1
|
Cerrito L, Ainora ME, Cuccia G, Galasso L, Mignini I, Esposto G, Garcovich M, Riccardi L, Gasbarrini A, Zocco MA. Dynamic Contrast-Enhanced Ultrasound in the Prediction of Advanced Hepatocellular Carcinoma Response to Systemic and Locoregional Therapies. Cancers (Basel) 2024; 16:551. [PMID: 38339302 PMCID: PMC10854581 DOI: 10.3390/cancers16030551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer and the sixth most common malignant tumor in the world, with an incidence of 2-8% per year in patients with hepatic cirrhosis or chronic hepatitis. Despite surveillance schedules, it is sometimes diagnosed at an advanced stage, requiring complex therapeutic efforts with both locoregional and systemic treatments. Traditional radiological tools (computed tomography and magnetic resonance) are used for the post-treatment follow-up of HCC. The first follow-up imaging is performed at 4 weeks after resection or locoregional treatments, or after 3 months from the beginning of systemic therapies, and subsequently every 3 months for the first 2 years. For this reason, these radiological methods do not grant the possibility of an early distinction between good and poor therapeutic response. Contrast-enhanced ultrasound (CEUS) and dynamic contrast-enhanced ultrasound (DCE-US) have gained the interest of several researchers for their potential role in the early assessment of response to locoregional treatments (chemoembolization) or antiangiogenic therapies in patients with advanced HCC. In fact, DCE-US, through a quantitative analysis performed by specific software, allows the construction of time-intensity curves, providing an evaluation of the parameters related to neoplastic tissue perfusion and its potential changes following therapies. It has the invaluable advantage of being easily repeatable, minimally invasive, and able to grant important evaluations regarding patients' survival, essential for well-timed therapeutic changes in case of unsatisfying response, and eventual further treatment planning.
Collapse
Affiliation(s)
- Lucia Cerrito
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy (M.E.A.); (G.C.); (G.E.); (M.G.); (L.R.); (A.G.)
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| | - Maria Elena Ainora
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy (M.E.A.); (G.C.); (G.E.); (M.G.); (L.R.); (A.G.)
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| | - Giuseppe Cuccia
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy (M.E.A.); (G.C.); (G.E.); (M.G.); (L.R.); (A.G.)
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| | - Linda Galasso
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| | - Irene Mignini
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| | - Giorgio Esposto
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy (M.E.A.); (G.C.); (G.E.); (M.G.); (L.R.); (A.G.)
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| | - Matteo Garcovich
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy (M.E.A.); (G.C.); (G.E.); (M.G.); (L.R.); (A.G.)
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| | - Laura Riccardi
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy (M.E.A.); (G.C.); (G.E.); (M.G.); (L.R.); (A.G.)
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| | - Antonio Gasbarrini
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy (M.E.A.); (G.C.); (G.E.); (M.G.); (L.R.); (A.G.)
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| | - Maria Assunta Zocco
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy (M.E.A.); (G.C.); (G.E.); (M.G.); (L.R.); (A.G.)
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (L.G.); (I.M.)
| |
Collapse
|
2
|
Li MH, Li WW, He L, Li JF, Zhang SY. Quantitative evaluation of colorectal tumour vasculature using contrast-enhanced ultrasound: Correlation with angiogenesis and prognostic significance. World J Gastrointest Surg 2023; 15:2052-2062. [PMID: 37901730 PMCID: PMC10600759 DOI: 10.4240/wjgs.v15.i9.2052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/11/2023] [Accepted: 07/26/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Ultrasound is a vital tool for the diagnosis and management of colorectal cancer (CRC). Contrast-enhanced ultrasound (CEUS) is a non-invasive, safe, and cost-effective method for evaluating tumour blood vessels, that play a crucial role in tumour growth and progression. AIM To explore CEUS's role in the quantitative evaluation of CRC blood vessels and their correlation with angiogenesis markers and prognosis. METHODS This study prospectively enrolled 100 patients with CRC confirmed by histopathology. All patients received preoperative CEUS examinations. Quantitative parameters, such as peak intensity (PI), time to peak (TTP), and area under the curve (AUC), were derived from time-intensity curve (TIC) analysis. Tumour tissue samples were obtained during surgery and examined immunohistochemically to assess the expression of angiogenesis markers, including vascular endothelial growth factor (VEGF) and microvessel density (MVD). The correlation between CEUS parameters, angiogenesis markers, and clinicopathological features was evaluated using appropriate statistical tests. RESULTS Quantitative CEUS parameters (PI, TTP, and AUC) showed significant correlations with VEGF expression (P < 0.001) and MVD (P < 0.001), indicating a strong link between tumour blood vessels and angiogenesis. Increased PI, reduced TTP, and expanded AUC values were significantly related to higher tumour stage (P < 0.001), lymph node metastasis (P < 0.001), and distant metastasis (P < 0.001). Furthermore, these parameters were recognized as independent predictors of overall survival and disease-free survival in multivariate analysis (P < 0.001). CONCLUSION CEUS has a high potential in guiding treatment planning and predicting patient outcomes. However, more comprehensive, multicentre studies are required to validate the clinical utility of CEUS in CRC management.
Collapse
Affiliation(s)
- Ming-Hui Li
- Department of Ultrasound, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, Xinjiang Uygur Autonomous Regions, China
| | - Wei-Wei Li
- Department of Ultrasound, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, Xinjiang Uygur Autonomous Regions, China
| | - Ling He
- Department of Ultrasound, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, Xinjiang Uygur Autonomous Regions, China
| | - Jian-Fang Li
- Department of Medical Imaging, Baoding Maternal and Child Health Hospital, Baoding 071023, Hebei Province, China
| | - Sun-Yan Zhang
- Department of Ultrasonography, Nantong Haimen District People’s Hospital, Nantong 226100, Jiangsu Province, China
| |
Collapse
|
3
|
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
|
4
|
Posada Calderon L, Eismann L, Reese SW, Reznik E, Hakimi AA. Advances in Imaging-Based Biomarkers in Renal Cell Carcinoma: A Critical Analysis of the Current Literature. Cancers (Basel) 2023; 15:cancers15020354. [PMID: 36672304 PMCID: PMC9856305 DOI: 10.3390/cancers15020354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Cross-sectional imaging is the standard diagnostic tool to determine underlying biology in renal masses, which is crucial for subsequent treatment. Currently, standard CT imaging is limited in its ability to differentiate benign from malignant disease. Therefore, various modalities have been investigated to identify imaging-based parameters to improve the noninvasive diagnosis of renal masses and renal cell carcinoma (RCC) subtypes. MRI was reported to predict grading of RCC and to identify RCC subtypes, and has been shown in a small cohort to predict the response to targeted therapy. Dynamic imaging is promising for the staging and diagnosis of RCC. PET/CT radiotracers, such as 18F-fluorodeoxyglucose (FDG), 124I-cG250, radiolabeled prostate-specific membrane antigen (PSMA), and 11C-acetate, have been reported to improve the identification of histology, grading, detection of metastasis, and assessment of response to systemic therapy, and to predict oncological outcomes. Moreover, 99Tc-sestamibi and SPECT scans have shown promising results in distinguishing low-grade RCC from benign lesions. Radiomics has been used to further characterize renal masses based on semantic and textural analyses. In preliminary studies, integrated machine learning algorithms using radiomics proved to be more accurate in distinguishing benign from malignant renal masses compared to radiologists' interpretations. Radiomics and radiogenomics are used to complement risk classification models to predict oncological outcomes. Imaging-based biomarkers hold strong potential in RCC, but require standardization and external validation before integration into clinical routines.
Collapse
Affiliation(s)
- Lina Posada Calderon
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lennert Eismann
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen W. Reese
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ed Reznik
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Abraham Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
5
|
Zhang Q, Wu G, Yang Q, Dai G, Li T, Chen P, Li J, Huang W. Survival rate prediction of nasopharyngeal carcinoma patients based on MRI and gene expression using a deep neural network. Cancer Sci 2022; 114:1596-1605. [PMID: 36541519 PMCID: PMC10067413 DOI: 10.1111/cas.15704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 12/24/2022] Open
Abstract
To achieve a better treatment regimen and follow-up assessment design for intensity-modulated radiotherapy (IMRT)-treated nasopharyngeal carcinoma (NPC) patients, an accurate progression-free survival (PFS) time prediction algorithm is needed. We propose developing a PFS prediction model of NPC patients after IMRT treatment using a deep learning method and comparing that with the traditional texture analysis method. One hundred and fifty-one NPC patients were included in this retrospective study. T1-weighted, proton density and dynamic contrast-enhanced magnetic resonance (MR) images were acquired. The expression level of five genes (HIF-1α, EGFR, PTEN, Ki-67, and VEGF) and infection of Epstein-Barr (EB) virus were tested. A residual network was trained to predict PFS from MR images. The output as well as patient characteristics were combined using a linear regression model to provide a final PFS prediction. The prediction accuracy was compared with that of the traditional texture analysis method. A regression model combining the deep learning output with HIF-1α expression and Epstein-Barr infection provides the best PFS prediction accuracy (Spearman correlation R2 = 0.53; Harrell's C-index = 0.82; receiver operative curve [ROC] analysis area under the curve [AUC] = 0.88; log-rank test hazard ratio [HR] = 8.45), higher than a regression model combining texture analysis with HIF-1α expression (Spearman correlation R2 = 0.14; Harrell's C-index =0.68; ROC analysis AUC = 0.76; log-rank test HR = 2.85). The deep learning method does not require a manually drawn tumor region of interest. MR image processing using deep learning combined with patient characteristics can provide accurate PFS prediction for nasopharyngeal carcinoma patients and does not rely on specific kernels or tumor regions of interest, which is needed for the texture analysis method.
Collapse
Affiliation(s)
- Qihao Zhang
- Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - Gang Wu
- Department of Radiotherapy, Hainan General Hospital, Hainan, China
| | - Qianyu Yang
- Department of Radiology, Hainan General Hospital, Hainan, China
| | - Ganmian Dai
- Department of Radiology, Hainan General Hospital, Hainan, China
| | - Tiansheng Li
- Department of Radiology, Hainan General Hospital, Hainan, China
| | - Pianpian Chen
- Department of Pathology, Hainan General Hospital, Hainan, China
| | - Jiao Li
- Department of Pathology, Hainan General Hospital, Hainan, China
| | - Weiyuan Huang
- Department of Radiology, Hainan General Hospital, Hainan, China
| |
Collapse
|
6
|
Albano D, Bruno F, Agostini A, Angileri SA, Benenati M, Bicchierai G, Cellina M, Chianca V, Cozzi D, Danti G, De Muzio F, Di Meglio L, Gentili F, Giacobbe G, Grazzini G, Grazzini I, Guerriero P, Messina C, Micci G, Palumbo P, Rocco MP, Grassi R, Miele V, Barile A. Dynamic contrast-enhanced (DCE) imaging: state of the art and applications in whole-body imaging. Jpn J Radiol 2022; 40:341-366. [PMID: 34951000 DOI: 10.1007/s11604-021-01223-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022]
Abstract
Dynamic contrast-enhanced (DCE) imaging is a non-invasive technique used for the evaluation of tissue vascularity features through imaging series acquisition after contrast medium administration. Over the years, the study technique and protocols have evolved, seeing a growing application of this method across different imaging modalities for the study of almost all body districts. The main and most consolidated current applications concern MRI imaging for the study of tumors, but an increasing number of studies are evaluating the use of this technique also for inflammatory pathologies and functional studies. Furthermore, the recent advent of artificial intelligence techniques is opening up a vast scenario for the analysis of quantitative information deriving from DCE. The purpose of this article is to provide a comprehensive update on the techniques, protocols, and clinical applications - both established and emerging - of DCE in whole-body imaging.
Collapse
Affiliation(s)
- Domenico Albano
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Dipartimento Di Biomedicina, Neuroscienze E Diagnostica Avanzata, Sezione Di Scienze Radiologiche, Università Degli Studi Di Palermo, via Vetoio 1L'Aquila, 67100, Palermo, Italy
| | - Federico Bruno
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy.
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Andrea Agostini
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Department of Clinical, Special and Dental Sciences, Department of Radiology, University Politecnica delle Marche, University Hospital "Ospedali Riuniti Umberto I - G.M. Lancisi - G. Salesi", Ancona, Italy
| | - Salvatore Alessio Angileri
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Radiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Benenati
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Dipartimento di Diagnostica per Immagini, Fondazione Policlinico Universitario A. Gemelli IRCCS, Oncologia ed Ematologia, RadioterapiaRome, Italy
| | - Giulia Bicchierai
- Diagnostic Senology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Michaela Cellina
- Department of Radiology, ASST Fatebenefratelli Sacco, Ospedale Fatebenefratelli, Milan, Italy
| | - Vito Chianca
- Ospedale Evangelico Betania, Naples, Italy
- Clinica Di Radiologia, Istituto Imaging Della Svizzera Italiana - Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Diletta Cozzi
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Department of Emergency Radiology, Careggi University Hospital, Florence, Italy
| | - Ginevra Danti
- Department of Emergency Radiology, Careggi University Hospital, Florence, Italy
| | - Federica De Muzio
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Letizia Di Meglio
- Postgraduation School in Radiodiagnostics, University of Milan, Milan, Italy
| | - Francesco Gentili
- Unit of Diagnostic Imaging, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Giuliana Giacobbe
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Giulia Grazzini
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Irene Grazzini
- Department of Radiology, Section of Neuroradiology, San Donato Hospital, Arezzo, Italy
| | - Pasquale Guerriero
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | | | - Giuseppe Micci
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Dipartimento Di Biomedicina, Neuroscienze E Diagnostica Avanzata, Sezione Di Scienze Radiologiche, Università Degli Studi Di Palermo, via Vetoio 1L'Aquila, 67100, Palermo, Italy
| | - Pierpaolo Palumbo
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Abruzzo Health Unit 1, Department of diagnostic Imaging, Area of Cardiovascular and Interventional Imaging, L'Aquila, Italy
| | - Maria Paola Rocco
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Roberto Grassi
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| | - Vittorio Miele
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Antonio Barile
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
7
|
Contrast-enhanced ultrasonography for blood flow detection in hepatocellular carcinoma during lenvatinib therapy. J Med Ultrason (2001) 2022; 49:425-432. [PMID: 35355122 DOI: 10.1007/s10396-022-01204-8] [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: 01/13/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE Blood flow reduction after initiation of lenvatinib therapy may not always indicate tumor necrosis. This study aimed to compare the blood flow detectability of contrast-enhanced ultrasonography (CEUS), contrast-enhanced computed tomography (CT), and contrast-enhanced magnetic resonance imaging (MRI) in hepatocellular carcinoma (HCC) during lenvatinib therapy. METHODS A total of 12 cases underwent CEUS and contrast-enhanced CT/MRI within 2 weeks during lenvatinib therapy. Vascularity on CEUS and CT/MRI was compared. RESULTS At the time of CEUS examination, the median period from the start of lenvatinib was 227 ± 210 (31-570) days. CEUS showed hyperenhancement in eight cases (66.7%), hypoenhancement in two cases (16.7%), and no enhancement in one case (8.3%), while CT/MRI showed hyperenhancement in one case (8.3%), ring enhancement in three cases (25.0%), and hypoenhancement in eight cases (66.7%) (p = 0.007). Transarterial chemoembolization (n = 3), radiofrequency ablation (n = 2), and stereotactic body radiation therapy (n = 2) were performed after blood flow detection by CEUS. CONCLUSIONS The viability of the HCC should be confirmed using CEUS when contrast-enhanced CT/MRI reveals lesion hypoenhancement during lenvatinib therapy.
Collapse
|
8
|
Faccia M, Garcovich M, Ainora ME, Riccardi L, Pompili M, Gasbarrini A, Zocco MA. Contrast-Enhanced Ultrasound for Monitoring Treatment Response in Different Stages of Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:481. [PMID: 35158749 PMCID: PMC8833342 DOI: 10.3390/cancers14030481] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 01/01/2023] Open
Abstract
The capacity of contrast-enhanced ultrasound (CEUS) to detect microvessel perfusion has received much attention in cancer imaging since it can be used to evaluate the enhancement patterns of the lesions during all vascular phases in real time, with higher temporal resolution as compared other imaging modalities. A rich body of literature has demonstrated the potential usefulness of CEUS in the assessment of HCC in response to both locoregional and systemic therapies. It is useful to evaluate the efficacy of ablation immediately after treatment to provide guidance for the retreatment of residual unablated tumors. In patients treated with transarterial chemoembolization (TACE), CEUS showed a high degree of concordance with computed tomography and magnetic resonance for the differentiation of responders from non-responders. Dynamic CEUS (D-CEUS) has emerged as a promising tool for the depicting changes in tumor perfusion during anti-angiogenetic treatment that can be associated with tumor response and clinical outcome. This article provides a general review of the current literature regarding the usefulness of CEUS in monitoring HCC response to therapy, highlighting the role of the procedure in different stages of the disease.
Collapse
Affiliation(s)
- Mariella Faccia
- Department of Internal Medicine, SS Annunziata Hospital Sulmona, 67039 Sulmona, Italy;
| | - Matteo Garcovich
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.G.); (M.E.A.); (L.R.); (M.P.); (A.G.)
| | - Maria Elena Ainora
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.G.); (M.E.A.); (L.R.); (M.P.); (A.G.)
| | - Laura Riccardi
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.G.); (M.E.A.); (L.R.); (M.P.); (A.G.)
| | - Maurizio Pompili
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.G.); (M.E.A.); (L.R.); (M.P.); (A.G.)
| | - Antonio Gasbarrini
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.G.); (M.E.A.); (L.R.); (M.P.); (A.G.)
| | - Maria Assunta Zocco
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Rome, 00168 Rome, Italy; (M.G.); (M.E.A.); (L.R.); (M.P.); (A.G.)
| |
Collapse
|
9
|
Nielsen MB, Søgaard SB, Bech Andersen S, Skjoldbye B, Hansen KL, Rafaelsen S, Nørgaard N, Carlsen JF. Highlights of the development in ultrasound during the last 70 years: A historical review. Acta Radiol 2021; 62:1499-1514. [PMID: 34791887 DOI: 10.1177/02841851211050859] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review looks at highlights of the development in ultrasound, ranging from interventional ultrasound and Doppler to the newest techniques like contrast-enhanced ultrasound and elastography, and gives reference to some of the valuable articles in Acta Radiologica. Ultrasound equipment is now available in any size and for any purpose, ranging from handheld devices to high-end devices, and the scientific societies include ultrasound professionals of all disciplines publishing guidelines and recommendations. Interventional ultrasound is expanding the field of use of ultrasound-guided interventions into nearly all specialties of medicine, from ultrasound guidance in minimally invasive robotic procedures to simple ultrasound-guided punctures performed by general practitioners. Each medical specialty is urged to define minimum requirements for equipment, education, training, and maintenance of skills, also for medical students. The clinical application of contrast-enhanced ultrasound and elastography is a topic often seen in current research settings.
Collapse
Affiliation(s)
- Michael Bachmann Nielsen
- Department of Radiology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stinne Byrholdt Søgaard
- Department of Radiology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofie Bech Andersen
- Department of Radiology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bjørn Skjoldbye
- Department of Radiology, Aleris-Hamlet Hospitals, Copenhagen Denmark
| | - Kristoffer Lindskov Hansen
- Department of Radiology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Rafaelsen
- Department of Radiology, University Hospital of Southern Denmark, Vejle, Denmark
- Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Nis Nørgaard
- Department of Urology, Herlev Gentofte Hospital, Copenhagen, Denmark
| | - Jonathan F. Carlsen
- Department of Radiology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Spek A, Graser A, Casuscelli J, Szabados B, Rodler S, Marcon J, Stief C, Staehler M. Dynamic contrast-enhanced CT-derived blood flow measurements enable early prediction of long term outcome in metastatic renal cell cancer patients on antiangiogenic treatment. Urol Oncol 2021; 40:13.e1-13.e8. [PMID: 34535355 DOI: 10.1016/j.urolonc.2021.08.012] [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: 05/15/2021] [Revised: 07/11/2021] [Accepted: 08/13/2021] [Indexed: 11/18/2022]
Abstract
PURPOSE To evaluate the role of dynamic contrast-enhanced CT (DCE-CT) as an independent non-invasive biomarker in predicting long term outcome in patients with metastatic renal cell carcinoma (mRCC) on antiangiogenic treatment. MATERIAL AND METHODS Eighty two mRCC patients were prospectively enrolled from 09/2011 to 04/2015, out of which 71 were included in the final data analysis; the population was observed until 12/2020 to obtain complete overall survival data. DCE-CT imaging was performed at baseline and 10 to 12 weeks after start of treatment with targeted therapy. DCE-CT included a dynamic acquisition after injection of 50 ml of nonionic contrast agent at 6 ml/s using a 4D spiral mode (10 cm z-axis coverage, acquisition time 43 sec, 100 kVp (abdomen), 80 kVp (chest), 80-100 mAs) on a dual source scanner (Definition FLASH, Siemens). Blood flow (BF) was calculated for target tumor volumes using a deconvolution model. Progression free survival (PFS) and overall survival (OS) were analyzed using Kaplan-Meier statistics (SPSS version 24). RESULTS Patients were treated with either sunitinib, pazopanib, sorafenib, tivozanib, axitinib, or cabozantinib. A cut-off value of 50% blood flow reduction at follow-up allowed for identification of patients with favorable long-term outcome: Median OS in n = 42 patients with an average blood flow reduction of >50% (mean, 79%) was 34 (range, 14-54) months, while n = 21 patients with an average reduction of less than 50% (mean, 28%) showed a median OS of 12 (range, 6-18) months, and n = 8 patients with an increase in blood flow survived for a median of 7 (range, 3-11) months. CONCLUSION Blood flow in metastases measured with DCE-CT at first follow-up is a strong predictor of overall survival in mRCC patients on antiangiogenic treatment.
Collapse
Affiliation(s)
- Annabel Spek
- Department of Urology, University Hospital, LMU Munich, Munich, Germany.
| | | | | | | | - Severin Rodler
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Julian Marcon
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Christian Stief
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Michael Staehler
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
11
|
Brloznik M, Kranjc Brezar S, Boc N, Knific T, Cemazar M, Milevoj N, Sersa G, Tozon N, Pavlin D. Results of Dynamic Contrast-Enhanced Ultrasound Correlate With Treatment Outcome in Canine Neoplasia Treated With Electrochemotherapy and Interleukin-12 Plasmid Electrotransfer. Front Vet Sci 2021; 8:679073. [PMID: 34095282 PMCID: PMC8173043 DOI: 10.3389/fvets.2021.679073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
Electrochemotherapy (ECT) and/or gene electrotransfer of plasmid DNA encoding interleukin-12 (GET pIL-12) are effective treatments for canine cutaneous, subcutaneous, and maxillofacial tumors. Despite the clinical efficacy of the combined treatments of ECT and GET, data on parameters that might predict the outcome of the treatments are still lacking. This study aimed to investigate whether dynamic contrast-enhanced ultrasound (DCE-US) results of subcutaneous tumors differ between tumors with complete response (CR) and tumors without complete response (non-CR) in dogs treated with ECT and GET pIL-12. Eight dogs with a total of 12 tumor nodules treated with ECT and GET pIL-12 were included. DCE-US examinations were performed in all animals before and immediately after therapy as well as 8 h and 1, 3, and 7 days later. Clinical follow-up examinations were performed 7 and 14 days, 1 and 6 months, and 1 year after treatment. Numerous significant differences in DCE-US parameters were noted between tumors with CR and non-CR tumors; perfusion and perfusion heterogeneity were lower in CR tumors than in non-CR tumors. Therefore, studies with larger numbers of patients are needed to investigate whether DCE-US results can be used to predict treatment outcomes and to make effective decisions about the need for repeated therapy or different treatment combinations in individual patients.
Collapse
Affiliation(s)
- Maja Brloznik
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| | - Simona Kranjc Brezar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Nina Boc
- Department of Radiology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Tanja Knific
- Institute of Food Safety, Feed and Environment, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia.,Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Nina Milevoj
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia.,Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
| | - Natasa Tozon
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| | - Darja Pavlin
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
12
|
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
|
13
|
Kumagawa M, Matsumoto N, Miura K, Ogawa M, Takahashi H, Hatta Y, Kondo R, Koizumi N, Takei M, Moriyama M. Correlation between alterations in blood flow of malignant lymphomas after induction chemotherapies and clinical outcomes: a pilot study utilising contrast-enhanced ultrasonography for early interim evaluation of lymphoma treatment. Clin Radiol 2021; 76:550.e9-550.e17. [PMID: 33691950 DOI: 10.1016/j.crad.2021.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
Abstract
AIM To clarify the utility of contrast-enhanced ultrasonography (CEUS) for interim evaluation of response to chemotherapy in lymphoma treatment. MATERIALS AND METHODS CEUS was performed both before (day 0) and after the treatment (7 and/or 14 days), and a time-intensity curve was obtained. The patients were divided into two groups (complete remission [CR] group and non-CR group) according to the results of conventional response evaluation, and peak enhancement (PE), time to peak enhancement, perfusion index (PI), the total area under the curve during wash-in (AUC-in), and the total AUC were compared between the groups. RESULTS Among 27 patients with various types of lymphoma, the median change ratio of PE and PI at day 7 evaluation were significantly different between the CR group and the non-CR group (0.81 versus 1.39, p=0.017 for PE and 0.92 versus 2.09, p=0.010 for PI). The change ratio of PE < 1.09 (specificity: 86%; sensitivity, 88%) and PI < 1.65 (specificity: 86%; sensitivity: 94%) distinguished CR from non-CR. Patients who achieved a PE change ratio <1.09 or a PI change ratio <1.65 had significantly better estimated progression-free survival (p<0.001). CONCLUSION The present study demonstrated that changes in tumour perfusion parameters evaluated with CEUS at 1 week after the treatment initiation were significantly different between lymphoma patients in CR group and non-CR group. Alterations in perfusion parameters evaluated via CEUS could impact the prognosis of lymphoma patients.
Collapse
Affiliation(s)
- M Kumagawa
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-ku, Tokyo, Japan
| | - N Matsumoto
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-ku, Tokyo, Japan.
| | - K Miura
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-ku, Tokyo, Japan.
| | - M Ogawa
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-ku, Tokyo, Japan
| | - H Takahashi
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-ku, Tokyo, Japan
| | - Y Hatta
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-ku, Tokyo, Japan
| | - R Kondo
- Department of Mechanical Engineering and Intelligent Systems, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1, Chofugaoka, Chofu-shi, Tokyo, Japan
| | - N Koizumi
- Department of Mechanical Engineering and Intelligent Systems, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1, Chofugaoka, Chofu-shi, Tokyo, Japan
| | - M Takei
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-ku, Tokyo, Japan
| | - M Moriyama
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-ku, Tokyo, Japan
| |
Collapse
|
14
|
Dietrich CF, Nolsøe CP, Barr RG, Berzigotti A, Burns PN, Cantisani V, Chammas MC, Chaubal N, Choi BI, Clevert DA, Cui X, Dong Y, D'Onofrio M, Fowlkes JB, Gilja OH, Huang P, Ignee A, Jenssen C, Kono Y, Kudo M, Lassau N, Lee WJ, Lee JY, Liang P, Lim A, Lyshchik A, Meloni MF, Correas JM, Minami Y, Moriyasu F, Nicolau C, Piscaglia F, Saftoiu A, Sidhu PS, Sporea I, Torzilli G, Xie X, Zheng R. Guidelines and Good Clinical Practice Recommendations for Contrast Enhanced Ultrasound (CEUS) in the Liver - Update 2020 - WFUMB in Cooperation with EFSUMB, AFSUMB, AIUM, and FLAUS. ULTRASCHALL IN DER MEDIZIN (STUTTGART, GERMANY : 1980) 2020; 41:562-585. [PMID: 32707595 DOI: 10.1055/a-1177-0530] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The present, updated document describes the fourth iteration of recommendations for the hepatic use of contrast enhanced ultrasound (CEUS), first initiated in 2004 by the European Federation of Societies for Ultrasound in Medicine and Biology (EFSUMB). The previous updated editions of the guidelines reflected changes in the available contrast agents and updated the guidelines not only for hepatic but also for non-hepatic applications.The 2012 guideline requires updating as previously the differences of the contrast agents were not precisely described and the differences in contrast phases as well as handling were not clearly indicated. In addition, more evidence has been published for all contrast agents. The update also reflects the most recent developments in contrast agents, including the United States Food and Drug Administration (FDA) approval as well as the extensive Asian experience, to produce a truly international perspective.These guidelines and recommendations provide general advice on the use of ultrasound contrast agents (UCA) and are intended to create standard protocols for the use and administration of UCA in liver applications on an international basis to improve the management of patients.
Collapse
Affiliation(s)
- Christoph F Dietrich
- Department Allgemeine Innere Medizin (DAIM), Kliniken Hirslanden Beau Site, Salem und Permanence, Bern, Switzerland
- Johann Wolfgang Goethe Universitätsklinik Frankfurt, Germany
| | - Christian Pállson Nolsøe
- Center for Surgical Ultrasound, Dep of Surgery, Zealand University Hospital, Køge. Copenhagen Academy for Medical Education and Simulation (CAMES). University of Copenhagen, Denmark
| | - Richard G Barr
- Department of Radiology, Northeastern Ohio Medical University, Rootstown, Ohio, USA and Southwoods Imaging, Youngstown, Ohio, USA
| | - Annalisa Berzigotti
- Hepatology, University Clinic for Visceral Surgery and Medicine, DBMR, Inselspital, University of Bern, Switzerland
| | - Peter N Burns
- Dept Medical Biophysics, University of Toronto, Imaging Research, Sunnybrook Research Institute, Toronto
| | - Vito Cantisani
- Uos Ecografia Internistico-chirurgica, Dipartimento di Scienze Radiologiche, Oncologiche, Anatomo-Patologiche, Policlinico Umberto I, Univ. Sapienza, Rome, Italy
| | - Maria Cristina Chammas
- Institute of Radiology, Hospital das Clínicas, School of Medicine, University of São Paulo, Brazil
| | - Nitin Chaubal
- Thane Ultrasound Centre, Jaslok Hospital and Research Centre, Mumbai, India
| | - Byung Ihn Choi
- Department of Radiology, Chung-Ang University Hospital, Seoul, Korea
| | - Dirk-André Clevert
- Interdisciplinary Ultrasound-Center, Department of Radiology, University of Munich-Grosshadern Campus, Munich, Germany
| | - Xinwu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan China
| | - Yi Dong
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mirko D'Onofrio
- Department of Radiology, G.B. Rossi University Hospital, University of Verona, Verona, Italy
| | - J Brian Fowlkes
- Basic Radiological Sciences Division, Department of Radiology, University of Michigan Health System, Ann Arbor, MI, United States
| | - Odd Helge Gilja
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, and Department of Clinical Medicine, University of Bergen, Norway
| | - Pintong Huang
- Department of Ultrasound in Medicine, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Andre Ignee
- Department of Internal Medicine 2, Caritas Krankenhaus, Bad Mergentheim, Germany
| | - Christian Jenssen
- Krankenhaus Märkisch Oderland, Department of Internal Medicine, Strausberg/Wriezen, Germany
| | - Yuko Kono
- Departments of Medicine and Radiology, University of California, San Diego, USA
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Nathalie Lassau
- Imaging Department. Gustave Roussy and BIOMAPS. Université Paris-Saclay, Villejuif, France
| | - Won Jae Lee
- Department of Radiology and Center For Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea. Departments of Health and Science and Technology and Medical Device Management and Research, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul, Korea
| | - Jae Young Lee
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Ping Liang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| | - Adrian Lim
- Department of Imaging, Imperial College London and Healthcare NHS Trust, Charing Cross Hospital Campus, London United Kingdom
| | - Andrej Lyshchik
- Department of Radiology, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | | | - Jean Michel Correas
- Service de Radiologie Adultes, Hôpital Necker, Université Paris Descartes, Paris, France
| | - Yasunori Minami
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Fuminori Moriyasu
- Center for Cancer Ablation Therapy, Sanno Hospital, International University of Health and Welfare, Tokyo, Japan
| | - Carlos Nicolau
- Radiology Department, Hospital Clinic. University of Barcelona, Barcelona, Spain
| | - Fabio Piscaglia
- Unit of Internal Medicine, Dept of Medical and Surgical Sciences, University of Bologna S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Adrian Saftoiu
- Research Center of Gastroenterology and Hepatology Craiova, University of Medicine and Pharmacy Craiova, Romania
| | - Paul S Sidhu
- Department of Radiology, King's College Hospital, King's College London, London
| | - Ioan Sporea
- Department of Gastroenterology and Hepatology, University of Medicine and Pharmacy "Victor Babes", Timisoara, Romania
| | - Guido Torzilli
- Department of Surgery, Division of Hepatobiliary & General Surgery, Humanitas University & Research Hospital, Rozzano, Milano, Italy
| | - Xiaoyan Xie
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Rongqin Zheng
- Department of Ultrasound, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Dietrich CF, Nolsøe CP, Barr RG, Berzigotti A, Burns PN, Cantisani V, Chammas MC, Chaubal N, Choi BI, Clevert DA, Cui X, Dong Y, D'Onofrio M, Fowlkes JB, Gilja OH, Huang P, Ignee A, Jenssen C, Kono Y, Kudo M, Lassau N, Lee WJ, Lee JY, Liang P, Lim A, Lyshchik A, Meloni MF, Correas JM, Minami Y, Moriyasu F, Nicolau C, Piscaglia F, Saftoiu A, Sidhu PS, Sporea I, Torzilli G, Xie X, Zheng R. Guidelines and Good Clinical Practice Recommendations for Contrast-Enhanced Ultrasound (CEUS) in the Liver-Update 2020 WFUMB in Cooperation with EFSUMB, AFSUMB, AIUM, and FLAUS. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:2579-2604. [PMID: 32713788 DOI: 10.1016/j.ultrasmedbio.2020.04.030] [Citation(s) in RCA: 270] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 05/14/2023]
Abstract
The present, updated document describes the fourth iteration of recommendations for the hepatic use of contrast-enhanced ultrasound, first initiated in 2004 by the European Federation of Societies for Ultrasound in Medicine and Biology. The previous updated editions of the guidelines reflected changes in the available contrast agents and updated the guidelines not only for hepatic but also for non-hepatic applications. The 2012 guideline requires updating as, previously, the differences in the contrast agents were not precisely described and the differences in contrast phases as well as handling were not clearly indicated. In addition, more evidence has been published for all contrast agents. The update also reflects the most recent developments in contrast agents, including U.S. Food and Drug Administration approval and the extensive Asian experience, to produce a truly international perspective. These guidelines and recommendations provide general advice on the use of ultrasound contrast agents (UCAs) and are intended to create standard protocols for the use and administration of UCAs in liver applications on an international basis to improve the management of patients.
Collapse
Affiliation(s)
- Christoph F Dietrich
- Department Allgemeine Innere Medizin (DAIM), Kliniken Hirslanden Beau Site, Salem und Permanence, Bern, Switzerland; Johann Wolfgang Goethe Universitätsklinik, Frankfurt, Germany.
| | - Christian Pállson Nolsøe
- Center for Surgical Ultrasound, Dep of Surgery, Zealand University Hospital, Køge. Copenhagen Academy for Medical Education and Simulation (CAMES). University of Copenhagen, Denmark
| | - Richard G Barr
- Department of Radiology, Northeastern Ohio Medical University, Rootstown, Ohio, USA; Southwoods Imaging, Youngstown, Ohio, USA
| | - Annalisa Berzigotti
- Hepatology, University Clinic for Visceral Surgery and Medicine, DBMR, Inselspital, University of Bern, Switzerland
| | - Peter N Burns
- Department of Medical Biophysics, University of Toronto, Imaging Research, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Vito Cantisani
- Uos Ecografia Internistico-chirurgica, Dipartimento di Scienze Radiologiche, Oncologiche, Anatomo-Patologiche, Policlinico Umberto I, Univ. Sapienza, Rome, Italy
| | - Maria Cristina Chammas
- Institute of Radiology, Hospital das Clínicas, School of Medicine, University of São Paulo, Brazil
| | - Nitin Chaubal
- Thane Ultrasound Centre, Jaslok Hospital and Research Centre, Mumbai, India
| | - Byung Ihn Choi
- Department of Radiology, Chung-Ang University Hospital, Seoul, Korea
| | - Dirk-André Clevert
- Interdisciplinary Ultrasound-Center, Department of Radiology, University of Munich-Grosshadern Campus, Munich, Germany
| | - Xinwu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Dong
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mirko D'Onofrio
- Department of Radiology, G. B. Rossi University Hospital, University of Verona, Verona, Italy
| | - J Brian Fowlkes
- Basic Radiological Sciences Division, Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Odd Helge Gilja
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, and Department of Clinical Medicine, University of Bergen, Norway
| | - Pintong Huang
- Department of Ultrasound in Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Andre Ignee
- Department of Internal Medicine 2, Caritas Krankenhaus, Bad Mergentheim, Germany
| | - Christian Jenssen
- Krankenhaus Märkisch Oderland, Department of Internal Medicine, Strausberg/Wriezen, Germany
| | - Yuko Kono
- Departments of Medicine and Radiology, University of California, San Diego, California, USA
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Nathalie Lassau
- Imaging Department, Gustave Roussy and BIOMAPS, Université Paris-Saclay, Villejuif, France
| | - Won Jae Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Departments of Health and Science and Technology and Medical Device Management and Research, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul, Korea
| | - Jae Young Lee
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Ping Liang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| | - Adrian Lim
- Department of Imaging, Imperial College London and Healthcare NHS Trust, Charing Cross Hospital Campus, London, United Kingdom
| | - Andrej Lyshchik
- Department of Radiology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | | | - Jean Michel Correas
- Service de Radiologie Adultes, Hôpital Necker, Université Paris Descartes, Paris, France
| | - Yasunori Minami
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Fuminori Moriyasu
- Center for Cancer Ablation Therapy, Sanno Hospital, International University of Health and Welfare, Tokyo, Japan
| | - Carlos Nicolau
- Radiology Department, Hospital Clinic. University of Barcelona, Barcelona, Spain
| | - Fabio Piscaglia
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University of Bologna S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Adrian Saftoiu
- Research Center of Gastroenterology and Hepatology Craiova, University of Medicine and Pharmacy Craiova, Romania
| | - Paul S Sidhu
- Department of Radiology, King's College Hospital, King's College London, London, United Kingdom
| | - Ioan Sporea
- Department of Gastroenterology and Hepatology, University of Medicine and Pharmacy "Victor Babes", Timisoara, Romania
| | - Guido Torzilli
- Department of Surgery, Division of Hepatobiliary & General Surgery, Humanitas University & Research Hospital, Rozzano, Milan, Italy
| | - Xiaoyan Xie
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Rongqin Zheng
- Department of Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
16
|
Lamuraglia M, Barrois G, Le Guillou-Buffello D, Santin M, Kerbol A, Comperat E, Coron A, Lucidarme O, Bridal SL. Monitoring Dual VEGF Inhibition in Human Pancreatic Tumor Xenografts With Dynamic Contrast-Enhanced Ultrasound. Technol Cancer Res Treat 2020; 19:1533033819886896. [PMID: 32065066 PMCID: PMC7026814 DOI: 10.1177/1533033819886896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Association of drugs acting against different antiangiogenic mechanisms may increase therapeutic effect and reduce resistance. Noninvasive monitoring of changes in the antiangiogenic response of individual tumors could guide selection and administration of drug combinations. Noninvasive detection of early therapeutic response during dual, vertical targeting of the vascular endothelial growth factor pathway was investigated in an ectopic subcutaneous xenograft model for human pancreatic tumor. METHODS Dynamic contrast-enhanced ultrasound 12 MHz was used to monitor tumor-bearing Naval Medical Research Institute mice beginning 15 days after tumor implantation. Mice received therapy from 15 to 29 days with sorafenib (N = 9), ziv-aflibercept (N = 11), combined antiangiogenic agents (N = 11), and placebo control (N = 14). Sorafenib (BAY 43-9006; Nexavar), a multikinase inhibitor acting on Raf kinase and receptor tyrosine kinases-including vascular endothelial growth factor receptors 2 and 3-was administered daily (60 mg/kg, per os). Ziv-aflibercept (ZALTRAP), a high-affinity ligand trap blocking the activity of vascular endothelial growth factor A, vascular endothelial growth factor B, and placental growth factor was administered twice per week (40 mg/kg, intraperitoneally). RESULTS Functional evaluation with dynamic contrast-enhanced ultrasound indicated stable tumor vascularization for the control group while revealing significant and sustained reduction after 1 day of therapy in the combined group (P = .007). There was no survival benefit or penalty due to drug combination. The functional progression-free survival assessed with dynamic contrast-enhanced ultrasound was significantly higher for the 3 treated groups; whereas, the progression-free survival based on tumor size did not discriminate therapeutic effect. CONCLUSIONS Dynamic contrast-enhanced ultrasound, therefore, presents strong potential to monitor microvascular modifications during antiangiogenic therapy, a key role to monitoring antiangiogenic combining therapy to adapt dose range drug.
Collapse
Affiliation(s)
- Michele Lamuraglia
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), AP-HP, Hôpital Beaujon, Paris, France
| | - Guillaume Barrois
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| | | | - Mathieu Santin
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| | - Anne Kerbol
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), AP-HP, Hôpital Beaujon, Paris, France
| | - Eva Comperat
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| | - Alain Coron
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| | - Olivier Lucidarme
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - S Lori Bridal
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| |
Collapse
|
17
|
Lassau N. Advanced Ultrasound Imaging for Patients in Oncology: DCE-US. Recent Results Cancer Res 2020; 216:765-771. [PMID: 32594405 DOI: 10.1007/978-3-030-42618-7_23] [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: 06/11/2023]
Abstract
Neovascularization is a key stage in the growth of malignancies beyond 2-3 mm3. This neoangiogenesis is an important target for novel anticancer treatments [1], and many new antiangiogenesis or antivascular treatments aim at destroying or limiting the growth of tumor vessels [2].
Collapse
Affiliation(s)
- Nathalie Lassau
- Institut Gustave Roussy, Villejuif, France.
- Université of Paris-Saclay, Villejuif, France.
| |
Collapse
|
18
|
Kuorda H, Abe T, Fujiwara Y, Okamoto T, Yonezawa M, Sato H, Endo K, Oikawa T, Sawara K, Takikawa Y. Change in arterial tumor perfusion is an early biomarker of lenvatinib efficacy in patients with unresectable hepatocellular carcinoma. World J Gastroenterol 2019; 25:2365-2372. [PMID: 31148907 PMCID: PMC6529888 DOI: 10.3748/wjg.v25.i19.2365] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/12/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Lenvatinib is one of the first-line tyrosine kinase inhibitors used for unresectable hepatocellular carcinoma (HCC). In the present study, we evaluated the potential of early changes in the time-intensity curve (TIC) of arterial phase on contrast-enhanced ultrasound (CEUS) as early imaging biomarkers of lenvatinib efficacy.
AIM To evaluate the potential of the early changes in the TIC of CEUS as early imaging biomarkers of lenvatinib efficacy in patients with unresectable HCC.
METHODS We analyzed 20 consecutive patients with unresectable HCC treated with lenvatinib from March to November 2018. Tumor response at 8 wk was assessed by computed tomography using the modified Response Evaluation Criteria in Solid Tumors (mRECIST). CEUS was performed at baseline before treatment (Day 0) and on day 7 (Day 7), and the images were analyzed in the arterial phase for 20 seconds after the contrast agent arrived at the target tumor. Three perfusion parameters were extracted from the TICs: the slope of wash-in (Slope), time to peak (TTP) intensity, and the total area under the curve (AUC) during wash-in. The rate of change in the TIC parameters between Day 0 and Day 7 was compared between treatment responders and non-responders based on mRECIST.
RESULTS The rate of change for all TIC parameters showed significant differences between the responders (n = 9) and non-responders (n = 11) (Slope, P = 0.025; TTP, P = 0.004; and AUC, P = 0.0003). The area under the receiver operating curve values for slope, TTP, and AUC for the prediction of responders were 0.805, 0.869, and 0.939, respectively.
CONCLUSION CEUS may be useful for the early prediction of tumor response to lenvatinib therapy in patients with unresectable HCC.
Collapse
Affiliation(s)
- Hidekatsu Kuorda
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Tamami Abe
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Yudai Fujiwara
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Takuya Okamoto
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Miki Yonezawa
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Hiroki Sato
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Kei Endo
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Takayoshi Oikawa
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Kei Sawara
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Yasuhiro Takikawa
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| |
Collapse
|
19
|
Vaidya T, Agrawal A, Mahajan S, Thakur MH, Mahajan A. The Continuing Evolution of Molecular Functional Imaging in Clinical Oncology: The Road to Precision Medicine and Radiogenomics (Part II). Mol Diagn Ther 2019; 23:27-51. [PMID: 30387041 DOI: 10.1007/s40291-018-0367-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The present era of precision medicine sees "cancer" as a consequence of molecular derangements occurring at the commencement of the disease process, with morphological changes happening much later in the process of tumourigenesis. Conventional imaging techniques, such as computed tomography (CT), ultrasound (US) and magnetic resonance imaging (MRI) play an integral role in the detection of disease at the macroscopic level. However, molecular functional imaging (MFI) techniques entail the visualisation and quantification of biochemical and physiological processes occurring during tumourigenesis. MFI has the potential to play a key role in heralding the transition from the concept of "one-size-fits-all" treatment to "precision medicine". Integration of MFI with other fields of tumour biology such as genomics has spawned a novel concept called "radiogenomics", which could serve as an indispensable tool in translational cancer research. With recent advances in medical image processing, such as texture analysis, deep learning and artificial intelligence, the future seems promising; however, their clinical utility remains unproven at present. Despite the emergence of novel imaging biomarkers, the majority of these require validation before clinical translation is possible. In this two part review, we discuss the systematic collaboration across structural, anatomical and molecular imaging techniques that constitute MFI. Part I reviews positron emission tomography, radiogenomics, AI, and optical imaging, while part II reviews MRI, CT and ultrasound, their current status, and recent advances in the field of precision oncology.
Collapse
Affiliation(s)
- Tanvi Vaidya
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India
| | - Archi Agrawal
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Parel, Mumbai, Maharashtra, 400 012, India
| | - Shivani Mahajan
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India
| | - M H Thakur
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India
| | - Abhishek Mahajan
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Tata Memorial Centre, Room No 125, Dr E Borges Road, Parel, Mumbai, Maharashtra, 400012, India.
| |
Collapse
|
20
|
Rojas JD, Papadopoulou V, Czernuszewicz TJ, Rajamahendiran RM, Chytil A, Chiang YC, Chong DC, Bautch VL, Rathmell WK, Aylward S, Gessner RC, Dayton PA. Ultrasound Measurement of Vascular Density to Evaluate Response to Anti-Angiogenic Therapy in Renal Cell Carcinoma. IEEE Trans Biomed Eng 2018; 66:873-880. [PMID: 30059292 DOI: 10.1109/tbme.2018.2860932] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Functional and molecular changes often precede gross anatomical changes, so early assessment of a tumor's functional and molecular response to therapy can help reduce a patient's exposure to the side effects of ineffective chemotherapeutics or other treatment strategies. OBJECTIVE Our intent was to test the hypothesis that an ultrasound microvascular imaging approach might provide indications of response to therapy prior to assessment of tumor size. METHODS Mice bearing clear-cell renal cell carcinoma xenograft tumors were treated with antiangiogenic and Notch inhibition therapies. An ultrasound measurement of microvascular density was used to serially track the tumor response to therapy. RESULTS Data indicated that ultrasound-derived microvascular density can indicate response to therapy a week prior to changes in tumor volume and is strongly correlated with physiological characteristics of the tumors as measured by histology ([Formula: see text]). Furthermore, data demonstrated that ultrasound measurements of vascular density can determine response to therapy and classify between-treatment groups with high sensitivity and specificity. CONCLUSION/SIGNIFICANCE Results suggests that future applications utilizing ultrasound imaging to monitor tumor response to therapy may be able to provide earlier insight into tumor behavior from metrics of microvascular density rather than anatomical tumor size measurements.
Collapse
|
21
|
Czernuszewicz TJ, Papadopoulou V, Rojas JD, Rajamahendiran RM, Perdomo J, Butler J, Harlacher M, O’Connell G, Zukić D, Aylward SR, Dayton PA, Gessner RC. A new preclinical ultrasound platform for widefield 3D imaging of rodents. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2018; 89:075107. [PMID: 30068108 PMCID: PMC6045495 DOI: 10.1063/1.5026430] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Noninvasive in vivo imaging technologies enable researchers and clinicians to detect the presence of disease and longitudinally study its progression. By revealing anatomical, functional, or molecular changes, imaging tools can provide a near real-time assessment of important biological events. At the preclinical research level, imaging plays an important role by allowing disease mechanisms and potential therapies to be evaluated noninvasively. Because functional and molecular changes often precede gross anatomical changes, there has been a significant amount of research exploring the ability of different imaging modalities to track these aspects of various diseases. Herein, we present a novel robotic preclinical contrast-enhanced ultrasound system and demonstrate its use in evaluating tumors in a rodent model. By leveraging recent advances in ultrasound, this system favorably compares with other modalities, as it can perform anatomical, functional, and molecular imaging and is cost-effective, portable, and high throughput, without using ionizing radiation. Furthermore, this system circumvents many of the limitations of conventional preclinical ultrasound systems, including a limited field-of-view, low throughput, and large user variability.
Collapse
Affiliation(s)
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
| | - Juan D. Rojas
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
| | | | - Jonathan Perdomo
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
| | - James Butler
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
| | - Max Harlacher
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
| | - Graeme O’Connell
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
| | - Dženan Zukić
- Kitware, Inc., Carrboro, North Carolina 27510, USA
| | | | - Paul A. Dayton
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
| | - Ryan C. Gessner
- SonoVol, Inc., Research Triangle Park, North Carolina 27709, USA
- Author to whom correspondence should be addressed: . Current address: First Flight Venture Center, 2 Davis Dr., Research Triangle Park, NC 27709-3169. Telephone: 844-766-6865 x707
| |
Collapse
|
22
|
Fukuda H, Numata K, Hara K, Nozaki A, Kondo M, Chuma M, Nakano M, Nozawa A, Maeda S, Tanaka K. Comparison of vascularity observed using contrast-enhanced 3D ultrasonography and pathological changes in patients with hepatocellular carcinoma after sorafenib treatment. J Cancer 2018; 9:2408-2414. [PMID: 30026837 PMCID: PMC6036707 DOI: 10.7150/jca.24236] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/15/2018] [Indexed: 12/12/2022] Open
Abstract
Aim: The aim of this study was to compare vascularity observed using contrast-enhanced 3D ultrasonography and pathological changes in human hepatocellular carcinoma (HCC) and surrounding non-tumorous areas after sorafenib treatment. Materials and methods: Twelve patients with HCC were enrolled in this clinical study. The maximum tumor diameter as measured using sonography ranged from 15 to 33 mm (mean, 24.0 mm; SD, 5.7 mm). Assessments using contrast-enhanced (0.2 mL of Sonazoid suspension; Daiichi Sankyo, Tokyo, Japan) 3D ultrasonography (LOGIQ 7; GE Healthcare, Milwaukee) were performed in all the patients before and 1 week after sorafenib treatment. The microvessel density (MVD) of the HCC and surrounding non-tumorous area was evaluated based on the immunohistochemical staining of microvessels using an antigen for CD34. Results: Blood flow in the tumor was decreased in all 12 cases after sorafenib treatment. The MVD of the tumorous area at 1 week after sorafenib administration (38.8 ± 5.2) was significantly lower than that observed before sorafenib administration (72.4 ± 13.0) (P < 0.01). Blood flow in the non-tumorous area had decreased in 6 cases at 1 week after sorafenib treatment and had not changed in the 6 other cases. In the reduced blood flow group, the MVD of the non-tumorous area at 1 week after sorafenib administration had decreased significantly, compared with the MVD of the non-tumorous area before sorafenib administration. However, in the group with no change in blood flow, the MVD of the non-tumorous area at 1 week after sorafenib treatment had not changed, compared with the MVD of the non-tumorous area before sorafenib treatment. Conclusion: Contrast-enhanced 3D ultrasonography studies showed a correlation between vascularity and pathological changes in human HCC and the surrounding non-tumorous area after sorafenib treatment.
Collapse
Affiliation(s)
- Hiroyuki Fukuda
- Gastroenterological Center, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, Kanagawa 232-0024, Japan
| | - Kazushi Numata
- Gastroenterological Center, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, Kanagawa 232-0024, Japan
| | - Koji Hara
- Gastroenterological Center, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, Kanagawa 232-0024, Japan
| | - Akito Nozaki
- Gastroenterological Center, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, Kanagawa 232-0024, Japan
| | - Masaaki Kondo
- Gastroenterological Center, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, Kanagawa 232-0024, Japan
| | - Makoto Chuma
- Gastroenterological Center, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, Kanagawa 232-0024, Japan
| | - Masayuki Nakano
- Gastroenterological Center, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, Kanagawa 232-0024, Japan
| | - Akinori Nozawa
- Department of Pathology, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, Kanagawa 232-0024, Japan
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Katsuaki Tanaka
- Gastroenterological Center, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, Kanagawa 232-0024, Japan
| |
Collapse
|
23
|
Contrast-enhanced ultrasonography for assessment of tubular atrophy/interstitial fibrosis in immunoglobulin A nephropathy: a preliminary clinical study. Abdom Radiol (NY) 2018; 43:1423-1431. [PMID: 29110052 DOI: 10.1007/s00261-017-1301-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE To investigate the potential of contrast-enhanced ultrasonography (CEUS) for evaluating the severity of tubular atrophy/interstitial fibrosis (TA/IF) in immunoglobulin A nephropathy (IgAN) patients. MATERIALS AND METHODS A total of 80 patients with IgAN and 33 healthy adults were investigated. Patients were divided into three groups according to the TA/IF (T) grade of the Oxford classification: T0 (n = 28), T1 (n = 35), and T2 (n = 17). Patients and control subjects underwent conventional ultrasound (US) and CEUS. Time-intensity curves of CEUS were drawn for regions of interest located in the renal cortex and medulla using QLab software. Conventional US and CEUS quantitative parameters were analyzed. One-way analysis of variance (ANOVA), binary logistic regression, and receiver operating characteristic (ROC) curves were used. RESULTS There were no significant differences in renal size, cortical thickness, and medullary perfusion parameters (P > 0.05), whereas the differences in peak intensity (PI), area under the time-intensity curve (AUC) and wash-in slope (WIS) of cortical perfusion parameters between the control subjects and patients were significant (P < 0.05). PI was significantly lower with the increasing degree of T (P < 0.05). PI was associated independently with the degree of T in IgAN patients (P < 0.05). ROC analysis revealed that using the optimal cutoff values of 15.38 dB for diagnosis of T0-T1 (sensitivity 83.30% and specificity 63.00%) and 14.69 dB for diagnosis of T2 (sensitivity 100.00% and specificity 66.70%), the corresponding areas under the ROC curve were found to be 0.782 and 0.952, respectively. CONCLUSIONS CEUS can potentially be used as a noninvasive imaging marker to evaluate the severity of TA/IF in IgAN patients.
Collapse
|
24
|
Malmstrøm ML, Săftoiu A, Riis LB, Hassan H, Klausen TW, Rahbek MS, Gögenur I, Vilmann P. Dynamic contrast-enhanced EUS for quantification of tumor perfusion in colonic cancer: a prospective cohort study. Gastrointest Endosc 2018; 87:1530-1538. [PMID: 29329991 DOI: 10.1016/j.gie.2018.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 01/02/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Dynamic contrast-enhanced EUS (CE-EUS) for quantification of perfusion in colonic tumors has not previously been reported in the literature. The aim of this study was to investigate correlations between perfusion parameters and vessel density assessed by immunohistochemical staining with antibodies toward CD31 and CD105. METHODS We conducted a prospective clinical study of 28 patients with left-sided colonic adenocarcinoma who underwent CE-EUS and left-sided hemicolectomy within 2 weeks. CE-EUS recordings were analyzed in 2 regions of interest: the entire tumor and the most enhanced area. Immunohistochemical staining with CD31 and CD105 was performed on tumor tissue sections. The slides were manually scanned for highly vascularized areas, and counting of vessels was performed in hotspots within the tumor and invasive front. New vasculature was assessed by CD105. Associations between CE-EUS and CD31 and CD105 were investigated using Spearman correlation. RESULTS We found significant P values for the correlation between CD31 and rise time (rho = .603 [95% confidence interval (95% CI), .238-.816]; P = .001) in tumor tissue and for the correlation between CD31 and rise time (rho = .50 [95% CI, .201-.695]; P = .008) and fall time (rho = .52 [95% CI, .204-.723]; P = .006) corresponding to the invasive front. We found no correlations between perfusion values evaluated by CE-EUS and CD105. CONCLUSIONS Our results show a significant correlation for vessel density evaluated by CD31 and perfusion parameters evaluated by CE-EUS. This may be the first step toward using real-time CE-EUS for monitoring antiangiogenic therapies in colonic cancer. (Clinical trial registration number: NCT02324023.).
Collapse
Affiliation(s)
- Marie Louise Malmstrøm
- Department of Surgery, Herlev Hospital, University of Copenhagen, Herlev, Denmark; Department of Surgery, Zealand University Hospital, University of Copenhagen, Køge, Denmark
| | - Adrian Săftoiu
- University of Medicine and Pharmacy, Research Centre of Gastroenterology and Hepatology, Craiova, Romania
| | - Lene Buhl Riis
- Department of Pathology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Hazem Hassan
- Department of Surgery, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | | | | | - Ismail Gögenur
- Department of Surgery, Zealand University Hospital, University of Copenhagen, Køge, Denmark
| | - Peter Vilmann
- Department of Surgery, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| |
Collapse
|
25
|
Peil-Grun A, Trenker C, Görg K, Neesse A, Haasenritter J, Görg C. Diagnostic accuracy and interobserver agreement of contrast-enhanced ultrasound in the evaluation of residual lesions after treatment for malignant lymphoma and testicular cancer: a retrospective pilot study in 52 patients. Leuk Lymphoma 2018; 59:2622-2627. [DOI: 10.1080/10428194.2018.1439170] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Anke Peil-Grun
- Interdisciplinary Center of Ultrasound, University Hospital Giessen and Marburg, Marburg, Germany
| | - Corinna Trenker
- Department of Haematology, Oncology and Immunology, University Hospital Giessen and Marburg, Philipps University Marburg, Marburg, Germany
| | - Konrad Görg
- Department of Haematology, Oncology and Immunology, University Hospital Giessen and Marburg, Philipps University Marburg, Marburg, Germany
| | - Albrecht Neesse
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Göttingen, Göttingen, Germany
| | - Jörg Haasenritter
- Department of General Practice and Family Medicine, Philipps University, Marburg, Germany
| | - Christian Görg
- Interdisciplinary Center of Ultrasound, University Hospital Giessen and Marburg, Marburg, Germany
| |
Collapse
|
26
|
Fournier L, Bellucci A, Vano Y, Bouaboula M, Thibault C, Elaidi R, Oudard S, Cuenod C. Imaging Response of Antiangiogenic and Immune-Oncology Drugs in Metastatic Renal Cell Carcinoma (mRCC): Current Status and Future Challenges. KIDNEY CANCER 2017; 1:107-114. [PMID: 30334012 PMCID: PMC6179123 DOI: 10.3233/kca-170011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This report aims to review criteria which have been proposed for treatment evaluation in mRCC under anti-angiogenic and immune-oncologic therapies and discuss future challenges for imagers. RECIST criteria seem to only partially reflect the clinical benefit derived from anti-angiogenic drugs in mRCC. New methods of analysis propose to better evaluate response to these drugs, including a new threshold for size criteria (-10%), attenuation (Choi and modified Choi criteria), functional imaging techniques (perfusion CT, ultrasound or MRI), and new PET radiotracers. Imaging of progression is one of the main future challenges facing imagers. It is progression and not response that will trigger changes in therapy, therefore it is tumour progression that should be identified by imaging techniques to guide the oncologist on the most appropriate time to change therapy. Yet little is known on dynamics of tumour progression, and much data still needs to be accrued to understand it. Finally, as immunotherapies develop, flare or pseudo-progression phenomena are observed. Studies need to be performed to determine whether imaging can distinguish between patients undergoing pseudo-progression for which therapy should be continued, or true progression for which the treatment must be changed.
Collapse
Affiliation(s)
- Laure Fournier
- Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Radiology Department, Paris, France.,Université Paris Descartes Sorbonne Paris Cité, INSERM UMRS970, Paris, France
| | - Alexandre Bellucci
- Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Radiology Department, Paris, France.,Université Paris Descartes Sorbonne Paris Cité, INSERM UMRS970, Paris, France
| | - Yann Vano
- Université Paris Descartes Sorbonne Paris Cité, INSERM UMRS970, Paris, France.,Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Oncology Department, Paris, France
| | - Mehdi Bouaboula
- Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Radiology Department, Paris, France
| | - Constance Thibault
- Université Paris Descartes Sorbonne Paris Cité, INSERM UMRS970, Paris, France.,Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Oncology Department, Paris, France
| | - Reza Elaidi
- ARTIC (Association pour la Recherche sur les Thérapeutique Innovantes en Cancérologie), Paris, France
| | - Stephane Oudard
- Université Paris Descartes Sorbonne Paris Cité, INSERM UMRS970, Paris, France.,Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Oncology Department, Paris, France
| | - Charles Cuenod
- Université Paris Descartes Sorbonne Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Radiology Department, Paris, France.,Université Paris Descartes Sorbonne Paris Cité, INSERM UMRS970, Paris, France
| |
Collapse
|
27
|
Shinagare AB, Krajewski KM, Braschi-Amirfarzan M, Ramaiya NH. Advanced Renal Cell Carcinoma: Role of the Radiologist in the Era of Precision Medicine. Radiology 2017; 284:333-351. [DOI: 10.1148/radiol.2017160343] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Atul B. Shinagare
- From the Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215; and Department of Radiology, Brigham and Women’s Hospital, Boston, Mass
| | - Katherine M. Krajewski
- From the Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215; and Department of Radiology, Brigham and Women’s Hospital, Boston, Mass
| | - Marta Braschi-Amirfarzan
- From the Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215; and Department of Radiology, Brigham and Women’s Hospital, Boston, Mass
| | - Nikhil H. Ramaiya
- From the Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215; and Department of Radiology, Brigham and Women’s Hospital, Boston, Mass
| |
Collapse
|
28
|
Eschbach RS, Clevert DA, Hirner-Eppeneder H, Ingrisch M, Moser M, Schuster J, Tadros D, Schneider M, Kazmierczak PM, Reiser M, Cyran CC. Contrast-Enhanced Ultrasound with VEGFR2-Targeted Microbubbles for Monitoring Regorafenib Therapy Effects in Experimental Colorectal Adenocarcinomas in Rats with DCE-MRI and Immunohistochemical Validation. PLoS One 2017; 12:e0169323. [PMID: 28060884 PMCID: PMC5217974 DOI: 10.1371/journal.pone.0169323] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/15/2016] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES To investigate contrast-enhanced ultrasound (CEUS) with VEGFR2-targeted microbubbles for monitoring therapy effects of regorafenib on experimental colon carcinomas in rats with correlation to dynamic contrast-enhanced MRI (DCE-MRI) and immunohistochemistry. MATERIALS AND METHODS Human colorectal adenocarcinoma xenografts (HT-29) were implanted subcutaneously in n = 21 (n = 11 therapy group; n = 10 control group) female athymic nude rats (Hsd: RH-Foxn1rnu). Animals were imaged at baseline and after a one-week daily treatment with regorafenib or a placebo (10 mg/kg bodyweight), using CEUS with VEGFR2-targeted microbubbles and DCE-MRI. In CEUS tumor perfusion was assessed during an early vascular phase (wash-in area under the curve = WiAUC) and VEGFR2-specific binding during a late molecular phase (signal intensity after 8 (SI8min) and 10 minutes (SI10min)), using a conventional 15L8 linear transducer (transmit frequency 7 MHz, dynamic range 80 dB, depth 25 mm). In DCE-MRI functional parameters plasma flow (PF) and plasma volume (PV) were quantified. For validation purposes, CEUS parameters were correlated with DCE-MRI parameters and immunohistochemical VEGFR2, CD31, Ki-67 and TUNEL stainings. RESULTS CEUS perfusion parameter WiAUC decreased significantly (116,989 ± 77,048 a.u. to 30,076 ± 27,095a.u.; p = 0.005) under therapy with no significant changes (133,932 ± 65,960 a.u. to 84,316 ± 74,144 a.u.; p = 0.093) in the control group. In the therapy group, the amount of bound microbubbles in the late phase was significantly lower in the therapy than in the control group on day 7 (SI8min: 283 ± 191 vs. 802 ± 460 a.u.; p = 0.006); SI10min: 226 ± 149 vs. 645 ± 461 a.u.; p = 0.009). PF and PV decreased significantly (PF: 147 ± 58 mL/100 mL/min to 71 ± 15 mL/100 mL/min; p = 0.003; PV: 13 ± 3% to 9 ± 4%; p = 0.040) in the therapy group. Immunohistochemistry revealed significantly fewer VEGFR2 (7.2 ± 1.8 vs. 17.8 ± 4.6; p < 0.001), CD31 (8.1 ± 3.0 vs. 20.8 ± 5.7; p < 0.001) and Ki-67 (318.7 ± 94.0 vs. 468.0 ± 133.8; p = 0.004) and significantly more TUNEL (672.7 ± 194.0 vs. 357.6 ± 192.0; p = 0.003) positive cells in the therapy group. CEUS parameters showed significant (p < 0.05) correlations to DCE-MRI parameters and immunohistochemistry. CONCLUSIONS CEUS with VEGFR2-targeted microbubbles allowed for monitoring regorafenib functional and molecular therapy effects on experimental colorectal adenocarcinomas with a significant decline of CEUS and DCE-MRI perfusion parameters as well as a significant reduction of specifically bound microbubbles under therapy, consistent with a reduced expression of VEGFR2.
Collapse
Affiliation(s)
- Ralf Stefan Eschbach
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
- * E-mail:
| | - Dirk-Andre Clevert
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Heidrun Hirner-Eppeneder
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Michael Ingrisch
- Josef Lissner Laboratory for Biomedical Imaging, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Matthias Moser
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Jessica Schuster
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Dina Tadros
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Moritz Schneider
- Josef Lissner Laboratory for Biomedical Imaging, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Philipp Maximilian Kazmierczak
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Maximilian Reiser
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Clemens C. Cyran
- Laboratory for Experimental Radiology, Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital, Munich, Germany
| |
Collapse
|
29
|
Smith AD, Zhang X, Bryan J, Souza F, Roda M, Sirous R, Zhang H, Vasanji A, Griswold M. Vascular Tumor Burden as a New Quantitative CT Biomarker for Predicting Metastatic RCC Response to Antiangiogenic Therapy. Radiology 2016; 281:484-498. [DOI: 10.1148/radiol.2016160143] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
30
|
Turco S, Wijkstra H, Mischi M. Mathematical Models of Contrast Transport Kinetics for Cancer Diagnostic Imaging: A Review. IEEE Rev Biomed Eng 2016; 9:121-47. [PMID: 27337725 DOI: 10.1109/rbme.2016.2583541] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Angiogenesis plays a fundamental role in cancer growth and the formation of metastasis. Novel cancer therapies aimed at inhibiting angiogenic processes and/or disrupting angiogenic tumor vasculature are currently being developed and clinically tested. The need for earlier and improved cancer diagnosis, and for early evaluation and monitoring of therapeutic response to angiogenic treatment, have led to the development of several imaging methods for in vivo noninvasive assessment of angiogenesis. The combination of dynamic contrast-enhanced imaging with mathematical modeling of the contrast agent kinetics enables quantitative assessment of the structural and functional changes in the microvasculature that are associated with tumor angiogenesis. In this paper, we review quantitative imaging of angiogenesis with dynamic contrast-enhanced magnetic resonance imaging, computed tomography, and ultrasound.
Collapse
|
31
|
Lo GM, Al Zahrani H, Jang HJ, Menezes R, Hudson J, Burns P, McNamara MG, Kandel S, Khalili K, Knox J, Rogalla P, Kim TK. Detection of Early Tumor Response to Axitinib in Advanced Hepatocellular Carcinoma by Dynamic Contrast Enhanced Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:1303-1311. [PMID: 27033332 DOI: 10.1016/j.ultrasmedbio.2016.01.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/27/2016] [Accepted: 01/30/2016] [Indexed: 06/05/2023]
Abstract
This study aimed to evaluate the utility of dynamic contrast-enhanced ultrasound (DCE-US) in measuring early tumor response of advanced hepatocellular carcinoma to axitinib. Twenty patients were enrolled (aged 18-78 y; median 65). DCE-US was performed with bolus injection and infusion/disruption replenishment. Median overall survival was 7.1 mo (1.8-27.3) and progression free survival was 3.6 mo (1.8-17.4). Fifteen patients completed infusion scans and 12 completed bolus scans at 2 wk. Among the perfusion parameters, fractional blood volume at infusion (INFBV) decreased at 2 wk in 10/15 (16%-81% of baseline, mean 47%) and increased in 5/15 (116%-535%, mean 220%). This was not significantly associated with progression free survival (p = 0.310) or progression at 16 wk (p = 0.849), but was borderline statistically significant (p = 0.050) with overall survival, limited by a small sample size. DCE-US is potentially useful in measuring early tumor response of advanced hepatocellular carcinoma to axitinib, but a larger trial is needed.
Collapse
Affiliation(s)
- Glen M Lo
- Medical Imaging, University of Toronto, Toronto, ON, Canada; Department of Radiology, Sir Charles Gairdner Hospital, QEII Medical Centre, Perth, Western Australia
| | | | - Hyun Jung Jang
- Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Ravi Menezes
- Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - John Hudson
- Department of Medical Biophysics, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Peter Burns
- Department of Medical Biophysics, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Mairéad G McNamara
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada; Department of Medical Oncology, The Christie NHS Foundation Trust/University of Manchester, Institute of Cancer Sciences, Manchester, UK
| | - Sonja Kandel
- Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Korosh Khalili
- Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Jennifer Knox
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Patrik Rogalla
- Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Tae Kyoung Kim
- Medical Imaging, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
32
|
Sirous R, Henegan JC, Zhang X, Howard CM, Souza F, Smith AD. Metastatic renal cell carcinoma imaging evaluation in the era of anti-angiogenic therapies. Abdom Radiol (NY) 2016; 41:1086-99. [PMID: 27193601 DOI: 10.1007/s00261-016-0742-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During the last decade, the arsenal of anti-angiogenic (AAG) agents used to treat metastatic renal cell carcinoma (RCC) has grown and revolutionized the treatment of metastatic RCC, leading to improved overall survival compared to conventional chemotherapy and traditional immunotherapy agents. AAG agents include inhibitors of vascular endothelial growth factor receptor signaling pathways and mammalian target of rapamycin inhibitors. Both of these classes of targeted agents are considered cytostatic rather than cytotoxic, inducing tumor stabilization rather than marked tumor shrinkage. As a result, decreases in tumor size alone are often minimal and/or occur late in the course of successful AAG therapy, while tumor devascularization is a distinct feature of AAG therapy. In successful AAG therapy, tumor devascularization manifests on computed tomography images as a composite of a decrease in tumor size, a decrease in tumor attenuation, and the development of tumor necrosis. In this article, we review Response Evaluation Criteria in Solid Tumors (RECIST)-the current standard of care for tumor treatment response assessment which is based merely on changes in tumor length-and its assessment of metastatic RCC tumor response in the era of AAG therapies. We then review the features of an ideal tumor imaging biomarker for predicting metastatic RCC response to a particular AAG agent and serving as a longitudinal tumor response assessment tool. Finally, a discussion of the more recently proposed imaging response criteria and new imaging trends in metastatic RCC response assessment will be reviewed.
Collapse
Affiliation(s)
- Reza Sirous
- Department of Radiology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - John C Henegan
- Department of Hematology/Oncology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Xu Zhang
- Center for Biostatistics and Bioinformatics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Candace M Howard
- Department of Radiology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Frederico Souza
- Department of Radiology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Andrew D Smith
- Department of Radiology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
33
|
Cidon EU, Alonso P, Masters B. Markers of Response to Antiangiogenic Therapies in Colorectal Cancer: Where Are We Now and What Should Be Next? Clin Med Insights Oncol 2016; 10:41-55. [PMID: 27147901 PMCID: PMC4849423 DOI: 10.4137/cmo.s34542] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/15/2016] [Accepted: 03/13/2016] [Indexed: 12/17/2022] Open
Abstract
Despite advances in the treatment of colorectal cancer (CRC), it remains the second most common cause of cancer-related death in the Western world. Angiogenesis is a complex process that involves the formation of new blood vessels from preexisting vessels. It is essential for promoting cancer survival, growth, and dissemination. The inhibition of angiogenesis has been shown to prevent tumor progression experimentally, and several chemotherapeutic targets of tumor angiogenesis have been identified. These include anti-vascular endothelial growth factor (VEGF) treatments, such as bevacizumab (a VEGF-specific binding antibody) and anti-VEGF receptor tyrosine kinase inhibitors, although antiangiogenic therapy has been shown to be effective in the treatment of several cancers, including CRC. However, it is also associated with its own side effects and financial costs. Therefore, the identification of biomarkers that are able to identify patients who are more likely to benefit from antiangiogenic treatment is very important. This article intends to be a concise summary of the potential biomarkers that can predict or prognosticate the benefit of antiangiogenic treatments in CRC, and also what we can expect in the near future.
Collapse
Affiliation(s)
- E. Una Cidon
- Department of Medical Oncology, Royal Bournemouth Hospital NHS Foundation Trust, Bournemouth, UK
| | - P. Alonso
- Department of Clinical Oncology, Clinical University Hospital, Valladolid, Spain
| | - B. Masters
- Department of Oncology, Nottingham City Hospital, Nottingham, UK
| |
Collapse
|
34
|
Use of Quantitative Dynamic Contrast-Enhanced Ultrasound to Assess Response to Antiangiogenic Therapy in Children and Adolescents With Solid Malignancies: A Pilot Study. AJR Am J Roentgenol 2016; 206:933-9. [PMID: 26999488 DOI: 10.2214/ajr.15.15789] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The purpose of this study was to investigate contrast-enhanced ultrasound assessment of tumor response to antiangiogenic therapy in children and adolescents with solid malignancies. SUBJECTS AND METHODS Children with recurrent solid tumors who were enrolled in an institutional phase 1 study of antiangiogenic therapy underwent contrast-enhanced ultrasound of target lesions before therapy, on therapy days 3 and 7, and at the end of course 1. Acoustic data from target lesion ROIs were used to measure peak enhancement, time to peak, rate of enhancement, total AUC, AUC during wash-in (AUC1), and AUC during washout (AUC2). The Cox regression model was used to assess the association between changes in parameters from baseline to follow-up time points and time to tumor progression. Values of p ≤ 0.050 were considered significant. RESULTS Target lesion sites included liver (n = 3), pleura (n = 2), and supraclavicular mass, soft-tissue component of bone metastasis, lung, retroperitoneum, peritoneum, lymph node, muscle mass, and perineum (n = 1 each). Hazard ratios for changes from baseline to end of course 1 for peak enhancement (1.17, p = 0.034), rate of enhancement (3.25, p = 0.029), and AUC1 (1.02, p = 0.040) were significantly associated with time to progression. Greater decreases in these parameters correlated with longer time to progression. CONCLUSION Contrast-enhanced ultrasound measurements of tumor peak enhancement, rate of enhancement, and AUC1 were early predictors of time to progression in a cohort of children and adolescents with recurrent solid tumors treated with antiangiogenic therapy. Further investigation of these findings in a larger population is warranted.
Collapse
|
35
|
Flaherty KT, Hamilton BK, Rosen MA, Amaravadi RK, Schuchter LM, Gallagher M, Chen H, Sehgal C, O'Dwyer PJ. Phase I/II Trial of Imatinib and Bevacizumab in Patients With Advanced Melanoma and Other Advanced Cancers. Oncologist 2015; 20:952-9. [PMID: 26084808 DOI: 10.1634/theoncologist.2015-0108] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/21/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor and platelet-derived growth factor signaling in the tumor microenvironment appear to cooperate in promoting tumor angiogenesis. PATIENTS AND METHODS We conducted a phase I trial combining bevacizumab (i.v. every 2 weeks) and imatinib (oral daily). Once a recommended phase II dose combination was established, a phase II trial was initiated in patients with metastatic melanoma. A Simon 2-stage design was used with 23 patients required in the first stage and 41 patients in total should the criteria to proceed be met. We required that 50% of the patients be progression-free at 16 weeks. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and power Doppler ultrasonography were performed in patients with metastatic tumors amenable to imaging with these methods at baseline and after 4 weeks. RESULTS A total of 17 patients were accrued to 4 dose and combination levels. Bevacizumab 10 mg/kg every 2 weeks could be safely combined with imatinib 800 mg daily. Common toxicities included fatigue, nausea, vomiting, edema, proteinuria, and anemia, but were not commonly severe. A total of 23 patients with metastatic melanoma (48% with American Joint Commission on Cancer stage M1c; median age, 63 years) were enrolled in the first stage of phase II. The 16-week progression-free survival rate was 35%, leading to termination of phase II after the first stage. In the small subset of patients who remained on study with lesions evaluable by DCE-MRI, significant decreases in tumor vascular permeability were noted, despite early disease progression using the Response Evaluation Criteria In Solid Tumors. CONCLUSION Bevacizumab and imatinib can be safely combined at the maximum doses used for each agent. We did not observe significant clinical activity with this regimen in melanoma patients.
Collapse
Affiliation(s)
- Keith T Flaherty
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Betty K Hamilton
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark A Rosen
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Ravi K Amaravadi
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Lynn M Schuchter
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Maryann Gallagher
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Helen Chen
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Chandra Sehgal
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Peter J O'Dwyer
- Developmental Therapeutics Program, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
36
|
Dynamic contrast-enhanced computed tomography as a potential biomarker in patients with metastatic renal cell carcinoma: preliminary results from the Danish Renal Cancer Group Study-1. Invest Radiol 2015; 49:601-7. [PMID: 24691140 DOI: 10.1097/rli.0000000000000058] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVES The aim of this study was to explore the impact of dynamic contrast-enhanced (DCE) computer tomography (CT) as a biomarker in metastatic renal cell carcinoma (mRCC). MATERIALS AND METHODS Twelve patients with favorable or intermediate Memorial Sloan Kettering Cancer Center risk group and clear cell mRCC participating in an ongoing prospective randomized phase II trial comprising interleukin-2-based immunotherapy and bevacizumab were included in this preliminary analysis. All patients had a follow-up time of at least 2 years. Interpretation of DCE-CT (max slope method) was performed blinded to treatment group. The DCE-CT scans were performed at baseline, at weeks 5 and 10, and thereafter every third month. Blood flow (BF; mL/min/100 mL), peak enhancement (Hounsfield units), time to peak (seconds), and blood volume (BV; mL/100 g) were calculated. Parameters for DCE-CT were correlated with sum of diameters (defined by Response Evaluation Criteria in Solid Tumors 1.1), progression-free survival (PFS), and overall survival (OS) using Wilcoxon, Man-Whitney, Kaplan-Meier, and log rank statistics, as appropriate. RESULTS Blood flow at baseline ranged from 4.9 to 148.1 mL/min/100 mL (median, 62.2; 25th percentile, 25.8; 75th percentile, 110.0). Patients with high baseline BF (using quartiles as cutoffs) had significantly longer OS (not reached vs 5.2 months, P = 0.011) and longer PFS (not reached vs 3.9 months, P = 0.026). Blood volume at baseline ranged from 8.8 to 74.1 mL/100 g tissue (median, 21.5), and at week 5, from 4.9 to 34.7 mL/100 g (median, 17.2). Relative changes in BV between baseline and week 5 ranged from -64% to +68% (median, -16%; 25th percentile, -41%; 75th percentile, +2%) and were significantly associated with OS using quartiles as cutoffs (5.2 months vs not reached, P = 0.038) and PFS using the median as cutoff (5.3 months vs not reached, P = 0.009), with larger reductions associated with longer survival. Using medians as cutoffs, relative changes in both BF and BV between baseline and week 10 were significantly associated with OS (for both, 8.6 months vs not reached, P = 0.031). CONCLUSIONS Dynamic contrast-enhanced CT is a potential biomarker in patients with mRCC. High baseline BF and reductions in BF and BV during early treatment are associated with improved outcome. Large-scale studies are required.
Collapse
|
37
|
Fröhlich E, Muller R, Cui XW, Schreiber-Dietrich D, Dietrich CF. Dynamic contrast-enhanced ultrasound for quantification of tissue perfusion. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2015; 34:179-196. [PMID: 25614391 DOI: 10.7863/ultra.34.2.179] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 05/13/2014] [Indexed: 05/23/2023]
Abstract
Dynamic contrast-enhanced ultrasound (US) imaging, a technique that uses microbubble contrast agents with diagnostic US, has recently been technically summarized and reviewed by a European Federation of Societies for Ultrasound in Medicine and Biology position paper. However, the practical applications of this imaging technique were not included. This article reviews and discusses the published literature on the clinical use of dynamic contrast-enhanced US. This review finds that dynamic contrast-enhanced US imaging is the most sensitive cross-sectional real-time method for measuring the perfusion of parenchymatous organs noninvasively. It can measure parenchymal perfusion and therefore can differentiate between benign and malignant tumors. The most important routine clinical role of dynamic contrast-enhanced US is the prediction of tumor responses to chemotherapy within a very short time, shorter than using Response Evaluation Criteria in Solid Tumors criteria. Other applications found include quantifying the hepatic transit time, diabetic kidneys, transplant grafts, and Crohn disease. In addition, the problems involved in using dynamic contrast-enhanced US are discussed.
Collapse
Affiliation(s)
- Eckhart Fröhlich
- Department of Internal Medicine I, Karl-Olga-Krankenhaus Stuttgart, Academic Teaching Hospital of the University of Ulm, Germany (E.F.); Tropical Health Solutions Pty, Ltd, and Anton-Breinl Center, James Cook University, Townsville City, Queensland, Australia (R.M.); Sino-German Research Center of Ultrasound in Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China, and Department of Internal Medicine II, Caritas-Krankenhaus Bad Mergentheim, Academic Teaching Hospital of the University of Würzburg, Bad Mergentheim, Germany (X.-W.C., D.S.-D., C.F.D.)
| | - Reinhold Muller
- Department of Internal Medicine I, Karl-Olga-Krankenhaus Stuttgart, Academic Teaching Hospital of the University of Ulm, Germany (E.F.); Tropical Health Solutions Pty, Ltd, and Anton-Breinl Center, James Cook University, Townsville City, Queensland, Australia (R.M.); Sino-German Research Center of Ultrasound in Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China, and Department of Internal Medicine II, Caritas-Krankenhaus Bad Mergentheim, Academic Teaching Hospital of the University of Würzburg, Bad Mergentheim, Germany (X.-W.C., D.S.-D., C.F.D.)
| | - Xin-Wu Cui
- Department of Internal Medicine I, Karl-Olga-Krankenhaus Stuttgart, Academic Teaching Hospital of the University of Ulm, Germany (E.F.); Tropical Health Solutions Pty, Ltd, and Anton-Breinl Center, James Cook University, Townsville City, Queensland, Australia (R.M.); Sino-German Research Center of Ultrasound in Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China, and Department of Internal Medicine II, Caritas-Krankenhaus Bad Mergentheim, Academic Teaching Hospital of the University of Würzburg, Bad Mergentheim, Germany (X.-W.C., D.S.-D., C.F.D.)
| | - Dagmar Schreiber-Dietrich
- Department of Internal Medicine I, Karl-Olga-Krankenhaus Stuttgart, Academic Teaching Hospital of the University of Ulm, Germany (E.F.); Tropical Health Solutions Pty, Ltd, and Anton-Breinl Center, James Cook University, Townsville City, Queensland, Australia (R.M.); Sino-German Research Center of Ultrasound in Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China, and Department of Internal Medicine II, Caritas-Krankenhaus Bad Mergentheim, Academic Teaching Hospital of the University of Würzburg, Bad Mergentheim, Germany (X.-W.C., D.S.-D., C.F.D.)
| | - Christoph F Dietrich
- Department of Internal Medicine I, Karl-Olga-Krankenhaus Stuttgart, Academic Teaching Hospital of the University of Ulm, Germany (E.F.); Tropical Health Solutions Pty, Ltd, and Anton-Breinl Center, James Cook University, Townsville City, Queensland, Australia (R.M.); Sino-German Research Center of Ultrasound in Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China, and Department of Internal Medicine II, Caritas-Krankenhaus Bad Mergentheim, Academic Teaching Hospital of the University of Würzburg, Bad Mergentheim, Germany (X.-W.C., D.S.-D., C.F.D.).
| |
Collapse
|
38
|
Depth of remission is a prognostic factor for survival in patients with metastatic renal cell carcinoma. Eur Urol 2015; 67:952-8. [PMID: 25577718 DOI: 10.1016/j.eururo.2014.12.036] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 12/22/2014] [Indexed: 11/23/2022]
Abstract
BACKGROUND Response remains an important endpoint in clinical cancer trials. However, the prognostic utility of best tumor response in metastatic renal cell carcinoma (mRCC) remains vague. OBJECTIVE To define the prognostic relevance of the depth of remission in mRCC. DESIGN, SETTING, AND PARTICIPANTS Pooled data from the Pfizer database for 2749 patients from phase 2 and 3 clinical trials in mRCC were analyzed. Tumor shrinkage was categorized according to the best percentage change in the sum of the largest diameter of target lesions. Outcome was computed using Kaplan-Meier curves and correlation was assessed via Cox regression, including a 6-mo landmark. INTERVENTION Sunitinib, sorafenib, axitinib, temsirolimus, or temsirolimus and interferon-α. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Categorized tumor shrinkage, overall survival (OS), progression free survival (PFS). RESULTS AND LIMITATIONS Major tumor shrinkage of 60% or more occurred in approximately 10% of patients and was associated with median OS of 54.5 mo. OS expectations steadily decreased with depth of remission (26.4, 16.6, 10.4, and 7.3 mo). The association was maintained when stratified by type of therapy, line of therapy, and performance status. Cox proportional regression analyses for the 6-mo landmark confirmed the prognostic relevance of major tumor shrinkage (hazard ratio 0.29, 95% confidence interval 0.22-0.39; p<0.001). The major limitation of our study is the variability of imaging intervals among studies. CONCLUSIONS This is the first and largest analysis of best tumor response in mRCC. We demonstrate that depth of remission is an independent prognostic factor in mRCC. PATIENT SUMMARY It remains unknown whether tumor shrinkage during therapy is needed to achieve clinical activity in metastatic renal cell carcinoma. Our analysis shows that the magnitude of tumor shrinkage correlates with better survival in patients. This observation may be used as a clinical research tool in future trials. TRIAL REGISTRATION NCT00054886, NCT00077974, NCT00267748, NCT00338884, NCT00137423, NCT00083889, NCT00065468, NCT00678392.
Collapse
|
39
|
McNamara MG, Le LW, Horgan AM, Aspinall A, Burak KW, Dhani N, Chen E, Sinaei M, Lo G, Kim TK, Rogalla P, Bathe OF, Knox JJ. A phase II trial of second-line axitinib following prior antiangiogenic therapy in advanced hepatocellular carcinoma. Cancer 2015; 121:1620-7. [PMID: 25565269 DOI: 10.1002/cncr.29227] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 11/07/2014] [Accepted: 11/25/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Second-line treatment options in advanced hepatocellular carcinoma (HCC) are limited. Axitinib, a selective potent tyrosine kinase inhibitor (TKI) of vascular endothelial growth factor VEGF) receptors 1, 2, and 3, merits exploration in HCC. METHODS This was a single-arm phase II trial of axitinib in advanced HCC. Eligible patients were Child-Pugh A/B7, with measurable progressive disease after TKIs/antiangiogenic drugs. Axitinib was started at 5 mg twice daily orally, titrated from 2 to 10 mg twice daily as tolerated. The primary end point was tumor control at 16 weeks by RECIST1.1; secondary end points were response rate, comparing response by RECIST1.1 to Choi and modified RECIST, exploring dynamic contrast-enhanced imaging models, safety, progression-free (PFS), and overall survival (OS). RESULTS Thirty patients were treated. Of 26 patients evaluable for response, there were 3 partial responses (PR) per RECIST1.1; 13 PR by Choi, 6 PR and 1 complete response by modified RECIST. Tumor control rate at 16 weeks was 42.3%. Two-week perfusion changes were noted on functional imaging. Of 21 patients with evaluable α-fetoprotein response, 43% had >50% decrease from baseline. Most common axitinib-related grade 3/4 adverse events (AEs) were hypertension, thrombocytopenia and diarrhea. Of 11 patients with any grade hypertension, 7 had disease control >36 wks. Four patients discontinued treatment due to AEs. Median PFS was 3.6 months. Median OS was 7.1 months. CONCLUSIONS With 42.3% tumor control at 16 weeks, primary endpoint was met. Axitinib has shown encouraging tolerable clinical activity in VEGF-pretreated HCC patients but further study should be in a selected population incorporating potential biomarkers of response.
Collapse
Affiliation(s)
- Mairéad G McNamara
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; The Christie NHS Foundation Trust/University of Manchester, Withington, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lassau N, Bonastre J, Kind M, Vilgrain V, Lacroix J, Cuinet M, Taieb S, Aziza R, Sarran A, Labbe-Devilliers C, Gallix B, Lucidarme O, Ptak Y, Rocher L, Caquot LM, Chagnon S, Marion D, Luciani A, Feutray S, Uzan-Augui J, Coiffier B, Benastou B, Koscielny S. Validation of dynamic contrast-enhanced ultrasound in predicting outcomes of antiangiogenic therapy for solid tumors: the French multicenter support for innovative and expensive techniques study. Invest Radiol 2014; 49:794-800. [PMID: 24991866 PMCID: PMC4222794 DOI: 10.1097/rli.0000000000000085] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Dynamic contrast-enhanced ultrasound (DCE-US) has been used in single-center studies to evaluate tumor response to antiangiogenic treatments: the change of area under the perfusion curve (AUC), a criterion linked to blood volume, was consistently correlated with the Response Evaluation Criteria in Solid Tumors response. The main objective here was to do a multicentric validation of the use of DCE-US to evaluate tumor response in different solid tumor types treated by several antiangiogenic agents. A secondary objective was to evaluate the costs of the procedure. MATERIALS AND METHODS This prospective study included patients from 2007 to 2010 in 19 centers (8 teaching hospitals and 11 comprehensive cancer centers). All patients treated with antiangiogenic therapy were eligible. Dynamic contrast-enhanced ultrasound examinations were performed at baseline as well as on days 7, 15, 30, and 60. For each examination, a perfusion curve was recorded during 3 minutes after injection of a contrast agent. Change from baseline at each time point was estimated for each of 7 fitted criteria. The main end point was freedom from progression (FFP). Criterion/time-point combinations with the strongest correlation with FFP were analyzed further to estimate an optimal cutoff point. RESULTS A total of 1968 DCE-US examinations in 539 patients were analyzed. The median follow-up was 1.65 years. Variations from baseline were significant at day 30 for several criteria, with AUC having the most significant association with FFP (P = 0.00002). Patients with a greater than 40% decrease in AUC at day 30 had better FFP (P = 0.005) and overall survival (P = 0.05). The mean cost of each DCE-US was 180&OV0556;, which corresponds to $250 using the current exchange rate. CONCLUSIONS Dynamic contrast-enhanced ultrasound is a new functional imaging technique that provides a validated criterion, namely, the change of AUC from baseline to day 30, which is predictive of tumor progression in a large multicenter cohort. Because of its low cost, it should be considered in the routine evaluation of solid tumors treated with antiangiogenic therapy.
Collapse
Affiliation(s)
- Nathalie Lassau
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Julia Bonastre
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Michèle Kind
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Valérie Vilgrain
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Joëlle Lacroix
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Marie Cuinet
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Sophie Taieb
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Richard Aziza
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Antony Sarran
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Catherine Labbe-Devilliers
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Benoit Gallix
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Olivier Lucidarme
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Yvette Ptak
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Laurence Rocher
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Louis-Michel Caquot
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Sophie Chagnon
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Denis Marion
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Alain Luciani
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Sylvaine Feutray
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Joëlle Uzan-Augui
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Benedicte Coiffier
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Baya Benastou
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| | - Serge Koscielny
- From the *Integrated Research Cancer Institute, Research Department, Villejuif; †Service Biostatistique et Épidémiologie, Gustave Roussy, Villejuif; ‡Imaging Department, Institut Bergonié, Bordeaux; §Department of Radiology, Assistance Publique-Hôpitaux de Paris, Hôpital Beaujon, Clichy, and Université Paris Diderot, Sorbonne Paris Cité; ∥Department of Radiology, Centre François Baclesse, Caen; ¶Department of Radiology, Centre Léon Bérard, Lyon; #Imaging Department, Centre Oscar Lambret, Lille; **Radiodiagnostics Department, Centre Claudius Regaud, Toulouse; ††Imaging Department, Institut Paoli Calmettes, Marseille; ‡‡Radiodiagnostics Department, Centre R Gauducheau, Institut de Cancérologie de l’Ouest Nantes; §§Department of Abdominal and Digestive Imaging, Hôpital Saint-Éloi, Montpellier; ∥∥Radiology Department, Centre Hospitalier Universitaire La Pitié-Salpêtrière, Paris; ¶¶Radiodiagnostics Department, Centre Jean Perrin, Clermont-Ferrand; ##Radiology Department, Centre Hospitalier Universitaire Bicêtre, Le Kremlin-Bicêtre; ***Radiodiagnostics and Imaging Department, Institut Jean Godinot, Reims; †††Ultrasonography Department, Hôpital Ambroise Paré, Boulogne-Billancourt; ‡‡‡Radiology Department, Centre Hospitalier Universitaire Hôtel-Dieu, Lyon; §§§Radiology Department, Centre Hospitalier Universitaire Henri Mondor, Créteil; ∥∥∥Imaging Department, Centre Georges-François Leclerc, Dijon Cedex; and ¶¶¶Radiology Department, Hôpital Cochin, Paris, France
| |
Collapse
|
41
|
De Marchi A, Prever EBD, Cavallo F, Pozza S, Linari A, Lombardo P, Comandone A, Piana R, Faletti C. Perfusion pattern and time of vascularisation with CEUS increase accuracy in differentiating between benign and malignant tumours in 216 musculoskeletal soft tissue masses. Eur J Radiol 2014; 84:142-150. [PMID: 25454097 DOI: 10.1016/j.ejrad.2014.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 09/24/2014] [Accepted: 10/03/2014] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Musculoskeletal Soft Tissue Tumours (STT) are frequent heterogeneous lesions. Guidelines consider a mass larger than 5 cm and deep with respect to the deep fascia potentially malignant. Contrast Enhanced Ultrasound (CEUS) can detect both vascularity and tumour neoangiogenesis. We hypothesised that perfusion patterns and vascularisation time could improve the accuracy of CEUS in discriminating malignant tumours from benign lesions. MATERIALS AND METHODS 216 STT were studied: 40% benign lesions, 60% malignant tumours, 56% in the lower limbs. Seven CEUS perfusion patterns and three types of vascularisation (arterial-venous uptake, absence of uptake) were applied. Accuracy was evaluated by comparing imaging with the histological diagnosis. Univariate and multivariate analysis, Chi-square test and t-test for independent variables were applied; significance was set at p<0.05 level, 95% computed CI. RESULTS CEUS pattern 6 (inhomogeneous perfusion), arterial uptake and location in the lower limb were associated with high risk of malignancy. CEUS pattern has PPV 77%, rapidity of vascularisation PPV 69%; location in the limbs is the most sensitive indicator, but NPV 52%, PPV 65%. The combination of CEUS-pattern and vascularisation has 74% PPV, 60% NPV, 70% sensitivity. No correlation with size and location in relation to the deep fascia was found. CONCLUSION US with CEUS qualitative analysis could be an accurate technique to identify potentially malignant STT, for which second line imaging and biopsy are indicated in Referral Centers. Intense inhomogeneous enhancement with avascular areas and rapid vascularisation time could be useful in discriminating benign from malignant SST, overall when the lower limbs are involved.
Collapse
Affiliation(s)
- Armanda De Marchi
- Department of Imaging, Azienda Ospedaliera Città della Salute e della Scienza, CTO Hospital, Via Zuretti 29, 10126 Torino, Italy.
| | - Elena Brach Del Prever
- Department of OrthopaedicOncology and ReconstructiveSurgery, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, CTO Hospital, Via Zuretti 29, 10126 Torino, Italy.
| | - Franco Cavallo
- Department of Public health and Paediatrics, University of Turin, Via Santena 5-bis, 10126 Torino, Italy.
| | - Simona Pozza
- Department of Imaging, Azienda Ospedaliera Città della Salute e della Scienza, CTO Hospital, Via Zuretti 29, 10126 Torino, Italy.
| | - Alessandra Linari
- Department of Pathology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Regina Margherita Hospital, Piazza Polonia, 10126 Torino, Italy.
| | - Paolo Lombardo
- Department of DiagnosticImaging and Radiotherapy of the University of Turin, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Via Genova 3, 10126 Torino, Italy.
| | - Alessandro Comandone
- Department of Oncology, Gradenigo Hospital, Corso Regina Margherita, 8/10.10153 Torino, Italy.
| | - Raimondo Piana
- Department of OrthopaedicOncology and ReconstructiveSurgery, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, CTO Hospital, Via Zuretti 29, 10126 Torino, Italy.
| | - Carlo Faletti
- Department of Imaging, Azienda Ospedaliera Città della Salute e della Scienza, CTO Hospital, Via Zuretti 29, 10126 Torino, Italy. falettic.@hotmail.it
| |
Collapse
|
42
|
van der Mijn JC, Mier JW, Broxterman HJ, Verheul HM. Predictive biomarkers in renal cell cancer: insights in drug resistance mechanisms. Drug Resist Updat 2014; 17:77-88. [PMID: 25457974 DOI: 10.1016/j.drup.2014.10.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION VEGF-targeted therapy is currently the first line treatment for patients with metastatic clear cell renal cell carcinoma (ccRCC), but most patients either display primary (intrinsic) resistance or acquire drug resistance. In recent years multiple mechanisms of resistance to VEGF-targeted therapy emerged from preclinical research, but it is currently unknown to what extent these drug resistance modalities play a role in the clinic. Here we reviewed the current literature on biomarkers that predict treatment outcome in patients with ccRCC to gain insight in clinical drug resistance mechanisms. METHODS A search syntax was compiled by combining different synonyms of "biomarker" AND "renal" AND "cancer". MEDLINE was accessed through PubMed, where this syntax was entered and used to search titles and abstracts of publications. Articles were selected based on three criteria: (1) description of patients with clear cell RCC, (2) treatment with VEGF targeted therapy and (3) discussion of biomarkers that were studied for potential association with treatment response. RESULTS The literature search was performed on March 4th 2014 and yielded 1882 articles. After carefully reading the titles and abstracts based on the three previously mentioned criteria, 103 publications were evaluated. Backward citation screening was performed on all eligible studies and revealed another 24 articles. This search revealed that (1) High glucose uptake and low contrast enhancement on PET- and CT-imaging before start of treatment may correlate with poor response to therapy, (2) Low dose intensity due to treatment intolerance is related to shorter progression free survival. (3) Acquired resistance appears to be associated with rebound vascularization based on both longitudinal monitoring of contrast enhancement by CT and blood vessel counts in tumor tissue, and (4) Based on plasma cytokine and single nucleotide polymorphism (SNP) studies, interleukin-8, VEGFR-3, FGFR2 and HGF/MET emerged as potential clinical markers for chemoresistance. CONCLUSION Low dose intensity, specific tumor-imaging techniques and potential biological biomarkers may be predictive for response to VEGF-targeted therapy in ccRCC. Some of these plausible biomarkers may also provide more insight into the underlying mechanisms of resistance such as altered glucose metabolism and rapid rebound vascularization.
Collapse
Affiliation(s)
- Johannes C van der Mijn
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands; Department of Hematology/Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - James W Mier
- Department of Hematology/Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Henk J Broxterman
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Henk M Verheul
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Radiological evaluation of response to treatment: Application to metastatic renal cancers receiving anti-angiogenic treatment. Diagn Interv Imaging 2014; 95:527-39. [DOI: 10.1016/j.diii.2013.01.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
44
|
Assessing the Response to Targeted Therapies in Renal Cell Carcinoma: Technical Insights and Practical Considerations. Eur Urol 2014; 65:766-77. [DOI: 10.1016/j.eururo.2013.11.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 11/18/2013] [Indexed: 12/21/2022]
|
45
|
Vano YA, Tartour E, Fournier LS, Beuselinck B, Mejean A, Oudard S. Prognostic factors in patients with advanced renal cell carcinoma treated with VEGF-targeted agents. Expert Rev Anticancer Ther 2014; 14:523-42. [DOI: 10.1586/14737140.2014.882773] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
46
|
Multimodal microvascular imaging reveals that selective inhibition of class I PI3K is sufficient to induce an antivascular response. Neoplasia 2014; 15:694-711. [PMID: 23814482 DOI: 10.1593/neo.13470] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 04/16/2013] [Accepted: 04/22/2013] [Indexed: 12/31/2022]
Abstract
The phosphatidylinositol 3-kinase (PI3K) pathway is a central mediator of vascular endothelial growth factor (VEGF)-driven angiogenesis. The discovery of small molecule inhibitors that selectively target PI3K or PI3K and mammalian target of rapamycin (mTOR) provides an opportunity to pharmacologically determine the contribution of these key signaling nodes in VEGF-A-driven tumor angiogenesis in vivo. This study used an array of micro-vascular imaging techniques to monitor the antivascular effects of selective class I PI3K, mTOR, or dual PI3K/mTOR inhibitors in colorectal and prostate cancer xenograft models. Micro-computed tomography (micro-CT) angiography, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), vessel size index (VSI) MRI, and DCE ultrasound (DCE-U/S) were employed to quantitatively evaluate the vascular (structural and physiological) response to these inhibitors. GDC-0980, a dual PI3K/mTOR inhibitor, was found to reduce micro-CT angiography vascular density, while VSI MRI demonstrated a significant reduction in vessel density and an increase in mean vessel size, consistent with a loss of small functional vessels and a substantial antivascular response. DCE-MRI showed that GDC-0980 produces a strong functional response by decreasing the vascular permeability/perfusion-related parameter, K (trans). Interestingly, comparable antivascular effects were observed for both GDC-980 and GNE-490 (a selective class I PI3K inhibitor). In addition, mTOR-selective inhibitors did not affect vascular density, suggesting that PI3K inhibition is sufficient to generate structural changes, characteristic of a robust antivascular response. This study supports the use of noninvasive microvascular imaging techniques (DCE-MRI, VSI MRI, DCE-U/S) as pharmacodynamic assays to quantitatively measure the activity of PI3K and dual PI3K/mTOR inhibitors in vivo.
Collapse
|
47
|
Lawrentschuk N, Scott AM, Davis ID. Potential of imaging biomarkers for characterization of renal masses. Expert Rev Anticancer Ther 2014; 10:781-6. [DOI: 10.1586/era.10.69] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
48
|
León L, García-Figueiras R, García-Figueras R, Suárez C, Arjonilla A, Puente J, Vargas B, Méndez Vidal MJ, Sebastiá C. Recommendations for the clinical and radiological evaluation of response to treatment in metastatic renal cell cancer. Target Oncol 2013; 9:9-24. [PMID: 24338498 DOI: 10.1007/s11523-013-0304-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 11/28/2013] [Indexed: 12/21/2022]
Abstract
The evaluation of response to treatment is a critical step for determining the effectiveness of oncology drugs. Targeted therapies such as tyrosine kinase inhibitors and mammalian target of rapamycin inhibitors are active drugs in patients with metastatic renal cell carcinoma (mRCC). However, treatment with this type of drugs may not result in significant reductions in tumor size, so standard evaluation criteria based on tumor size, such as Response Evaluation Criteria in Solid Tumors (RECIST), may be inappropriate for evaluating response to treatment in patients with mRCC. In fact, targeted therapies apparently yield low response rates that do not reflect increased disease control they may cause and, consequently, the benefit in terms of time to progression. To improve the clinical and radiological evaluation of response to treatment in patients with mRCC treated with targeted drugs, a group of 32 experts in this field have reviewed different aspects related to this issue and have put together a series of recommendations with the intention of providing guidance to clinicians on this matter.
Collapse
Affiliation(s)
- Luís León
- Medical Oncology Department, Complejo Hospitalario Universitario de Santiago, A Coruña, Spain,
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Bernardin L, O'Flynn EAM, Desouza NM. Functional imaging biomarkers for assessing response to treatment in liver and lung metastases. Cancer Imaging 2013; 13:482-94. [PMID: 24334562 PMCID: PMC3864224 DOI: 10.1102/1470-7330.2013.0047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2013] [Indexed: 01/15/2023] Open
Abstract
Management of patients with metastatic cancer and development of new treatments rely on imaging to provide non-invasive biomarkers of tumour response and progression. The widely used size-based criteria have increasingly become inadequate where early measures of response are required to avoid toxicity of ineffective treatments, as biological, physiologic, and molecular modifications in tumours occur before changes in gross tumour size. A multiparametric approach with the current range of imaging techniques allows functional aspects of tumours to be simultaneously interrogated. Appropriate use of these imaging techniques and their timing in relation to the treatment schedule, particularly in the context of clinical trials, is fundamental. There is a lack of consensus regarding which imaging parameters are most informative for a particular disease site and the best time to image so that, despite an increasing body of literature, open questions on these aspects remain. In addition, standardization of these new parameters is required. This review summarizes the published literature over the last decade on functional and molecular imaging techniques in assessing treatment response in liver and lung metastases.
Collapse
Affiliation(s)
- Livia Bernardin
- Clinical Magnetic Resonance Group, Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, UK
| | - Elizabeth A M O'Flynn
- Clinical Magnetic Resonance Group, Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, UK
| | - Nandita M Desouza
- Clinical Magnetic Resonance Group, Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, UK
| |
Collapse
|
50
|
[Pharmacological therapy of urogenital cancer: rational routine diagnostic imaging]. Urologe A 2013; 52:1564-73. [PMID: 24197084 DOI: 10.1007/s00120-013-3253-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Imaging studies are an integral and important diagnostic modality to stage, monitor, and follow-up patients with metastatic urogenital cancer. The currently available guidelines on diagnosis and treatment of urogenital cancer do not provide the clinician with evidence-based recommendations for daily routine. It is the aim of the current manuscript to develop scientifically valid recommendations with regard to the most appropriate imaging technique and the most useful time interval in metastatic urogenital cancer patients undergoing systemic therapy. RESULTS Therapeutic response of soft tissue metastases is evaluated with the use of the RECIST criteria. In skeletal metastases, bone scans with validated algorithms must be performed to assess response. In patients with testicular germ cell tumors, computed tomography (CT) of the chest, the retroperitoneum, and the abdomen represents the standard imaging technique of choice usually performed prior to and at the end of systemic chemotherapy. Only in seminomas with residual tumors > 3 cm in diameter should FDG-PET/CT be performed about 6 weeks after chemotherapy. Metastatic renal cell carcinomas treated with molecular targeted therapies are routinely evaluated by CT scans at 3 month intervals. In specific cases, FDG-PET/CT is able to predict responses as early as 8 weeks after initiation of treatment. In patients with metastatic urothelial carcinomas, imaging studies should be performed after every second cycle of cytotoxic therapy. In patients with metastatic prostate cancer, the modality and the frequency of imaging studies depends on the type of the treatment. In men undergoing androgen deprivation therapy, no routine imaging studies are recommended except for patients with new onset symptoms or significant PSA progression prior to change of treatment. In men with metastatic castration-resistant PCA who are treated with cytotoxic regimes, routine imaging studies in the presence of decreasing or stable PSA serum concentrations are not indicated. In men treated with lyase inhibitor or inhibitors of the androgen receptor signaling cascade, imaging studies should be performed at 3 month intervals due to the low correlation of PSA serum concentrations with clinical response. CONCLUSIONS Imaging studies to assess therapeutic response to systemic treatment in metastatic cancers of the urogenital tract must be chosen depending on the treatment regime, primary organ, and potential consequences of the findings. Routine imaging studies without specific clinical or therapeutic relevance are not justified.
Collapse
|