1
|
Schmidt R, Rueger C, Xu H, He Y, Yilmaz EY, Heidemann L, Sulejmani O, Liu Y, Noack L, Hesse F, Ruppel R, Abosabie SA, Hamm CA, Penzkofer T, Gebauer B, Savic LJ. Comparing the Prognostic Value of Quantitative Response Assessment Tools and LIRADS Treatment Response Algorithm in Patients with Hepatocellular Carcinoma Following Interstitial High-Dose-Rate Brachytherapy and Conventional Transarterial Chemoembolization. Cancers (Basel) 2025; 17:1275. [PMID: 40282451 PMCID: PMC12025668 DOI: 10.3390/cancers17081275] [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: 02/15/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The aim of this study was to investigate the prognostic value of established response assessment tools for hepatocellular carcinoma (HCC) treated with high-dose-rate interstitial brachytherapy (iBT) alone or with transarterial chemoembolization (cTACE). Methods: (Non-)responders were categorized using size-based RECIST 1.1 and WHO criteria, enhancement-based mRECIST and EASL criteria, and the LI-RADS Treatment Response Algorithm (LR-TRA). The outcomes were the overall survival (OS), progression-free survival (PFS), and time to progression (TTP). The statistics used included Fisher's exact test, a t-test, the Mann-Whitney-U test, and a Kaplan-Meier analysis. The median OS, PFS, and TTP were higher in patients following iBT (26.3, 9.1, and 13.0 months) than following cTACE/iBT (23.3, 7.6, and 9.2 months). Results: The enhancement-based criteria identified more responders and predicted PFS and TTP better compared to the size-based criteria. At two months, the cTACE/iBT responders showed improved PFS (mRECIST and EASL: 11.3 vs. 2.3 and 11.0 vs. 2.3, p < 0.01) and TTP (mRECIST and EASL: 11.9 vs. 2.4 months, p < 0.01) by the enhancement-based criteria. An EASL assessment at five months predicted improved survival following both cTACE/iBT (PFS: 11.9 vs. 5.1 months, p = 0.03; TTP: 12.4 vs. 5.0, p < 0.01) and iBT (11.1 vs. 5.1 months, p = 0.04; 13.0 vs. 5.3, p < 0.01). The LR-TRA showed OS benefits at five months for cTACE/iBT responders. Size-based criteria were not prognostic. Conclusions: Extending follow-up post-iBT or post-iBT/cTACE may improve responder stratification and prognostication.
Collapse
Affiliation(s)
- Robin Schmidt
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Experimental Clinical Research Center (ECRC) at Charité-Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Christopher Rueger
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Han Xu
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Yubei He
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Experimental Clinical Research Center (ECRC) at Charité-Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Emine Yaren Yilmaz
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Experimental Clinical Research Center (ECRC) at Charité-Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Luisa Heidemann
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Experimental Clinical Research Center (ECRC) at Charité-Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Ornela Sulejmani
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Experimental Clinical Research Center (ECRC) at Charité-Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Yu Liu
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Experimental Clinical Research Center (ECRC) at Charité-Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Lasse Noack
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Friederike Hesse
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Richard Ruppel
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sara A. Abosabie
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Charlie Alexander Hamm
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Tobias Penzkofer
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Bernhard Gebauer
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Lynn Jeanette Savic
- Department of Radiology, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Experimental Clinical Research Center (ECRC) at Charité-Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
2
|
Yang TS, Gong XH, Wang L, Zhang S, Shi YP, Ren HN, Yan YQ, Zhu L, Lv L, Dai YM, Qian LJ, Xu JR, Zhou Y. Comparison of automated with manual 3D qEASL assessment based on MR imaging in hepatocellular carcinoma treated with conventional TACE. Abdom Radiol (NY) 2025; 50:1180-1188. [PMID: 39297930 DOI: 10.1007/s00261-024-04571-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/21/2024]
Affiliation(s)
- Tian Shu Yang
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xu Hua Gong
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Li Wang
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Shan Zhang
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yao Ping Shi
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- Interventional Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Hai Nan Ren
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yun Qi Yan
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Li Zhu
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lei Lv
- ShuKun (Beijing) Technology Co. Ltd, Beijing, China
| | | | - Li Jun Qian
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Jian Rong Xu
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Yan Zhou
- Diagnostic Radiology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Sobirey R, Matuschewski N, Gross M, Lin M, Kao T, Kasolowsky V, Strazzabosco M, Stein S, Savic LJ, Gebauer B, Jaffe A, Duncan J, Madoff DC, Chapiro J. Tumor response assessment in hepatocellular carcinoma treated with immunotherapy: imaging biomarkers for clinical decision-making. Eur Radiol 2025; 35:73-83. [PMID: 39033181 DOI: 10.1007/s00330-024-10955-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/23/2024] [Accepted: 04/22/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVE To compare the performance of 1D and 3D tumor response assessment for predicting median overall survival (mOS) in patients who underwent immunotherapy for hepatocellular carcinoma (HCC). METHODS Patients with HCC who underwent immunotherapy between 2017 and 2023 and received multi-phasic contrast-enhanced MRIs pre- and post-treatment were included in this retrospective study. Tumor response was measured using 1D, RECIST 1.1, and mRECIST, and 3D, volumetric, and percentage quantitative EASL (vqEASL and %qEASL). Patients were grouped into disease control vs progression and responders vs non-responders. Kaplan-Meier curves analyzed with log-rank tests assessed the predictive value for mOS. Cox regression modeling evaluated the association of clinical baseline parameters with mOS. RESULTS This study included 37 patients (mean age, 69.1 years [SD, 8.0]; 33 men). The mOS was 16.9 months. 3D vqEASL and %qEASL successfully stratified patients into disease control and progression (vqEASL: HR 0.21, CI: 0.55-0.08, p < 0.001; %qEASL: HR 0.18, CI: 0.83-0.04, p = 0.013), as well as responder and nonresponder (vqEASL: HR 0.25, CI: 0.08-0.74, p = 0.007; %qEASL: HR 0.17, CI: 0.04-0.72, p = 0.007) for predicting mOS. The 1D criteria, mRECIST stratified into disease control and progression only (HR 0.24, CI: 0.65-0.09, p = 0.002), and RECIST 1.1 showed no predictive value in either stratification. Multivariate Cox regression identified alpha-fetoprotein > 500 ng/mL as a predictor for poor mOS (p = 0.04). CONCLUSION The 3D quantitative enhancement-based response assessment tool qEASL can predict overall survival in patients undergoing immunotherapy for HCC and could identify non-responders. CLINICAL RELEVANCE STATEMENT Using 3D quantitative enhancement-based tumor response criteria (qEASL), radiologists' predictions of tumor response in patients undergoing immunotherapy for HCC can be further improved. KEY POINTS MRI-based tumor response criteria predict immunotherapy survival benefits in HCC patients. 3D tumor response assessment methods surpass current evaluation criteria in predicting overall survival during HCC immunotherapy. Enhancement-based 3D tumor response criteria are robust prognosticators of survival for HCC patients on immunotherapy.
Collapse
Affiliation(s)
- Rabea Sobirey
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, Berlin, Germany
| | - Nickolai Matuschewski
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, Berlin, Germany
| | - Moritz Gross
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, Berlin, Germany
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Visage Imaging Inc., San Diego, CA, USA
| | - Tabea Kao
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, Berlin, Germany
| | - Victor Kasolowsky
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, Berlin, Germany
| | - Mario Strazzabosco
- Department of Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Stacey Stein
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Lynn Jeanette Savic
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, Berlin, Germany
| | - Bernhard Gebauer
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, Berlin, Germany
| | - Ariel Jaffe
- Department of Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - James Duncan
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - David C Madoff
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT, USA
- Department of Surgery, Section of Surgical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA.
- Department of Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
4
|
Ghabili K, Windham-Herman AM, Konstantinidis M, Murali N, Borde T, Adam LC, Laage-Gaupp F, Lin M, Chapiro J, Georgiades C, Nezami N. Outcomes of repeat conventional transarterial chemoembolization in patients with liver metastases. Ann Hepatol 2024; 29:101529. [PMID: 39033928 PMCID: PMC11558520 DOI: 10.1016/j.aohep.2024.101529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 06/18/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION AND OBJECTIVES Although unlimited sessions of conventional transarterial chemoembolization (cTACE) may be performed for liver metastases, there is no data indicating when treatment becomes ineffective. This study aimed to determine the optimal number of repeat cTACE sessions for nonresponding patients before abandoning cTACE in patients with liver metastases. MATERIALS AND METHODS In this retrospective, single-institutional analysis, patients with liver metastases from neuroendocrine tumors (NET), colorectal carcinoma (CRC), and lung cancer who underwent consecutive cTACE sessions from 2001 to 2015 were studied. Quantitative European Association for Study of the Liver (qEASL) criteria were utilized for response assessment. The association between the number of cTACE and 2-year, 5-year, and overall survival was evaluated to estimate the optimal number of cTACE for each survival outcome. RESULTS Eighty-five patients underwent a total of 186 cTACE sessions for 117 liver metastases, of which 30.7 % responded to the first cTACE. For the target lesions that did not respond to the first, second, and third cTACE sessions, response rates after the second, third, and fourth cTACE sessions were 33.3 %, 23 %, and 25 %, respectively. The fourth cTACE session was the optimal number for 2-year survival (HR 0.40; 95 %CI: 0.16-0.97; p = 0.04), 5-year survival (HR 0.31; 95 %CI: 0.11-0.87; p = 0.02), and overall survival (HR 0.35; 95 %CI: 0.13-0.89; p = 0.02). CONCLUSIONS Repeat cTACE in the management of liver metastases from NET, CRC, and lung cancer was associated with improved patient survival. We recommend at least four cTACE sessions before switching to another treatment for nonresponding metastatic liver lesions.
Collapse
Affiliation(s)
- Kamyar Ghabili
- Department of Radiology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA; Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Austin-Marley Windham-Herman
- Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical imaging, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Interventional Radiology, University of California San Diego, La Jolla, California, USA
| | - Menelaos Konstantinidis
- Institute of Health Policy, Management and Evaluation, University of Toronto, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Canada
| | - Nikitha Murali
- Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical imaging, Yale University School of Medicine, New Haven, Connecticut, USA; Section of Interventional Radiology, Department of Radiology, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tabea Borde
- Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical imaging, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology and Experimental Neurology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Charité Campus Benjamin Franklin (CBF), Berlin, Germany
| | - Lucas C Adam
- Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical imaging, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology and Experimental Neurology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Charité Campus Benjamin Franklin (CBF), Berlin, Germany
| | - Fabian Laage-Gaupp
- Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - MingDe Lin
- Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Julius Chapiro
- Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Christos Georgiades
- Division of Vascular and Interventional Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Nariman Nezami
- Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical imaging, Yale University School of Medicine, New Haven, Connecticut, USA; Division of Vascular and Interventional Radiology, Department of Radiology, Medstar Georgetown Hospital, Washington, DC, USA; Georgetown University School of Medicine, Washington, DC, USA; Lombardi Comprehensive Cancer Center, Washington, DC, USA.
| |
Collapse
|
5
|
Rebillard E, De Abreu N, Buchard B, Muti L, Boulin M, Pereira B, Magnin B, Abergel A. AFP-DIAM Score to Predict Survival in Patients with Hepatocellular Carcinoma Before TACE: A French Multicenter Study. Dig Dis Sci 2024; 69:4259-4267. [PMID: 39322806 DOI: 10.1007/s10620-024-08639-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 09/05/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Transarterial chemoembolization (TACE) is recommended as a palliative treatment for patients of the B stage of the Barcelona Clinic Liver Cancer (BCLC) classification. AIMS To identify clinical, biological, and radiological predictors of survival in patients undergoing TACE and develop a pre-therapeutic prognostic score. METHODS 191 adult cirrhotic patients treated for HCC with TACE at the University Hospital (UH) of Clermont-Ferrand (France) from 2007-2017 were retrospectively included. We investigated the impact of baseline liver function, patient characteristics, and tumor burden on overall survival and developed a prognostic score. RESULTS Patients had a median age of 66 years and 126 patients were Child A. The AFP-DIAM score distinguishes two groups with a significant difference in survival time (median OS 28.3 months in patients with a score = 0 versus 17.7 months in patients with a score > 0). AFP-DIAM was validated on an external cohort, is well calibrated, and has the best discrimination capacity (C-index) as compared to NIACE, HAP, STATE, and SIX TO TWELVE. AFP-DIAM and SIX TO TWELVE are the more easy-to-use scores. When AFP-DIAM and the SIX TO TWELVE scores were tested in the same statistical model, results confirmed a better AFP-DIAM performance. CONCLUSIONS The AFP-DIAM is an easy-to-use score which allows to distinguish two groups with different prognosis before the first TACE session. Its use could provide further support to BCLC system to guide the therapeutic strategy of patients with HCC.
Collapse
Affiliation(s)
- Estelle Rebillard
- Médecine Digestive et Hépato-Biliaire, Centre Hospitalier Universitaire de Clermont-Ferrand, 1 Place Lucie et Raymond Aubrac, 63003, Clermont-Ferrand Cedex 1, France
| | - Nicolas De Abreu
- Radiologie, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Benjamin Buchard
- Médecine Digestive et Hépato-Biliaire, Centre Hospitalier Universitaire de Clermont-Ferrand, 1 Place Lucie et Raymond Aubrac, 63003, Clermont-Ferrand Cedex 1, France
| | - Léon Muti
- Médecine Digestive et Hépato-Biliaire, Centre Hospitalier Universitaire de Clermont-Ferrand, 1 Place Lucie et Raymond Aubrac, 63003, Clermont-Ferrand Cedex 1, France
| | - Mathieu Boulin
- Pharmacie, Centre Hospitalier Universitaire de Dijon, Dijon, France
| | - Bruno Pereira
- Direction de la Recherche Clinique et Innovation, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Benoit Magnin
- Radiologie, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Armand Abergel
- Médecine Digestive et Hépato-Biliaire, Centre Hospitalier Universitaire de Clermont-Ferrand, 1 Place Lucie et Raymond Aubrac, 63003, Clermont-Ferrand Cedex 1, France.
- Radiologie, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France.
- Pharmacie, Centre Hospitalier Universitaire de Dijon, Dijon, France.
- Direction de la Recherche Clinique et Innovation, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France.
- Université Clermont Auvergne, CHU, CNRS, Clermont Auvergne INP, Institut Pascal, 63000, Clermont-Ferrand, France.
| |
Collapse
|
6
|
Mohseni A, Baghdadi A, Madani SP, Shahbazian H, Mirza-Aghazadeh-Attari M, Borhani A, Afyouni S, Zandieh G, Baretti M, Kim AK, Yarchoan M, Kamel IR. Predicting survival of patients with advanced hepatocellular carcinoma receiving combination targeted immunotherapy: an evaluation of volumetric imaging parameters. Abdom Radiol (NY) 2024; 49:2595-2605. [PMID: 38546828 DOI: 10.1007/s00261-024-04257-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 08/06/2024]
Abstract
PURPOSE To evaluate the potential of volumetric imaging in predicting survival of advanced hepatocellular carcinoma (HCC) patients receiving immunotherapy. METHODS Retrospective analysis included 40 patients with advanced HCC who received targeted immunotherapy. Baseline and follow-up contrast-enhanced abdominal computed tomography (CT) scans were analyzed. The largest tumor was chosen as the index lesion. Viable tumor volume (qViable) and percentage tumor viability (%Viability) were calculated. Response Evaluation Criteria in Solid Tumors (RECIST) and Tumor volume change after treatment (qRECIST) were measured. Associations with overall survival (OS) were assessed. Cox regression analysis assessed the association between variables and overall survival (OS). A new prognostic stratification system was attempted to categorize patients based on significant predictors of OS. Patients with a baseline %viability > 69% and %viability reduction ≥ 8% were classified as better prognosis. Patients were stratified into better, intermediate and worse prognosis groups based on baseline %viability > 69% and ≥ 8% %viability reduction (better prognosis); baseline %viability ≤ 69% and < 8% %viability reduction (worse prognosis); remainder were intermediate prognosis. RESULTS Patients with baseline %Viability > 69% and %Viability reduction ≥ 8% showed significantly higher OS. Multivariate analysis confirmed %Viability and %Viability reduction as significant predictors of OS. A prognostic stratification system using these parameters stratified patients into better, intermediate and worse prognosis groups, with the better prognosis showing highest OS. Most patients (97.5%) had stable disease by RECIST while the prognostic model re-classified 47.5% as better prognosis, 37.5% intermediate prognosis, and 15% worse prognosis. CONCLUSION Volumetric parameters of %Viability and %Viability reduction predict OS in HCC patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Alireza Mohseni
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Azarakhsh Baghdadi
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Seyedeh Panid Madani
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Haneyeh Shahbazian
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Mohammad Mirza-Aghazadeh-Attari
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Ali Borhani
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Shadi Afyouni
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Ghazal Zandieh
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Marina Baretti
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy K Kim
- Department of Medicine, Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark Yarchoan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ihab R Kamel
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA.
| |
Collapse
|
7
|
Wesdorp NJ, Zeeuw JM, Postma SCJ, Roor J, van Waesberghe JHTM, van den Bergh JE, Nota IM, Moos S, Kemna R, Vadakkumpadan F, Ambrozic C, van Dieren S, van Amerongen MJ, Chapelle T, Engelbrecht MRW, Gerhards MF, Grunhagen D, van Gulik TM, Hermans JJ, de Jong KP, Klaase JM, Liem MSL, van Lienden KP, Molenaar IQ, Patijn GA, Rijken AM, Ruers TM, Verhoef C, de Wilt JHW, Marquering HA, Stoker J, Swijnenburg RJ, Punt CJA, Huiskens J, Kazemier G. Deep learning models for automatic tumor segmentation and total tumor volume assessment in patients with colorectal liver metastases. Eur Radiol Exp 2023; 7:75. [PMID: 38038829 PMCID: PMC10692044 DOI: 10.1186/s41747-023-00383-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/08/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND We developed models for tumor segmentation to automate the assessment of total tumor volume (TTV) in patients with colorectal liver metastases (CRLM). METHODS In this prospective cohort study, pre- and post-systemic treatment computed tomography (CT) scans of 259 patients with initially unresectable CRLM of the CAIRO5 trial (NCT02162563) were included. In total, 595 CT scans comprising 8,959 CRLM were divided into training (73%), validation (6.5%), and test sets (21%). Deep learning models were trained with ground truth segmentations of the liver and CRLM. TTV was calculated based on the CRLM segmentations. An external validation cohort was included, comprising 72 preoperative CT scans of patients with 112 resectable CRLM. Image segmentation evaluation metrics and intraclass correlation coefficient (ICC) were calculated. RESULTS In the test set (122 CT scans), the autosegmentation models showed a global Dice similarity coefficient (DSC) of 0.96 (liver) and 0.86 (CRLM). The corresponding median per-case DSC was 0.96 (interquartile range [IQR] 0.95-0.96) and 0.80 (IQR 0.67-0.87). For tumor segmentation, the intersection-over-union, precision, and recall were 0.75, 0.89, and 0.84, respectively. An excellent agreement was observed between the reference and automatically computed TTV for the test set (ICC 0.98) and external validation cohort (ICC 0.98). In the external validation, the global DSC was 0.82 and the median per-case DSC was 0.60 (IQR 0.29-0.76) for tumor segmentation. CONCLUSIONS Deep learning autosegmentation models were able to segment the liver and CRLM automatically and accurately in patients with initially unresectable CRLM, enabling automatic TTV assessment in such patients. RELEVANCE STATEMENT Automatic segmentation enables the assessment of total tumor volume in patients with colorectal liver metastases, with a high potential of decreasing radiologist's workload and increasing accuracy and consistency. KEY POINTS • Tumor response evaluation is time-consuming, manually performed, and ignores total tumor volume. • Automatic models can accurately segment tumors in patients with colorectal liver metastases. • Total tumor volume can be accurately calculated based on automatic segmentations.
Collapse
Affiliation(s)
- Nina J Wesdorp
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands.
| | - J Michiel Zeeuw
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands.
| | - Sam C J Postma
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Joran Roor
- Department of Health, SAS Institute B.V, Huizen, the Netherlands
| | - Jan Hein T M van Waesberghe
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Janneke E van den Bergh
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Irene M Nota
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Shira Moos
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Ruby Kemna
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Fijoy Vadakkumpadan
- Department of Computer Vision and Machine Learning, SAS Institute Inc, Cary, NC, USA
| | - Courtney Ambrozic
- Department of Computer Vision and Machine Learning, SAS Institute Inc, Cary, NC, USA
| | - Susan van Dieren
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | | | - Thiery Chapelle
- Department of Hepatobiliary, Transplantation, and Endocrine Surgery, Antwerp University Hospital, Antwerp, Belgium
| | - Marc R W Engelbrecht
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Dirk Grunhagen
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Thomas M van Gulik
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - John J Hermans
- Department of Medical Imaging, Radboud University Medical Center, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Koert P de Jong
- Department of HPB Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Joost M Klaase
- Department of HPB Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mike S L Liem
- Department of Surgery, Medical Spectrum Twente, Enschede, the Netherlands
| | - Krijn P van Lienden
- Department of Interventional Radiology, St Antonius Hospital, Nieuwegein, the Netherlands
| | - I Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Surgery, St Antonius Hospital, Nieuwegein, the Netherlands
| | - Gijs A Patijn
- Department of Surgery, Isala Hospital, Zwolle, the Netherlands
| | - Arjen M Rijken
- Department of Surgery, Amphia Hospital, Breda, the Netherlands
| | - Theo M Ruers
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Johannes H W de Wilt
- Department of Surgery, Radboud University Medical Center, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Henk A Marquering
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jaap Stoker
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Rutger-Jan Swijnenburg
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joost Huiskens
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| | - Geert Kazemier
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands
| |
Collapse
|
8
|
Elderkin J, Al Hallak N, Azmi AS, Aoun H, Critchfield J, Tobon M, Beal EW. Hepatocellular Carcinoma: Surveillance, Diagnosis, Evaluation and Management. Cancers (Basel) 2023; 15:5118. [PMID: 37958294 PMCID: PMC10647678 DOI: 10.3390/cancers15215118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 11/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranks fourth in cancer-related deaths worldwide. Semiannual surveillance of the disease for patients with cirrhosis or hepatitis B virus allows for early detection with more favorable outcomes. The current underuse of surveillance programs demonstrates the need for intervention at both the patient and provider level. Mail outreach along with navigation provision has proven to increase surveillance follow-up in patients, while provider-targeted electronic medical record reminders and compliance reports have increased provider awareness of HCC surveillance. Imaging is the primary mode of diagnosis in HCC with The Liver Imaging Reporting and Data System (LI-RADS) being a widely accepted comprehensive system that standardizes the reporting and data collection for HCC. The management of HCC is complex and requires multidisciplinary team evaluation of each patient based on their preference, the state of the disease, and the available medical and surgical interventions. Staging systems are useful in determining the appropriate intervention for HCC. Early-stage HCC is best managed by curative treatment modalities, such as liver resection, transplant, or ablation. For intermediate stages of the disease, transarterial local regional therapies can be applied. Advanced stages of the disease are treated with systemic therapies, for which there have been recent advances with new drug combinations. Previously sorafenib was the mainstay systemic treatment, but the recent introduction of atezolizumab plus bevacizumab proves to have a greater impact on overall survival. Although there is a current lack of improved outcomes in Phase III trials, neoadjuvant therapies are a potential avenue for HCC management in the future.
Collapse
Affiliation(s)
- Jessica Elderkin
- Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Najeeb Al Hallak
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (N.A.H.); (A.S.A.)
| | - Asfar S. Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (N.A.H.); (A.S.A.)
| | - Hussein Aoun
- Department of Radiology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.A.); (J.C.)
| | - Jeffrey Critchfield
- Department of Radiology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (H.A.); (J.C.)
| | - Miguel Tobon
- Department of Surgery, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Eliza W. Beal
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (N.A.H.); (A.S.A.)
- Department of Surgery, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| |
Collapse
|
9
|
Ronot M, Dioguardi Burgio M, Gregory J, Hentic O, Vullierme MP, Ruszniewski P, Zappa M, de Mestier L. Appropriate use of morphological imaging for assessing treatment response and disease progression of neuroendocrine tumors. Best Pract Res Clin Endocrinol Metab 2023; 37:101827. [PMID: 37858478 DOI: 10.1016/j.beem.2023.101827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Neuroendocrine tumors (NETs) are relatively rare neoplasms displaying heterogeneous clinical behavior, ranging from indolent to aggressive forms. Patients diagnosed with NETs usually receive a varied array of treatments, including somatostatin analogs, locoregional treatments (ablation, intra-arterial therapy), cytotoxic chemotherapy, peptide receptor radionuclide therapy (PRRT), and targeted therapies. To maximize therapeutic efficacy while limiting toxicity (both physical and economic), there is a need for accurate and reliable tools to monitor disease evolution and progression and to assess the effectiveness of these treatments. Imaging morphological methods, primarily relying on computed tomography (CT) and magnetic resonance imaging (MRI), are indispensable modalities for the initial evaluation and continuous monitoring of patients with NETs, therefore playing a pivotal role in gauging the response to treatment. The primary goal of assessing tumor response is to anticipate and weigh the benefits of treatments, especially in terms of survival gain. The World Health Organization took the pioneering step of introducing assessment criteria based on cross-sectional imaging. This initial proposal standardized the measurement of lesion sizes, laying the groundwork for subsequent criteria. The Response Evaluation Criteria in Solid Tumors (RECIST) subsequently refined and enhanced these standards, swiftly gaining acceptance within the oncology community. New treatments were progressively introduced, targeting specific features of NETs (such as tumor vascularization or expression of specific receptors), and achieving significant qualitative changes within tumors, although associated with minimal or paradoxical effects on tumor size. Several alternative criteria, adapted from those used in other cancer types and focusing on tumor viability, the slow growth of NETs, or refining the existing size-based RECIST criteria, have been proposed in NETs. This review article aims to describe and discuss the optimal utilization of CT and MRI for assessing the response of NETs to treatment; it provides a comprehensive overview of established and emerging criteria for evaluating tumor response, along with comparative analyses. Molecular imaging will not be addressed here and is covered in a dedicated article within this special issue.
Collapse
Affiliation(s)
- Maxime Ronot
- Université Paris-Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, FHU MOSAIC, Paris, France; Université Paris-Cité, Department of Radiology, Beaujon Hospital (APHP.Nord), Clichy, France.
| | - Marco Dioguardi Burgio
- Université Paris-Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, FHU MOSAIC, Paris, France; Université Paris-Cité, Department of Radiology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Jules Gregory
- Université Paris-Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, FHU MOSAIC, Paris, France; Université Paris-Cité, Department of Radiology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Olivia Hentic
- Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
| | | | - Philippe Ruszniewski
- Université Paris-Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, FHU MOSAIC, Paris, France; Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
| | - Magaly Zappa
- Department of Radiology, Cayenne University Hospital, Cayenne, Guyanne, France
| | - Louis de Mestier
- Université Paris-Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, FHU MOSAIC, Paris, France; Université Paris-Cité, Department of Pancreatology and Digestive Oncology, Beaujon Hospital (APHP.Nord), Clichy, France
| |
Collapse
|
10
|
Yang S, Zhang Z, Su T, Chen Q, Wang H, Jin L. Comparison of quantitative volumetric analysis and linear measurement for predicting the survival of Barcelona Clinic Liver Cancer 0- and A stage hepatocellular carcinoma after radiofrequency ablation. Diagn Interv Radiol 2023; 29:450-459. [PMID: 37154818 PMCID: PMC10679614 DOI: 10.4274/dir.2023.222055] [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: 12/12/2022] [Accepted: 04/13/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE The prognostic role of the tumor volume in patients with hepatocellular carcinoma (HCC) at the Barcelona Clinic Liver Cancer (BCLC) 0 and A stages remains unclear. This study aims to compare the volumetric measurement with linear measurement in early HCC burden profile and clarify the optimal cut-off value of the tumor volume. METHODS The consecutive patients diagnosed with HCC who underwent initial and curative-intent radiofrequency ablation (RFA) were included retrospectively. The segmentation was performed semi-automatically, and enhanced tumor volume (ETV) as well as total tumor volume (TTV) were obtained. The patients were categorized into high- and low-tumor burden groups according to various cutoff values derived from commonly used diameter values, X-tile software, and decision-tree analysis. The inter- and intra-reviewer agreements were measured using the intra-class correlation coefficient. Univariate and multivariate time-to-event Cox regression analyses were performed to identify the prognostic factors of overall survival. RESULTS A total of 73 patients with 81 lesions were analyzed in the whole cohort with a median follow-up of 31.0 (interquartile range: 16.0–36.3). In tumor segmentation, excellent consistency was observed in intra- and inter-reviewer assessments. There was a strong correlation between diameter-derived spherical volume and ETV as well as ETV and TTV. As opposed to all linear candidates and 4,188 mm3 (sphere equivalent to 2 cm in diameter), ETV >14,137 mm3 (sphere equivalent to 3 cm in diameter) or 23,000 mm3 (sphere equivalent to 3.5 cm in diameter) was identified as an independent risk factor of survival. Considering the value of hazard ratio and convenience to use, when ETV was at 23,000 mm3, it was regarded as the optimal volumetric cut-off value in differentiating survival risk. CONCLUSION The volumetric measurement outperforms linear measurement on tumor burden evaluation for survival stratification in patients at BCLC 0 and A stages HCC after RFA.
Collapse
Affiliation(s)
- Siwei Yang
- Department of Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhiyuan Zhang
- Department of Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tianhao Su
- Department of Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qiyang Chen
- Department of Ultrasound, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Haochen Wang
- Department of Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Long Jin
- Department of Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Xu J, Yin Y, Yang J, Chen L, Li Z, Shen J, Wang W, Ni C. Modified quantitative and volumetric response evaluation criteria for patients with hepatocellular carcinoma after transarterial chemoembolization. Front Oncol 2023; 13:957722. [PMID: 36761945 PMCID: PMC9905806 DOI: 10.3389/fonc.2023.957722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023] Open
Abstract
Objective This study aimed to investigate the cutoff value of quantitative and volumetric response evaluation criteria for patients with hepatocellular carcinoma (HCC) after transarterial chemoembolization (TACE) and compare the performance of the modified criteria to one-dimensional criteria in survival prediction. Methods A retrospective single-center study was performed for treatment-naive patients with HCC who underwent initial TACE between June 2015 and June 2019. Treatment response assessment was performed after the first observation by contrast CT or MRI, with the measurement of diameters by modified Response Evaluation Criteria in Solid Tumors (mRECIST) and volumes by quantitative European Association for Study of the Liver (qEASL). Overall survival (OS) was the primary endpoint of this study. The new cutoff value for volumetric response evaluation criteria was created using restricted cubic splines. The performance of modified qEASL (mqEASL, with the new cutoff value) and mRECIST on survival prediction was compared by Cox regression models in internal and external validation. Results A total of 129 patients (mean age, 60 years ± 11 [standard deviation]; 111 men) were included and divided into training (n=90) and validation (n=39) cohorts. The cutoff value for the viable volume reduction was set at 57.0%. The mqEASL enabled separation of non-responders and responders in terms of median OS (p<0.001), 11.2 months (95% CI, 8.5-17.2 months) vs. 31.5 months (95% CI, 25.5-44.0 months). Two multivariate models were developed with independent prognostic factors (tumor response, metastasis, portal vein tumor thrombus, and subsequent treatment) to predict OS. Model 2 (for mqEASL) had a greater Harrel's C index, higher time-dependent area under the receiving operator characteristic curve (AUROC), and more precise calibration on 6-month survival rates than Model 1 (for mRECIST). Conclusions With the modified cutoff value, the quantitative and volumetric response of HCC patients to TACE becomes a precise predictor of overall survival. Further studies are needed to verify this modification before application in clinical practice.
Collapse
|
12
|
Adam LC, Savic LJ, Chapiro J, Letzen B, Lin M, Georgiades C, Hong KK, Nezami N. Response assessment methods for patients with hepatic metastasis from rare tumor primaries undergoing transarterial chemoembolization. Clin Imaging 2022; 89:112-119. [PMID: 35777239 PMCID: PMC9470015 DOI: 10.1016/j.clinimag.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/17/2022] [Accepted: 06/19/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE This study assessed the response to conventional transarterial chemoembolization (cTACE) in patients with liver metastases from rare tumor primaries using one-dimensional (1D) and three-dimensional (3D) quantitative response assessment methods, and investigate the relationship of lipiodol deposition in predicting response. MATERIALS AND METHODS This retrospective bicentric study included 16 patients with hepatic metastases from rare tumors treated with cTACE between 2002 and 2017. Multi-phasic MR imaging obtained before and after cTACE was used for assessment of response. Response evaluation criteria in solid tumors (RECIST) and modified-RECIST (mRECIST) were utilized for 1D response assessment, and volumetric RECIST (vRECIST) and enhancement-based quantitative European Association for Study of the Liver EASL (qEASL) were used for 3D response assessment. The same day post-cTACE CT scan was analyzed to quantify intratumoral lipiodol deposition (%). RESULTS The mean and standard deviation (SD) of diameter of treated lesions per targeted area was 7.5 ± 5.4 cm, and the mean and SD of number of metastases in each targeted area was 4.2 ± 4.6. cTACE was technically successful in all patients, without major complications. While RECIST and vRECIST methods did not allocate patients with partial response, mRECIST and qEASL identified patients with partial response. Intratumoral lipiodol deposition significantly predicted treatment response according qEASL (R2 = 0.470, p < 0.01), while no association was shown between lipiodol deposition within treated tumor area and RECIST or mRECIST (p > 0.212). CONCLUSION 3D quantitative volumetric response analysis can be used for stratification of response to cTACE in patients with hepatic metastases originating from rare primary tumors. Lipiodol deposition could potentially be used as an early surrogate to predict response to cTACE.
Collapse
Affiliation(s)
- Lucas C Adam
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA; Institute of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Lynn J Savic
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA; Institute of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, 10117 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, Charitéplatz 1, 10117 Berlin, Germany
| | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Brian Letzen
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA; Visage Imaging, Inc., San Diego, CA, USA
| | - Christos Georgiades
- Division of Vascular and Interventional Radiology, Russel H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kelvin K Hong
- Division of Vascular and Interventional Radiology, Russel H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nariman Nezami
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA; Division of Vascular and Interventional Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Experimental Therapeutics Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
13
|
Zhao Y, Haroun RR, Sahu S, Schernthaner RE, Smolka S, Lin MD, Hong KK, Georgiades C, Duran R. Three-Dimensional Quantitative Tumor Response and Survival Analysis of Hepatocellular Carcinoma Patients Who Failed Initial Transarterial Chemoembolization: Repeat or Switch Treatment? Cancers (Basel) 2022; 14:cancers14153615. [PMID: 35892874 PMCID: PMC9329887 DOI: 10.3390/cancers14153615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVES The purpose of this study was to assess treatment responses and evaluate survival outcomes between responders and non-responders after each transarterial chemoembolization (TACE) session using the 3D quantitative criteria of the European Association for the Study of the Liver (qEASL) in hepatocellular carcinoma (HCC) patients. METHODS A total of 94 consecutive patients who underwent MR imaging before and after TACE were retrospectively included. Volumetric tumor enhancement (qEASL) was expressed in cubic centimeters (cm3). The Kaplan-Meier method with the log-rank test was used to calculate the overall survival (OS) for the non-/responders. RESULTS In total, 28 (29.8%) patients showed a response after the first TACE. These responders demonstrated a clear trend toward longer OS compared with the non-responders (36.7 vs. 21.5 months, p = 0.071). Of the 43 initial non-responders who underwent a second TACE within 3 months and had complete follow-up imaging, 15/43 (34.9%) achieved a response, and their median OS was significantly longer than that of the 28 non-responders to the second TACE (47.8 vs. 13.6 months, p = 0.01). Furthermore, there was no significant difference in OS between the 28 patients who achieved a response after the first TACE and the 15 initial non-responders who achieved a response after the second TACE (36.7 vs. 47.8 months, p = 0.701). The difference in OS between the responders and non-responders after the third TACE was not significant (11.4 months vs. 13.5 months, p = 0.986). CONCLUSION Our study quantitatively demonstrated that a second TACE can be beneficial in terms of tumor response and survival for HCC patients who do not initially respond to TACE.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Gastroenterology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China;
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Sheikh Zayed Tower, Ste 7203, 1800 Orleans St, Baltimore, MD 21287, USA; (R.R.H.); (S.S.); (R.E.S.); (K.K.H.); (C.G.)
| | - Reham R. Haroun
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Sheikh Zayed Tower, Ste 7203, 1800 Orleans St, Baltimore, MD 21287, USA; (R.R.H.); (S.S.); (R.E.S.); (K.K.H.); (C.G.)
- Department of Radiology, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109, USA
| | - Sonia Sahu
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Sheikh Zayed Tower, Ste 7203, 1800 Orleans St, Baltimore, MD 21287, USA; (R.R.H.); (S.S.); (R.E.S.); (K.K.H.); (C.G.)
| | - Ruediger E. Schernthaner
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Sheikh Zayed Tower, Ste 7203, 1800 Orleans St, Baltimore, MD 21287, USA; (R.R.H.); (S.S.); (R.E.S.); (K.K.H.); (C.G.)
| | - Susanne Smolka
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, TE 2-230, New Haven, CT 06520, USA; (S.S.); (M.-D.L.)
| | - Ming-De Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, TE 2-230, New Haven, CT 06520, USA; (S.S.); (M.-D.L.)
| | - Kelvin K. Hong
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Sheikh Zayed Tower, Ste 7203, 1800 Orleans St, Baltimore, MD 21287, USA; (R.R.H.); (S.S.); (R.E.S.); (K.K.H.); (C.G.)
| | - Christos Georgiades
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Sheikh Zayed Tower, Ste 7203, 1800 Orleans St, Baltimore, MD 21287, USA; (R.R.H.); (S.S.); (R.E.S.); (K.K.H.); (C.G.)
| | - Rafael Duran
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Sheikh Zayed Tower, Ste 7203, 1800 Orleans St, Baltimore, MD 21287, USA; (R.R.H.); (S.S.); (R.E.S.); (K.K.H.); (C.G.)
- Department of Radiology and Interventional Radiology, Lausanne University Hospital, University of Lausanne, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
- Correspondence: ; Tel.: +41-(21)-3144444; Fax: +41-(21)-3144443
| |
Collapse
|
14
|
Osman MF, Farag ASA, Samy HA, El-Baz TM, Elkholy SF. Role of multislice computed tomography 3D volumetric analysis in the assessment of the therapeutic response of hepatocellular carcinoma after transarterial chemoembolization. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2021. [DOI: 10.1186/s43055-021-00542-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Volumetric analysis is a novel radiological technique used in the measurement of target lesions in three dimensions in order to estimate the lesion’s volume. Recently, it has been used for evaluation of the remaining liver volume after partial hepatectomy and also for evaluation of the response of tumours to treatment. It has been proven to be more accurate than the standard one or two-dimensional measurements, and it is especially useful for the evaluation of complex tumours after radiological interventional methods when the use of standard methods is limited. In the current study, our aim was to evaluate the value of the three-dimensional (3D) volumetric method “Response Evaluation Criteria in Solid Tumours (vRECIST)” and to compare it with the non–three-dimensional methods (RECIST) and modified RECIST (mRECIST) in the assessment of the therapeutic response of hepatocellular carcinoma (HCC) after transarterial chemoembolization (TACE).
Results
A retrospective study was conducted on 50 patients with confirmed radiological or pathological diagnosis of hepatocellular carcinoma (HCC) who underwent TACE as the only interventional procedure and follwed up by triphasic CT 1 and 4 months after treatment. The study revealed a significant difference between mRECIST and vRECIST in the assessment of the therapeutic response of HCC after TACE, a weak agreement was found between both methods in the detection of complete response (CR), partial response (PR), stable disease (SD) or progressive disease (PD). Also, there was no significant agreement between mRECIST and vRECIST regarding the assessment by classifying the patients into responders or nonresponders.
Conclusion
Volumetric analysis is an effective method for measuring the HCC lesions and evaluating its response to locoregional treatment with a significant difference between vRECIST and mRECIST in the assessment of therapeutic response, which in turn help the interventional radiologist to decide the future treatments and change the therapeutic plans. Based on these results, we recommend vRECIST to be an essential part of the assessment of therapeutic response after locoregional therapy.
Collapse
|
15
|
Li Q, Che F, Wei Y, Jiang HY, Zhang Y, Song B. Role of noninvasive imaging in the evaluation of intrahepatic cholangiocarcinoma: from diagnosis and prognosis to treatment response. Expert Rev Gastroenterol Hepatol 2021; 15:1267-1279. [PMID: 34452581 DOI: 10.1080/17474124.2021.1974294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Intrahepatic cholangiocarcinoma is the second most common liver cancer. Desmoplastic stroma may be revealed as distinctive histopathologic findings favoring intrahepatic cholangiocarcinoma. Meanwhile, a range of imaging manifestations is often accompanied with rich desmoplastic stroma in intrahepatic cholangiocarcinoma, which can indicate large bile duct ICC, and a higher level of cancer-associated fibroblasts with poor prognosis and weak treatment response. AREAS COVERED We provide a comprehensive review of current state-of-the-art and recent advances in the imaging evaluation for diagnosis, staging, prognosis and treatment response of intrahepatic cholangiocarcinoma. In addition, we discuss precursor lesions, cells of origin, molecular mutation, which would cause the different histological classification. Moreover, histological classification and tumor microenvironment, which are related to the proportion of desmoplastic stroma with many imaging manifestations, would be also discussed. EXPERT OPINION The diagnosis, prognosis, treatment response of intrahepatic cholangiocarcinoma may be revealed as the presence and the proportion of desmoplastic stroma with a range of imaging manifestations. With the utility of radiomics and artificial intelligence, imaging is helpful for ICC evaluation. Multicentre, large-scale, prospective studies with external validation are in need to develop comprehensive prediction models based on clinical data, imaging findings, genetic parameters, molecular, metabolic, and immune biomarkers.
Collapse
Affiliation(s)
- Qian Li
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Feng Che
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Yi Wei
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Han-Yu Jiang
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Yun Zhang
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Bin Song
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| |
Collapse
|
16
|
Nezami N, VAN Breugel JMM, Konstantinidis M, Chapiro J, Savic LJ, Miszczuk MA, Rexha I, Lin M, Hong K, Georgiades C. Lipiodol Deposition and Washout in Primary and Metastatic Liver Tumors After Chemoembolization. In Vivo 2021; 35:3261-3270. [PMID: 34697157 DOI: 10.21873/invivo.12621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/18/2021] [Accepted: 09/06/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND/AIM Lipiodol is the key component of conventional trans-arterial chemoembolization. Our aim was to evaluate lipiodol deposition and washout rate after conventional trans-arterial chemoembolization in intrahepatic cholangiocarcinoma and hepatic metastases originating from neuroendocrine tumors and colorectal carcinoma. PATIENTS AND METHODS This was a retrospective analysis of 44 patients with intrahepatic cholangiocarcinoma and liver metastasis from neuroendocrine tumors or colorectal carcinoma who underwent conventional trans-arterial chemoembolization. Lipiodol volume (cm3) was analyzed on non-contrast computed tomography imaging obtained within 24 h post conventional trans-arterial chemoembolization, and 40-220 days after conventional trans-arterial chemoembolization using volumetric image analysis software. Tumor response was assessed on contrast-enhanced magnetic resonance imaging 1 month after conventional trans-arterial chemoembolization. RESULTS The washout rate was longer for neuroendocrine tumors compared to colorectal carcinoma, with half-lives of 54.61 days (p<0.00001) and 19.39 days (p<0.001), respectively, with no exponential washout among intrahepatic cholangiocarcinomas (p=0.83). The half-life for lipiodol washout was longer in tumors larger than 300 cm3 compared to smaller tumors (25.43 vs. 22.71 days). Lipiodol wash out half-life was 54.76 days (p<0.01) and 29.45 days (p<0.00001) for tumors with a contrast enhancement burden of 60% or more and less than 60%, respectively. A negative exponential relationship for lipiodol washout was observed in non-responders (p<0.00001). CONCLUSION Lipiodol washout is a time-dependent process, and occurs faster in colorectal carcinoma tumors, tumors smaller than 300 cm3, tumors with baseline contrast enhancement burden of less than 60%, and non-responding target lesions.
Collapse
Affiliation(s)
- Nariman Nezami
- Section of Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, U.S.A.; .,Division of Vascular and Interventional Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, U.S.A
| | - Johanna Maria Mijntje VAN Breugel
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, U.S.A.,Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands.,Medical faculty, Utrecht University, Utrecht, the Netherlands
| | - Menelaos Konstantinidis
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Julius Chapiro
- Section of Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, U.S.A.,Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, U.S.A
| | - Lynn Jeanette Savic
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, U.S.A.,Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Milena Anna Miszczuk
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, U.S.A
| | - Irvin Rexha
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, U.S.A.,Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mingde Lin
- Visage Imaging, Inc., San Diego, CA, U.S.A
| | - Kelvin Hong
- Division of Vascular and Interventional Radiology, Russel H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Christos Georgiades
- Division of Vascular and Interventional Radiology, Russel H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| |
Collapse
|
17
|
Ghani MA, Fereydooni A, Chen E, Letzen B, Laage-Gaupp F, Nezami N, Deng Y, Gan G, Thakur V, Lin M, Papademetris X, Schernthaner RE, Huber S, Chapiro J, Hong K, Georgiades C. Identifying enhancement-based staging markers on baseline MRI in patients with colorectal cancer liver metastases undergoing intra-arterial tumor therapy. Eur Radiol 2021; 31:8858-8867. [PMID: 34061209 DOI: 10.1007/s00330-021-08058-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/07/2021] [Accepted: 05/06/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVES To determine if three-dimensional whole liver and baseline tumor enhancement features on MRI can serve as staging biomarkers and help predict survival of patients with colorectal cancer liver metastases (CRCLM) more accurately than one-dimensional and non-enhancement-based features. METHODS This retrospective study included 88 patients with CRCLM, treated with transarterial chemoembolization or Y90 transarterial radioembolization between 2001 and 2014. Semi-automated segmentations of up to three dominant lesions were performed on pre-treatment MRI to calculate total tumor volume (TTV) and total liver volumes (TLV). Quantitative 3D analysis was performed to calculate enhancing tumor volume (ETV), enhancing tumor burden (ETB, calculated as ETV/TLV), enhancing liver volume (ELV), and enhancing liver burden (ELB, calculated as ELV/TLV). Overall and enhancing tumor diameters were also measured. A modified Kaplan-Meier method was used to determine appropriate cutoff values for each metric. The predictive value of each parameter was assessed by Kaplan-Meier survival curves and univariable and multivariable cox proportional hazard models. RESULTS All methods except whole liver (ELB, ELV) and one-dimensional/non-enhancement-based methods were independent predictors of survival. Multivariable analysis showed a HR of 2.1 (95% CI 1.3-3.4, p = 0.004) for enhancing tumor diameter, HR 1.7 (95% CI 1.1-2.8, p = 0.04) for TTV, HR 2.3 (95% CI 1.4-3.9, p < 0.001) for ETV, and HR 2.4 (95% CI 1.4-4.0, p = 0.001) for ETB. CONCLUSIONS Tumor enhancement of CRCLM on baseline MRI is strongly associated with patient survival after intra-arterial therapy, suggesting that enhancing tumor volume and enhancing tumor burden are better prognostic indicators than non-enhancement-based and one-dimensional-based markers. KEY POINTS • Tumor enhancement of colorectal cancer liver metastases on MRI prior to treatment with intra-arterial therapies is strongly associated with patient survival. • Three-dimensional, enhancement-based imaging biomarkers such as enhancing tumor volume and enhancing tumor burden may serve as the basis of a novel prognostic staging system for patients with liver-dominant colorectal cancer metastases.
Collapse
Affiliation(s)
- Mansur A Ghani
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA
| | - Arash Fereydooni
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA
| | - Evan Chen
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA
| | - Brian Letzen
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA
| | - Fabian Laage-Gaupp
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA
| | - Nariman Nezami
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA.,Russel H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD, USA.,Interventional Radiology and Image-Guided Medicine, Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Yanhong Deng
- Yale Center for Analytical Science, Yale School of Public Health, New Haven, CT, USA
| | - Geliang Gan
- Yale Center for Analytical Science, Yale School of Public Health, New Haven, CT, USA
| | - Vinayak Thakur
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA
| | - Xenophon Papademetris
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ruediger E Schernthaner
- Russel H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD, USA.,Department of Diagnostic and Interventional Radiology, Hospital Landstraße, Vienna, Austria
| | - Steffen Huber
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA
| | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT, USA. .,Russel H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Kelvin Hong
- Russel H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Christos Georgiades
- Russel H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD, USA
| |
Collapse
|
18
|
Letzen BS, Malpani R, Miszczuk M, de Ruiter QMB, Petty CW, Rexha I, Nezami N, Laage-Gaupp F, Lin M, Schlachter TR, Chapiro J. Lipiodol as an intra-procedural imaging biomarker for liver tumor response to transarterial chemoembolization: Post-hoc analysis of a prospective clinical trial. Clin Imaging 2021; 78:194-200. [PMID: 34022765 DOI: 10.1016/j.clinimag.2021.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 05/12/2021] [Accepted: 05/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The use of the ethiodized oil- Lipiodol in conventional trans-arterial chemoembolization (cTACE) ensures radiopacity to visualize drug delivery in the process of providing selective drug targeting to hepatic cancers and arterial embolization. Lipiodol functions as a carrier of chemo drugs for targeted therapy, as an embolic agent, augmenting the drug effect by efflux into the portal veins as well as a predictor for the tumor response and survival. PURPOSE To prospectively evaluate the role of 3D quantitative assessment of intra-procedural Lipiodol deposition in liver tumors on CBCT immediately after cTACE as a predictive biomarker for the outcome of cTACE. MATERIALS & METHODS This was a post-hoc analysis of data from an IRB-approved prospective clinical trial. Thirty-two patients with hepatocellular carcinoma or liver metastases underwent contrast enhanced CBCT obtained immediately after cTACE, unenhanced MDCT at 24 h after cTACE, and follow-up imaging 30-, 90- and 180-days post-procedure. Lipiodol deposition was quantified on CBCT after cTACE and was characterized by 4 ordinal levels: ≤25%, >25-50%, >50-75%, >75%. Tumor response was assessed on follow-up MRI. Lipiodol deposition on imaging, correlation between Lipiodol deposition and tumor response criteria, and correlation between Lipiodol coverage and median overall survival (MOS) were evaluated. RESULTS Image analysis demonstrated a high degree of agreement between the Lipiodol deposition on CBCT and the 24 h post-TACE CT, with a Bland-Altman plot of Lipiodol deposition on imaging demonstrated a bias of 2.75, with 95%-limits-of-agreement: -16.6 to 22.1%. An inverse relationship between Lipiodol deposition in responders versus non-responders for two-dimensional EASL reached statistical significance at 30 days (p = 0.02) and 90 days (p = 0.05). Comparing the Lipiodol deposition in Modified Response Evaluation Criteria in Solid Tumors (mRECIST) responders versus non-responders showed a statistically significant higher volumetric deposition in responders for European Association for the Study of the Liver (EASL)-30d, EASL-90d, and quantitative EASL-180d. The correlation between the relative Lipiodol deposition and the change in enhancing tumor volume showed a negative association post-cTACE (30-day: p < 0.001; rho = -0.63). A Kaplan-Meier analysis for patients with high vs. low Lipiodol deposition showed a MOS of 46 vs. 33 months (p = 0.05). CONCLUSION 3D quantification of Lipiodol deposition on intra-procedural CBCT is a predictive biomarker of outcome in patients with primary or metastatic liver cancer undergoing cTACE. There are spatial and volumetric agreements between 3D quantification of Lipiodol deposition on intra-procedural CBCT and 24 h post-cTACE MDCT. The spatial and volumetric agreement between Lipiodol deposition on intra-procedural CBCT and 24 h post-cTACE MDCT could suggest that acquiring MDCT 24 h after cTACE is redundant. Importantly, the demonstrated relationship between levels of tumor coverage with Lipiodol and degree and timeline of tumor response after cTACE underline the role of Lipiodol as an intra-procedural surrogate for tumor response, with potential implications for the prediction of survival.
Collapse
Affiliation(s)
- Brian S Letzen
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - Rohil Malpani
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - Milena Miszczuk
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA; Department of Radiology, Charité University School of Medicine, Charitépl. 1, 10117 Berlin, Germany
| | - Quirina M B de Ruiter
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA; Philips Healthcare, Image Guided Therapy, Amstelplein 2, Amsterdam 1096 BC, Netherlands
| | - Christopher W Petty
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - Irvin Rexha
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA; Department of Radiology, Charité University School of Medicine, Charitépl. 1, 10117 Berlin, Germany
| | - Nariman Nezami
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA; Division of Interventional Radiology and Image-Guided Medicine, Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1364 Clifton Road Northeast, Atlanta, GA 30322, USA
| | - Fabian Laage-Gaupp
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA; Visage Imaging Inc., 12625 High Bluff Drive, Suite 205, San Diego, CA 92130, USA
| | - Todd R Schlachter
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
19
|
Ding J, Xiao H, Deng W, Liu F, Zhu R, Ha R. Feasibility of quantitative and volumetric enhancement measurement to assess tumor response in patients with breast cancer after early neoadjuvant chemotherapy. J Int Med Res 2021; 49:300060521991017. [PMID: 33682494 PMCID: PMC7944542 DOI: 10.1177/0300060521991017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Objective To evaluate the feasibility of quantitative enhancing lesion volume (ELV) for evaluating the responsiveness of breast cancer patients to early neoadjuvant chemotherapy (NAC) using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Methods Seventy-five women with breast cancer underwent DCE-MRI before and after NAC. Lesions were assessed by ELV, response evaluation criteria in solid tumors 1.1 (RECIST 1.1), and total lesion volume (TLV). The diagnostic and pathological predictive performances of the methods were compared and color maps were compared with pathological results. Results ELV identified 29%, 67%, and 4% of cases with partial response, stable disease, and progressive disease, respectively. There was no significant difference in evaluation performances among the methods. The sensitivity, specificity, positive predictive value, negative predictive value (NPV), and accuracy of ELV for predicting pathologic response were 72%, 92%, 81.8%, 86.8%, and 85.3%, respectively, with the highest sensitivity, NPV, and accuracy of the three methods. The area under the receiver operating characteristic curve was also highest for ELV. Pre- and post-NAC color maps reflecting tumor activity were consistent with pathological necrosis. Conclusions ELV may help evaluate the responsiveness of breast cancer patients to NAC, and may provide a good tumor-response indicator through the ability to indicate tumor viability.
Collapse
Affiliation(s)
- Jie Ding
- Medical Imaging Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Hongyan Xiao
- The Pathology Department, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | | | - Fengjiao Liu
- Medical Imaging Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Rongrong Zhu
- Medical Imaging Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Ruoshui Ha
- Medical Imaging Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| |
Collapse
|
20
|
Role of 3D quantitative tumor analysis for predicting overall survival after conventional chemoembolization of intrahepatic cholangiocarcinoma. Sci Rep 2021; 11:9337. [PMID: 33927226 PMCID: PMC8085245 DOI: 10.1038/s41598-021-88426-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023] Open
Abstract
This study was designed to assess 3D vs. 1D and 2D quantitative tumor analysis for prediction of overall survival (OS) in patients with Intrahepatic Cholangiocarcinoma (ICC) who underwent conventional transarterial chemoembolization (cTACE). 73 ICC patients who underwent cTACE were included in this retrospective analysis between Oct 2001 and Feb 2015. The overall and enhancing tumor diameters and the maximum cross-sectional and enhancing tumor areas were measured on baseline images. 3D quantitative tumor analysis was used to assess total tumor volume (TTV), enhancing tumor volume (ETV), and enhancing tumor burden (ETB) (ratio between ETV and liver volume). Patients were divided into low (LTB) and high tumor burden (HTB) groups. There was a significant separation between survival curves of the LTB and HTB groups using enhancing tumor diameter (p = 0.003), enhancing tumor area (p = 0.03), TTV (p = 0.03), and ETV (p = 0.01). Multivariate analysis showed a hazard ratio of 0.46 (95%CI: 0.27–0.78, p = 0.004) for enhancing tumor diameter, 0.56 (95% CI 0.33–0.96, p = 0.04) for enhancing tumor area, 0.58 (95%CI: 0.34–0.98, p = 0.04) for TTV, and 0.52 (95%CI: 0.30–0.91, p = 0.02) for ETV. TTV and ETV, as well as the largest enhancing tumor diameter and maximum enhancing tumor area, reliably predict the OS of patients with ICC after cTACE and could identify ICC patients who are most likely to benefit from cTACE.
Collapse
|
21
|
Cost-Effectiveness of Imaging Tumor Response Criteria in Hepatocellular Cancer After Transarterial Chemoembolization. J Am Coll Radiol 2021; 18:927-934. [PMID: 33484726 DOI: 10.1016/j.jacr.2020.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Several tumor response criteria on cross-sectional imaging have been used in hepatocellular cancer after locoregional, intra-arterial therapy. The cost implications of their efficacy and accuracy are not well established. PURPOSE To evaluate cost-effectiveness of quantitative European Association for Study of the Liver (qEASL) compared with Response Evaluation Criteria in Solid Tumors (RECIST) and modified RECIST (mRECIST) response criteria. MATERIALS AND METHODS A Markov decision-analytic model was constructed, accounting for both costs and outcomes from a payor perspective. Three different tumor imaging response criteria were evaluated: (1) qEASL, (2) RECIST, and (3) mRECIST. Input parameters were derived from the most comprehensive literature available focusing on the assessment of liver tumor response after transarterial chemoembolization. Deterministic and probabilistic sensitivity analyses were performed. RESULTS Base case calculation showed qEASL to be the dominant strategy, with the highest effectiveness (1.06 quality-adjusted life years (QALY), as compared with 1.05 QALY in mRECIST and 1.02 QALY in RECIST). The expected costs of qEASL, mRECIST, and RECIST were $451,773, $460,489, and $459,004, respectively. qEASL was more cost-effective than RECIST in 71.50% of the 10,000 iterations and mRECIST in 69.26% of the iterations. One-way sensitivity analysis varying the cost showed that qEASL remained cost-effective until its additional cost was above $9,994. CONCLUSION Our study demonstrates qEASL to be the most cost-effective tumor response assessment criterion, with substantial cost savings as compared with RECIST and mRECIST for patients with hepatocellular carcinoma after transarterial chemoembolization.
Collapse
|
22
|
Stolley DL, Crouch AC, Özkan A, Seeley EH, Whitley EM, Rylander MN, Cressman ENK. Combining Chemistry and Engineering for Hepatocellular Carcinoma: Nano-Scale and Smaller Therapies. Pharmaceutics 2020; 12:E1243. [PMID: 33419304 PMCID: PMC7766014 DOI: 10.3390/pharmaceutics12121243] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022] Open
Abstract
Primary liver cancer, or hepatocellular carcinoma (HCC), is a major worldwide cause of death from carcinoma. Most patients are not candidates for surgery and medical therapies, including new immunotherapies, have not shown major improvements since the modest benefit seen with the introduction of sorafenib over a decade ago. Locoregional therapies for intermediate stage disease are not curative but provide some benefit. However, upon close scrutiny, there is still residual disease in most cases. We review the current status for treatment of intermediate stage disease, summarize the literature on correlative histopathology, and discuss emerging methods at micro-, nano-, and pico-scales to improve therapy. These include transarterial hyperthermia methods and thermoembolization, along with microfluidics model systems and new applications of mass spectrometry imaging for label-free analysis of pharmacokinetics and pharmacodynamics.
Collapse
Affiliation(s)
- Danielle L. Stolley
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA; (D.L.S.); (M.N.R.)
| | - Anna Colleen Crouch
- Interventional Radiology, M.D. Anderson Cancer Center, Houston, TX 77030, USA; (A.C.C.); (E.M.W.)
| | - Aliçan Özkan
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA;
| | - Erin H. Seeley
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712, USA;
| | - Elizabeth M. Whitley
- Interventional Radiology, M.D. Anderson Cancer Center, Houston, TX 77030, USA; (A.C.C.); (E.M.W.)
| | - Marissa Nichole Rylander
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA; (D.L.S.); (M.N.R.)
| | - Erik N. K. Cressman
- Interventional Radiology, M.D. Anderson Cancer Center, Houston, TX 77030, USA; (A.C.C.); (E.M.W.)
| |
Collapse
|
23
|
Zaid M, Widmann L, Dai A, Sun K, Zhang J, Zhao J, Hurd MW, Varadhachary GR, Wolff RA, Maitra A, Katz MHG, Herman JM, Wang H, Knopp MV, Williams TM, Bhosale P, Tamm EP, Koay EJ. Predictive Modeling for Voxel-Based Quantification of Imaging-Based Subtypes of Pancreatic Ductal Adenocarcinoma (PDAC): A Multi-Institutional Study. Cancers (Basel) 2020; 12:3656. [PMID: 33291471 PMCID: PMC7762105 DOI: 10.3390/cancers12123656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 01/19/2023] Open
Abstract
Previously, we characterized qualitative imaging-based subtypes of pancreatic ductal adenocarcinoma (PDAC) on computed tomography (CT) scans. Conspicuous (high delta) PDAC tumors are more likely to have aggressive biology and poorer clinical outcomes compared to inconspicuous (low delta) tumors. Here, we developed a quantitative classification of this imaging-based subtype (quantitative delta; q-delta). Retrospectively, baseline pancreatic protocol CT scans of three cohorts (cohort#1 = 101, cohort#2 = 90 and cohort#3 = 16 [external validation]) of patients with PDAC were qualitatively classified into high and low delta. We used a voxel-based method to volumetrically quantify tumor enhancement while referencing normal-pancreatic-parenchyma and used machine learning-based analysis to build a predictive model. In addition, we quantified the stromal content using hematoxylin- and eosin-stained treatment-naïve PDAC sections. Analyses revealed that PDAC quantitative enhancement values are predictive of the qualitative delta scoring and were used to build a classification model (q-delta). Compared to high q-delta, low q-delta tumors were associated with improved outcomes, and the q-delta class was an independent prognostic factor for survival. In addition, low q-delta tumors had higher stromal content and lower cellularity compared to high q-delta tumors. Our results suggest that q-delta classification provides a clinically and biologically relevant tool that may be integrated into ongoing and future clinical trials.
Collapse
Affiliation(s)
- Mohamed Zaid
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Lauren Widmann
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Annie Dai
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Kevin Sun
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Jie Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Jun Zhao
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.Z.); (M.W.H.)
| | - Mark W. Hurd
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.Z.); (M.W.H.)
| | - Gauri R. Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.R.V.); (R.A.W.)
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.R.V.); (R.A.W.)
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.M.); (H.W.)
| | - Matthew H. G. Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Joseph M. Herman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.M.); (H.W.)
| | - Michael V. Knopp
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Terence M. Williams
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Priya Bhosale
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.B.); (E.P.T.)
| | - Eric P. Tamm
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.B.); (E.P.T.)
| | - Eugene J. Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| |
Collapse
|
24
|
King MJ, Tong A, Dane B, Huang C, Zhan C, Shanbhogue K. Response assessment of hepatocellular carcinoma treated with yttrium-90 radioembolization: inter-reader variability, comparison with 3D quantitative approach, and role in the prediction of clinical outcomes. Eur J Radiol 2020; 133:109351. [DOI: 10.1016/j.ejrad.2020.109351] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/05/2020] [Accepted: 10/11/2020] [Indexed: 12/26/2022]
|
25
|
Tegel BR, Huber S, Savic LJ, Lin M, Gebauer B, Pollak J, Chapiro J. Quantification of contrast-uptake as imaging biomarker for disease progression of renal cell carcinoma after tumor ablation. Acta Radiol 2020; 61:1708-1716. [PMID: 32216452 DOI: 10.1177/0284185120909964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The prognosis of patients with renal cell carcinoma (RCC) depends greatly on the presence of extra-renal metastases. PURPOSE To investigate the value of total tumor volume (TTV) and enhancing tumor volume (ETV) as three-dimensional (3D) quantitative imaging biomarkers for disease aggressiveness in patients with RCC. MATERIAL AND METHODS Retrospective, HIPAA-compliant, IRB-approved study including 37 patients with RCC treated with image-guided thermal ablation during 2007-2015. TNM stage, RENAL Nephrometry Score, largest tumor diameter, TTV, and ETV were assessed on cross-sectional imaging at baseline and correlated with outcome measurements. The primary outcome was time-to-occurrence of extra-renal metastases and the secondary outcome was progression-free survival (PFS). Correlation was assessed using a Cox regression model and differences in outcomes were shown by Kaplan-Meier plots with significance and odds ratios (OR) calculated by Log-rank test/generalized Wilcoxon and continuity-corrected Woolf logit method. RESULTS Patients with a TTV or ETV > 5 cm3 were more likely to develop distant metastases compared to patients with TTV (OR 6.69, 95% confidence interval [CI] 0.33-134.4, P=0.022) or ETV (OR 8.48, 95% CI 0.42-170.1, P=0.016) < 5 cm3. Additionally, PFS was significantly worse in patients with larger ETV (P = 0.039; median PFS 51.87 months vs. 69.97 months). In contrast, stratification by median value of the established, caliper-based measurements showed no significant correlation with outcome parameters. CONCLUSION ETV, as surrogate of lesion vascularity, is a sensitive imaging biomarker for occurrence of extra-renal metastatic disease and PFS in patients with RCC.
Collapse
Affiliation(s)
- Bruno R Tegel
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität Berlin and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - Steffen Huber
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| | - Lynn J Savic
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität Berlin and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - MingDe Lin
- U/S Imaging and Interventions, Philips Research North America, Cambridge, MA, USA
| | - Bernhard Gebauer
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität Berlin and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - Jeffrey Pollak
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| | - Julius Chapiro
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| |
Collapse
|
26
|
Domaratius C, Settmacher U, Malessa C, Teichgräber U. Transarterial chemoembolization with drug-eluting beads in patients with hepatocellular carcinoma: response analysis with mRECIST. Diagn Interv Radiol 2020; 27:85-93. [PMID: 33135664 DOI: 10.5152/dir.2020.19439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE According to the Barcelona Clinic Liver Cancer (BCLC) staging classification, transarterial chemoembolization (TACE) is the treatment of choice for intermediate hepatocellular carcinoma (HCC). Thereby, the use of drug-eluting beads (DEB) as embolic agents has been recently established in clinical practice. The aim of this study was to evaluate tumor response after DEB-TACE. METHODS This retrospective study was approved by the institutional ethics committee. Overall, 89 patients with HCC (Child Pugh A or B) receiving DEB-TACE as palliative treatment option or as bridging before liver transplantation were included in the study. Tumor response was assessed by modified response evaluation criteria in solid tumors (mRECIST) and a tumor growth rate. Survival analysis was performed using Kaplan-Meier estimator with log-rank testing and Cox proportional hazards. RESULTS A total of 188 TACE procedures were performed between 2006 and 2010. After the last intervention, 18% achieved complete response, 45% achieved partial response, 28% had stable disease and 9% had progressive disease. Using the tumor growth rate, 90% of all patients showed a tumor reduction between first and final response evaluation. The 6-month, 1-, 2- and 3-year overall survival rates were 86.5%, 67.4%, 47.2%, and 33.7%, with a median survival of 45, 24, 15, and 14 months for complete response, partial response, stable disease, and progressive disease, respectively. Tumor reduction showed a positive effect on survival. CONCLUSION DEB-TACE offers conclusive response results with mRECIST and proves a strong tendency of tumor reduction on survival benefits. Therefore, tumor growth rate represents a possible parameter to predict survival.
Collapse
Affiliation(s)
- Claudia Domaratius
- Department of Radiology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Utz Settmacher
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Christina Malessa
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Ulf Teichgräber
- Department of Radiology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
27
|
Reliable prediction of survival in advanced-stage hepatocellular carcinoma treated with sorafenib: comparing 1D and 3D quantitative tumor response criteria on MRI. Eur Radiol 2020; 31:2737-2746. [PMID: 33123796 DOI: 10.1007/s00330-020-07381-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/07/2020] [Accepted: 10/06/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES To compare 1D and 3D quantitative tumor response criteria applied to DCE-MRI in patients with advanced-stage HCC undergoing sorafenib therapy to predict overall survival (OS) early during treatment. METHODS This retrospective analysis included 29 patients with advanced-stage HCC who received sorafenib for at least 60 days. All patients underwent baseline and follow-up DCE-MRI at 81.5 ± 29.3 days (range 35-140 days). Response to sorafenib was assessed in 46 target lesions using 1D criteria RECIST1.1 and mRECIST. In addition, a segmentation-based 3D quantification of absolute enhancing lesion volume (vqEASL) was performed on the arterial phase MRI, and the enhancement fraction of total tumor volume (%qEASL) was calculated. Accordingly, patients were stratified into groups of disease control (DC) and disease progression (DP). OS was evaluated using Kaplan-Meier curves with log-rank test and Cox proportional hazards regression model. RESULTS The Kaplan-Meier analysis revealed that stratification of patients in DC vs. DP according to mRECIST (p = 0.0371) and vqEASL (p = 0.0118) successfully captured response and stratified OS, while stratification according to RECIST and %qEASL did not correlate with OS (p = 0.6273 and p = 0.7474, respectively). Multivariable Cox regression identified tumor progression according to mRECIST and qEASL as independent risk factors of decreased OS (p = 0.039 and p = 0.006, respectively). CONCLUSIONS The study identified enhancement-based vqEASL and mRECIST as reliable predictors of patient survival early after initiation of treatment with sorafenib. This data provides evidence for potential advantages 3D quantitative, enhancement-based tumor response analysis over conventional techniques regarding early identification of treatment success or failure. KEY POINTS • Tumor response criteria on MRI can be used to predict survival benefit of sorafenib therapy in patients with advanced HCC. • Stratification into DC and DP using mRECIST and vqEASL significantly correlates with OS (p = 0.0371 and p = 0.0118, respectively) early after initiation of sorafenib, while stratification according to RECIST and %qEASL did not correlate with OS (p = 0.6273 and p = 0.7474, respectively). • mRECIST (HR = 0.325, p = 0.039. 95%CI 0.112-0.946) and qEASL (HR = 0.183, p = 0.006, 95%CI 0.055-0.613) are independent prognostic factors of survival in HCC patients undergoing sorafenib therapy.
Collapse
|
28
|
Automated feature quantification of Lipiodol as imaging biomarker to predict therapeutic efficacy of conventional transarterial chemoembolization of liver cancer. Sci Rep 2020; 10:18026. [PMID: 33093524 PMCID: PMC7582153 DOI: 10.1038/s41598-020-75120-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023] Open
Abstract
Conventional transarterial chemoembolization (cTACE) is a guideline-approved image-guided therapy option for liver cancer using the radiopaque drug-carrier and micro-embolic agent Lipiodol, which has been previously established as an imaging biomarker for tumor response. To establish automated quantitative and pattern-based image analysis techniques of Lipiodol deposition on 24 h post-cTACE CT as biomarker for treatment response. The density of Lipiodol deposits in 65 liver lesions was automatically quantified using Hounsfield Unit thresholds. Lipiodol deposition within the tumor was automatically assessed for patterns including homogeneity, sparsity, rim, and peripheral deposition. Lipiodol deposition was correlated with enhancing tumor volume (ETV) on baseline and follow-up MRI. ETV on baseline MRI strongly correlated with Lipiodol deposition on 24 h CT (p < 0.0001), with 8.22% ± 14.59 more Lipiodol in viable than necrotic tumor areas. On follow-up, tumor regions with Lipiodol showed higher rates of ETV reduction than areas without Lipiodol (p = 0.0475) and increasing densities of Lipiodol enhanced this effect. Also, homogeneous (p = 0.0006), non-sparse (p < 0.0001), rim deposition within sparse tumors (p = 0.045), and peripheral deposition (p < 0.0001) of Lipiodol showed improved response. This technical innovation study showed that an automated threshold-based volumetric feature characterization of Lipiodol deposits is feasible and enables practical use of Lipiodol as imaging biomarker for therapeutic efficacy after cTACE.
Collapse
|
29
|
Ghosn M, Derbel H, Kharrat R, Oubaya N, Mulé S, Chalaye J, Regnault H, Amaddeo G, Itti E, Luciani A, Kobeiter H, Tacher V. Prediction of overall survival in patients with hepatocellular carcinoma treated with Y-90 radioembolization by imaging response criteria. Diagn Interv Imaging 2020; 102:35-44. [PMID: 33012693 DOI: 10.1016/j.diii.2020.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/20/2020] [Accepted: 09/07/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE To evaluate the potential of imaging criteria in predicting overall survival of patients with hepatocellular carcinoma (HCC) after a first transcatheter arterial yttrium-90 radioembolization (TARE) MATERIALS AND METHODS: From October 2013 to July 2017, 37 patients with HCC were retrospectively included. There were 34 men and 3 women with a mean age of 60.5±10.2 (SD) years (range: 32.7-78.9 years). Twenty-five patients (68%) were Barcelona Clinic Liver Cancer (BCLC) C and 12 (32%) were BCLC B. Twenty-four primary index tumors (65%) were>5cm. Three radiologists evaluated tumor response on pre- and 4-7 months post-TARE magnetic resonance imaging or computed tomography examinations, using Response Evaluation Criteria in Solid Tumors (RECIST) 1.1, modified RECIST (mRECIST), European Association for Study of the Liver (EASL), volumetric RECIST (vRECIST), quantitative EASL (qEASL) and the Liver Imaging Reporting and Data System treatment response algorithm. Kaplan-Meier survival curves were used to compare responders and non-responders for each criterion. Univariate and multivariate Cox proportional hazard ratio (HR) analysis were used to identify covariates associated with overall survival. Fleiss kappa test was used to assess interobserver agreement. RESULTS At multivariate analysis, RECIST 1.1 (HR: 0.26; 95% confidence interval [95% CI]: 0.09-0.75; P=0.01), mRECIST (HR: 0.22; 95% CI: 0.08-0.59; P=0.003), EASL (HR: 0.22; 95% CI: 0.07-0.63; P=0.005), and qEASL (HR: 0.30; 95% CI: 0.12-0.80; P=0.02) showed a significant difference in overall survival between responders and nonresponders. RECIST 1.1 had the highest interobserver reproducibility. CONCLUSION RECIST and mRECIST seem to be the best compromise between reproducibility and ability to predict overall survival in patients with HCC treated with TARE.
Collapse
Affiliation(s)
- M Ghosn
- Department of Medical Imaging, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France.
| | - H Derbel
- Department of Medical Imaging, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France; Unité Inserm 955, équipe 18, IMRB, University of Paris Est Créteil, 94010 Créteil, France
| | - R Kharrat
- Department of Medical Imaging, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - N Oubaya
- Public Health Department, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - S Mulé
- Department of Medical Imaging, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France; Unité Inserm 955, équipe 18, IMRB, University of Paris Est Créteil, 94010 Créteil, France
| | - J Chalaye
- Department of Nuclear Medicine, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du-Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - H Regnault
- Department of Hepatology, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France; Unité Inserm 955, équipe 18, IMRB, University of Paris Est Créteil, 94010 Créteil, France
| | - G Amaddeo
- Department of Hepatology, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France; Unité Inserm 955, équipe 18, IMRB, University of Paris Est Créteil, 94010 Créteil, France
| | - E Itti
- Department of Nuclear Medicine, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du-Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - A Luciani
- Department of Medical Imaging, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France; Unité Inserm 955, équipe 18, IMRB, University of Paris Est Créteil, 94010 Créteil, France
| | - H Kobeiter
- Department of Medical Imaging, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France; Unité Inserm 955, Équipe 8, IMRB, University of Paris Est Créteil, 94010 Créteil, France
| | - V Tacher
- Department of Medical Imaging, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France; Unité Inserm 955, équipe 18, IMRB, University of Paris Est Créteil, 94010 Créteil, France
| |
Collapse
|
30
|
Liu C, Smolka S, Papademetris X, Minh DD, Gan G, Deng Y, Lin M, Chapiro J, Wang X, Georgiades C, Hong K. Predicting Infiltrative Hepatocellular Carcinoma Patient Outcome Post-TACE: MR Bias Field Correction Effect on 3D-quantitative Image Analysis. J Clin Transl Hepatol 2020; 8:292-298. [PMID: 33083252 PMCID: PMC7562808 DOI: 10.14218/jcth.2020.00054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/13/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022] Open
Abstract
Background and Aims: To investigate the impact of MR bias field correction on response determination and survival prediction using volumetric tumor enhancement analysis in patients with infiltrative hepatocellular carcinoma, after transcatheter arterial chemoembolization (TACE). Methods: This study included 101 patients treated with conventional or drug-eluting beads TACE between the years of 2001 and 2013. Semi-automated 3D quantification software was used to segment and calculate the enhancing tumor volume (ETV) of the liver with and without bias-field correction on multi-phasic contrast-enhanced MRI before and 1-month after initial TACE. ETV (expressed as cm3) at baseline imaging and the relative change in ETV (as % change, ETV%) before and after TACE were used to predict response and survival, respectively. Statistical survival analyses included Kaplan-Meier curve generation and Cox proportional hazards modeling. Q statistics were calculated and used to identify the best cut-off value for ETV to separate responders and non-responders (ETV cm3). The difference in survival was evaluated between responders and non-responders using Kaplan-Meier and Cox models. Results: MR bias field correction correlated with improved response calculation from baseline MR as well as survival after TACE; using a 415 cm3 cut-off for ETV at baseline (hazard ratio: 2.00, 95% confidence interval: 1.23-3.26, p=0.01) resulted in significantly improved response prediction (median survival in patients with baseline ETV <415 cm3: 19.66 months vs. ≥415 cm3: 9.21 months, p<0.001, log-rank test). A ≥41% relative decrease in ETV (hazard ratio: 0.58, 95%confidence interval: 0.37-0.93, p=0.02) was significant in predicting survival (ETV ≥41%: 19.20 months vs. ETV <41%: 8.71 months, p=0.008, log-rank test). Without MR bias field correction, response from baseline ETV could be predicted but survival after TACE could not. Conclusions: MR bias field correction improves both response assessment and accuracy of survival prediction using whole liver tumor enhancement analysis from baseline MR after initial TACE in patients with infiltrative hepatocellular carcinoma.
Collapse
Affiliation(s)
- Cuihong Liu
- Department of Ultrasound Diagnosis and Treatment, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Susanne Smolka
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Diagnostic and Interventional Radiology, Charité University Hospital, Berlin, Germany
| | | | - Duc Do Minh
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Diagnostic and Interventional Radiology, Charité University Hospital, Berlin, Germany
| | - Geliang Gan
- School of Public Health, Yale University, New Haven, CT, USA
| | - Yanhong Deng
- School of Public Health, Yale University, New Haven, CT, USA
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Christos Georgiades
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Kelvin Hong
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, MD, USA
| |
Collapse
|
31
|
Gregory J, Dioguardi Burgio M, Corrias G, Vilgrain V, Ronot M. Evaluation of liver tumour response by imaging. JHEP Rep 2020; 2:100100. [PMID: 32514496 PMCID: PMC7267412 DOI: 10.1016/j.jhepr.2020.100100] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
The goal of assessing tumour response on imaging is to identify patients who are likely to benefit - or not - from anticancer treatment, especially in relation to survival. The World Health Organization was the first to develop assessment criteria. This early score, which assessed tumour burden by standardising lesion size measurements, laid the groundwork for many of the criteria that followed. This was then improved by the Response Evaluation Criteria in Solid Tumours (RECIST) which was quickly adopted by the oncology community. At the same time, many interventional oncology treatments were developed to target specific features of liver tumours that result in significant changes in tumours but have little effect on tumour size. New criteria focusing on the viable part of tumours were therefore designed to provide more appropriate feedback to guide patient management. Targeted therapy has resulted in a breakthrough that challenges conventional response criteria due to the non-linear relationship between response and tumour size, requiring the development of methods that emphasize the appearance of tumours. More recently, research into functional and quantitative imaging has created new opportunities in liver imaging. These results have suggested that certain parameters could serve as early predictors of response or could predict later tumour response at baseline. These approaches have now been extended by machine learning and deep learning. This clinical review focuses on the progress made in the evaluation of liver tumours on imaging, discussing the rationale for this approach, addressing challenges and controversies in the field, and suggesting possible future developments.
Collapse
Key Words
- (c)TACE, (conventional) transarterial chemoembolisation
- (m)RECIST, (modified) Response Evaluation Criteria in Solid Tumours
- 18F-FDG, 18F-fluorodeoxyglucose
- 90Y, yttrium-90
- ADC, apparent diffusion coefficient
- APHE, arterial phase hyperenhancement
- CEUS, contrast-enhanced ultrasound
- CRLM, colorectal liver metastases
- DWI, diffusion-weighted imaging
- EASL
- EASL, European Association for the Study of the Liver criteria
- GIST, gastrointestinal stromal tumours
- HCC, hepatocellular carcinoma
- HU, Hounsfield unit
- Imaging
- LI-RADS
- LI-RADS, Liver Imaging Reporting And Data System
- Liver
- Metastases
- PD, progressive disease
- PET, positron emission tomography
- PR, partial response
- RECIST
- SD, stable disease
- SIRT, selective internal radiotherapy
- TR, treatment response
- Tumours
- WHO, World Health Organization
- mRECIST
Collapse
Affiliation(s)
- Jules Gregory
- Department of Radiology, APHP, University Hospitals Paris Nord Val de Seine, Beaujon, Clichy, France
- University of Paris, Paris, France
- INSERM U1149, CRI, Paris, France
| | - Marco Dioguardi Burgio
- Department of Radiology, APHP, University Hospitals Paris Nord Val de Seine, Beaujon, Clichy, France
- University of Paris, Paris, France
- INSERM U1149, CRI, Paris, France
| | - Giuseppe Corrias
- Department of Radiology, APHP, University Hospitals Paris Nord Val de Seine, Beaujon, Clichy, France
- University of Paris, Paris, France
- INSERM U1149, CRI, Paris, France
| | - Valérie Vilgrain
- Department of Radiology, APHP, University Hospitals Paris Nord Val de Seine, Beaujon, Clichy, France
- University of Paris, Paris, France
- INSERM U1149, CRI, Paris, France
| | - Maxime Ronot
- Department of Radiology, APHP, University Hospitals Paris Nord Val de Seine, Beaujon, Clichy, France
- University of Paris, Paris, France
- INSERM U1149, CRI, Paris, France
| |
Collapse
|
32
|
Moawad AW, Fuentes D, Khalaf AM, Blair KJ, Szklaruk J, Qayyum A, Hazle JD, Elsayes KM. Feasibility of Automated Volumetric Assessment of Large Hepatocellular Carcinomas' Responses to Transarterial Chemoembolization. Front Oncol 2020; 10:572. [PMID: 32457831 PMCID: PMC7221016 DOI: 10.3389/fonc.2020.00572] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is the most common liver malignancy and the leading cause of death in patients with cirrhosis. Various treatments for HCC are available, including transarterial chemoembolization (TACE), which is the commonest intervention performed in HCC. Radiologic tumor response following TACE is an important prognostic factor for patients with HCC. We hypothesized that, for large HCC tumors, assessment of treatment response made with automated volumetric response evaluation criteria in solid tumors (RECIST) might correlate with the assessment made with the more time- and labor-intensive unidimensional modified RECIST (mRECIST) and manual volumetric RECIST (M-vRECIST) criteria. Accordingly, we undertook this retrospective study to compare automated volumetric RECIST (A-vRECIST) with M-vRECIST and mRESIST for the assessment of large HCC tumors' responses to TACE. Methods:We selected 42 pairs of contrast-enhanced computed tomography (CT) images of large HCCs. Images were taken before and after TACE, and in each of the images, the HCC was segmented using both a manual contouring tool and a convolutional neural network. Three experienced radiologists assessed tumor response to TACE using mRECIST criteria. The intra-class correlation coefficient was used to assess inter-reader reliability in the mRECIST measurements, while the Pearson correlation coefficient was used to assess correlation between the volumetric and mRECIST measurements. Results:Volumetric tumor assessment using automated and manual segmentation tools showed good correlation with mRECIST measurements. For A-vRECIST and M-vRECIST, respectively, r = 0.597 vs. 0.622 in the baseline studies; 0.648 vs. 0.748 in the follow-up studies; and 0.774 vs. 0.766 in the response assessment (P < 0.001 for all). The A-vRECIST evaluation showed high correlation with the M-vRECIST evaluation (r = 0.967, 0.937, and 0.826 in baseline studies, follow-up studies, and response assessment, respectively, P < 0.001 for all). Conclusion:Volumetric RECIST measurements are likely to provide an early marker for TACE monitoring, and automated measurements made with a convolutional neural network may be good substitutes for manual volumetric measurements.
Collapse
Affiliation(s)
- Ahmed W. Moawad
- Imaging Physics Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David Fuentes
- Imaging Physics Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ahmed M. Khalaf
- Diagnostic Radiology Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katherine J. Blair
- Diagnostic Radiology Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Janio Szklaruk
- Diagnostic Radiology Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aliya Qayyum
- Diagnostic Radiology Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John D. Hazle
- Imaging Physics Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Khaled M. Elsayes
- Diagnostic Radiology Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
33
|
Aykan NF, Özatlı T. Objective response rate assessment in oncology: Current situation and future expectations. World J Clin Oncol 2020; 11:53-73. [PMID: 32133275 PMCID: PMC7046919 DOI: 10.5306/wjco.v11.i2.53] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 11/05/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023] Open
Abstract
The tumor objective response rate (ORR) is an important parameter to demonstrate the efficacy of a treatment in oncology. The ORR is valuable for clinical decision making in routine practice and a significant end-point for reporting the results of clinical trials. World Health Organization and Response Evaluation Criteria in Solid Tumors (RECIST) are anatomic response criteria developed mainly for cytotoxic chemotherapy. These criteria are based on the visual assessment of tumor size in morphological images provided by computed tomography (CT) or magnetic resonance imaging. Anatomic response criteria may not be optimal for biologic agents, some disease sites, and some regional therapies. Consequently, modifications of RECIST, Choi criteria and Morphologic response criteria were developed based on the concept of the evaluation of viable tumors. Despite its limitations, RECIST v1.1 is validated in prospective studies, is widely accepted by regulatory agencies and has recently shown good performance for targeted cancer agents. Finally, some alternatives of RECIST were developed as immune-specific response criteria for checkpoint inhibitors. Immune RECIST criteria are based essentially on defining true progressive disease after a confirmatory imaging. Some graphical methods may be useful to show longitudinal change in the tumor burden over time. Tumor tissue is a tridimensional heterogenous mass, and tumor shrinkage is not always symmetrical; thus, metabolic response assessments using positron emission tomography (PET) or PET/CT may reflect the viability of cancer cells or functional changes evolving after anticancer treatments. The metabolic response can show the benefit of a treatment earlier than anatomic shrinkage, possibly preventing delays in drug approval. Computer-assisted automated volumetric assessments, quantitative multimodality imaging in radiology, new tracers in nuclear medicine and finally artificial intelligence have great potential in future evaluations.
Collapse
Affiliation(s)
- Nuri Faruk Aykan
- Department of Medical Oncology, Istinye University Medical School, Bahcesehir Liv Hospital, Istanbul 34510, Turkey
| | - Tahsin Özatlı
- Department of Medical Oncology, Istinye University Medical School, Bahcesehir Liv Hospital, Istanbul 34510, Turkey
| |
Collapse
|
34
|
Comparing HCC arterial tumour vascularisation on baseline imaging and after lipiodol cTACE: how do estimations of enhancing tumour volumes differ on contrast-enhanced MR and CT? Eur Radiol 2019; 30:1601-1608. [PMID: 31811428 DOI: 10.1007/s00330-019-06430-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/05/2019] [Accepted: 08/27/2019] [Indexed: 01/11/2023]
Abstract
OBJECTIVES In this study, pre-treatment target lesion vascularisation in either contrast-enhanced (CE) CT or MRI and post-treatment lipiodol deposition in native CT scans were compared in HCC patients who underwent their first cTACE treatment. We analysed the impact of stratification according to cTACE selectivity on these correlations. METHODS Seventy-eight HCC patients who underwent their first cTACE procedure were retrospectively included. Pre-treatment tumour vascularisation in arterial contrast phase and post-treatment lipiodol deposition in native CT scans were evaluated using the qEASL (quantitative tumour enhancement) method. Correlations were analysed using scatter plots, the Pearson correlation coefficient (PCC) and linear regression analysis. Subgroup analysis was performed according to lobar, segmental and subsegmental execution of cTACE. RESULTS Arterial tumour volumes in both baseline CE CT (R2 = 0.83) and CE MR (R2 = 0.82) highly correlated with lipiodol deposition after cTACE. The regression coefficient between lipiodol deposition and enhancing tumour volume was 1.39 for CT and 0.33 for MR respectively, resulting in a ratio of 4.24. After stratification according to selectivity of cTACE, the regression coefficient was 0.94 (R2 = 1) for lobar execution, 1.38 (R2 = 0.96) for segmental execution and 1.88 (R2 = 0.89) for subsegmental execution in the CE CT group. CONCLUSIONS Volumetric lipiodol deposition can be used as a reference to compare different imaging modalities in detecting vital tumour volumes. That approach proved CE MRI to be more sensitive than CE CT. Selectivity of cTACE significantly impacts the respective regression coefficients which allows for an innovative approach to the assessment of technical success after cTACE with a multitude of possible applications. KEY POINTS • Lipiodol deposition after cTACE highly correlates with pre-treatment tumour vascularisation and can be used as a reference to compare different imaging modalities in detecting vital tumour volumes. • Lipiodol deposition also correlates with the selectivity of cTACE and can therefore be used to quantify the technical success of the intervention.
Collapse
|
35
|
Savic LJ, Schobert IT, Peters D, Walsh JJ, Laage-Gaupp FM, Hamm CA, Tritz N, Doemel LA, Lin M, Sinusas A, Schlachter T, Duncan JS, Hyder F, Coman D, Chapiro J. Molecular Imaging of Extracellular Tumor pH to Reveal Effects of Locoregional Therapy on Liver Cancer Microenvironment. Clin Cancer Res 2019; 26:428-438. [PMID: 31582517 DOI: 10.1158/1078-0432.ccr-19-1702] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/24/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE To establish magnetic resonance (MR)-based molecular imaging paradigms for the noninvasive monitoring of extracellular pH (pHe) as a functional surrogate biomarker for metabolic changes induced by locoregional therapy of liver cancer. EXPERIMENTAL DESIGN Thirty-two VX2 tumor-bearing New Zealand white rabbits underwent longitudinal imaging on clinical 3T-MRI and CT scanners before and up to 2 weeks after complete conventional transarterial chemoembolization (cTACE) using ethiodized oil (lipiodol) and doxorubicin. MR-spectroscopic imaging (MRSI) was employed for pHe mapping. Multiparametric MRI and CT were performed to quantify tumor enhancement, diffusion, and lipiodol coverage of the tumor posttherapy. In addition, incomplete cTACE with reduced chemoembolic doses was applied to mimic undertreatment and exploit pHe mapping to detect viable tumor residuals. Imaging findings were correlated with histopathologic markers indicative of metabolic state (HIF-1α, GLUT-1, and LAMP-2) and viability (proliferating cell nuclear antigen and terminal deoxynucleotidyl-transferase dUTP nick-end labeling). RESULTS Untreated VX2 tumors demonstrated a significantly lower pHe (6.80 ± 0.09) than liver parenchyma (7.19 ± 0.03, P < 0.001). Upregulation of HIF-1α, GLUT-1, and LAMP-2 confirmed a hyperglycolytic tumor phenotype and acidosis. A gradual tumor pHe increase toward normalization similar to parenchyma was revealed within 2 weeks after complete cTACE, which correlated with decreasing detectability of metabolic markers. In contrast, pHe mapping after incomplete cTACE indicated both acidic viable residuals and increased tumor pHe of treated regions. Multimodal imaging revealed durable tumor devascularization immediately after complete cTACE, gradually increasing necrosis, and sustained lipiodol coverage of the tumor. CONCLUSIONS MRSI-based pHe mapping can serve as a longitudinal monitoring tool for viable tumors. As most liver tumors are hyperglycolytic creating microenvironmental acidosis, therapy-induced normalization of tumor pHe may be used as a functional biomarker for positive therapeutic outcome.
Collapse
Affiliation(s)
- Lynn Jeanette Savic
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.,Institute of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, Berlin, Germany
| | - Isabel Theresa Schobert
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.,Institute of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, Berlin, Germany
| | - Dana Peters
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - John J Walsh
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Fabian Max Laage-Gaupp
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Charlie Alexander Hamm
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.,Institute of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, Berlin, Germany
| | - Nina Tritz
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Luzie A Doemel
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.,Institute of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, Berlin, Germany
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.,Visage Imaging, Inc., San Diego, California
| | - Albert Sinusas
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.,Department of Internal Medicine (Cardiology), Yale School of Medicine, New Haven, Connecticut
| | - Todd Schlachter
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - James S Duncan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.,Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Daniel Coman
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
36
|
Imber BS, Lin AL, Zhang Z, Keshavamurthy KN, Deipolyi AR, Beal K, Cohen MA, Tabar V, DeAngelis LM, Geer EB, Yang TJ, Young RJ. Comparison of Radiographic Approaches to Assess Treatment Response in Pituitary Adenomas: Is RECIST or RANO Good Enough? J Endocr Soc 2019; 3:1693-1706. [PMID: 31528829 PMCID: PMC6735764 DOI: 10.1210/js.2019-00130] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/25/2019] [Indexed: 12/29/2022] Open
Abstract
Context Pituitary adenomas (PA) are often irregularly shaped, particularly posttreatment. There are no standardized radiographic criteria for assessing treatment response, substantially complicating interpretation of prospective outcome data. Existing imaging frameworks for intracranial tumors assume perfectly spherical targets and may be suboptimal. Objective To compare a three-dimensional (3D) volumetric approach against accepted surrogate measurements to assess PA posttreatment response (PTR). Design Retrospective review of patients with available pre- and postradiotherapy (RT) imaging. A neuroradiologist determined tumor sizes in one dimensional (1D) per Response Evaluation in Solid Tumors (RECIST) criteria, two dimensional (2D) per Response Assessment in Neuro-Oncology (RANO) criteria, and 3D estimates assuming a perfect sphere or perfect ellipsoid. Each tumor was manually segmented for 3D volumetric measurements. The Hakon Wadell method was used to calculate sphericity. Setting Tertiary cancer center. Patients or Other Participants Patients (n = 34, median age = 50 years; 50% male) with PA and MRI scans before and after sellar RT. Interventions Patients received sellar RT for intact or surgically resected lesions. Main Outcome Measures Radiographic PTR, defined as percent tumor size change. Results Using 3D volumetrics, mean sphericity = 0.63 pre-RT and 0.60 post-RT. With all approaches, most patients had stable disease on post-RT scan. PTR for 1D, 2D, and 3D spherical measurements were moderately well correlated with 3D volumetrics (e.g., for 1D: 0.66, P < 0.0001) and were superior to 3D ellipsoid. Intraclass correlation coefficient demonstrated moderate to good reliability for 1D, 2D, and 3D sphere (P < 0.001); 3D ellipsoid was inferior (P = 0.009). 3D volumetrics identified more potential partially responding and progressive lesions. Conclusions Although PAs are irregularly shaped, 1D and 2D approaches are adequately correlated with volumetric assessment.
Collapse
Affiliation(s)
- Brandon S Imber
- Department of Radiation Oncology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Andrew L Lin
- Department of Neurology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Zhigang Zhang
- Department of Epidemiology & Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Krishna Nand Keshavamurthy
- Department of Radiology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Amy Robin Deipolyi
- Department of Radiology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Kathryn Beal
- Department of Radiation Oncology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Marc A Cohen
- Department of Surgery, Head & Neck Service, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Viviane Tabar
- Department of Neurosurgery, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Lisa M DeAngelis
- Department of Neurology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Eliza B Geer
- Department of Endocrinology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - T Jonathan Yang
- Department of Radiation Oncology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Robert J Young
- Department of Radiology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
37
|
van Breugel JMM, Geschwind JF, Mirpour S, Savic LJ, Zhang X, Duran R, Lin M, Miszczuk M, Liapi E, Chapiro J. Theranostic application of lipiodol for transarterial chemoembolization in a VX2 rabbit liver tumor model. Theranostics 2019; 9:3674-3686. [PMID: 31281506 PMCID: PMC6587357 DOI: 10.7150/thno.32943] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED The goal of this study was to investigate the role of Lipiodol as a tumor-specific imaging biomarker to determine therapeutic efficacy of cTACE and investigate its inter-dependency with tumor perfusion using radiological-pathological correlation in an animal model of liver cancer. METHODS A total of N=36 rabbits were implanted in the left lobe of the liver with VX2 tumors, treated with cTACE using doxorubicin suspended in Lipiodol, and randomly sacrificed at 24 h, 7 days, or 20 days post-TACE. Unenhanced and contrast-enhanced CT scans including a perfusion protocol were obtained before cTACE and immediately before sacrifice. Tumor vascularity and Lipiodol deposition within tumors and hepatic tissue (non-target deposits) were quantified using 3D quantitative assessment tools and measurements of arterial flow, portal flow, and perfusion index (PI). After sacrifice histologic staining, including hematoxylin and eosin (H&E), CD31, and Oil Red O (ORO) were performed on tumor and liver samples to evaluate necrosis, microvascular density (MVD), and Lipiodol retention over time. Transmission electron microscopy (TEM) was performed to assess Lipiodol deposition and clearance over time. RESULTS All cTACE procedures were carried out successfully except for one, which was excluded from further analysis. Twenty-four hours post-TACE, tumor PI (p=0.04) was significantly decreased, which was maintained at 7 days (p=0.003), but not at 20 days (p=0.4). A strong correlation (R2 = 0.894) was found between the volume of enhancing tumor tissue at baseline and Lipiodol-positive tumor volume post-TACE. Both ORO and TEM showed deposition of Lipiodol across all imaging time points within the VX2 tumors. However, gradual and ultimately near-complete Lipiodol washout was observed over time in the non-tumoral liver. MVD decreased between 24 h and 7 days post-TACE, and then increased 20 days post-TACE (both p<0.01). CONCLUSIONS Our data provide radiology-pathology evidence for the function of Lipiodol as a theranostic, tumor-specific drug delivery agent because it is both imageable and tumor-seeking, whereby it is preferentially taken up and retained by tumor cells. Those tumor-specific functions also enable Lipiodol to act as an imaging biomarker for the therapeutic efficacy of cTACE. Together with volumetric quantification of tumor vascularization on CT, Lipiodol could be used as a predictor of a patient's response to cTACE and contribute to the therapeutic management of patients with liver cancer.
Collapse
Affiliation(s)
- Johanna Maria Mijntje van Breugel
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Medical faculty, Utrecht University, Utrecht, The Netherlands
| | | | - Sahar Mirpour
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Lynn Jeanette Savic
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Xuchen Zhang
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Rafael Duran
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Milena Miszczuk
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Eleni Liapi
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
38
|
Automated Volumetric Assessment of Hepatocellular Carcinoma Response to Sorafenib: A Pilot Study. J Comput Assist Tomogr 2019; 43:499-506. [PMID: 31082956 DOI: 10.1097/rct.0000000000000866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE This pilot study evaluates the feasibility of automated volumetric quantification of hepatocellular carcinoma (HCC) as an imaging biomarker to assess treatment response for sorafenib. METHODS In this institutional review board-approved, Health Insurance Portability and Accountability Act-compliant retrospective study, a training database of manually labeled background liver, enhancing and nonenhancing tumor tissue was established using pretherapy and first posttherapy multiphasic computed tomography images from a registry of 13 HCC patients. For each patient, Hounsfield density and geometry-based feature images were generated from registered multiphasic computed tomography data sets and used as the input for a random forest-based classifier of enhancing and nonenhancing tumor tissue. Leave-one-out cross-validation of the dice similarity measure was applied to quantify the classifier accuracy. A Cox regression model was used to confirm volume changes as predictors of time to progression (TTP) of target lesions for both manual and automatic methods. RESULTS When compared with manual labels, an overall classification accuracy of dice similarity coefficient of 0.71 for pretherapy and 0.66 posttherapy enhancing tumor labels and 0.45 for pretherapy and 0.59 for posttherapy nonenhancing tumor labels was observed. Automated methods for quantifying volumetric changes in the enhancing lesion agreed with manual methods and were observed as a significant predictor of TTP. CONCLUSIONS Automated volumetric analysis was determined to be feasible for monitoring HCC response to treatment. The information extracted using automated volumetrics is likely to reproduce labor-intensive manual data and provide a good predictor for TTP. Further work will extend these studies to additional treatment modalities and larger patient populations.
Collapse
|
39
|
Patella F, Pesapane F, Fumarola E, Zannoni S, Brambillasca P, Emili I, Costa G, Anderson V, Levy EB, Carrafiello G, Wood BJ. Assessment of the response of hepatocellular carcinoma to interventional radiology treatments. Future Oncol 2019; 15:1791-1804. [PMID: 31044615 DOI: 10.2217/fon-2018-0747] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
According to Barcelona Clinic Liver Cancer (BCLC) guidelines, interventional radiology procedures are valuable treatment options for many hepatocellular carcinomas (HCCs) that are not amenable to resection or transplantation. Accurate assessment of the efficacy of therapies at earlier stages enables completion of treatment, optimal follow-up and to prevent potentially unnecessary treatments, side effects and costly failure. The goal of this review is to summarize and describe the radiological strategies that have been proposed to predict survival and to stratify HCC responses after interventional radiology therapies. New techniques currently in development are also described.
Collapse
Affiliation(s)
- Francesca Patella
- Postgraduate School of Radiodiagnostics, University of Milan, Milan, Italy.,Center for Interventional Oncology, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Filippo Pesapane
- Postgraduate School of Radiodiagnostics, University of Milan, Milan, Italy.,Center for Interventional Oncology, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Enrico Fumarola
- Postgraduate School of Radiodiagnostics, University of Milan, Milan, Italy.,Center for Interventional Oncology, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Stefania Zannoni
- Postgraduate School of Radiodiagnostics, University of Milan, Milan, Italy
| | | | - Ilaria Emili
- Postgraduate School of Radiodiagnostics, University of Milan, Milan, Italy
| | - Guido Costa
- Università degli Studi di Milano, Postgraduate School of General Surgery, Milan, Italy
| | - Victoria Anderson
- Center for Interventional Oncology, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Elliot B Levy
- Center for Interventional Oncology, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Bradford J Wood
- Center for Interventional Oncology, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
40
|
Abajian A, Murali N, Savic LJ, Laage-Gaupp FM, Nezami N, Duncan JS, Schlachter T, Lin M, Geschwind JF, Chapiro J. Predicting Treatment Response to Image-Guided Therapies Using Machine Learning: An Example for Trans-Arterial Treatment of Hepatocellular Carcinoma. J Vis Exp 2018. [PMID: 30371657 DOI: 10.3791/58382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intra-arterial therapies are the standard of care for patients with hepatocellular carcinoma who cannot undergo surgical resection. The objective of this study was to develop a method to predict response to intra-arterial treatment prior to intervention. The method provides a general framework for predicting outcomes prior to intra-arterial therapy. It involves pooling clinical, demographic and imaging data across a cohort of patients and using these data to train a machine learning model. The trained model is applied to new patients in order to predict their likelihood of response to intra-arterial therapy. The method entails the acquisition and parsing of clinical, demographic and imaging data from N patients who have already undergone trans-arterial therapies. These data are parsed into discrete features (age, sex, cirrhosis, degree of tumor enhancement, etc.) and binarized into true/false values (e.g., age over 60, male gender, tumor enhancement beyond a set threshold, etc.). Low-variance features and features with low univariate associations with the outcome are removed. Each treated patient is labeled according to whether they responded or did not respond to treatment. Each training patient is thus represented by a set of binary features and an outcome label. Machine learning models are trained using N - 1 patients with testing on the left-out patient. This process is repeated for each of the N patients. The N models are averaged to arrive at a final model. The technique is extensible and enables inclusion of additional features in the future. It is also a generalizable process that may be applied to clinical research questions outside of interventional radiology. The main limitation is the need to derive features manually from each patient. A popular modern form of machine learning called deep learning does not suffer from this limitation, but requires larger datasets.
Collapse
Affiliation(s)
- Aaron Abajian
- Department of Radiology and Biomedical Imaging, Yale School of Medicine
| | - Nikitha Murali
- Department of Radiology and Biomedical Imaging, Yale School of Medicine
| | - Lynn Jeanette Savic
- Department of Radiology and Biomedical Imaging, Yale School of Medicine; Department of Diagnostic and Interventional Radiology, Universitätsmedizin Charité Berlin
| | | | - Nariman Nezami
- Department of Radiology and Biomedical Imaging, Yale School of Medicine
| | - James S Duncan
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science
| | - Todd Schlachter
- Department of Radiology and Biomedical Imaging, Yale School of Medicine
| | | | | | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale School of Medicine;
| |
Collapse
|
41
|
Vande Lune P, Abdel Aal AK, Klimkowski S, Zarzour JG, Gunn AJ. Hepatocellular Carcinoma: Diagnosis, Treatment Algorithms, and Imaging Appearance after Transarterial Chemoembolization. J Clin Transl Hepatol 2018; 6:175-188. [PMID: 29951363 PMCID: PMC6018317 DOI: 10.14218/jcth.2017.00045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 11/02/2017] [Accepted: 12/02/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common cause of cancer-related death, with incidence increasing worldwide. Unfortunately, the overall prognosis for patients with HCC is poor and many patients present with advanced stages of disease that preclude curative therapies. Diagnostic and interventional radiologists play a key role in the management of patients with HCC. Diagnostic radiologists can use contrast-enhanced computed tomography (CT), magnetic resonance imaging, and ultrasound to diagnose and stage HCC, without the need for pathologic confirmation, by following established criteria. Once staged, the interventional radiologist can treat the appropriate patients with percutaneous ablation, transarterial chemoembolization, or radioembolization. Follow-up imaging after these liver-directed therapies for HCC can be characterized according to various radiologic response criteria; although, enhancement-based criteria, such as European Association for the Study of the Liver and modified Response Evaluation Criteria in Solid Tumors, are more reflective of treatment effect in HCC. Newer imaging technologies like volumetric analysis, dual-energy CT, cone beam CT and perfusion CT may provide additional benefits for patients with HCC.
Collapse
Affiliation(s)
- Patrick Vande Lune
- University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Ahmed K. Abdel Aal
- Division of Vascular and Interventional Radiology, Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sergio Klimkowski
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jessica G. Zarzour
- Division of Abdominal Imaging, Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew J. Gunn
- Division of Vascular and Interventional Radiology, Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
- *Correspondence to: Andrew J. Gunn, Division of Vascular and Interventional Radiology, Department of Radiology, University of Alabama at Birmingham, 619 19 St S, NHB 623, Birmingham, AL 35249, USA. Tel: +1-205-975-4850, Fax: +1-205-975-5257, E-mail:
| |
Collapse
|
42
|
Zhu LZ, Xu S, Qian HL. Transarterial embolization and low-dose continuous hepatic arterial infusion chemotherapy with oxaliplatin and raltitrexed for hepatocellular carcinoma with major portal vein tumor thrombus. World J Gastroenterol 2018; 24:2501-2507. [PMID: 29930471 PMCID: PMC6010942 DOI: 10.3748/wjg.v24.i23.2501] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/09/2018] [Accepted: 03/29/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To determine the efficacy and safety of transarterial embolization and low-dose continuous hepatic arterial infusion chemotherapy with oxaliplatin and raltitrexed in hepatocellular carcinoma (HCC) with major portal vein tumor thrombus (MPVTT).
METHODS Eighty-six patients with MPVTT accepted routine embolization. The catheter was kept in the hepatic artery and oxaliplatin (50 mg in 250 mL of glucose) was infused by pump for 4 h, followed by raltitrexed (2 mg in 100 mL of 0.9% saline) infusion by pump for the next 1 h. The efficacy and safety were evaluated after the transarterial chemoembolization (TACE).
RESULTS Full or partial embolization was achieved in 86 cases, where all the cases received low dose continuous hepatic arterial infusion chemotherapy. Complete responses (CRs), partial responses (PRs), stable disease (SD), and disease progression (PD) for intrahepatic disease were observed in 0, 45, 20, and 21 patients, respectively. The 1-, 2-and 3-year overall survival rates of the 86 patients were 40.7%, 22.1%, and 8.1% respectively, and the median survival time was 8.7 mo. Complication was limited.
CONCLUSION TACE with low dose continuous hepatic arterial infusion of oxaliplatin and raltitrexed could be an option in MPVTT patient; it was shown to be effective in patients with advanced HCC with MPVTT with less toxicity.
Collapse
Affiliation(s)
- Lin-Zhong Zhu
- Department of Interventional Therapy, Beijing Cancer Hospital, Beijing 100142, China
| | - Song Xu
- Department of Radiology, Yunnan Second People’s Hospital, Kunming 650021, Yunnan Province, China
| | - Hai-Long Qian
- Interventional Therapy, Baotou Cancer Hospital, Baotou 014030, Inner Mongolia, China
| |
Collapse
|
43
|
Abajian A, Murali N, Savic LJ, Laage-Gaupp FM, Nezami N, Duncan JS, Schlachter T, Lin M, Geschwind JF, Chapiro J. Predicting Treatment Response to Intra-arterial Therapies for Hepatocellular Carcinoma with the Use of Supervised Machine Learning-An Artificial Intelligence Concept. J Vasc Interv Radiol 2018; 29:850-857.e1. [PMID: 29548875 DOI: 10.1016/j.jvir.2018.01.769] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/08/2018] [Accepted: 01/11/2018] [Indexed: 02/06/2023] Open
Abstract
PURPOSE To use magnetic resonance (MR) imaging and clinical patient data to create an artificial intelligence (AI) framework for the prediction of therapeutic outcomes of transarterial chemoembolization by applying machine learning (ML) techniques. MATERIALS AND METHODS This study included 36 patients with hepatocellular carcinoma (HCC) treated with transarterial chemoembolization. The cohort (age 62 ± 8.9 years; 31 men; 13 white; 24 Eastern Cooperative Oncology Group performance status 0, 10 status 1, 2 status 2; 31 Child-Pugh stage A, 4 stage B, 1 stage C; 1 Barcelona Clinic Liver Cancer stage 0, 12 stage A, 10 stage B, 13 stage C; tumor size 5.2 ± 3.0 cm; number of tumors 2.6 ± 1.1; and 30 conventional transarterial chemoembolization, 6 with drug-eluting embolic agents). MR imaging was obtained before and 1 month after transarterial chemoembolization. Image-based tumor response to transarterial chemoembolization was assessed with the use of the 3D quantitative European Association for the Study of the Liver (qEASL) criterion. Clinical information, baseline imaging, and therapeutic features were used to train logistic regression (LR) and random forest (RF) models to predict patients as treatment responders or nonresponders under the qEASL response criterion. The performance of each model was validated using leave-one-out cross-validation. RESULTS Both LR and RF models predicted transarterial chemoembolization treatment response with an overall accuracy of 78% (sensitivity 62.5%, specificity 82.1%, positive predictive value 50.0%, negative predictive value 88.5%). The strongest predictors of treatment response included a clinical variable (presence of cirrhosis) and an imaging variable (relative tumor signal intensity >27.0). CONCLUSIONS Transarterial chemoembolization outcomes in patients with HCC may be predicted before procedures by combining clinical patient data and baseline MR imaging with the use of AI and ML techniques.
Collapse
Affiliation(s)
- Aaron Abajian
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520
| | - Nikitha Murali
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520
| | - Lynn Jeanette Savic
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520; Department of Diagnostic and Interventional Radiology, Universitätsmedizin Charité Berlin, Berlin, Germany
| | - Fabian Max Laage-Gaupp
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520
| | - Nariman Nezami
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520
| | - James S Duncan
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut
| | - Todd Schlachter
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520
| | - MingDe Lin
- Philips Research North America, Cambridge, Massachusetts
| | | | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520.
| |
Collapse
|
44
|
Riaz A, Gabr A, Abouchaleh N, Ali R, Al Asadi A, Mora R, Kulik L, Desai K, Thornburg B, Mouli S, Hickey R, Miller FH, Yaghmai V, Ganger D, Lewandowski RJ, Salem R. Radioembolization for hepatocellular carcinoma: Statistical confirmation of improved survival in responders by landmark analyses. Hepatology 2018; 67:873-883. [PMID: 28833344 DOI: 10.1002/hep.29480] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/07/2017] [Accepted: 08/14/2017] [Indexed: 12/22/2022]
Abstract
UNLABELLED Does imaging response predict survival in hepatocellular carcinoma (HCC)? We studied the ability of posttherapeutic imaging response to predict overall survival. Over 14 years, 948 patients with HCC were treated with radioembolization. Patients with baseline metastases, vascular invasion, multifocal disease, Child-Pugh > B7, and transplanted/resected were excluded. This created our homogeneous study cohort of 134 patients with Child-Pugh ≤ B7 and solitary HCC. Response (using European Association for Study of the Liver [EASL] and Response Evaluation Criteria in Solid Tumors 1.1 [RECIST 1.1] criteria) was associated with survival using Landmark and risk-of-death methodologies after reviewing 960 scans. In a subanalysis, survival times of responders were compared to those of patients with stable disease (SD) and progressive disease (PD). Uni/multivariate survival analyses were performed at each Landmark. At the 3-month Landmark, responders survived longer than nonresponders by EASL (hazard ratio [HR], 0.46; confidence interval [CI], 0.26-0.82; P = 0.002) but not RECIST 1.1 criteria (HR, 0.70; CI, 0.37-1.32; P = 0.32). At the 6-month Landmark, responders survived longer than nonresponders by EASL (HR, 0.32; CI, 0.15-0.77; P < 0.001) and RECIST 1.1 criteria (HR, 0.50; CI, 0.29-0.87; P = 0.021). At the 12-month Landmark, responders survived longer than nonresponders by EASL (HR, 0.34; CI, 0.15-0.77; P < 0.001) and RECIST 1.1 criteria (HR, 0.52; CI 0.27-0.98; P = 0.049). At 6 months, risk of death was lower for responders by EASL (P < 0.001) and RECIST 1.1 (P = 0.0445). In subanalyses, responders lived longer than patients with SD or PD. EASL response was a significant predictor of survival at 3-, 6-, and 12-month Landmarks on uni/multivariate analyses. CONCLUSION Response to radioembolization in patients with solitary HCC can prognosticate improved survival. EASL necrosis criteria outperformed RECIST 1.1 size criteria in predicting survival. The therapeutic objective of radioembolization should be radiologic response and not solely to prevent progression. (Hepatology 2018;67:873-883).
Collapse
Affiliation(s)
- Ahsun Riaz
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Ahmed Gabr
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Nadine Abouchaleh
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Rehan Ali
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Ali Al Asadi
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Ronald Mora
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Laura Kulik
- Department of Medicine, Division of Hepatology, Northwestern University, Chicago, IL
| | - Kush Desai
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Bartley Thornburg
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Samdeep Mouli
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Ryan Hickey
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Frank H Miller
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Vahid Yaghmai
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Daniel Ganger
- Department of Medicine, Division of Hepatology, Northwestern University, Chicago, IL
| | - Robert J Lewandowski
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Riad Salem
- Department of Radiology, Section of Interventional Radiology, Northwestern Memorial Hospital, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL.,Department of Surgery, Division of Transplantation, Comprehensive Transplant Center, Northwestern University, Chicago, IL.,Department of Medicine, Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| |
Collapse
|
45
|
Pupulim L, Ronot M, Paradis V, Chemouny S, Vilgrain V. Volumetric measurement of hepatic tumors: Accuracy of manual contouring using CT with volumetric pathology as the reference method. Diagn Interv Imaging 2018; 99:83-89. [DOI: 10.1016/j.diii.2017.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/31/2017] [Accepted: 11/19/2017] [Indexed: 01/16/2023]
|
46
|
Amer AM, Zaid M, Chaudhury B, Elganainy D, Lee Y, Wilke CT, Cloyd J, Wang H, Maitra A, Wolff RA, Varadhachary G, Overman MJ, Lee JE, Fleming JB, Tzeng CW, Katz MH, Holliday EB, Krishnan S, Minsky BD, Herman JM, Taniguchi CM, Das P, Crane CH, Le O, Bhosale P, Tamm EP, Koay EJ. Imaging-based biomarkers: Changes in the tumor interface of pancreatic ductal adenocarcinoma on computed tomography scans indicate response to cytotoxic therapy. Cancer 2018; 124:1701-1709. [PMID: 29370450 PMCID: PMC5891375 DOI: 10.1002/cncr.31251] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/22/2017] [Accepted: 12/21/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The assessment of pancreatic ductal adenocarcinoma (PDAC) response to therapy remains challenging. The objective of this study was to investigate whether changes in the tumor/parenchyma interface are associated with response. METHODS Computed tomography (CT) scans before and after therapy were reviewed in 4 cohorts: cohort 1 (99 patients with stage I/II PDAC who received neoadjuvant chemoradiation and surgery); cohort 2 (86 patients with stage IV PDAC who received chemotherapy), cohort 3 (94 patients with stage I/II PDAC who received protocol‐based neoadjuvant gemcitabine chemoradiation), and cohort 4 (47 patients with stage I/II PDAC who received neoadjuvant chemoradiation and were prospectively followed in a registry). The tumor/parenchyma interface was visually classified as either a type I response (the interface remained or became well defined) or a type II response (the interface became poorly defined) after therapy. Consensus (cohorts 1‐3) and individual (cohort 4) visual scoring was performed. Changes in enhancement at the interface were quantified using a proprietary platform. RESULTS In cohort 1, type I responders had a greater probability of achieving a complete or near‐complete pathologic response (21% vs 0%; P = .01). For cohorts 1, 2, and 3, type I responders had significantly longer disease‐free and overall survival, independent of traditional covariates of outcomes and of baseline and normalized cancer antigen 19‐9 levels. In cohort 4, 2 senior radiologists achieved a κ value of 0.8, and the interface score was associated with overall survival. The quantitative method revealed high specificity and sensitivity in classifying patients as type I or type II responders (with an area under the receiver operating curve of 0.92 in cohort 1, 0.96 in cohort 2, and 0.89 in cohort 3). CONCLUSIONS Changes at the PDAC/parenchyma interface may serve as an early predictor of response to therapy. Cancer 2018;124:1701‐9. © 2018 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution‐NonCommercial‐NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made. An imaging feature of pancreatic cancer is identified that indicates a response to cytotoxic therapies. This may be helpful as an early predictor of response for clinical trials and for deciding whether to change therapy.
Collapse
Affiliation(s)
- Ahmed M Amer
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mohamed Zaid
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Baishali Chaudhury
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dalia Elganainy
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yeonju Lee
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher T Wilke
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jordan Cloyd
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gauri Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffery E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa Bay, Florida
| | - Ching Wei Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Matthew H Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emma B Holliday
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunil Krishnan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bruce D Minsky
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph M Herman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cullen M Taniguchi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher H Crane
- Department of Radiation Oncology, Memorial Sloan Cancer Center, New York, New York
| | - Ott Le
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Priya Bhosale
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric P Tamm
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
47
|
Hasdemir DB, Dávila LA, Schweitzer N, Meyer BC, Koch A, Vogel A, Wacker F, Rodt T. Evaluation of CT vascularization patterns for survival prognosis in patients with hepatocellular carcinoma treated by conventional TACE. Diagn Interv Radiol 2018; 23:217-222. [PMID: 28256449 DOI: 10.5152/dir.2016.16006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE Transarterial chemoembolization (TACE) is an established treatment for intermediate stage hepatocellular carcinoma (HCC). The aim of this retrospective study was to evaluate the power of lesion vascularization criteria based on computed tomography for prognosis of overall survival before initiation of treatment. METHODS A total of 59 patients with intermediate stage HCC treated with TACE as first-line treatment were retrospectively evaluated. TACE procedures were performed using doxorubicin, cisplatin, and lipiodol. Response evaluation criteria in solid tumors version 1.1 (RECIST 1.1) were used to determine the initial tumor response. Four vascularization patterns (VP) of the largest target lesion (homogeneous vascularization [VP1], homogeneous vascularization with additional arterial hypervascularization [VP2], heterogeneous vascularization with [VP3] and without zones of hypervascularization [VP4]) were assessed prior to the first TACE and correlated to survival. RESULTS Kaplan-Meier analysis yielded a median overall survival of 608 days (standard error [SE], 120.5 days). Survival analysis showed significant differences depending on the vascularization patterns (P = 0.012; hazard ratio, 0.327): patients with homogeneously vascularized lesions (VP1, VP2) had a median overall survival of 1091 days (SE, 235.5 days). Patients with heterogeneous vascularization of the lesion (VP3 and VP4) showed a median overall survival of 508 days (SE, 113.9 days). CONCLUSION The vascularization pattern of the largest HCC lesion is helpful for survival prognosis under TACE treatment and therefore has the potential to be used as an additional parameter for treatment stratification.
Collapse
Affiliation(s)
- Davut B Hasdemir
- Institutes of Diagnostic and Interventional Radiology, The Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Lewis H, Ghasabeh M, Khoshpouri P, Kamel I, Pawlik T. Functional hepatic imaging as a biomarker of primary and secondary tumor response to loco-regional therapies. Surg Oncol 2017; 26:411-422. [DOI: 10.1016/j.suronc.2017.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023]
|
49
|
Zhao Y, Duran R, Bai W, Sahu S, Wang W, Kabus S, Lin M, Han G, Geschwind JF. Which Criteria Applied in Multi-Phasic CT Can Predict Early Tumor Response in Patients with Hepatocellular Carcinoma Treated Using Conventional TACE: RECIST, mRECIST, EASL or qEASL? Cardiovasc Intervent Radiol 2017; 41:433-442. [PMID: 29086058 DOI: 10.1007/s00270-017-1829-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 10/19/2017] [Indexed: 12/27/2022]
Abstract
PURPOSE Our study aimed to evaluate quantitative tumor response assessment (quantitative EASL-[qEASL]) on computed tomography (CT) images in patients with hepatocellular carcinoma (HCC) treated using conventional transarterial chemoembolization (cTACE), compared to existing 1-dimensional and 2-dimensional methods (RECIST, mRECIST, EASL). MATERIALS AND METHODS In this IRB-approved, single-institution retrospective cohort study, 52 consecutive patients with intermediate-stage HCC were consecutively included. All patients underwent contrast-enhanced CT scan at baseline and 4 weeks after cTACE. RESULTS Median follow-up period was 13.5 months (range 1.2-54.1). RECIST, mRECIST and EASL identified progression in 2 (4%), 1 (2%) and 1 (2%) patients, respectively, whereas qEASL identified 10 (19%) patients. qEASL was the only tumor response method able to predict survival among different tumor response groups (P < 0.05), whereas RECIST, mRECIST and EASL did not (P > 0.05). Both EASL and qEASL were able to identify responders and non-responders and were predictive of survival (P < 0.05). Multivariate analysis showed that progression was an independent predictor of overall survival with hazard ratio of 1.9 (P = 0.025). Patients who demonstrated progression with qEASL had significantly shorter survival than those with non-progression (7.6 vs. 20.4 months, P = 0.012). Similar multivariate analysis using RECIST, mRECIST and EASL could not be performed because too few patients were categorized as progressive disease. CONCLUSION qEASL could be applied on CT images to assess tumor response following cTACE and is a more sensitive biomarker to predict survival and identify tumor progression than RECIST, mRECIST and EASL at an early time point. LEVEL OF EVIDENCE Level 2a, retrospective cohort study.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Liver Disease and Digestive Interventional Radiology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 27 West Changle Road, Xi'an, 710032, China.,Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Rafael Duran
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Wei Bai
- Department of Liver Disease and Digestive Interventional Radiology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 27 West Changle Road, Xi'an, 710032, China
| | - Sonia Sahu
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Wenjun Wang
- Department of Liver Disease and Digestive Interventional Radiology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 27 West Changle Road, Xi'an, 710032, China
| | - Sven Kabus
- Philips Research Hamburg, Hamburg, Germany
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA.,U/S Imaging and Interventions (UII), Philips Research North America, Cambridge, MS, USA
| | - Guohong Han
- Department of Liver Disease and Digestive Interventional Radiology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, No. 27 West Changle Road, Xi'an, 710032, China.
| | - Jean-François Geschwind
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA.,PreScience Labs LLC, Westport, CT, USA
| |
Collapse
|
50
|
Tovoli F, Renzulli M, Granito A, Golfieri R, Bolondi L. Radiologic criteria of response to systemic treatments for hepatocellular carcinoma. Hepat Oncol 2017; 4:129-137. [PMID: 30191059 PMCID: PMC6096444 DOI: 10.2217/hep-2017-0018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 10/16/2017] [Indexed: 02/07/2023] Open
Abstract
Sorafenib has been the only approved systemic therapy for hepatocellular carcinoma until very recently. However, the radiologic assessment of its biological activity is a disputed matter as at least five different criteria have been proposed. In this review, we describe the characteristic of the Response Evaluation Criteria In Solid Tumors (RECIST), European Association for the Study of The Liver (EASL), modified RECIST (mRECIST), Response Evaluation Criteria In the Cancer of the Liver (RECICL) and Choi criteria. The existing comparative studies are reported together with recent pieces of evidence, analyzing the reasons behind the split between recommendations of the scientific societies and regulatory agencies. Future perspectives in the wake of the impending results of the immunotherapy trials are also discussed.
Collapse
Affiliation(s)
- Francesco Tovoli
- Department of Medical & Surgical Sciences, University of Bologna, Italy
- *Author for correspondence: Tel.: +39 051 214 2214; Fax: +39 051 214 2725;
| | - Matteo Renzulli
- Radiology Unit, S.Orsola-Malpighi Bologna University Hospital, Italy
| | | | - Rita Golfieri
- Radiology Unit, S.Orsola-Malpighi Bologna University Hospital, Italy
| | - Luigi Bolondi
- Department of Medical & Surgical Sciences, University of Bologna, Italy
| |
Collapse
|