F.Y.L., J.Y., P.L.); Department of Cadet Corps, Chinese PLA Medical School, Beijing, China (F.X.); Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (M.X.X.); Department of Ultrasound, Third Xiangya Hospital, Central South University, Hunan, China (P.Z.); Department of Ultrasound, Aero-space Center Hospital, Beijing, China (L.L.); Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin, China (G.M.Z.); Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C.); Department of Ultrasound, Yantai Hospital of Shandong Wendeng Orthopaedics & Traumatology, Yantai, China (S.R.W.); Department of Ultrasound, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China (C.L.); and Department of Ultrasound, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China (Z.B.C.)
| |
Collapse
|
11
|
Yan M, Li R, Hu D, Zhao P. Detection of Subchondral Bone Microcirculatory Perfusion in Adults with Early Osteonecrosis of the Femoral Head Using Contrast-Enhanced Ultrasound: A Prospective Study. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:635-644. [PMID: 36336550 DOI: 10.1016/j.ultrasmedbio.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/13/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
The aim of this study was to quantitatively assess subchondral bone microcirculation perfusion in adults with early osteonecrosis of the femoral head (ONFH) using contrast-enhanced ultrasound (CEUS) and to evaluate its correlation with the Association Research Circulation Osseous (ARCO) stage. We investigated 97 adult patients with definite ONFH by imaging a total of 155 hips, performing CEUS, storing images of CEUS processes at different ARCO stages and generating CEUS time-intensity curves (TICs) to obtain perfusion parameters. Differences in CEUS parameters at different ARCO stages were analyzed, and correlations were explored. A logistic regression model was constructed by incorporating the meaningful CEUS indicators. The CEUS parameters time to peak (TTP), peak intensity (PI), enhanced intensity (EI), ascending slope (AS), descending slope (DS) and area under the receiver operating characteristic curve (AUC) were significantly different in ARCO stage Ⅰ compared with stage ⅢA, and the same results were obtained in stage Ⅱ compared with stage ⅢA. However, there were no significant differences between stages Ⅰ and Ⅱ. The MTT (mean transit time) assay was not significantly different between the different stages. The receiver operating characteristic curve analysis of TTP, PI, EI, AS, DS and AUC in stages Ⅰ and ⅢA had a certain diagnostic efficacy, similar to the results in stages Ⅱ and ⅢA. The diagnostic performance of DS was less accurate in stages Ⅰ and ⅢA, while the diagnostic performance of TTP was less accurate in stages Ⅱ and ⅢA. ARCO stage was independently and negatively correlated with TTP and DS and independently and positively correlated with PI, EI, AS and AUC. The MTT assay was not correlated with ARCO stage. Logistic regression models containing statistically significant TTP, EI and AUC values were constructed, and all three values were closely related to the ARCO stage. In patients with different ARCO stages of ONFH, CEUS can effectively assess subchondral bone perfusion of the femoral head and is expected to become an effective imaging method for the diagnosis of early ONFH.
Collapse
Affiliation(s)
- Meijun Yan
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruoyu Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Die Hu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ping Zhao
- Department of Ultrasound, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
12
|
Wu R, Wang K, Gai Y, Li M, Wang J, Wang C, Zhang Y, Xiao Z, Jiang D, Gao Z, Xia X. Nanomedicine for renal cell carcinoma: imaging, treatment and beyond. J Nanobiotechnology 2023; 21:3. [PMID: 36597108 PMCID: PMC9809106 DOI: 10.1186/s12951-022-01761-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023] Open
Abstract
The kidney is a vital organ responsible for maintaining homeostasis in the human body. However, renal cell carcinoma (RCC) is a common malignancy of the urinary system and represents a serious threat to human health. Although the overall survival of RCC has improved substantially with the development of cancer diagnosis and management, there are various reasons for treatment failure. Firstly, without any readily available biomarkers, timely diagnosis has been greatly hampered. Secondly, the imaging appearance also varies greatly, and its early detection often remains difficult. Thirdly, chemotherapy has been validated as unavailable for treating renal cancer in the clinic due to its intrinsic drug resistance. Concomitant with the progress of nanotechnological methods in pharmaceuticals, the management of kidney cancer has undergone a transformation in the recent decade. Nanotechnology has shown many advantages over widely used traditional methods, leading to broad biomedical applications ranging from drug delivery, prevention, diagnosis to treatment. This review focuses on nanotechnologies in RCC management and further discusses their biomedical translation with the aim of identifying the most promising nanomedicines for clinical needs. As our understanding of nanotechnologies continues to grow, more opportunities to improve the management of renal cancer are expected to emerge.
Collapse
Affiliation(s)
- Ruolin Wu
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Keshan Wang
- grid.33199.310000 0004 0368 7223Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yongkang Gai
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Mengting Li
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Jingjing Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Chenyang Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Yajing Zhang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zhiwei Xiao
- grid.413247.70000 0004 1808 0969Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dawei Jiang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zairong Gao
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Xiaotian Xia
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| |
Collapse
|
13
|
Early Assessment of Chemoradiotherapy Response for Locally Advanced Pancreatic Ductal Adenocarcinoma by Dynamic Contrast-Enhanced Ultrasound. Diagnostics (Basel) 2022; 12:diagnostics12112662. [PMID: 36359506 PMCID: PMC9689529 DOI: 10.3390/diagnostics12112662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Objective: To evaluate the value of dynamic contrast-enhanced ultrasound (DCE-US) and quantitative parameters in early prediction of tumor response to chemoradiotherapy (CRT) in patients with locally advanced pancreatic ductal adenocarcinoma (LAPC). Patients and Methods: In this prospective study, patients with biopsy-proved and histopathologically proved LAPC who underwent regular CRT were recruited. DCE-US evaluations were performed before and four months after CRT. SonoVue-enhanced contrast-enhanced ultrasound (CEUS) was performed by an ultrasound system (ACUSON Sequoia; Siemens Medical Solutions, USA) equipped with a 5C1 MHz convex array transducer. Time−intensity curves were created by VueBox software (Bracco, Italy), and various DCE-US quantitative parameters were obtained. Taking Response Evaluation Criteria in Solid Tumors (RECIST) based on computed tomography (CT) or magnetic resonance imaging (MRI) as the gold standard, DCE-US parameters were compared between the treatment responder group (RG) and non-responder group (NRG). The correlation between the DCE-US parameters and the serum carbohydrate antigen 19-9 (CA 19-9) level was also analyzed. Results: Finally, 21 LAPC patients (mean age 59.3 ± 7.2 years) were included. In comparing the RG (n = 18) and NRG (n = 3), no significant change could be found among the mean size of the lesions (31.2 ± 8.1 mm vs. 27.2 ± 8.3 mm, p = 0.135). In comparing the TICs between the two groups, the LAPC lesions in the RG took a longer time to reach peak enhancement and to wash out. Among all the DCE-US parameters, RT (rise time), WiAUC (wash-in area under the curve), WoAUC (wash-out area under the curve) and WiWoAUC (wash-in and wash-out area under the curve) decreased significantly after CRT in the RG (p < 0.05). The RT ratio, WiAUC ratio, WoAUC ratio and WiWoAUC ratio were closely correlated with the change in serum CA 19-9 level in the RG (p < 0.05). Conclusion: DCE-US might be a potential imaging method for non-invasive follow-up for early response in LAPC patients treated by CRT.
Collapse
|
14
|
Avry F, Mousset C, Oujagir E, Bouakaz A, Gouilleux-Gruart V, Thépault RA, Renault S, Marouillat S, Machet L, Escoffre JM. Microbubble-Assisted Ultrasound for Imaging and Therapy of Melanoma Skin Cancer: A Systematic Review. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:2174-2198. [PMID: 36050232 DOI: 10.1016/j.ultrasmedbio.2022.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023]
Abstract
Recent technological developments in ultrasound (US) imaging and ultrasound contrast agents (UCAs) have improved diagnostic confidence in echography. In the clinical management of melanoma, contrast-enhanced ultrasound (CEUS) imaging complements conventional US imaging (i.e., high-resolution US and Doppler imaging) for clinical examination and therapeutic follow-up. These developments have set into motion the combined use of ultrasound and UCAs as a new modality for drug delivery. This modality, called sonoporation, has emerged as a non-invasive, targeted and safe method for the delivery of therapeutic drugs into melanoma. This review focuses on the results and prospects of using US and UCAs as dual modalities for CEUS imaging and melanoma treatment.
Collapse
Affiliation(s)
- François Avry
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
| | - Coralie Mousset
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France; GICC EA 7501, Université de Tours, Tours, France
| | - Edward Oujagir
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
| | | | | | | | | | - Laurent Machet
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France; Department of Dermatology, Tours University Hospital, Tours, France
| | | |
Collapse
|
15
|
Girot C, Volk A, Walczak C, Lassau N, Pitre-Champagnat S. New method for quantification of intratumoral heterogeneity: a feasibility study on K trans maps from preclinical DCE-MRI. MAGMA (NEW YORK, N.Y.) 2021; 34:845-857. [PMID: 34091826 DOI: 10.1007/s10334-021-00930-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/31/2022]
Abstract
OBJECT To develop new imaging biomarkers of therapeutic efficacy through the quantification of intratumoral microvascular heterogeneity. MATERIALS AND METHODS The described method was a combination of non-supervised clustering and extraction of intratumoral complexity features (ICF): number of non-connected objects, volume fraction. It was applied to a set of 3D DCE-MRI Ktrans maps acquired previously on tumor bearing mice prior to and on day 4 of anti-angiogenic treatment. Evolutions of ICF were compared to conventional summary statistics (CSS) and to heterogeneity related whole tumor texture features (TF) on treated (n = 9) and control (n = 6) mice. RESULTS Computed optimal number of clusters per tumor was 4. Several intratumoral features extracted from the clusters were able to monitor a therapy effect. Whereas no feature significantly changed for the control group, 6 features significantly changed for the treated group (4 ICF, 2 CSS). Among these, 5 also significantly differentiated the two groups (3 ICF, 2 CSS). TF failed in demonstrating differences within and between the two groups. DISCUSSION ICF are potential imaging biomarkers for anti-angiogenic therapy assessment. The presented method may be expected to have advantages with respect to texture analysis-based methods regarding interpretability of results and setup of standardized image analysis protocols.
Collapse
Affiliation(s)
- Charly Girot
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France.
| | - Andreas Volk
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Christine Walczak
- Institut Curie, Inserm, Université Paris-Saclay, CNRS, 91405, Orsay, France
| | - Nathalie Lassau
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France.,Département de Radiologie, Gustave Roussy, 94805, Villejuif, France
| | - Stéphanie Pitre-Champagnat
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| |
Collapse
|
16
|
Hwang M, Barnewolt CE, Jüngert J, Prada F, Sridharan A, Didier RA. Contrast-enhanced ultrasound of the pediatric brain. Pediatr Radiol 2021; 51:2270-2283. [PMID: 33599780 PMCID: PMC11458139 DOI: 10.1007/s00247-021-04974-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/10/2020] [Accepted: 01/14/2021] [Indexed: 12/16/2022]
Abstract
Brain contrast-enhanced ultrasound (CEUS) is an emerging application that can complement gray-scale US and yield additional insights into cerebral flow dynamics. CEUS uses intravenous injection of ultrasound contrast agents (UCAs) to highlight tissue perfusion and thus more clearly delineate cerebral pathologies including stroke, hypoxic-ischemic injury and focal lesions such as tumors and vascular malformations. It can be applied not only in infants with open fontanelles but also in older children and adults via a transtemporal window or surgically created acoustic window. Advancements in CEUS technology and post-processing methods for quantitative analysis of UCA kinetics further elucidate cerebral microcirculation. In this review article we discuss the CEUS examination protocol for brain imaging in children, current clinical applications and future directions for research and clinical uses of brain CEUS.
Collapse
Affiliation(s)
- Misun Hwang
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Carol E Barnewolt
- Department of Radiology, Boston Children's Hospital, Harvard University, Boston, MA, USA
| | - Jörg Jüngert
- Department of Pediatrics, Friedrich-Alexander University Erlangen - Nürnberg, Erlangen, Germany
| | - Francesco Prada
- Acoustic Neuroimaging and Therapy Laboratory, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, VA, USA
- Focused Ultrasound Foundation, Charlottesville, VA, USA
| | - Anush Sridharan
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Ryne A Didier
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
Li Y, Wang X, Ren W, Xiao Y, Yu X, Tan X. Cardiac thrombotic stability determined by contrast-enhanced echocardiography: investigative protocol and preliminary results. BMC Cardiovasc Disord 2021; 21:267. [PMID: 34058996 PMCID: PMC8167997 DOI: 10.1186/s12872-021-02085-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/27/2021] [Indexed: 11/11/2022] Open
Abstract
Objective This study’s intent was to test a new system for scoring cardiac thrombotic stability, based on contrast-enhanced ultrasound (CEUS). Methods We used human whole blood for an in vitro thrombotic model involving 1-h (T1h) and 7-day (T7d) subsets. The T1h group was monitored for 1 h continuously to observe for the formation of a new thrombus on the original thrombus base. Changes in thrombotic CEUS images, histologic features, and shear wave elastography were recorded over time. We also studied 28 patients diagnosed with cardiac thrombi, each examined by transthoracic echocardiography and CEUS.Thrombi were scored for substrate (Ts) and hardness (Th) based on the visualized degree of contrast penetration into the thrombi. Statistical analyses of Ts and Th reflected thrombolytic time and risk of embolism to other organs. Results Histologically, the loosely constructed ends of in vitro thrombi solidified over time. In addition, the average Young’s modulus of thrombi over time indicated a progressive increase in hardness. Contrast-enhancing agents were able to penetrate fresh, loose thrombi only, not chronic, stable thrombi. As Ts and Th increased, prolonged thrombolytic time and greater risk of embolism to other organs were apparent. Conclusions Our data suggest that this new CEUS scoring system correlates well with cardiac thrombotic hardness and the quality of its underlying substrate, serving to quantify thrombotic stability.
Collapse
Affiliation(s)
- Ying Li
- Department of Ultrasound, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang City, 110004, China
| | - Xin Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang City, 110004, China.
| | - Weidong Ren
- Department of Ultrasound, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang City, 110004, China
| | - Yangjie Xiao
- Department of Ultrasound, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang City, 110004, China
| | - Xiaona Yu
- Department of Ultrasound, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang City, 110004, China
| | - Xueying Tan
- Department of Ultrasound, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Heping District, Shenyang City, 110004, China
| |
Collapse
|
18
|
Wang H, Ding L, Xu F, He L, Ye L, Huang L, Zhang L, Luo B. Construction of novel amphiphilic chitosan-polylactide graft copolymer nanodroplets for contrast enhanced ultrasound tumor imaging. J Biomater Appl 2021; 36:613-625. [PMID: 33899561 DOI: 10.1177/08853282211011766] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In this experiment, a new amphiphilic chitosan-poly(lactide) graft copolymer was synthesized and characterized by IR, 1H-NMR, XRD, TGA. The obtained chitosan-poly (lactide) graft copolymer was used as the matrix material to prepare nanodroplets (NDs) encapsulating with liquid PFP by double-emulsion and solvent evaporation method. The resulting NDs were characterized by photon correlation spectroscopy and transmission electron microscopy (TEM). The biocompatibility was explored by cytotoxicity assay, cell migration assay and blood biochemistry analysis. The experiments of ultrasonic imaging in vitro and in vivo were carried out with a B-mode clinical ultrasound imaging system. The results of FI-IR and 1H-NMR confirmed the successful grafting reaction of polylactic acid(PLLA) to chitosan with a graft rate of 365%. The average size of the NDs was 101.1 ± 2.7 nm, with the polydispersity index (PDI) of 0.127 ± 0.020, and the zeta potential was -31.8 ± 1.5 mV. From the TEM results, NDs were highly dispersed and had a spherical shape with a distinct capsule structure. The NDs exhibited good stability during storage at 4°C. The NDs solution with different concentrations did not affect cell growth and showed good biocompatibility in cytotoxicity, cell migration and blood biochemistry studies. Under the irradiation of ultrasonic waves, the NDs formed an ultrasonic high signal, which could significantly enhance the ultrasound imaging of tumor tissue in vivo. Taken together, the NDs hold great potential for ultrasound imaging as a nanosized contrast agent.
Collapse
Affiliation(s)
- Huili Wang
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Liqiong Ding
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Fengnan Xu
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Liu He
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Lin Ye
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Lingping Huang
- Department of Medical Ultrasound, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Ling Zhang
- School of Biomedical Engineering, Hubei University of Science and Technology, Xianning, China
| | - Binhua Luo
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
19
|
Wan P, Chen F, Shao W, Liu C, Zhang Y, Wen B, Kong W, Zhang D. Irregular Respiratory Motion Compensation for Liver Contrast-Enhanced Ultrasound via Transport-Based Motion Estimation. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:1117-1130. [PMID: 33108284 DOI: 10.1109/tuffc.2020.3033984] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Contrast-enhanced ultrasound (CEUS) imaging has been widely applied for the detection and characterization of focal liver lesions (FLLs), for its ability to visualize the blood flow in real time. However, cyclic liver motion poses a great challenge to the recovery of perfusion curves as well as quantitative kinetic parameters estimation. Recently, a few gating methods have been proposed to eliminate unexpected intensity fluctuations by the breathing phase estimation, with the assumption that each breathing phase corresponds to a specific lesion position strictly. While practical liver motion tends to be irregular due to changes in the patient's underlying physiologic status, that is, the same phase might not correspond to the same position. To tackle the challenge of motion irregularity, we present a novel motion estimation-based respiratory compensation method, named RCME, which first estimates salient motion information through the framework of optimal transport (OT) by jointly modeling pixel intensity as well as their locations and then employs sparse subspace clustering (SSC) to identify the subset of frames acquired at the same position. Our proposed method is evaluated on 15 clinical CEUS sequences in both qualitative and quantitative manners. Experimental results demonstrate good performance on irregular liver motion compensation.
Collapse
|
20
|
Taiji R, Nishiofuku H, Tanaka T, Minamiguchi K, Fukuoka Y, Saito N, Taguchi H, Matsumoto T, Marugami N, Hirai T, Kichikawa K. Useful Parameters in Dynamic Contrast-enhanced Ultrasonography for Identifying Early Response to Chemotherapy in a Rat Liver Tumor Model. J Clin Imaging Sci 2021; 11:15. [PMID: 33767907 PMCID: PMC7981939 DOI: 10.25259/jcis_6_2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
Objectives The objective of the study is to determine a parameter on the time-intensity curve (TIC) of dynamic contrast-enhanced ultrasonography (DCE-US) that best correlates with tumor growth and to evaluate whether the parameter could correlate with the early response to irinotecan in a rat liver tumor model. Material and Methods Twenty rats with tumors were evaluated (control: Saline, n = 6; treatment: Irinotecan, n = 14) regarding four parameters from TIC: Peak intensity (PI), k value, slope (PI × k), and time to peak (TTP). Relative changes in maximum tumor diameter between day 0 and 10, and parameters in the first 3 days were evaluated. The Mann-Whitney U-test was used to compare differences in tumor size and other parameters. Pearson's correlation coefficients (r) between tumor size and parameters in the control group were calculated. In the treatment group, relative changes of parameters in the first 3 days were compared between responder and non-responder (<20% and ≥20% increase in size on day 10, respectively). Results PI, k value, PI × k, and TTP significantly correlated with tumor growth (r = 0.513, 0.911, 0.665, and 0.741, respectively). The mean RC in k value among responders (n = 6) was significantly lower than non-responders (n = 8) (mean k value, 4.96 vs. 72.5; P = 0.003). Conclusion Parameters of DCE-US could be a useful parameter for identifying early response to irinotecan.
Collapse
Affiliation(s)
- Ryosuke Taiji
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | | | - Toshihiro Tanaka
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | | | - Yasushi Fukuoka
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | - Natsuhiko Saito
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | - Hidehiko Taguchi
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | - Takeshi Matsumoto
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | - Nagaaki Marugami
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | - Toshiko Hirai
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| | - Kimihiko Kichikawa
- Department of Radiology, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
21
|
Wang D, Liu D, Sang Y, Zhang Y, Wan M, Diederich CJ. In vivo Nakagami-m parametric imaging of microbubble-enhanced ultrasound regulated by RF and VF processing techniques. Med Phys 2020; 47:5659-5668. [PMID: 32965033 DOI: 10.1002/mp.14474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/28/2020] [Accepted: 08/19/2020] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Application of the Nakagami statistical model and associated m parameter has the potential to suppress artifacts from adjustable system parameters and operator selections typical in echo amplitude-coded microbubble-enhanced ultrasound (MEUS). However, the feasibility of applying m estimation and determination of the associated Nakagami distribution features for in vivo MEUS remain to be investigated. Sensitivity and discriminability of m-coded MEUS are often limited since raw envelopes are regulated by complex radiofrequency (RF) and video-frequency (VF) processing. This study aims to develop an improved imaging approach for the m parameter estimation which can overcome the above limitations in in vivo condition. METHOD The regulation effects of RF processing of pulse-inversion (PI) harmonic detection techniques and VF processing of logarithmic compression in Nakagami distributions were investigated in MEUS. A window-modulated compounding moment estimator was developed to estimate the MEUS m values. The sensitivity and discriminability of m-coded MEUS were quantified with contrast-to-tissue ratio (CTR), contrast-to-noise ratio (CNR), and axial and lateral resolutions, which were validated through in vivo perfusion experiments on rabbit kidneys. RESULTS Regulated by RF and VF processing, the distributions of MEUS obeyed the Nakagami statistical model. The Nakagami-fitted correlation coefficient was 0.996 ± 0.003 (P < 0.05 in the t test and P < 0.001 in the Kolmogorov-Smirnov test). Among each of the m-coded MEUS methods, the logarithmic m-coded PI-MEUS scheme effectively characterized the peripheral rim perfusion features and details within the renal cortex. The CTR and CNR in this region reached 7.9 ± 1.5 dB and 34.4 ± 1.7 dB, respectively, which were higher than those of standard amplitude-coded MEUS; and the axial and lateral resolutions were 1.02 ± 0.02 and 0.91 ± 0.02 mm, respectively, which were slightly longer than those of amplitude-coded MEUS. CONCLUSIONS The Nakagami statistical model could characterize MEUS even when the envelope distributions were regulated by RF and VF processing. The logarithmic m-coded PI-MEUS scheme significantly improved the sensitivity, discriminability, and robustness of m estimation in MEUS. The scheme provides an option to remove artifacts in echo amplitude-coded MEUS and to distinctly characterize the inherent microvasculature enhanced by microbubbles, with potential to improve and expand the role of MEUS in diagnostic ultrasound.
Collapse
Affiliation(s)
- Diya Wang
- Department of Radiation Oncology, University of California, San Francisco, CA, 94115, USA.,Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, P. R. China
| | - Dong Liu
- Department of Radiation Oncology, University of California, San Francisco, CA, 94115, USA
| | - Yuchao Sang
- Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yu Zhang
- Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, P. R. China
| | - Mingxi Wan
- Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, P. R. China
| | - Chris J Diederich
- Department of Radiation Oncology, University of California, San Francisco, CA, 94115, USA
| |
Collapse
|
22
|
Park AY, Kwon M, Woo OH, Cho KR, Park EK, Cha SH, Song SE, Lee JH, Cha J, Son GS, Seo BK. A Prospective Study on the Value of Ultrasound Microflow Assessment to Distinguish Malignant from Benign Solid Breast Masses: Association between Ultrasound Parameters and Histologic Microvessel Densities. Korean J Radiol 2020; 20:759-772. [PMID: 30993927 PMCID: PMC6470080 DOI: 10.3348/kjr.2018.0515] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/22/2018] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To investigate the value of ultrasound (US) microflow assessment in distinguishing malignant from benign solid breast masses as well as the association between US parameters and histologic microvessel density (MVD). MATERIALS AND METHODS Ninety-eight breast masses (57 benign and 41 malignant) were examined using Superb Microvascular Imaging (SMI) and contrast-enhanced US (CEUS) before biopsy. Two radiologists evaluated the quantitative and qualitative vascular parameters on SMI (vascular index, morphology, distribution, and penetration) and CEUS (time-intensity curve analysis and enhancement characteristics). US parameters were compared between benign and malignant masses and the diagnostic performance was compared between SMI and CEUS. Subgroup analysis was performed according to lesion size. The effect of vascular parameters on downgrading Breast Imaging Reporting and Data System (BI-RADS) category 4A masses was evaluated. The association between histologic MVD and US parameters was analyzed. RESULTS Malignant masses were associated with a higher vascular index (15.1 ± 7.3 vs. 5.9 ± 5.6), complex vessel morphology (82.9% vs. 42.1%), central vascularity (95.1% vs. 59.6%), penetrating vessels (80.5% vs. 31.6%) on SMI (all, p < 0.001), as well as higher peak intensity (37.1 ± 25.7 vs. 17.0 ± 15.8, p < 0.001), slope (10.6 ± 11.2 vs. 3.9 ± 4.2, p = 0.001), area (1035.7 ± 726.9 vs. 458.2 ± 410.2, p < 0.001), hyperenhancement (95.1% vs. 70.2%, p = 0.005), centripetal enhancement (70.7% vs. 45.6%, p = 0.023), penetrating vessels (65.9% vs. 22.8%, p < 0.001), and perfusion defects (31.7% vs. 3.5%, p < 0.001) on CEUS (p ≤ 0.023). The areas under the receiver operating characteristic curve (AUCs) of SMI and CEUS were 0.853 and 0.841, respectively (p = 0.803). In 19 masses measuring < 10 mm, central vascularity on SMI was associated with malignancy (100% vs. 38.5%, p = 0.018). Considering all benign SMI parameters on the BI-RADS assessment, unnecessary biopsies could be avoided in 12 category 4A masses with improved AUCs (0.500 vs. 0.605, p < 0.001). US vascular parameters associated with malignancy showed higher MVD (p ≤ 0.016). MVD was higher in malignant masses than in benign masses, and malignant masses negative for estrogen receptor or positive for Ki67 had higher MVD (p < 0.05). CONCLUSION US microflow assessment using SMI and CEUS is valuable in distinguishing malignant from benign solid breast masses, and US vascular parameters are associated with histologic MVD.
Collapse
Affiliation(s)
- Ah Young Park
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea.,Department of Radiology, Bundang CHA Medical Center, CHA University, Seongnam, Korea
| | - Myoungae Kwon
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Ok Hee Woo
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Kyu Ran Cho
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Eun Kyung Park
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Sang Hoon Cha
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Sung Eun Song
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Ju Han Lee
- Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - JaeHyung Cha
- Medical Science Research Center, Korea University Ansan Hospital, Ansan, Korea
| | - Gil Soo Son
- Division of Breast Endocrine Surgery, Department of General Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Bo Kyoung Seo
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea.
| |
Collapse
|
23
|
Butler M, Perperidis A, Zahra JLM, Silva N, Averkiou M, Duncan WC, McNeilly A, Sboros V. Differentiation of Vascular Characteristics Using Contrast-Enhanced Ultrasound Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:2444-2455. [PMID: 31208880 DOI: 10.1016/j.ultrasmedbio.2019.05.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 05/02/2019] [Accepted: 05/10/2019] [Indexed: 05/09/2023]
Abstract
Ultrasound contrast imaging has been used to assess tumour growth and regression by assessing the flow through the macro- and micro-vasculature. Our aim was to differentiate the blood kinetics of vessels such as veins, arteries and microvasculature within the limits of the spatial resolution of contrast-enhanced ultrasound imaging. The highly vascularised ovine ovary was used as a biological model. Perfusion of the ovary with SonoVue was recorded with a Philips iU22 scanner in contrast imaging mode. One ewe was treated with prostaglandin to induce vascular regression. Time-intensity curves (TIC) for different regions of interest were obtained, a lognormal model was fitted and flow parameters calculated. Parametric maps of the whole imaging plane were generated for 2 × 2 pixel regions of interest. Further analysis of TICs from selected locations helped specify parameters associated with differentiation into four categories of vessels (arteries, veins, medium-sized vessels and micro-vessels). Time-dependent parameters were associated with large veins, whereas intensity-dependent parameters were associated with large arteries. Further development may enable automation of the technique as an efficient way of monitoring vessel distributions in a clinical setting using currently available scanners.
Collapse
Affiliation(s)
- Mairead Butler
- Heriot-Watt University, Institute of Biochemistry, Biological Physics and Bio Engineering, Riccarton, Edinburgh, UK.
| | - Antonios Perperidis
- Heriot-Watt University, Institute of Signals, Sensors and Systems, Riccarton, Edinburgh, UK
| | | | - Nadia Silva
- Centre for Marine Sciences, University of Algarve Faro, Portugal
| | - Michalakis Averkiou
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - W Colin Duncan
- Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Alan McNeilly
- Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Vassilis Sboros
- Heriot-Watt University, Institute of Biochemistry, Biological Physics and Bio Engineering, Riccarton, Edinburgh, UK
| |
Collapse
|
24
|
Kanoulas E, Butler M, Rowley C, Voulgaridou V, Diamantis K, Duncan WC, McNeilly A, Averkiou M, Wijkstra H, Mischi M, Wilson RS, Lu W, Sboros V. Super-Resolution Contrast-Enhanced Ultrasound Methodology for the Identification of In Vivo Vascular Dynamics in 2D. Invest Radiol 2019; 54:500-516. [PMID: 31058661 PMCID: PMC6661242 DOI: 10.1097/rli.0000000000000565] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The aim of this study was to provide an ultrasound-based super-resolution methodology that can be implemented using clinical 2-dimensional ultrasound equipment and standard contrast-enhanced ultrasound modes. In addition, the aim is to achieve this for true-to-life patient imaging conditions, including realistic examination times of a few minutes and adequate image penetration depths that can be used to scan entire organs without sacrificing current super-resolution ultrasound imaging performance. METHODS Standard contrast-enhanced ultrasound was used along with bolus or infusion injections of SonoVue (Bracco, Geneva, Switzerland) microbubble (MB) suspensions. An image analysis methodology, translated from light microscopy algorithms, was developed for use with ultrasound contrast imaging video data. New features that are tailored for ultrasound contrast image data were developed for MB detection and segmentation, so that the algorithm can deal with single and overlapping MBs. The method was tested initially on synthetic data, then with a simple microvessel phantom, and then with in vivo ultrasound contrast video loops from sheep ovaries. Tracks detailing the vascular structure and corresponding velocity map of the sheep ovary were reconstructed. Images acquired from light microscopy, optical projection tomography, and optical coherence tomography were compared with the vasculature network that was revealed in the ultrasound contrast data. The final method was applied to clinical prostate data as a proof of principle. RESULTS Features of the ovary identified in optical modalities mentioned previously were also identified in the ultrasound super-resolution density maps. Follicular areas, follicle wall, vessel diameter, and tissue dimensions were very similar. An approximately 8.5-fold resolution gain was demonstrated in vessel width, as vessels of width down to 60 μm were detected and verified (λ = 514 μm). Best agreement was found between ultrasound measurements and optical coherence tomography with 10% difference in the measured vessel widths, whereas ex vivo microscopy measurements were significantly lower by 43% on average. The results were mostly achieved using video loops of under 2-minute duration that included respiratory motion. A feasibility study on a human prostate showed good agreement between density and velocity ultrasound maps with the histological evaluation of the location of a tumor. CONCLUSIONS The feasibility of a 2-dimensional contrast-enhanced ultrasound-based super-resolution method was demonstrated using in vitro, synthetic and in vivo animal data. The method reduces the examination times to a few minutes using state-of-the-art ultrasound equipment and can provide super-resolution maps for an entire prostate with similar resolution to that achieved in other studies.
Collapse
Affiliation(s)
- Evangelos Kanoulas
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| | - Mairead Butler
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| | - Caitlin Rowley
- Department of Physics, Heriot-Watt University, Riccarton
| | - Vasiliki Voulgaridou
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| | | | - William Colin Duncan
- Center for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Alan McNeilly
- Center for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Massimo Mischi
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; and
| | - Rhodri Simon Wilson
- **Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Weiping Lu
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| | - Vassilis Sboros
- From the Institute of Biochemistry, Biological Physics, and Bio Engineering, and
| |
Collapse
|
25
|
Arteaga-Marrero N, Mainou-Gomez JF, Brekke Rygh C, Lutay N, Roehrich D, Reed RK, Olsen DR. Radiation treatment monitoring with DCE-US in CWR22 prostate tumor xenografts. Acta Radiol 2019; 60:788-797. [PMID: 30231620 DOI: 10.1177/0284185118798167] [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] [Indexed: 01/10/2023]
Abstract
BACKGROUND Longitudinal monitoring of potential radiotherapy treatment effects can be determined by dynamic contrast-enhanced ultrasound (DCE-US). PURPOSE To assess functional parameters by means of DCE-US in a murine subcutaneous model of human prostate cancer, and their relationship to dose deposition and time-frame after treatment. A special focus has been placed to evaluate the vascular heterogeneity of the tumor and on the most suitable data analysis approach that reflects this heterogeneity. MATERIAL AND METHODS In vivo DCE-US was acquired 24 h and 48 h after radiation treatment with a single dose of 7.5 Gy and 10 Gy, respectively. Tumor vasculature was characterized pixelwise using the Brix pharmacokinetic analysis of the time-intensity curves. RESULTS Longitudinal changes were detected ( P < 0.001) at 24 h and 48 h after treatment. At 48 h, the eliminating rate constant of the contrast agent from the plasma, kel, was correlated ( P ≤ 0.05) positively with microvessel density (MVD; rτ = 0.7) and negatively with necrosis (rτ = -0.6) for the treated group. Furthermore, Akep, a parameter related to transcapillary transport properties, was also correlated to MVD (rτ = 0.6, P ≤ 0.05). CONCLUSION DCE-US has been shown to detect vascular changes at a very early stage after radiotherapy, which is a great advantage since DCE-US is non-invasive, available at most hospitals, and is low in cost compared to other techniques used in clinical practice.
Collapse
Affiliation(s)
- Natalia Arteaga-Marrero
- Department of Physics and Technology, University of Bergen, Bergen, Norway
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | | | - Cecilie Brekke Rygh
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Health Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Nataliya Lutay
- Imagene-iT AB, Medicon Village Scheelevägen 2, Lund, Sweden
| | - Dieter Roehrich
- Department of Physics and Technology, University of Bergen, Bergen, Norway
| | - Rolf K Reed
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Center for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
| | - Dag R Olsen
- Department of Physics and Technology, University of Bergen, Bergen, Norway
| |
Collapse
|
26
|
Wang D, Xu S, Zhang K, Zhang X, Yang X, Xiao M, Su Q, Wan M. A fast scheme for renal microvascular perfusion functional imaging: Assessed by an imaging quality evaluation model. Med Phys 2018; 46:738-745. [PMID: 30585642 DOI: 10.1002/mp.13358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/14/2018] [Accepted: 12/14/2018] [Indexed: 11/09/2022] Open
Abstract
PURPOSE This study aimed to develop a fast scheme of multiparametric perfusion functional imaging (PFI) based on dynamic contrast-enhanced ultrasound (DCEUS) for assessing renal microvascular hemodynamics. METHOD The flow process in the DCEUS-based PFI was modified step-by-step to improve its operational efficiency, which was validated through in vivo renal perfusion experiments. A multiparametric model with a comprehensive coefficient of imaging quality (CIQ) was then built on four terms of the average information entropy, contrast, gray, and noise coefficient of PFIs to evaluate the sacrifice of imaging quality during modifications of DCEUS-based PFI. RESULTS The multiparametric model successfully evaluated modifications of DCEUS-based PFI from multiple perspectives (R2 = 0.73, P < 0.01). Compared with the raw scheme in the renal sagittal and coronal planes, the fast PFI scheme significantly improved its operational efficiency by 62.82 ± 1.07% (P < 0.01) and the nine PFIs simultaneously maintained a similar CIQ of 0.26 ± 0.06. CONCLUSIONS The inhomogeneous hemodynamic distributions with a ring-like feature in the renal microvasculature were accurately and efficiently characterized by the fast PFI scheme. The fast PFI scheme can be applied for early diagnosis, follow-up evaluation and monitoring treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Diya Wang
- Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China.,Department of Radiology, Radio-Oncology and Nuclear Medicine, Institute of Biomedical Engineering, University of Montreal, Montreal, QC, H2X 0A9, Canada
| | - Shanshan Xu
- Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| | - Kejia Zhang
- Department of Plastic and Cosmetic Surgery, The Eastern Division of The First Hospital of Jilin University, Changchun, 130031, China
| | - Xinyu Zhang
- Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| | - Xuan Yang
- Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| | - Mengnan Xiao
- Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| | - Qiang Su
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, 1000050, China
| | - Mingxi Wan
- Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| |
Collapse
|
27
|
Wang D, Su Z, Su Q, Zhang X, Qu Z, Wang N, Zong Y, Yang Y, Wan M. Evaluation of accuracy of automatic out-of-plane respiratory gating for DCEUS-based quantification using principal component analysis. Comput Med Imaging Graph 2018; 70:155-164. [DOI: 10.1016/j.compmedimag.2018.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 09/01/2018] [Accepted: 10/18/2018] [Indexed: 01/24/2023]
|
28
|
Mori N, Mugikura S, Miyashita M, Kudo Y, Suzuki M, Li L, Mori Y, Takahashi S, Takase K. Perfusion contrast-enhanced ultrasound to predict early lymph-node metastasis in breast cancer. Jpn J Radiol 2018; 37:145-153. [PMID: 30460444 DOI: 10.1007/s11604-018-0792-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE To evaluate whether quantitative analysis of perfusion contrast-enhanced ultrasound (CE-US) could predict early lymph-node (LN) metastasis in clinically node-negative breast cancer. MATERIALS AND METHODS In this prospective study, 64 breast cancer patients were selected for perfusion CE-US imaging. Regions of interest were placed where the strongest and weakest signal increases were found to obtain peak intensities (PIs; PImax and PImin, respectively) for time-intensity curve analyzes. The PI difference and PI ratio were calculated as follows: PI difference = PImax-PImin; PI ratio = PImax/PImin. RESULTS Forty-seven cases were histologically diagnosed as negative for LN metastasis and 17 were positive. There was a significant difference in PImin and the PI ratio between the LN-negative and -positive metastasis groups (p = 0.0053 and 0.0082, respectively). Receiver-operating curve analysis revealed that the area under the curve of PImin and the PI ratio were 0.73 and 0.72, respectively. The most effective threshold for the PI ratio was 1.52, and the sensitivity, specificity, positive predictive value, and negative predictive value were 59% (10/17), 87% (41/47), 63% (10/16), and 85% (41/48), respectively. CONCLUSIONS Parameters from the quantitative analysis of perfusion CE-US imaging showed significant differences between the LN-negative and -positive metastasis groups in clinically node-negative breast cancer.
Collapse
Affiliation(s)
- Naoko Mori
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Seiryo 1-1, Sendai, 980-8574, Japan.
| | - Shunji Mugikura
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Seiryo 1-1, Sendai, 980-8574, Japan
| | - Minoru Miyashita
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, Seiryo 1-1, Sendai, 980-8574, Japan
| | - Yumiko Kudo
- Department of Physiological Laboratory Center, Tohoku University Hospital, Seiryo 1-1, Sendai, 980-8574, Japan
| | - Mikiko Suzuki
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Seiryo 1-1, Sendai, 980-8574, Japan
| | - Li Li
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Seiryo 1-1, Sendai, 980-8574, Japan
| | - Yu Mori
- Department of Orthopedic Surgery, Tohoku University Graduate School of Medicine, Seiryo 1-1, Sendai, 980-8574, Japan
| | - Shoki Takahashi
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Seiryo 1-1, Sendai, 980-8574, Japan
| | - Kei Takase
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Seiryo 1-1, Sendai, 980-8574, Japan
| |
Collapse
|
29
|
Wischhusen J, Wilson KE, Delcros JG, Molina-Peña R, Gibert B, Jiang S, Ngo J, Goldschneider D, Mehlen P, Willmann JK, Padilla F. Ultrasound molecular imaging as a non-invasive companion diagnostic for netrin-1 interference therapy in breast cancer. Theranostics 2018; 8:5126-5142. [PMID: 30429890 PMCID: PMC6217066 DOI: 10.7150/thno.27221] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023] Open
Abstract
In ultrasound molecular imaging (USMI), ligand-functionalized microbubbles (MBs) are used to visualize vascular endothelial targets. Netrin-1 is upregulated in 60% of metastatic breast cancers and promotes tumor progression. A novel netrin-1 interference therapy requires the assessment of netrin-1 expression prior to treatment. In this study, we studied netrin-1 as a target for USMI and its potential as a companion diagnostic in breast cancer models. Methods: To verify netrin-1 expression and localization, an in vivo immuno-localization approach was applied, in which anti-netrin-1 antibody was injected into living mice 24 h before tumor collection, and revealed with secondary fluorescent antibody for immunofluorescence analysis. Netrin-1 interactions with the cell surface were studied by flow cytometry. Netrin-1-targeted MBs were prepared using MicroMarker Target-Ready (VisualSonics), and validated in in vitro binding assays in static conditions or in a flow chamber using purified netrin-1 protein or netrin-1-expressing cancer cells. In vivo USMI of netrin-1 was validated in nude mice bearing human netrin-1-positive SKBR7 tumors or weakly netrin-1-expressing MDA-MB-231 tumors using the Vevo 2100 small animal imaging device (VisualSonics). USMI feasibility was further tested in transgenic murine FVB/N Tg(MMTV/PyMT634Mul) (MMTV-PyMT) mammary tumors. Results: Netrin-1 co-localized with endothelial CD31 in netrin-1-positive breast tumors. Netrin-1 binding to the surface of endothelial HUVEC and cancer cells was partially mediated by heparan sulfate proteoglycans. MBs targeted with humanized monoclonal anti-netrin-1 antibody bound to netrin-1-expressing cancer cells in static and dynamic conditions. USMI signal was significantly increased with anti-netrin-1 MBs in human SKBR7 breast tumors and transgenic murine MMTV-PyMT mammary tumors compared to signals recorded with either isotype control MBs or after blocking of netrin-1 with humanized monoclonal anti-netrin-1 antibody. In weakly netrin-1-expressing human tumors and normal mammary glands, no difference in imaging signal was observed with anti-netrin-1- and isotype control MBs. Ex vivo analysis confirmed netrin-1 expression in MMTV-PyMT tumors. Conclusions: These results show that USMI allowed reliable detection of netrin-1 on the endothelium of netrin-1-positive human and murine tumors. Significant differences in USMI signal for netrin-1 reflected the significant differences in netrin-1 mRNA & protein expression observed between different breast tumor models. The imaging approach was non-invasive and safe, and provided the netrin-1 expression status in near real-time. Thus, USMI of netrin-1 has the potential to become a companion diagnostic for the stratification of patients for netrin-1 interference therapy in future clinical trials.
Collapse
|
30
|
Opacic T, Dencks S, Theek B, Piepenbrock M, Ackermann D, Rix A, Lammers T, Stickeler E, Delorme S, Schmitz G, Kiessling F. Motion model ultrasound localization microscopy for preclinical and clinical multiparametric tumor characterization. Nat Commun 2018. [PMID: 29670096 DOI: 10.1101/203935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
Super-resolution imaging methods promote tissue characterization beyond the spatial resolution limits of the devices and bridge the gap between histopathological analysis and non-invasive imaging. Here, we introduce motion model ultrasound localization microscopy (mULM) as an easily applicable and robust new tool to morphologically and functionally characterize fine vascular networks in tumors at super-resolution. In tumor-bearing mice and for the first time in patients, we demonstrate that within less than 1 min scan time mULM can be realized using conventional preclinical and clinical ultrasound devices. In this context, next to highly detailed images of tumor microvascularization and the reliable quantification of relative blood volume and perfusion, mULM provides multiple new functional and morphological parameters that discriminate tumors with different vascular phenotypes. Furthermore, our initial patient data indicate that mULM can be applied in a clinical ultrasound setting opening avenues for the multiparametric characterization of tumors and the assessment of therapy response.
Collapse
Affiliation(s)
- Tatjana Opacic
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, CMBS, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Stefanie Dencks
- Chair for Medical Engineering, Department of Electrical Engineering and Information Technology, Ruhr University Bochum, Universitätsstr. 150, 44780, Bochum, Germany
| | - Benjamin Theek
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, CMBS, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Marion Piepenbrock
- Chair for Medical Engineering, Department of Electrical Engineering and Information Technology, Ruhr University Bochum, Universitätsstr. 150, 44780, Bochum, Germany
| | - Dimitri Ackermann
- Chair for Medical Engineering, Department of Electrical Engineering and Information Technology, Ruhr University Bochum, Universitätsstr. 150, 44780, Bochum, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, CMBS, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, CMBS, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Elmar Stickeler
- Department of Obstetrics and Gynecology, University Clinic Aachen, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Stefan Delorme
- Department of Radiology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Georg Schmitz
- Chair for Medical Engineering, Department of Electrical Engineering and Information Technology, Ruhr University Bochum, Universitätsstr. 150, 44780, Bochum, Germany.
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Clinic Aachen, RWTH Aachen University, CMBS, Forckenbeckstr. 55, 52074, Aachen, Germany.
| |
Collapse
|
31
|
Motion model ultrasound localization microscopy for preclinical and clinical multiparametric tumor characterization. Nat Commun 2018; 9:1527. [PMID: 29670096 PMCID: PMC5906644 DOI: 10.1038/s41467-018-03973-8] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 03/27/2018] [Indexed: 12/21/2022] Open
Abstract
Super-resolution imaging methods promote tissue characterization beyond the spatial resolution limits of the devices and bridge the gap between histopathological analysis and non-invasive imaging. Here, we introduce motion model ultrasound localization microscopy (mULM) as an easily applicable and robust new tool to morphologically and functionally characterize fine vascular networks in tumors at super-resolution. In tumor-bearing mice and for the first time in patients, we demonstrate that within less than 1 min scan time mULM can be realized using conventional preclinical and clinical ultrasound devices. In this context, next to highly detailed images of tumor microvascularization and the reliable quantification of relative blood volume and perfusion, mULM provides multiple new functional and morphological parameters that discriminate tumors with different vascular phenotypes. Furthermore, our initial patient data indicate that mULM can be applied in a clinical ultrasound setting opening avenues for the multiparametric characterization of tumors and the assessment of therapy response. The vascular structure of tumors impacts diagnosis, prognosis and drug response; however, imaging methods to analyse this important feature have been hindered by spatial resolution limitations. Here the authors present a tool called motion model ultrasound localization microscopy to morphologically and functionally characterize fine vascular networks in tumors at super-resolution.
Collapse
|
32
|
Wang D, Su Z, Zhang Y, Zhao X, Wang S, Wan M. DCEUS-based multiparametric perfusion imaging using pulse-inversion Bubblet decorrelation. Med Phys 2018; 45:2509-2517. [PMID: 29611197 DOI: 10.1002/mp.12897] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/22/2018] [Accepted: 03/23/2018] [Indexed: 11/09/2022] Open
Abstract
PURPOSE This study aimed to clarify the influences of composite dynamic contrast-enhanced ultrasound (DCEUS) on multiparametric perfusion imaging (PPI) and to develop a novel PPI scheme through pulse-inversion Bubblet decorrelation (PIBD) to improve its contrast and detailed discriminability. METHOD In in vivo perfusion experiments on rabbit kidneys, a pair of phase-inverted "Bubblets" was constructed. Phase-inverted raw radiofrequency echoes were reconstructed by using the maximum coefficients obtained from Bubblet decorrelation analysis and summed to form DCEUS loops. Nine perfusion parameters were estimated from these loops and color coded to create the corresponding PIBD-based PPIs. RESULTS In addition to time-related PPIs, the contrast and detailed discriminability quantified by the average contrast and information entropy of intensity- and ratio-related PPIs were proportional to the microbubble detection sensitivity and microvascular discriminability evaluated by CTR in DCEUS techniques. Compared with the second harmonic, the CTR of DCEUS and the average contrast and information entropy of PPI were significantly improved by 9.03 ± 5.39 dB (P < 0.01), 6.39 ± 1.38 dB (P < 0.01), and 0.57 ± 0.15 (P < 0.05) in PIBD technique, respectively. CONCLUSIONS As a multiparametric functional imaging technique, these improvements in the proposed scheme can be beneficial to accurately quantify and depict the hemodynamic perfusion features and details of tumor angiogenesis, and further can also assist clinicians in making a confirmed diagnosis.
Collapse
Affiliation(s)
- Diya Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, China.,Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Quebec, H2X 0A9, Canada
| | - Zhe Su
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, China
| | - Yu Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, China
| | - Xiaoyan Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, China
| | - Supin Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, China
| | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
33
|
Wang D, Xiao M, Zhang Y, Wan M. Abdominal parametric perfusion imaging with respiratory motion-compensation based on contrast-enhanced ultrasound: In-vivo validation. Comput Med Imaging Graph 2018; 65:11-21. [DOI: 10.1016/j.compmedimag.2017.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/03/2017] [Accepted: 06/19/2017] [Indexed: 10/19/2022]
|
34
|
Wang D, Xiao M, Zhang Y, Su Z, Zong Y, Wang S, Wan M. In-vitro evaluation of accuracy of dynamic contrast-enhanced ultrasound (DCEUS)-based parametric perfusion imaging with respiratory motion-compensation. Med Phys 2018; 45:2119-2128. [PMID: 29574795 DOI: 10.1002/mp.12872] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 02/14/2018] [Accepted: 03/01/2018] [Indexed: 01/24/2023] Open
Abstract
PURPOSE The accuracy of multi-parametric perfusion imaging (PPI) based on dynamic contrast-enhanced ultrasound is disturbed by the respiratory motion in some cases, especially during characterizing hemodynamic features of abdominal tumor angiogenesis. This study aimed to effectively remove those disturbances on PPI and evaluate its accuracy. METHOD The respiratory motion-compensation (rMoCo) strategy in PPI was modified by employing non-negative matrix factorization combined with phase-by-phase compensation. According to the known and controllable ground truths in in-vitro perfusion experiments, the accuracy of the modified rMoCo strategy was further evaluated from multiple perspectives in a simulated dual-vessel flow phantom. RESULTS Compared with that of PPIs without rMoCo, the mean correlation coefficient between six PPIs with rMoCo and the corresponding static PPIs was up to 0.98 ± 0.01 and improved by 0.17 ± 0.04 (P < 0.05). The estimated error of vascular diameter decreased from 87.85% (P < 0.05) to 7.25% (P < 0.05) after rMoCo. PPIs with rMoCo were significantly consistent with static PPIs without respiratory motion disturbances. CONCLUSIONS These quantitative results illustrated the disturbances induced by respiratory motion were effectively removed and the accuracy of PPIs was significantly improved. The partial parabolic and bimodal hemodynamic characteristics and the anatomical structures and sizes were accurately quantified and depicted by PPIs with rMoCo. The modified method can benefit physicians in providing accurate diagnoses and in developing appropriate therapeutic strategies for abdominal diseases.
Collapse
Affiliation(s)
- Diya Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China.,Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Quebec, H2X 0A9, Canada
| | - Mengnan Xiao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| | - Yu Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| | - Zhe Su
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| | - Yujin Zong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| | - Supin Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, 710049, China
| |
Collapse
|
35
|
Rix A, Lederle W, Theek B, Lammers T, Moonen C, Schmitz G, Kiessling F. Advanced Ultrasound Technologies for Diagnosis and Therapy. J Nucl Med 2018; 59:740-746. [DOI: 10.2967/jnumed.117.200030] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/20/2018] [Indexed: 12/27/2022] Open
|
36
|
Wang D, Xiao M, Hu H, Zhang Y, Su Z, Xu S, Zong Y, Wan M. DCEUS-based focal parametric perfusion imaging of microvessel with single-pixel resolution and high contrast. ULTRASONICS 2018; 84:392-403. [PMID: 29245119 DOI: 10.1016/j.ultras.2017.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 11/23/2017] [Accepted: 11/29/2017] [Indexed: 06/07/2023]
Abstract
This study aimed to develop a focal microvascular contrast-enhanced ultrasonic parametric perfusion imaging (PPI) scheme to overcome the tradeoff between the resolution, contrast, and accuracy of focal PPI in the tumor. Its resolution was limited by the low signal-to-clutter ratio (SCR) of time-intensity-curves (TICs) induced by multiple limitations, which deteriorated the accuracy and contrast of focal PPI. The scheme was verified by the in-vivo perfusion experiments. Single-pixel TICs were first extracted to ensure PPI with the highest resolution. The SCR of focal TICs in the tumor was improved using respiratory motion compensation combined with detrended fluctuation analysis. The entire and focal PPIs of six perfusion parameters were then accurately created after filtrating the valid TICs and targeted perfusion parameters. Compared with those of the conventional PPIs, the axial and lateral resolutions of focal PPIs were improved by 30.29% (p < .05) and 32.77% (p < .05), respectively; the average contrast and accuracy evaluated by SCR improved by 7.24 ± 4.90 dB (p < .05) and 5.18 ± 1.28 dB (p < .05), respectively. The edge, morphostructure, inhomogeneous hyper-enhanced distribution, and ring-like perfusion features in intratumoral microvessel were accurately distinguished and highlighted by the focal PPIs. The developed focal PPI can assist clinicians in making confirmed diagnoses and in providing appropriate therapeutic strategies for liver tumor.
Collapse
Affiliation(s)
- Diya Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, PR China; Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, QC, Canada
| | - Mengnan Xiao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, PR China
| | - Hong Hu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, PR China
| | - Yu Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, PR China
| | - Zhe Su
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, PR China
| | - Shanshan Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, PR China
| | - Yujin Zong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, PR China
| | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, PR China.
| |
Collapse
|
37
|
Dynamic contrast-enhanced ultrasonography (D-CEUS) for the early prediction of bevacizumab efficacy in patients with metastatic colorectal cancer. Eur Radiol 2018; 28:2969-2978. [PMID: 29417252 DOI: 10.1007/s00330-017-5254-5] [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: 05/31/2017] [Revised: 11/30/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To investigate early changes in tumour perfusion parameters by dynamic contrast-enhanced ultrasonography (D-CEUS) and to identify any correlation with survival and tumour response in patients with metastatic colorectal cancer (CRC) treated with bevacizumab (B). METHODS Thirty-seven patients randomized to either chemotherapy (C) plus B or C alone were considered for this study. D-CEUS was performed at baseline and after the first treatment cycle (day 15). Four D-CEUS perfusion parameters were considered: derived peak intensity (DPI), area under the curve (AUC), slope of wash-in (A) and time to peak intensity (TPI). RESULTS In patients treated with C plus B, a ≥22.5 % reduction in DPI, ≥20 % increase in TPI and ≥10 % reduction in AUC were correlated with higher progression-free survival in the C+B arm (p = 0.048, 0.024 and 0.010, respectively) but not in the C arm. None of the evaluated parameter modifications had a correlation with tumour response or overall survival. CONCLUSIONS D-CEUS could be useful for detecting and quantifying dynamic changes in tumour vascularity as early as 15 days after the start of B-based therapy. Although these changes may be predictive of progression-free survival, no correlation with response or overall survival was found. KEY POINTS • D-CEUS showed early changes in liver metastasis perfusion in colorectal cancer. • A decrease in tumour perfusion was associated with longer progression-free survival. • The decrease in perfusion was not correlated with higher overall survival.
Collapse
|
38
|
Haimerl M, Brünn K, Poelsterl S, Beyer L, Wiesinger I, Stroszczynski C, Jung EM, Wiggermann P. Quantitative evaluation of real-time maximum liver capacity (LiMAx) and time intensity curve (TIC) analysis in CEUS-based microperfusion. Clin Hemorheol Microcirc 2017; 67:373-382. [DOI: 10.3233/ch-179217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- M. Haimerl
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - K Brünn
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - S. Poelsterl
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - L.P. Beyer
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - I. Wiesinger
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - C. Stroszczynski
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - E.-M. Jung
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - P. Wiggermann
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
39
|
Zhu L, Guo Y, Wang L, Fan X, Xiong X, Fang K, Xu D. Construction of ultrasonic nanobubbles carrying CAIX polypeptides to target carcinoma cells derived from various organs. J Nanobiotechnology 2017; 15:63. [PMID: 28962657 PMCID: PMC5622542 DOI: 10.1186/s12951-017-0307-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/23/2017] [Indexed: 01/01/2023] Open
Abstract
Background Ultrasound molecular imaging is a novel diagnostic approach for tumors, whose key link is the construction of targeted ultrasound contrast agents. However, available targeted ultrasound contrast agents for molecular imaging of tumors are only achieving imaging in blood pool or one type tumor. No targeted ultrasound contrast agents have realized targeted ultrasound molecular imaging of tumor parenchymal cells in a variety of solid tumors so far. Carbonic anhydrase IX (CAIX) is highly expressed on cell membranes of various malignant solid tumors, so it’s a good target for ultrasound molecular imaging. Here, targeted nanobubbles carrying CAIX polypeptides for targeted binding to a variety of malignant tumors were constructed, and targeted binding ability and ultrasound imaging effect in different types of tumors were evaluated. Results The mean diameter of lipid targeted nanobubbles was (503.7 ± 78.47) nm, and the polypeptides evenly distributed on the surfaces of targeted nanobubbles, which possessed the advantages of homogenous particle size, high stability, and good safety. Targeted nanobubbles could gather around CAIX-positive cells (786-O and Hela cells), while they cannot gather around CAIX-negative cells (BxPC-3 cells) in vitro, and the affinity of targeted nanobubbles to CAIX-positive cells were significantly higher than that to CAIX-negative cells (P < 0.05). Peak intensity and duration time of targeted nanobubbles and blank nanobubbles were different in CAIX-positive transplanted tumor tissues in vivo (P < 0.05). Moreover, targeted nanobubbles in CAIX-positive transplanted tumor tissues produced higher peak intensity and longer duration time than those in CAIX-negative transplanted tumor tissues (P < 0.05). Finally, immunofluorescence not only confirmed targeted nanobubbles could pass through blood vessels to enter in tumor tissue spaces, but also clarified imaging differences of targeted nanobubbles in different types of transplanted tumor tissues. Conclusions Targeted nanobubbles carrying CAIX polypeptides can specifically enhance ultrasound imaging in CAIX-positive transplanted tumor tissues and could potentially be used in early diagnosis of a variety of solid tumors derived from various organs. Electronic supplementary material The online version of this article (doi:10.1186/s12951-017-0307-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lianhua Zhu
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
| | - Luofu Wang
- Department of Urology, Daping Hospital, Third Military Medical University, 10 Changjiang Zhi Road, Yuzhong District, Chongqing, 400038, China
| | - Xiaozhou Fan
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xingyu Xiong
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Kejing Fang
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Dan Xu
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| |
Collapse
|
40
|
Automated Generation of Reliable Blood Velocity Parameter Maps from Contrast-Enhanced Ultrasound Data. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:2098324. [PMID: 29097912 PMCID: PMC5612675 DOI: 10.1155/2017/2098324] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/18/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023]
Abstract
Objectives The purpose of this study was the automated generation and validation of parametric blood flow velocity maps, based on contrast-enhanced ultrasound (CEUS) scans. Materials and Methods Ethical approval for animal experiments was obtained. CEUS destruction-replenishment sequences were recorded in phantoms and three different tumor xenograft mouse models. Systematic pixel binning and intensity averaging was performed to generate parameter maps of blood flow velocities with different pixel resolution. The 95% confidence interval of the mean velocity, calculated on the basis of the whole tumor segmentation, served as ground truth for the different parameter maps. Results In flow phantoms the measured mean velocity values were only weakly influenced by the pixel resolution and correlated with real velocities (r2 ≥ 0.94, p < 0.01). In tumor xenografts, however, calculated mean velocities varied significantly (p < 0.0001), depending on the parameter maps' resolution. Pixel binning was required for all in vivo measurements to obtain reliable parameter maps and its degree depended on the tumor model. Conclusion Systematic pixel binning allows the automated identification of optimal pixel resolutions for parametric maps, supporting textural analysis of CEUS data. This approach is independent from the ultrasound setup and can be implemented in the software of other (clinical) ultrasound devices.
Collapse
|
41
|
Mueller S, Wendl CM, Ettl T, Klingelhöffer C, Geis S, Prantl L, Reichert TE, Jung EM. Contrast-enhanced ultrasonography as a new method for assessing autonomization of pedicled and microvascular free flaps in head and neck reconstructive surgery. Clin Hemorheol Microcirc 2017; 65:317-325. [DOI: 10.3233/ch-16194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Steffen Mueller
- Department of Cranio-Maxillofacial Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Christina M. Wendl
- Institute of Radiology, University Medical Center Regensburg, Regensburg, Germany
| | - Tobias Ettl
- Department of Cranio-Maxillofacial Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Christoph Klingelhöffer
- Department of Cranio-Maxillofacial Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Sebastian Geis
- Department of Plastic-, Hand-, and Reconstructive Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic-, Hand-, and Reconstructive Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Torsten E. Reichert
- Department of Cranio-Maxillofacial Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Ernst Michael Jung
- Institute of Radiology, University Medical Center Regensburg, Regensburg, Germany
| |
Collapse
|
42
|
Mori N, Mugikura S, Takahashi S, Ito K, Takasawa C, Li L, Miyashita M, Kasajima A, Mori Y, Ishida T, Kodama T, Takase K. Quantitative Analysis of Contrast-Enhanced Ultrasound Imaging in Invasive Breast Cancer: A Novel Technique to Obtain Histopathologic Information of Microvessel Density. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:607-614. [PMID: 28041744 DOI: 10.1016/j.ultrasmedbio.2016.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/27/2016] [Accepted: 11/14/2016] [Indexed: 06/06/2023]
Abstract
We examined whether enhancement area ratios obtained by the new bubble detection method correlate with histologic microvessel density in invasive breast cancer. Forty consecutive patients with invasive breast cancer lesions underwent contrast-enhanced ultrasound. The ratio of enhanced area to manually segmented tumor area (enhancement area ratio) was obtained with the new method at peak and delayed phases (50-54, 55-59, 60-64 and 65-69 s). We also analyzed time-intensity curves to obtain peak intensity and area under curve. Enhancement area ratios in both peak and delayed phases (50-54, 55-59, 60-64 and 65-69 s) were significantly correlated with microvessel density (r = 0.57, 0.62, 0.68, 0.61 and 0.58; p = 0.0001, <0.0001, <.0001, <.0001 and 0.0001, respectively). In time-intensity curve analysis, peak intensity was significantly correlated (r = 0.43, p = 0.0073), whereas area under the curve was not (r = 0.29, p = 0.0769). Enhancement area ratios obtained by the new method were correlated with microvessel density in invasive breast cancer.
Collapse
Affiliation(s)
- Naoko Mori
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Shunji Mugikura
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shoki Takahashi
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koichi Ito
- Department of Information Science, Tohoku University Graduate School, Sendai, Japan
| | - Chiaki Takasawa
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Li Li
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Minoru Miyashita
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsuko Kasajima
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Mori
- Department of Orthopedic Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takanori Ishida
- Department of Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Kodama
- Department of Biomedical Engineering, Tohoku University Graduate School, Sendai, Japan
| | - Kei Takase
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
43
|
Haimerl M, Poelsterl S, Beyer L, Wiesinger I, Nießen C, Stroszczynski C, Wiggermann P, Jung EM. Chronic liver disease: Quantitative MRI vs CEUS-based microperfusion. Clin Hemorheol Microcirc 2017; 64:435-446. [DOI: 10.3233/ch-168112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Sun D, Wei C, Li Y, Lu Q, Zhang W, Hu B. Contrast-Enhanced Ultrasonography with Quantitative Analysis allows Differentiation of Renal Tumor Histotypes. Sci Rep 2016; 6:35081. [PMID: 27725761 PMCID: PMC5057121 DOI: 10.1038/srep35081] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/20/2016] [Indexed: 02/08/2023] Open
Abstract
Totally 85 patients with 93 renal lesions who underwent contrast-enhanced ultrasound (CEUS) were retrospectively studied with quantitative analysis to evaluate its value in the differential diagnosis of renal tumor histotypes. CEUS characteristics were analysed including the enhancement patterns, peak intensity, homogeneity of enhancement, and pseudocapsule. Quantitative parameters of peak intensity (P) and time to peak (TP) were measured with QontraXt software, and the index “relative enhancement percentage” ΔP% and “difference in TP between tumor and cortex” ΔTP were used to quantify the CEUS features of renal tumors. There are significant difference in CEUS features between the 46 clear cell renal cell carcinoma (CCRCC) and other types of renal tumors, including 17 low malignant lesions, 11 urothelial carcinoma of the renal pelvis, and 19 renal angiomyolipoma. The differences lie in the peak intensity, the homogeneity, the time of wash-in, peak, clearance and presence of pseudocapsule. The ΔTP and ΔP% of the CCRCC is significantly different from other tumors. With “fast to peak + high peak intensity” as the main criterion, assisted with “heterogeneous enhancement” and “fast wash-in” as the secondary criteria, the diagnostic accuracy of CCRCC is 91.4%, demonstrating quantitative CEUS imaging is highly valuable in differentiating CCRCC from other tumors.
Collapse
Affiliation(s)
- Di Sun
- Department of Ultrasound in Medicine, Shanghai Jiao tong University Affiliated Sixth People's Hospital, China.,Shanghai Institute of Ultrasound in Medicine, China
| | - Cong Wei
- Department of Ultrasound in Medicine, Shanghai Jiao tong University Affiliated Sixth People's Hospital, China.,Shanghai Institute of Ultrasound in Medicine, China
| | - Yi Li
- Department of Ultrasound in Medicine, Shanghai Jiao tong University Affiliated Sixth People's Hospital, China.,Shanghai Institute of Ultrasound in Medicine, China
| | - Qijie Lu
- Department of Ultrasound in Medicine, Shanghai Jiao tong University Affiliated Sixth People's Hospital, China.,Shanghai Institute of Ultrasound in Medicine, China
| | - Wei Zhang
- Department of Ultrasound in Medicine, Shanghai Jiao tong University Affiliated Sixth People's Hospital, China.,Shanghai Institute of Ultrasound in Medicine, China
| | - Bing Hu
- Department of Ultrasound in Medicine, Shanghai Jiao tong University Affiliated Sixth People's Hospital, China.,Shanghai Institute of Ultrasound in Medicine, China
| |
Collapse
|
45
|
Kan X, Zhang Y, Zheng C, Li L, Chen J, Wu Y, Guo T, Xiong B. Stress Test of Contrast-Enhanced US with Phenylephrine in a Rabbit VX2 Liver Tumor Model: Differentiating Benign Periablational Enhancement from Residual Tumor after Radiofrequency Ablation. J Vasc Interv Radiol 2016; 27:1077-1085.e2. [PMID: 27117950 DOI: 10.1016/j.jvir.2016.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To differentiate benign periablational enhancement (BPE) from residual tumor after radiofrequency (RF) ablation by using a stress contrast-enhanced ultrasonography (US) test with phenylephrine in a rabbit VX2 liver tumor model. MATERIALS AND METHODS VX2 tumors were implanted in the livers of 40 rabbits for two experiments. In experiment one, liver tumors from 32 animals were completely ablated. On days 2, 7, 14, and 21 after RF ablation, eight animals were randomly chosen for contrast-enhanced US before and after phenylephrine administration, and the microvessel density (MVD) of BPE at these four time points was assessed. In experiment two, liver tumors from eight animals were partly ablated, and each animal underwent contrast-enhanced US before and after phenylephrine administration on day 7 after RF ablation. Perfusion parameters were observed, including maximum intensity (IMAX), rise time (ie, time between 10% and 90% of IMAX), time to peak, mean transit time, and area under the curve (AUC), along with the profile of time-intensity curves (TICs) in BPE and residual tumor in response to phenylephrine. RESULTS Among the four time points after ablation, the IMAX and AUC before phenylephrine administration and the MVD of BPE were greatest on day 7 (P < .05). The profile of TICs and the corresponding perfusion parameters in residual tumor did not change significantly in response to phenylephrine. However, those from BPE changed significantly (P < .05). CONCLUSIONS Contrast-enhanced US with phenylephrine stress may be helpful in differentiating BPE from residual tumor after RF ablation in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Rd., Wuhan, Hubei 430022, China
| | - Yanrong Zhang
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Rd., Wuhan, Hubei 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Rd., Wuhan, Hubei 430022, China
| | - Lin Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Rd., Wuhan, Hubei 430022, China
| | - Juan Chen
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Rd., Wuhan, Hubei 430022, China
| | - Yihua Wu
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Rd., Wuhan, Hubei 430022, China
| | - Tao Guo
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Rd., Wuhan, Hubei 430022, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Rd., Wuhan, Hubei 430022, China.
| |
Collapse
|
46
|
Strachinaru M, Damry N, Duttmann R, Wauthy P, Catez E, Lutea M, Costescu I, Morissens M. Ultrasound Contrast Quantification for the Diagnosis of Intracardiac Masses. JACC Cardiovasc Imaging 2016; 9:747-50. [PMID: 27085435 DOI: 10.1016/j.jcmg.2015.06.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/08/2015] [Accepted: 06/11/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Mihai Strachinaru
- Department of Cardiology, Brugmann University Hospital, Brussels, Belgium.
| | - Nasroolla Damry
- Department of Medical Imaging, Brugmann University Hospital, Brussels, Belgium
| | - Ruth Duttmann
- Department of Pathology, Brugmann University Hospital, Brussels, Belgium
| | - Pierre Wauthy
- Department of Cardiac Surgery, Brugmann University Hospital, Brussels, Belgium
| | - Emmanuel Catez
- Department of Cardiology, Brugmann University Hospital, Brussels, Belgium
| | - Mirela Lutea
- Department of Cardiology, Brugmann University Hospital, Brussels, Belgium
| | - Isabela Costescu
- Department of Internal Medicine, St. Pierre University Hospital, Brussels, Belgium
| | - Marielle Morissens
- Department of Cardiology, Brugmann University Hospital, Brussels, Belgium
| |
Collapse
|
47
|
Klibanov AL, Hossack JA. Ultrasound in Radiology: From Anatomic, Functional, Molecular Imaging to Drug Delivery and Image-Guided Therapy. Invest Radiol 2015; 50:657-70. [PMID: 26200224 PMCID: PMC4580624 DOI: 10.1097/rli.0000000000000188] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During the past decade, ultrasound has expanded medical imaging well beyond the "traditional" radiology setting: a combination of portability, low cost, and ease of use makes ultrasound imaging an indispensable tool for radiologists as well as for other medical professionals who need to obtain imaging diagnosis or guide a therapeutic intervention quickly and efficiently. Ultrasound combines excellent ability for deep penetration into soft tissues with very good spatial resolution, with only a few exceptions (ie, those involving overlying bone or gas). Real-time imaging (up to hundreds and thousands of frames per second) enables guidance of therapeutic procedures and biopsies; characterization of the mechanical properties of the tissues greatly aids with the accuracy of the procedures. The ability of ultrasound to deposit energy locally brings about the potential for localized intervention encompassing the following: tissue ablation, enhancing penetration through the natural barriers to drug delivery in the body and triggering drug release from carrier microparticles and nanoparticles. The use of microbubble contrast agents brings the ability to monitor and quantify tissue perfusion, and microbubble targeting with ligand-decorated microbubbles brings the ability to obtain molecular biomarker information, that is, ultrasound molecular imaging. Overall, ultrasound has become the most widely used imaging modality in modern medicine; it will continue to grow and expand.
Collapse
Affiliation(s)
- Alexander L Klibanov
- From the *Cardiovascular Division, Robert M. Berne Cardiovascular Research Center, School of Medicine, and †Department of Biomedical Engineering, University of Virginia, Charlottesville VA
| | | |
Collapse
|
48
|
Wang H, Hristov D, Qin J, Tian L, Willmann JK. Three-dimensional Dynamic Contrast-enhanced US Imaging for Early Antiangiogenic Treatment Assessment in a Mouse Colon Cancer Model. Radiology 2015. [PMID: 26020439 DOI: 10.1148/radiol.2015142824]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE To evaluate feasibility and reproducibility of three-dimensional (3D) dynamic contrast material-enhanced (DCE) ultrasonographic (US) imaging by using a clinical matrix array transducer to assess early antiangiogenic treatment effects in human colon cancer xenografts in mice. MATERIALS AND METHODS Animal studies were approved by the Institutional Administrative Panel on Laboratory Animal Care at Stanford University. Three-dimensional DCE US imaging with two techniques (bolus and destruction-replenishment) was performed in human colon cancer xenografts (n = 38) by using a clinical US system and transducer. Twenty-one mice were imaged twice to assess reproducibility. Seventeen mice were scanned before and 24 hours after either antiangiogenic (n = 9) or saline-only (n = 8) treatment. Data sets of 3D DCE US examinations were retrospectively segmented into consecutive 1-mm imaging planes to simulate two-dimensional (2D) DCE US imaging. Six perfusion parameters (peak enhancement [PE], area under the time-intensity curve [AUC], time to peak [TTP], relative blood volume [rBV], relative blood flow [rBF], and blood flow velocity) were measured on both 3D and 2D data sets. Percent area of blood vessels was quantified ex vivo with immunofluorescence. Statistical analyses were performed with the Wilcoxon rank test by calculating intraclass correlation coefficients and by using Pearson correlation analysis. RESULTS Reproducibility of both 3D DCE US imaging techniques was good to excellent (intraclass correlation coefficient, 0.73-0.86). PE, AUC, rBV, and rBF significantly decreased (P ≤ .04) in antiangiogenic versus saline-treated tumors. rBV (r = 0.74; P = .06) and rBF (r = 0.85; P = .02) correlated with ex vivo percent area of blood vessels, although the statistical significance of rBV was not reached, likely because of small sample size. Overall, 2D DCE-US overestimated and underestimated treatment effects from up to 125-fold to170-fold compared with 3D DCE US imaging. If the central tumor plane was assessed, treatment response was underestimated up to threefold or overestimated up to 57-fold on 2D versus 3D DCE US images. CONCLUSION Three-dimensional DCE US imaging with a clinical matrix array transducer is feasible and reproducible to assess tumor perfusion in human colon cancer xenografts in mice and allows for assessment of early treatment response after antiangiogenic therapy.
Collapse
Affiliation(s)
- Huaijun Wang
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Dimitre Hristov
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Jiale Qin
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Lu Tian
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Jürgen K Willmann
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| |
Collapse
|
49
|
Wang H, Hristov D, Qin J, Tian L, Willmann JK. Three-dimensional Dynamic Contrast-enhanced US Imaging for Early Antiangiogenic Treatment Assessment in a Mouse Colon Cancer Model. Radiology 2015; 277:424-34. [PMID: 26020439 DOI: 10.1148/radiol.2015142824] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE To evaluate feasibility and reproducibility of three-dimensional (3D) dynamic contrast material-enhanced (DCE) ultrasonographic (US) imaging by using a clinical matrix array transducer to assess early antiangiogenic treatment effects in human colon cancer xenografts in mice. MATERIALS AND METHODS Animal studies were approved by the Institutional Administrative Panel on Laboratory Animal Care at Stanford University. Three-dimensional DCE US imaging with two techniques (bolus and destruction-replenishment) was performed in human colon cancer xenografts (n = 38) by using a clinical US system and transducer. Twenty-one mice were imaged twice to assess reproducibility. Seventeen mice were scanned before and 24 hours after either antiangiogenic (n = 9) or saline-only (n = 8) treatment. Data sets of 3D DCE US examinations were retrospectively segmented into consecutive 1-mm imaging planes to simulate two-dimensional (2D) DCE US imaging. Six perfusion parameters (peak enhancement [PE], area under the time-intensity curve [AUC], time to peak [TTP], relative blood volume [rBV], relative blood flow [rBF], and blood flow velocity) were measured on both 3D and 2D data sets. Percent area of blood vessels was quantified ex vivo with immunofluorescence. Statistical analyses were performed with the Wilcoxon rank test by calculating intraclass correlation coefficients and by using Pearson correlation analysis. RESULTS Reproducibility of both 3D DCE US imaging techniques was good to excellent (intraclass correlation coefficient, 0.73-0.86). PE, AUC, rBV, and rBF significantly decreased (P ≤ .04) in antiangiogenic versus saline-treated tumors. rBV (r = 0.74; P = .06) and rBF (r = 0.85; P = .02) correlated with ex vivo percent area of blood vessels, although the statistical significance of rBV was not reached, likely because of small sample size. Overall, 2D DCE-US overestimated and underestimated treatment effects from up to 125-fold to170-fold compared with 3D DCE US imaging. If the central tumor plane was assessed, treatment response was underestimated up to threefold or overestimated up to 57-fold on 2D versus 3D DCE US images. CONCLUSION Three-dimensional DCE US imaging with a clinical matrix array transducer is feasible and reproducible to assess tumor perfusion in human colon cancer xenografts in mice and allows for assessment of early treatment response after antiangiogenic therapy.
Collapse
Affiliation(s)
- Huaijun Wang
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Dimitre Hristov
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Jiale Qin
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Lu Tian
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Jürgen K Willmann
- From the Department of Radiology, Molecular Imaging Program at Stanford (H.W., J.Q., J.K.W.), Department of Radiation Oncology (D.H.), and Department of Health, Research & Policy (L.T.), Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| |
Collapse
|
50
|
Quantitative Imaging. Invest Radiol 2015; 50:187. [DOI: 10.1097/rli.0000000000000139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|