1
|
Haefliger L, Chapellier P, Vietti Violi N, Ledoux JB, Mantziari S, Schäfer M, Dromain C. Advancing Esophageal Cancer Staging and Restaging: The Role of MRI in Precision Diagnosis. Cancers (Basel) 2025; 17:1351. [PMID: 40282527 PMCID: PMC12026097 DOI: 10.3390/cancers17081351] [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: 02/11/2025] [Revised: 04/09/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
This review provides an in-depth analysis and comprehensive overview of recent advancements in MRI techniques for evaluating esophageal cancer (EC). It discusses the specific MRI acquisition protocols and parameters that enhance image quality and diagnostic accuracy. The review highlights MRI's role and performance in the initial TNM staging and its potential to refine treatment strategies by improving tumor delineation and characterization. Additionally, the paper explores MRI utility in restaging after NAT, focusing on its accuracy in assessing treatment response and detecting residual or recurrent disease. Comparisons with other imaging modalities currently used-such as endoscopic ultrasound (EUS), contrast-enhanced computed tomography (CE-CT), and 18F-fluorodeoxyglucose (FDG) positron emission tomography/CT (PET/CT)-are included to highlight the strengths and limitations of each method. Illustrated with numerous Figures, this article proposes a novel MRI-based strategy for EC staging and restaging. It aims to integrate MRI into clinical practice by leveraging its superior soft-tissue contrast and functional imaging capabilities to enhance diagnostic precision and improve patient outcomes. Through this comprehensive evaluation, the review underscores the potential of MRI to become a cornerstone in the precision diagnosis and management of EC.
Collapse
Affiliation(s)
- Laura Haefliger
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Pauline Chapellier
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Naik Vietti Violi
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Jean-Baptiste Ledoux
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
- CIBM Center for Biomedical Imaging, CH-1015 Lausanne, Switzerland
| | - Styliani Mantziari
- Department of Surgery, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Markus Schäfer
- Department of Surgery, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Clarisse Dromain
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| |
Collapse
|
2
|
Chapellier P, Fasquelle F, Saglietti C, Kinj R, Mantziari S, Schäfer M, Haefliger L, Jreige M, Vietti Violi N, Sempoux C, Dromain C. Prospective evaluation of MR-TRG (Tumor Regression Grade) in esophageal cancer after neo-adjuvant therapy: Preliminary results. Eur J Radiol 2024; 171:111263. [PMID: 38159523 DOI: 10.1016/j.ejrad.2023.111263] [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: 09/13/2023] [Revised: 12/03/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE To develop MRI-based criteria to assess tumor response to neoadjuvant therapies (NAT) of esophageal cancers (EC) and to evaluate its diagnostic performance in predicting the pathological Tumor Regression Grade (pTRG). METHOD From 2018 to 2022, patients with newly diagnosed locally advanced EC underwent MRI examinations for initial staging and restaging after NAT. Magnetic Resonance TRG (MR-TRG), equivalent to the Mandard and Becker classifications, were developed and independently assessed by two radiologists, blinded to pTRG, using T2W and DW-MR Images. All patients underwent surgery and benefited from a blinded pTRG evaluation by two pathologists. The agreement between readers and between MR-TRG and pTRG were assessed with Cohen's Kappa. The correlation of MR-TRG and pTRG was determined using Spearman's correlation. RESULTS 28 patients were included. Interrater agreement was substantial between radiologists, improved when grouping grade 1 and 2 (κ = 0.78 rose to 0,84 for Mandard and 0.68 to 0,78 for Becker score). Agreement between pTRG and MR-TRG was moderate with a percentaged agreement (p) = 87.5 %, kappa (κ) = 0.54 and p = 83.3 %, κ = 0.49 for Mandard and Becker, respectively. Agreement was improved to substantial when grouping grades 1-2 for Mandard and 1a-1b for Becker with p = 89.3 %, κ = 0.65 and p = 85.2 %, κ = 0.65 respectively. Sensitivity and specificity of MR-TRG in predicting pTRG were 88.2 % and 72.7 % for Mandard system (scores 1-2 versus 3-5), and 83.3 % and 80 % for Becker system (scores 1a-1b versus 2-3). CONCLUSION A substantial agreement between MR-TRG and pTRG was achieved when grouping grade 1-2. Hence, MR-TRG could be used as a surrogate of complete and near-complete pTRG.
Collapse
Affiliation(s)
- Pauline Chapellier
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - François Fasquelle
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Chiara Saglietti
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Rémy Kinj
- Service of radiation oncology, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Styliani Mantziari
- Department of Visceral Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Markus Schäfer
- Department of Visceral Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laura Haefliger
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Mario Jreige
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Naïk Vietti Violi
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christine Sempoux
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Clarisse Dromain
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
3
|
Lu S, Wang C, Liu Y, Chu F, Jia Z, Zhang H, Wang Z, Lu Y, Wang S, Yang G, Qu J. The MRI radiomics signature can predict the pathologic response to neoadjuvant chemotherapy in locally advanced esophageal squamous cell carcinoma. Eur Radiol 2024; 34:485-494. [PMID: 37540319 DOI: 10.1007/s00330-023-10040-4] [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: 03/08/2023] [Revised: 05/26/2023] [Accepted: 06/19/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVES To investigate the MRI radiomics signatures in predicting pathologic response among patients with locally advanced esophageal squamous cell carcinoma (ESCC), who received neoadjuvant chemotherapy (NACT). METHODS Patients who underwent NACT from March 2015 to October 2019 were prospectively included. Each patient underwent esophageal MR scanning within one week before NACT and within 2-3 weeks after completion of NACT, prior to surgery. Radiomics features extracted from T2-TSE-BLADE were randomly split into the training and validation sets at a ratio of 7:3. According to the progressive tumor regression grade (TRG), patients were stratified into two groups: good responders (GR, TRG 0 + 1) and poor responders (non-GR, TRG 2 + 3). We constructed the Pre/Post-NACT model (Pre/Post-model) and the Delta-NACT model (Delta-model). Kruskal-Wallis was used to select features, logistic regression was used to develop the final model. RESULTS A total of 108 ESCC patients were included, and 3/2/4 out of 107 radiomics features were selected for constructing the Pre/Post/Delta-model, respectively. The selected radiomics features were statistically different between GR and non-GR groups. The highest area under the curve (AUC) was for the Delta-model, which reached 0.851 in the training set and 0.831 in the validation set. Among the three models, Pre-model showed the poorest performance in the training and validation sets (AUC, 0.466 and 0.596), and the Post-model showed better performance than the Pre-model in the training and validation sets (AUC, 0.753 and 0.781). CONCLUSIONS MRI-based radiomics models can predict the pathological response after NACT in ESCC patients, with the Delta-model exhibiting optimal predictive efficacy. CLINICAL RELEVANCE STATEMENT MRI radiomics features could be used as a useful tool for predicting the efficacy of neoadjuvant chemotherapy in esophageal carcinoma patients, especially in selecting responders among those patients who may be candidates to benefit from neoadjuvant chemotherapy. KEY POINTS • The MRI radiomics features based on T2WI-TSE-BLADE could potentially predict the pathologic response to NACT among ESCC patients. • The Delta-model exhibited the best predictive ability for pathologic response, followed by the Post-model, which similarly had better predictive ability, while the Pre-model performed less well in predicting TRG.
Collapse
Affiliation(s)
- Shuang Lu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Chenglong Wang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200062, China
| | - Yun Liu
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200062, China
| | - Funing Chu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Zhengyan Jia
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Hongkai Zhang
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Zhaoqi Wang
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Yanan Lu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Shuting Wang
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Guang Yang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200062, China.
| | - Jinrong Qu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No. 127 Dongming Road, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
4
|
Veziant J, Bouché O, Aparicio T, Barret M, El Hajbi F, Lepilliez V, Lesueur P, Maingon P, Pannier D, Quero L, Raoul JL, Renaud F, Seitz JF, Serre AA, Vaillant E, Vermersch M, Voron T, Tougeron D, Piessen G. Esophageal cancer - French intergroup clinical practice guidelines for diagnosis, treatments and follow-up (TNCD, SNFGE, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO, ACHBT, SFP, RENAPE, SNFCP, AFEF, SFR). Dig Liver Dis 2023; 55:1583-1601. [PMID: 37635055 DOI: 10.1016/j.dld.2023.07.015] [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: 04/19/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION This document is a summary of the French intergroup guidelines regarding the management of esophageal cancer (EC) published in July 2022, available on the website of the French Society of Gastroenterology (SNFGE) (www.tncd.org). METHODS This collaborative work was conducted under the auspices of several French medical and surgical societies involved in the management of EC. Recommendations were graded in three categories (A, B and C), according to the level of evidence found in the literature until April 2022. RESULTS EC diagnosis and staging evaluation are mainly based on patient's general condition assessment, endoscopy plus biopsies, TAP CT-scan and 18F FDG-PET. Surgery alone is recommended for early-stage EC, while locally advanced disease (N+ and/or T3-4) is treated with perioperative chemotherapy (FLOT) or preoperative chemoradiation (CROSS regimen) followed by immunotherapy for adenocarcinoma. Preoperative chemoradiation (CROSS regimen) followed by immunotherapy or definitive chemoradiation with the possibility of organ preservation are the two options for squamous cell carcinoma. Salvage surgery is recommended for incomplete response or recurrence after definitive chemoradiation and should be performed in an expert center. Treatment for metastatic disease is based on systemic therapy including chemotherapy, immunotherapy or combined targeted therapy according to biomarkers testing such as HER2 status, MMR status and PD-L1 expression. CONCLUSION These guidelines are intended to provide a personalised therapeutic strategy for daily clinical practice and are subject to ongoing optimization. Each individual case should be discussed by a multidisciplinary team.
Collapse
Affiliation(s)
- Julie Veziant
- Department of Digestive and Oncological Surgery, Claude Huriez Hospital, CHU Lille, University of Lille, Lille F-59000, France.
| | - Olivier Bouché
- Department of Digestive Oncology, CHU Reims, Reims, France
| | - T Aparicio
- Department of Gastroenterology and Digestive Oncology, AP-HP, Saint-Louis Hospital, Paris, France
| | - M Barret
- Gastroenterology Department, Cochin Hospital, APHP, Paris, France
| | - F El Hajbi
- Department of Oncology, Centre Oscar Lambret, Lille, France
| | - V Lepilliez
- Gastroenterology Department, Jean Mermoz Private Hospital, Ramsay Santé, Lyon, France
| | - P Lesueur
- Department of Radiation Oncology, Centre Guillaume le Conquérant, Le Havre, France
| | - P Maingon
- Department of Radiation Oncology, La Pitié-Salpêtrière, APHP, Sorbonne University, Paris, France
| | - D Pannier
- Department of Oncology, Centre Oscar Lambret, Lille, France
| | - L Quero
- Department of Radiation Oncology, Saint-Louis Hospital, APHP, Paris, France
| | - J L Raoul
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - F Renaud
- Department of Pathology, La Pitié-Salpêtrière, APHP, Sorbonne University, Paris, France
| | - J F Seitz
- Department of Digestive Oncology, La Timone, Aix Marseille Université, Marseille, France
| | - A A Serre
- Department of Radiotherapy, Centre Léon Bérard, Lyon, France
| | | | - M Vermersch
- Medical Imaging Department, Valencienne Hospital Centre, Valencienne 59300, France
| | - T Voron
- Department of General and Digestive Surgery, Sorbonne Université, AP-HP, Hôpital Saint Antoine, 184 rue du faubourg Saint-Antoine, Paris 75012, France
| | - D Tougeron
- Department of Gastro-Enterology and Hepatology, Poitiers University Hospital, Poitiers, France
| | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, Claude Huriez Hospital, CHU Lille, University of Lille, Lille F-59000, France
| |
Collapse
|
5
|
Zhou Y, Song L, Xia J, Liu H, Xing J, Gao J. Radiomics model based on contrast-enhanced CT texture features for pretreatment prediction of overall survival in esophageal neuroendocrine carcinoma. Front Oncol 2023; 13:1225180. [PMID: 37664013 PMCID: PMC10473874 DOI: 10.3389/fonc.2023.1225180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/25/2023] [Indexed: 09/05/2023] Open
Abstract
Background Limited studies have observed the prognostic value of CT images for esophageal neuroendocrine carcinoma (NEC) due to rare incidence and low treatment experience in clinical. In this study, the pretreatment enhanced CT texture features and clinical characteristics were investigated to predict the overall survival of esophageal NEC. Methods This retrospective study included 89 patients with esophageal NEC. The training and testing cohorts comprised 61 (70%) and 28 (30%) patients, respectively. A total of 402 radiomics features were extracted from the tumor region that segmented pretreatment venous phase CT images. The least absolute shrinkage and selection operator (LASSO) Cox regression was applied to feature dimension reduction, feature selection, and radiomics signature construction. A radiomics nomogram was constructed based on the radiomics signature and clinical risk factors using a multivariable Cox proportional regression. The performance of the nomogram for the pretreatment prediction of overall survival (OS) was evaluated for discrimination and calibration. Results Only the enhancement degree was an independent factor in clinical variable influenced OS. The radiomics signatures demonstrated good predictability for prognostic status discrimination. The radiomics nomogram integrating texture signatures was slightly superior to the nomogram derived from the combined model with a C-index of 0.844 (95%CI: 0.783-0.905) and 0.847 (95% CI: 0.782-0.912) in the training set, and 0.805 (95%CI: 0.707-0.903) and 0.745 (95% CI: 0.639-0.851) in the testing set, respectively. Conclusion The radiomics nomogram based on pretreatment CT radiomics signature had better prognostic power and predictability of the overall survival in patients with esophageal NEC than the model using combined variables.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijie Song
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin Xia
- Department of Oncology, Anyang Tumor Hospital, Anyang, China
| | - Huan Liu
- Advanced Analytics Team, GE Healthcare, Shanghai, China
| | - Jingjing Xing
- Department of Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianbo Gao
- Department of Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
A CT-Based Radiomics Nomogram Model for Differentiating Primary Malignant Melanoma of the Esophagus from Esophageal Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2023; 2023:6057196. [PMID: 36860814 PMCID: PMC9970707 DOI: 10.1155/2023/6057196] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/06/2023] [Accepted: 02/02/2023] [Indexed: 02/22/2023]
Abstract
Objective The diagnosis of primary malignant melanoma of the esophagus (PMME) before treatment is essential for clinical decision-making. However, PMME may be misdiagnosed as esophageal squamous cell carcinoma (ESCC) sometimes. This research is aimed at devising a radiomics nomogram model of CT for distinguishing PMME from ESCC. Methods In this retrospective analysis, 122 individuals with proven pathologically PMME (n = 28) and ESCC (n = 94) were registered from our hospital. PyRadiomics was applied to derive radiomics features from plain and enhanced CT images after resampling image into an isotropic resolution of 0.625 × 0.625 × 0.625 mm3. The diagnostic efficiency of the model was evaluated by an independent validation group. Results For the purpose of differentiation between PMME and ESCC, a radiomics model was constructed using 5 radiomics features obtained from nonenhanced CT and 4 radiomics features derived from enhanced CT. A radiomics model including multiple radiomics features showed excellent discrimination efficiency with AUCs of 0.975 and 0.906 in the primary and validation cohorts, respectively. Then, a radiomics nomogram model was developed. The decision curve analysis has shown remarkable performance of this nomogram model for distinguishing PMME from ESCC. Conclusions The proposed radiomics nomogram model based on CT could be used for distinguishing PMME from ESCC. Moreover, this model also contributed to helping clinicians determine an appropriate treatment strategy for esophageal neoplasms.
Collapse
|
7
|
Shi YJ, Liu C, Wei YY, Li XT, Shen L, Lu ZH, Sun YS. Quantitative CT analysis to predict esophageal fistula in patients with advanced esophageal cancer treated by chemotherapy or chemoradiotherapy. Cancer Imaging 2022; 22:62. [PMCID: PMC9636691 DOI: 10.1186/s40644-022-00490-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/06/2022] [Indexed: 11/06/2022] Open
Abstract
Background Esophageal fistula is one of the most serious complications of chemotherapy or chemoradiotherapy (CRT) for advanced esophageal cancer. This study aimed to evaluate the performance of quantitative computed tomography (CT) analysis and to establish a practical imaging model for predicting esophageal fistula in esophageal cancer patients treated with chemotherapy or chemoradiotherapy. Methods This study retrospectively enrolled 204 esophageal cancer patients (54 patients with fistula, 150 patients without fistula) and all patients were allocated to the primary and validation cohorts according to the time of inclusion in a 1:1 ratio. Ulcer depth, tumor thickness and length, and minimum and maximum enhanced CT values of esophageal cancer were measured in pretreatment CT imaging. Logistic regression analysis was used to evaluate the associations of CT quantitative measurements with esophageal fistula. Receiver operating characteristic curve (ROC) analysis was also used. Results Logistic regression analysis showed that independent predictors of esophageal fistula included tumor thickness [odds ratio (OR) = 1.167; p = 0.037], the ratio of ulcer depth to adjacent tumor thickness (OR = 164.947; p < 0.001), and the ratio of minimum to maximum enhanced CT value (OR = 0.006; p = 0.039) in the primary cohort at baseline CT imaging. These predictors were used to establish a predictive model for predicting esophageal fistula, with areas under the receiver operating characteristic curves (AUCs) of 0.946 and 0.841 in the primary and validation cohorts, respectively. The quantitative analysis combined with T stage for predicting esophageal fistula had AUCs of 0.953 and 0.917 in primary and validation cohorts, respectively. Conclusion Quantitative pretreatment CT analysis has excellent performance for predicting fistula formation in esophageal cancer patients who treated by chemotherapy or chemoradiotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s40644-022-00490-2.
Collapse
Affiliation(s)
- Yan-Jie Shi
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No.52 Fu Cheng Road, Hai Dian District, Beijing, 100142 China
| | - Chang Liu
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No.52 Fu Cheng Road, Hai Dian District, Beijing, 100142 China
| | - Yi-Yuan Wei
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No.52 Fu Cheng Road, Hai Dian District, Beijing, 100142 China
| | - Xiao-Ting Li
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No.52 Fu Cheng Road, Hai Dian District, Beijing, 100142 China
| | - Lin Shen
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No.52 Fu Cheng Road, Hai Dian District, Beijing, 100142 China
| | - Zhi-Hao Lu
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, No.52 Fu Cheng Road, Hai Dian District, Beijing, 100142 China
| | - Ying-Shi Sun
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No.52 Fu Cheng Road, Hai Dian District, Beijing, 100142 China
| |
Collapse
|
8
|
Raptis CA, Goldstein A, Henry TS, Porter KK, Catenacci D, Kelly AM, Kuzniewski CT, Lai AR, Lee E, Long JM, Martin MD, Morris MF, Sandler KL, Sirajuddin A, Surasi DS, Wallace GW, Kamel IR, Donnelly EF. ACR Appropriateness Criteria® Staging and Follow-Up of Esophageal Cancer. J Am Coll Radiol 2022; 19:S462-S472. [PMID: 36436970 DOI: 10.1016/j.jacr.2022.09.008] [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: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/27/2022]
Abstract
This document provides recommendations regarding the role of imaging in the staging and follow-up of esophageal cancer. For initial clinical staging, locoregional extent and nodal disease are typically assessed with esophagogastroduodenoscopy and esophageal ultrasound. FDG-PET/CT or CT of the chest and abdomen is usually appropriate for use in initial clinical staging as they provide additional information regarding distant nodal and metastatic disease. The detection of metastatic disease is critical in the initial evaluation of patients with esophageal cancer because it will direct patients to a treatment pathway centered on palliative radiation rather than surgery. For imaging during treatment, particularly neoadjuvant chemotherapy, FDG-PET/CT is usually appropriate, because some studies have found that it can provide information regarding primary lesion response, but more importantly it can be used to detect metastases that have developed since the induction of treatment. For patients who have completed treatment, FDG-PET/CT or CT of the chest and abdomen is usually appropriate for evaluating the presence and extent of metastases in patients with no suspected or known recurrence and in those with a suspected or known recurrence. The ACR Appropriateness Criteria are evidence-based guidelines for specific clinical conditions that are reviewed annually by a multidisciplinary expert panel. The guideline development and revision process support the systematic analysis of the medical literature from peer reviewed journals. Established methodology principles such as Grading of Recommendations Assessment, Development, and Evaluation or GRADE are adapted to evaluate the evidence. The RAND/UCLA Appropriateness Method User Manual provides the methodology to determine the appropriateness of imaging and treatment procedures for specific clinical scenarios. In those instances where peer reviewed literature is lacking or equivocal, experts may be the primary evidentiary source available to formulate a recommendation.
Collapse
Affiliation(s)
| | - Alan Goldstein
- Division Chief, Abdominal Imaging, Director of CT Colonography, UMass Medical School, Worcester, Massachusetts
| | - Travis S Henry
- Panel Chair; Division Chief of Cardiothoracic Imaging, Duke University, Durham, North Carolina; Co-Director, ACR Education Center HRCT Course
| | - Kristin K Porter
- Panel Chair, University of Alabama Medical Center, Birmingham, Alabama; ACR Council Steering Committee
| | - Daniel Catenacci
- The University of Chicago, Chicago, Illinois; American Society of Clinical Oncology
| | - Aine Marie Kelly
- Assistant Program Director Radiology Residency, Emory University Hospital, Atlanta, Georgia
| | | | - Andrew R Lai
- Hospitalist; University of California San Francisco (UCSF), San Francisco, California; Former Director of the UCSF Hospitalist Procedure Service; Former Director of the UCSF Division of Hospital Medicine's Case Review Committee; Former Director of Procedures/Quality Improvement Rotation for the UCSF Internal Medicine Residency
| | - Elizabeth Lee
- Director, M1 Radiology Education, University of Michigan Medical School; Associate Program Director, Diagnostic Radiology, Michigan Medicine; Director of Residency Education Cardiothoracic Division, Michigan Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Jason M Long
- Director of Robotic Thoracic Surgery, Director of Lung Cancer Screening, University of North Carolina Hospital, Chapel Hill, North Carolina; The Society of Thoracic Surgeons
| | - Maria D Martin
- Director, Diversity and Inclusion, Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Michael F Morris
- Director of Cardiac CT and MRI, University of Arizona College of Medicine, Phoenix, Arizona
| | - Kim L Sandler
- Co-Director Vanderbilt Lung Screening Program, Vanderbilt University Medical Center, Nashville, Tennessee; Imaging Chair, Thoracic Committee, ECOG-ACRIN; Co-Chair, Lung Screening 2.0 Steering Committee
| | | | - Devaki Shilpa Surasi
- Patient Safety and Quality Officer, Department of Nuclear Medicine, Chair-Elect, Junior Faculty Committee, The University of Texas MD Anderson Cancer Center, Houston, Texas; Commission on Nuclear Medicine and Molecular Imaging
| | | | - Ihab R Kamel
- Specialty Chair, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edwin F Donnelly
- Specialty Chair; Chief of Thoracic Radiology, Interim Vice Chair of Academic Affairs, Department of Radiology, Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
9
|
Lai MY, Kang SY, Sun YT, Quan TT, Lu SX, He CY, Zhou ZW, Yang LQ, Luo HY, Wang FH, Li YH, Xu RH, Guan WL, Qiu MZ. Comparison of response evaluation criteria in solid tumors and tumor regression grade in evaluating the effect of preoperative systemic therapy of gastric cancer. BMC Cancer 2022; 22:1031. [PMID: 36183074 PMCID: PMC9526302 DOI: 10.1186/s12885-022-10125-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 09/23/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Both Response Evaluation Criteria in Solid Tumors (RECIST) and tumor regression grade (TRG) play key roles in evaluating tumor response. We analyzed the consistency of TRG and RECIST 1.1 for gastric cancer (GC) patients and compared their prognostic values. METHODS Patients with GC who received preoperative chemotherapy or chemoimmunotherapy and had records of TRG from December 2013 to October 2021 were enrolled retrospectively. TRG 0-1 and 2-3 are considered as corresponding to complete response (CR)/partial response (PR) and stable disease (SD)/progress disease (PD) in RECIST 1.1, respectively. The primary endpoints were disease-free survival (DFS) and overall survival (OS). The consistency of RECIST and TRG was examined by kappa statistics. Survival analysis was performed using the Kaplan Meier method. RESULT One hundred fifty seven GC patients were enrolled, including 125 with preoperative chemotherapy and 32 with chemoimmunotherapy. Among them, 56 patients had measurable lesions. Only 19.6% (11/56) of the patients had consistent results between RECIST 1.1 and TRG. TRG was correlated with both OS and DFS (P = 0.02 and 0.03, respectively) while response according to RECIST1.1 was not (P = 0.86 and 0.23, respectively). The median DFS had not reached in the TRG 0-1 group and was 16.13 months in TRG 2-3 group. TRG 2-3 was associated with young age and peritoneal or liver metastasis. Besides, preoperative chemoimmunotherapy had a significantly higher pCR rate than chemotherapy alone (34.4% vs 8.0%, P < 0.001). CONCLUSION TRG was in poor agreement with RECIST 1.1. TRG was better than RECIST 1.1 in predicting DFS and OS for GC patients who received preoperative therapy.
Collapse
Affiliation(s)
- Ming-Yu Lai
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Shi-Yang Kang
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yu-Ting Sun
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Ting-Ting Quan
- Department of Image, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Shi-Xun Lu
- Department of Pathology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Cai-Yun He
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Li-Qiong Yang
- Department of Experiment, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Hui-Yan Luo
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Feng-Hua Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yu-Hong Li
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Wen-Long Guan
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Miao-Zhen Qiu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
10
|
Shi YJ, Yang X, Yan S, Li XT, Wei YY, Zhang XY, Sun YS. Primary malignant melanoma of the esophagus: differentiation from esophageal squamous cell carcinoma and leiomyoma using dynamic contrast-enhanced CT findings. Abdom Radiol (NY) 2022; 47:2747-2759. [PMID: 35668195 PMCID: PMC9300547 DOI: 10.1007/s00261-022-03556-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE This study aimed to summarize the computed tomography (CT) findings of PMME and differentiate it from esophageal SCC and leiomyoma using CT analysis. METHODS This was a retrospective study including 23 patients with PMME, 69 patients with SCC, and 21 patients with leiomyoma in our hospital. Qualitative CT morphological characteristics of each lesion included the location, tumor range, ulcer, enhanced pattern, and so on. For quantitative CT analysis, thickness, length and area of tumor, size of largest lymph node, number of metastatic lymph node, and CT value of tumor in plain, arterial, and delayed phases were measured. The associated factors for differentiating PMME from SCC and leiomyoma were examined with univariate and multivariate analysis. Receive operating characteristic curve (ROC) was used to determine the performance of CT models in discriminating PMME from SCC and leiomyoma. RESULTS The thickness, mean CT value in arterial phase, and range of tumor were the independent factors for diagnosing PMME from SCC. These parameters were used to establish a diagnostic CT model with area under the ROC (AUC) of 0.969, and accuracy of 90.2%. In pathology, interstitial vessels in PMME were more abundant than that of SCC, and the stromal fibrosis was more obvious in SCC. PMME commonly exhibited intraluminal expansively growth pattern and SCC often showed infiltrative pattern. The postcontrast attenuation difference in maximum CT attenuation value between plain and arterial phases was the independent factor for diagnosing PMME from leiomyoma. This parameter was applied to differentiate PMME from leiomyoma with AUC of 0.929 and accuracy of 86.4%. CONCLUSION The qualitative and quantitative CT analysis had excellent performance for differentiating PMME from SCC and esophageal leiomyoma.
Collapse
Affiliation(s)
- Yan-Jie Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Shuo Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Xiao-Ting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Yi-Yuan Wei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Xiao-Yan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, No. 52 Fu Cheng Road, Hai Dian District, Beijing, 100142, China.
| |
Collapse
|
11
|
Impact of CT-assessed changes in tumor size after neoadjuvant chemotherapy on pathological response and survival of patients with esophageal squamous cell carcinoma. Langenbecks Arch Surg 2022; 407:965-974. [PMID: 34989856 DOI: 10.1007/s00423-022-02430-x] [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: 09/21/2021] [Accepted: 01/01/2022] [Indexed: 12/09/2022]
Abstract
PURPOSE Neoadjuvant chemotherapy (NAC) followed by surgery is the standard treatment for advanced esophageal squamous cell carcinoma (ESCC) in Japan. Computed tomography (CT) is usually used to assess the therapeutic effect of NAC; however, there are no reliable criteria for predicting pathological response or patient prognosis. METHODS We included 84 patients who underwent esophagectomy between January 2009 and December 2018 and retrospectively reviewed their CT scans performed before and after NAC. The reduction rate of the largest tumor area (TA), long diameter (LD), and short diameter (SD) were measured on a transverse CT image. The pathological response and cutoff values were calculated using the receiver operating characteristic curve, and the most suitable ones for determining the effect were examined. RESULTS The areas under the curve for predicting responders to NAC based on the reduction rate of the TA, LD, and SD were 0.755, 0.761, and 0.781, respectively. The optimal cutoff value of the SD reduction rate for predicting responders to NAC was 22%. An SD reduction ≥ 22% was an independent prognostic factor for overall survival in univariate (p = 0.005, hazard ratio [HR] = 2.755) and multivariate analyses (p = 0.030 HR 2.690). Furthermore, an SD reduction of ≥ 22% was also an independent prognostic factor for relapse-free survival in the univariate (p = 0.007, HR = 2.491) and multivariate analyses (p = 0.007, HR = 0.030). CONCLUSIONS The reduction rate of the tumor SD is a simple predictor of pathological response and patient prognosis.
Collapse
|
12
|
Does a high Mandard score really define a poor response to chemotherapy in oesophageal adenocarcinoma? Br J Cancer 2021; 124:1653-1660. [PMID: 33742143 PMCID: PMC8110771 DOI: 10.1038/s41416-021-01290-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/16/2020] [Accepted: 01/29/2021] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND A high Mandard score implies a non-response to chemotherapy in oesophageal adenocarcinoma. However, some patients exhibit tumour volume reduction and a nodal response despite a high score. This study examines survival and recurrence patterns in these patients. METHODS Clinicopathological factors were analysed using multivariable Cox regression assessing time to death and recurrence. Computed tomography-estimated tumour volume change was examined in a subgroup of consecutive patients. RESULTS Five hundred and fifty-five patients were included. Median survival was 55 months (Mandard 1-3) and 21 months (Mandard 4 and 5). In the Mandard 4 and 5 group (332 patients), comparison between complete nodal responders and persistent nodal disease showed improved survival (90 vs 18 months), recurrence rates (locoregional 14.75 vs 28.74%, systemic 24.59 vs 48.42%) and circumferential resection margin positivity (22.95 vs 68.11%). Complete nodal response independently predicted improved survival (hazard ratio 0.34 (0.16-0.74). Post-chemotherapy tumour volume reduction was greater in patients with a complete nodal response (-16.3 vs -7.7 cm3, p = 0.033) with no significant difference between Mandard groups. CONCLUSION Patients with a complete nodal response to chemotherapy have significantly improved outcomes despite a poor Mandard score. High Mandard score does not correspond with a non-response to chemotherapy in all cases and patients with nodal downstaging may still benefit from adjuvant chemotherapy.
Collapse
|
13
|
Song T, Yao Q, Qu J, Zhang H, Zhao Y, Qin J, Feng W, Zhang S, Han X, Wang S, Yan X, Li H. The value of intravoxel incoherent motion diffusion-weighted imaging in predicting the pathologic response to neoadjuvant chemotherapy in locally advanced esophageal squamous cell carcinoma. Eur Radiol 2020; 31:1391-1400. [PMID: 32901300 DOI: 10.1007/s00330-020-07248-z] [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: 05/02/2020] [Revised: 07/05/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To explore the value of intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) for the prediction of pathologic response to neoadjuvant chemotherapy (NAC) in locally advanced esophageal squamous cell carcinoma (ESCC). MATERIAL AND METHODS Forty patients with locally advanced ESCC who were treated with NAC followed by radical resection were prospectively enrolled from September 2015 to May 2018. MRI and IVIM were performed within 1 week before and 2-3 weeks after NAC, prior to surgery. Parameters including apparent diffusion coefficient (ADC), true diffusion coefficient (D), pseudodiffusion coefficient (D*), and pseudodiffusion fraction (f) before and after NAC were measured. Pathologic response was evaluated according to the AJCC tumor regression grade (TRG) system. The changes in IVIM values before and after therapy in different TRG groups were assessed. Receiver operating characteristic (ROC) curves analysis was used to determine the best cutoff value for predicting the pathologic response to NAC. RESULTS Twenty-two patients were identified as TRG 2 (responders), and eighteen as TRG 3 (non-responders) in pathologic evaluation. The ADC, D, and f values increased significantly after NAC. The post-NAC D and ΔD values of responders were significantly higher than those of non-responders. The area under the curve (AUC) was 0.722 for post-NAC D and 0.859 for ΔD in predicting pathologic response. The cutoff values of post-NAC D and ΔD were 1.685 × 10-3 mm2/s and 0.350 × 10-3 mm2/s, respectively. CONCLUSION IVIM-DWI may be used as an effective functional imaging technique to predict pathologic response to NAC in locally advanced ESCC. KEY POINTS • The optimal cutoff values of post-NAC D and ΔD for predicting pathologic response to NAC in locally advanced ESCC were 1.685 × 10-3 mm2/s and 0.350 × 10-3 mm2/s, respectively. • Pathologic response to NAC in locally advanced ESCC was favorable in patients with post-NAC D and ΔD values that were higher than the optimal cutoff values. • IVIM-DWI can potentially be used to preoperatively predict pathologic response to NAC in esophageal carcinoma. Accurate quantification of the D value derived from IVIM-DWI may eventually translate into an effective and non-invasive marker to predict therapeutic efficacy.
Collapse
Affiliation(s)
- Tao Song
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Qi Yao
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Jinrong Qu
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China.
| | - Hongkai Zhang
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Yan Zhao
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Jianjun Qin
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Wen Feng
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Shouning Zhang
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Xianhua Han
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Shaoyu Wang
- MR Scientific Marketing, Siemens Healthineers, XI'an, 710065, China
| | - Xu Yan
- MR Scientific Marketing, Siemens Healthineers, Shanghai, 201318, China
| | - Hailiang Li
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| |
Collapse
|
14
|
Zhuo ZG, Zhu YK, Deng HY, Li G, Luo J, Alai GH, Lin YD. Predictive Value of Excision Repair Cross-Complementation Group 1 in the Response to Platinum-Based Chemotherapy in Esophageal Cancer: A Meta-Analysis. Oncol Res Treat 2020; 43:160-169. [PMID: 31958797 DOI: 10.1159/000505378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 12/10/2019] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Platinum is widely used in the treatment of esophageal cancer. In clinical practice, it is significant to distinguish patients who respond to platinum from those who do not. Excision repair cross-complementation group 1 (ERCC1) is thought to be the key in the resistance to platinum. However, whether it is related to the platinum-based chemotherapy response on real esophageal cancer patients is controversial. We conducted this meta-analysis to explore the association between ERCC1 polymorphisms, its expression levels and platinum-based chemotherapy response, and identify the most sensitive genotypes. METHODS The study was carried out according to the Cochrane handbook for systemic reviews of intervention. The study protocol has been registered on PROSPERO. RESULTS Three studies were included in the analysis of C8092A polymorphisms, 5 in the C118T, and another 6 in ERCC1 expression levels. In C118T polymorphisms, compared to wild genotype, patients with mutant genotypes had a significantly higher response rate. As for C8092A polymorphisms, the mutant genotypes also presented a better response than the wild genotype. The pooled analysis indicated a significantly higher response rate in patients with a low expression of ERCC1. CONCLUSIONS ERCC1 is a valuable biomarker for platinum-based chemotherapy in esophageal cancer. Patients with ERCC1 mutations or low-level ERCC1 expression are more sensitive to platinum-based chemotherapy.
Collapse
Affiliation(s)
- Ze-Guo Zhuo
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yun-Ke Zhu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Han-Yu Deng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Luo
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Gu-Ha Alai
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yi-Dan Lin
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China,
| |
Collapse
|
15
|
de Gouw DJJM, Klarenbeek BR, Driessen M, Bouwense SAW, van Workum F, Fütterer JJ, Rovers MM, Ten Broek RPG, Rosman C. Detecting Pathological Complete Response in Esophageal Cancer after Neoadjuvant Therapy Based on Imaging Techniques: A Diagnostic Systematic Review and Meta-Analysis. J Thorac Oncol 2019; 14:1156-1171. [PMID: 30999111 DOI: 10.1016/j.jtho.2019.04.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 03/14/2019] [Accepted: 03/17/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Up to 32% of patients with esophageal cancer show a pathological complete response (ypCR) after neoadjuvant therapy. To prevent overtreatment, the indication to perform esophagectomy in these patients should be reconsidered. Implementing an organ-preserving strategy for patients with ypCR requires an accurate assessment of residual disease after neoadjuvant treatment. The aim of this study was to systematically review the effectiveness of imaging techniques used for detection of ypCR after neoadjuvant therapy but before resection in patients with esophageal cancer. METHODS A systematic literature search of the Medline, Embase, and Cochrane Library databases was performed from January 1, 2000, to December 13, 2017. Eligible studies were diagnostic studies that compared results of imaging modalities after neoadjuvant therapy to histopathological findings in the resection specimen after esophagectomy. Methodological quality was assessed by the Cochrane Quality Assessment of Diagnostic Accuracy Studies, version 2, model. Primary outcome measures were true positive, false-positive, false-negative, and true negative values of imaging techniques predicting ypCR. A meta-analysis was performed by pooling sensitivities and specificities by using a bivariate model. RESULTS A total of 4420 articles were identified. After exclusion of irrelevant titles and abstracts, 360 articles were reviewed in full text. In total, four imaging modalities (computed tomography [CT], positron emission tomography [PET-CT], endoscopic ultrasound [EUS], and magnetic resonance imaging [MRI]) were used for restaging. The meta-analysis was conducted with data from 56 studies involving 3625 patients. The pooled sensitivities of CT, PET-CT, EUS, and MRI for detecting ypCR were 0.35, 0.62, 0.01 and 0.80, respectively, whereas the pooled specificities were 0.83, 0.73, 0.99, and 0.83, respectively. The positive predictive value in detecting ypCR was 0.47 for CT, 0.41 for PET-CT, not applicable for EUS, and 0.61 for MRI. CONCLUSION Current imaging modalities such as CT, PET-CT, and EUS seem to be insufficiently accurate to identify complete responders. More accurate diagnostic tests are needed to improve restaging accuracy for patients with esophageal cancer.
Collapse
Affiliation(s)
- Didi J J M de Gouw
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands.
| | | | - Mitchell Driessen
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stefan A W Bouwense
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frans van Workum
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jurgen J Fütterer
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maroeska M Rovers
- Department of Health Evidence and Operating Rooms, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Camiel Rosman
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Djuric-Stefanovic A, Jankovic A, Saponjski D, Micev M, Stojanovic-Rundic S, Cosic-Micev M, Pesko P. Analyzing the post-contrast attenuation of the esophageal wall on routine contrast-enhanced MDCT examination can improve the diagnostic accuracy in response evaluation of the squamous cell esophageal carcinoma to neoadjuvant chemoradiotherapy in comparison with the esophageal wall thickness. Abdom Radiol (NY) 2019; 44:1722-1733. [PMID: 30758534 DOI: 10.1007/s00261-019-01911-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE To evaluate the accuracy of the multidetector computed tomography (MDCT) in the response evaluation of the esophageal squamous cell carcinoma (ESCC) to neoadjuvant chemoradiotherapy (nCRT) by analyzing the thickness and post-contrast attenuation of the esophageal wall after the nCRT. METHODS Contrast-enhanced (CE)-MDCT examinations in portal venous phase of one hundred patients with locally advanced ESCC who received nCRT and underwent esophageal resection and histopathology assessment of tumor regression grade (TRG) were retrospectively analyzed by measuring the maximal thickness and mean density of the esophageal wall in the segment involved by tumor and visually searching for hyperdense foci within it. Diagnostic performance was evaluated using the ROC analysis. RESULTS Average attenuation of the esophageal wall had stronger diagnostic performance for predicting pathologic complete regression (pCR) (AUC = 0.994; p < 0.001) in relation to maximal esophageal wall thickness (AUC = 0.731; p < 0.001). Maximal esophageal wall thickness ≤ 9 mm and average attenuation of the esophageal wall ≤ 64 HU predicted pCR with the sensitivity, specificity, and overall accuracy of 62.5%, 77.9%, and 73%, and 96.9%, 98.5%, and 98%, respectively. Combination of both cutoff values enabled correct assessment of pCR with the 100% accuracy. Visual detection of the hyperdense focus within the esophageal wall predicted pCR with the sensitivity, specificity, and overall accuracy values of 100%, 94.1%, and 96%, respectively. CONCLUSION Visual analysis and measurement of post-contrast attenuation of the esophageal wall after the nCRT can improve diagnostic accuracy of MDCT in the response evaluation of the ESCC to nCRT in comparison with measuring the esophageal wall thickness.
Collapse
|
17
|
Vollenbrock SE, Voncken FEM, van Dieren JM, Lambregts DMJ, Maas M, Meijer GJ, Goense L, Mook S, Hartemink KJ, Snaebjornsson P, Ter Beek LC, Verheij M, Aleman BMP, Beets-Tan RGH, Bartels-Rutten A. Diagnostic performance of MRI for assessment of response to neoadjuvant chemoradiotherapy in oesophageal cancer. Br J Surg 2019; 106:596-605. [PMID: 30802305 PMCID: PMC6594024 DOI: 10.1002/bjs.11094] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/12/2018] [Accepted: 11/26/2018] [Indexed: 01/03/2023]
Abstract
Background Patients with a pathological complete response (pCR) after neoadjuvant chemoradiotherapy (nCRT) for oesophageal cancer may benefit from non‐surgical management. The aim of this study was to determine the diagnostic performance of visual response assessment of the primary tumour after nCRT on T2‐weighted (T2W) and diffusion‐weighted (DW) MRI. Methods Patients with locally advanced oesophageal cancer who underwent T2W‐ and DW‐MRI (1·5 T) before and after nCRT in two hospitals, between July 2013 and September 2017, were included in this prospective study. Three radiologists evaluated T2W images retrospectively using a five‐point score for the assessment of residual tumour in a blinded manner and immediately rescored after adding DW‐MRI. Histopathology of the resection specimen was used as the reference standard; ypT0 represented a pCR. Sensitivity, specificity, area under the receiver operating characteristic (ROC) curve (AUC) and interobserver agreement were calculated. Results Twelve of 51 patients (24 per cent) had a pCR. The sensitivity and specificity of T2W‐MRI for detection of residual tumour ranged from 90 to 100 and 8 to 25 per cent respectively. Respective values for T2W + DW‐MRI were 90–97 and 42–50 per cent. AUCs for the three readers were 0·65, 0·66 and 0·68 on T2W‐MRI, and 0·71, 0·70 and 0·70 on T2W + DW‐MRI (P = 0·441, P = 0·611 and P = 0·828 for readers 1, 2 and 3 respectively). The κ value for interobserver agreement improved from 0·24–0·55 on T2W‐MRI to 0·55–0·71 with DW‐MRI. Conclusion Preoperative assessment of residual tumour on MRI after nCRT for oesophageal cancer is feasible with high sensitivity, reflecting a low chance of missing residual tumour. However, the specificity was low; this results in overstaging of complete responders as having residual tumour and, consequently, overtreatment.
Collapse
Affiliation(s)
- S E Vollenbrock
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - F E M Voncken
- Department of Radiation Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - J M van Dieren
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - D M J Lambregts
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - M Maas
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - G J Meijer
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - L Goense
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - S Mook
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - K J Hartemink
- Department of Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - P Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - L C Ter Beek
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - M Verheij
- Department of Radiation Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - B M P Aleman
- Department of Radiation Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - R G H Beets-Tan
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - A Bartels-Rutten
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| |
Collapse
|
18
|
White DB, Hora MJ, Jenkins SM, Marks RS, Garces YI, Cassivi SD, Roden AC. Efficacy of chest computed tomography prediction of the pathological TNM stage of thymic epithelial tumours. Eur J Cardiothorac Surg 2019; 56:5316016. [PMID: 30753469 DOI: 10.1093/ejcts/ezz013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/06/2019] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The aim of this study is to evaluate the efficacy of chest computed tomography (CT) to predict the pathological stage of thymic epithelial tumours (TET) using the recently introduced tumour, node and metastasis (TNM) staging with comparison to the modified Masaoka staging. METHODS Preoperative chest CT examinations in cases of resected TET with sampled lymph nodes (2006-2016) were retrospectively reviewed by 2 thoracic radiologists and radiologically (r) staged using both staging systems. A thoracic pathologist reviewed all cases for the pathological (p) stage. Concordance between r-staging and p-staging was assessed by % agreement and unweighted kappa statistics. Associations between r-stage and p-stage with outcomes were assessed using the Cox proportional hazards regression. RESULTS Sixty patients with TET were included (47 thymomas, 12 thymic carcinomas and 1 atypical carcinoid tumour). Sixteen patients (26.7%) had received neoadjuvant therapy. Fifty-four patients (90.0%) had complete resection. The overall agreement between the r-stage and p-stage was 66.7% (κ = 0.46) for TNM staging and 46.7% (κ = 0.30) for modified Masaoka staging. Agreement between r-assessment and p-assessment of the T, N and M components of the TNM stage was 61.7% (κ = 0.28), 86.7% (κ = 0.48) and 98.3% (κ = 0.88), respectively. CT overstaged 12 patients (20.0%) for TNM staging and 12 patients (20.0%) for modified Masaoka staging and understaged 8 (13.3%) and 20 (33.3%) patients for TNM staging modified Masaoka staging, respectively. The r-TNM staging accuracy was lower for patients with neoadjuvant therapy (50.0% with vs 72.7% without). During a median follow-up of 2.6 years (range 0.1-10.5 years), 12 patients had metastases and/or recurrence; 11 patients died (4 of disease). The r-TNM stage and modified Masaoka stage were associated with overall survival and progression-free survival (P < 0.001). CONCLUSIONS Preoperative chest CT is able to accurately predict p-TNM stage in two-thirds of surgically resected TET, with an agreement between radiological staging and pathological staging superior to the modified Masaoka staging.
Collapse
Affiliation(s)
- Darin B White
- Department of Radiology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Megan J Hora
- Department of Radiology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Sarah M Jenkins
- Department of Health Sciences Research, Mayo Clinic Rochester, Rochester, MN, USA
| | - Randolph S Marks
- Division of Medical Oncology, Department of Oncology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Yolanda I Garces
- Department of Radiation-Oncology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Stephen D Cassivi
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic Rochester, Rochester, MN, USA
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN, USA
| |
Collapse
|
19
|
A Diagnostic Algorithm That Combines Quantitative 18F-FDG PET Parameters and Contrast-Enhanced CT Improves Posttherapeutic Locoregional Restaging and Prognostication of Survival in Patients With Esophageal Cancer. Clin Nucl Med 2019; 44:e13-e21. [PMID: 30418211 DOI: 10.1097/rlu.0000000000002366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE The aim of this study was to determine whether the combination of contrast-enhanced CT (CE-CT) and quantitative F-FDG PET parameters improves locoregional restaging in esophageal cancer (EC) after neoadjuvant therapy. METHODS Eighty-eight consecutive patients with locally advanced esophageal cancer, who underwent restaging after neoadjuvant chemotherapy or chemoradiotherapy before esophagectomy, were included in this retrospective study. The diagnostic accuracy of CE-CT, visual F-FDG PET/CT (vPET/CT), and quantitative PET parameters was assessed for T and N staging. Histopathology was used as the reference standard. The prognostic value for recurrence-free survival, cancer-specific survival, and overall survival was assessed using Cox regression analysis. RESULTS Sensitivity, positive predictive value, and accuracy were 78.8%, 70.2%, and 59.0% (CE-CT), and 81.1%, 81.1%, and 68.2% (vPET/CT) for T staging as well as 59.5%, 75.9%, and 50.0% (CE-CT), and 70.2%, 93.7%, and 67.0% (vPET/CT) for N staging, respectively. Tumor length and metabolic tumor volume (MTV) exhibited an incremental increase with advancing T stages (P = 0.002 and 0.038). Contrast-enhanced CT had the highest sensitivity to differentiate advanced T stages (T3/4 vs 0-2; area under the receiver operating curve [AUC], 0.86; P < 0.001), whereas MTV at a threshold of 5.8 mL had the highest sensitivity to detect complete response (T0 vs 1-4; AUC, 0.77; P = 0.002). Contrast-enhanced CT and MTV combined had an even superior accuracy to predict complete response (AUC, 0.82; P < 0.001). The imaging American Joint Committee on Cancer stage provided a better prognostication of recurrence-free survival, cancer-specific survival, and overall survival than either T stage, N stage derived from CE-CT or vPET/CT, or quantitative PET parameters alone. CONCLUSIONS Combined CE-CT and MTV had the highest diagnostic accuracy to identify the posttherapeutic T stage, allowing for robust prediction of recurrence and survival.
Collapse
|
20
|
Zhou Y, Liu D, Hou P, Zha KJ, Wang F, Zhou K, He W, Gao JB. Low-dose spectral insufflation computed tomography protocol preoperatively optimized for T stage esophageal cancer - preliminary research experience. World J Gastroenterol 2018; 24:4197-4207. [PMID: 30271084 PMCID: PMC6158477 DOI: 10.3748/wjg.v24.i36.4197] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/28/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the T stage of esophageal squamous cell carcinoma (ESCC) using preoperative low-dose esophageal insufflation computed tomography (EICT).
METHODS One hundred and twenty ESCC patients confirmed by surgery or esophagoscopy were divided into three groups. Groups B and C were injected with 300 mgI/kg contrast medium for automatic spectral imaging assist (GSI assist), while group A underwent a conventional 120 kVp computed tomography (CT) scan with a 450 mgI/kg contrast medium injection. EICT was performed in group C. Group A was reconstructed with filtered back projection, and groups B and C were reconstructed with 50% adaptive statistical iterative reconstruction. The contrast-to-noise ratio of lesion-to-mediastinal adipose tissue and the radiation dose were measured. Specific imaging features were observed, and T stage ESCCs were evaluated.
RESULTS The sensitivity and accuracy of the T1/2 stage were higher in group C than in groups A and B (sensitivity: 43.75% vs 31.82% and 33.33%; accuracy: 54.29% vs 46.67% and 52.50%, respectively). With regard to the T3 stage, the sensitivity and specificity in group C were higher than those in groups A and B (sensitivity: 56.25% vs 41.17% and 44.44%; specificity: 73.68% vs 67.86% and 63.64%, respectively). The diagnostic sensitivity, specificity and accuracy of the T4 stage were similar among all groups. There were no significant differences in volume CT dose index [(5.91 ± 2.57) mGy vs (3.24 ± 1.20) vs (3.65 ± 1.77) mGy], dose-length product [(167.10 ± 99.08) mGy•cm vs (113.24 ± 54.46) mGy•cm vs (117.98 ± 32.32) mGy•cm] and effective dose [(2.52 ± 1.39) vs (1.63 ± 0.76) vs (1.73 ± 0.44) mSv] among the groups (P > 0.05). However, groups B and C received similar effective doses but lower iodine loads than group A [(300 vs 450) mgI/kg].
CONCLUSION EICT combined with GSI assist allows differential diagnosis between the T1/2 and T3 stages. The ability to differentially diagnose the T3 and T4 stages of medullary ESCC can be improved by quantitatively and qualitatively analyzing the adipose tissue in front of the vertebral body.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Dan Liu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Ping Hou
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Kai-Ji Zha
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Kun Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Wei He
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jian-Bo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
21
|
Usefulness of computed tomography density of a tumor in predicting the response of advanced esophageal cancer to preoperative chemotherapy. Surgery 2017; 162:823-835. [DOI: 10.1016/j.surg.2017.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/07/2017] [Accepted: 06/02/2017] [Indexed: 11/23/2022]
|
22
|
Hamburg-Glasgow classification: preoperative staging by combination of disseminated tumour load and systemic inflammation in oesophageal carcinoma. Br J Cancer 2017; 117:612-618. [PMID: 28704837 PMCID: PMC5572176 DOI: 10.1038/bjc.2017.219] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/10/2017] [Accepted: 06/15/2017] [Indexed: 12/24/2022] Open
Abstract
Background: The aim of this study was to establish a new preoperative staging classification and evaluate its comparability to the post-operative tumour stage, lymph node invasion and metastasis (TNM) classification. To date, adequate, preoperative staging in patients with oesophageal carcinoma (EC) is still missing but urgently needed. Systemic inflammation and disseminated tumour load have a pivotal role in recurrence and oncological outcome. To improve the clinical staging, we merged the Glasgow Prognostic Score (GPS) and disseminated tumour cells (DTC) into a new sufficient preoperative staging classification, the Hamburg-Glasgow classification (HGC). Methods: In this prospective, single-centre study, 326 patients following curative oesophagectomy were included. From all patients preoperative bone marrow was aspirated from the iliac crest to detect DTCs by immunostaining with the pan-keratin antibody A45-B/B3. HGC was subdefined into four prognostic groups on the basis of C-reactive protein (CRP), albumin and DTC. The three prognostic groups of the GPS were supplemented by DTC detection status. Results were correlated with clinicopathological parameters and clinical outcome. Results: Increasing HGC significantly correlated with lymph node invasion (P=0.022), post-operative pathohistological TNM staging (P=0.001) and tumour recurrence (P=0.001). The four HGC prognostic groups displayed a gradual decrease in overall as well as disease-free survival (P<0.001, each). Hamburg-Glasgow classification was a strong, significant independent predictor of overall survival and disease-free survival (P<0.001, both) in multivariate analysis. Conclusions: Hamburg-Glasgow classification seems to be a promising preoperative additive staging classification for accurate and simple outcome stratification.
Collapse
|
23
|
Findlay JM, Gillies RS, Franklin JM, Teoh EJ, Jones GE, di Carlo S, Gleeson FV, Maynard ND, Bradley KM, Middleton MR. Restaging oesophageal cancer after neoadjuvant therapy with (18)F-FDG PET-CT: identifying interval metastases and predicting incurable disease at surgery. Eur Radiol 2016; 26:3519-33. [PMID: 26883329 DOI: 10.1007/s00330-016-4227-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/08/2016] [Accepted: 01/15/2016] [Indexed: 01/17/2023]
Abstract
OBJECTIVES It is unknown whether restaging oesophageal cancer after neoadjuvant therapy with positron emission tomography-computed tomography (PET-CT) is more sensitive than contrast-enhanced CT for disease progression. We aimed to determine this and stratify risk. METHODS This was a retrospective study of patients staged before neoadjuvant chemotherapy (NAC) by (18)F-FDG PET-CT and restaged with CT or PET-CT in a single centre (2006-2014). RESULTS Three hundred and eighty-three patients were restaged (103 CT, 280 PET-CT). Incurable disease was detected by CT in 3 (2.91 %) and PET-CT in 17 (6.07 %). Despite restaging unsuspected incurable disease was encountered at surgery in 34/336 patients (10.1 %). PET-CT was more sensitive than CT (p = 0.005, McNemar's test). A new classification of FDG-avid nodal stage (mN) before NAC (plus tumour FDG-avid length) predicted subsequent progression, independent of conventional nodal stage. The presence of FDG-avid nodes after NAC and an impassable tumour stratified risk of incurable disease at surgery into high (75.0 %; both risk factors), medium (22.4 %; either), and low risk (3.87 %; neither) groups (p < 0.001). Decision theory supported restaging PET-CT. CONCLUSIONS PET-CT is more sensitive than CT for detecting interval progression; however, it is insufficient in at least higher risk patients. mN stage and response (mNR) plus primary tumour characteristics can stratify this risk simply. KEY POINTS • Restaging (18) F-FDG-PET-CT after neoadjuvant chemotherapy identifies metastases in 6 % of patients • Restaging (18) F-FDG-PET-CT is more sensitive than CT for detecting interval progression • Despite this, at surgery 10 % of patients had unsuspected incurable disease • New concepts (FDG-avid nodal stage and response) plus tumour impassability stratify risk • Higher risk (if not all) patients may benefit from additional restaging modalities.
Collapse
Affiliation(s)
- John M Findlay
- Oxford OesophagoGastric Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LJ, UK.
- NIHR Oxford Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK.
| | - Richard S Gillies
- Oxford OesophagoGastric Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LJ, UK
| | - James M Franklin
- Department of Nuclear Medicine, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Eugene J Teoh
- Department of Nuclear Medicine, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Greg E Jones
- Oxford OesophagoGastric Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LJ, UK
- Royal Berkshire Hospital, Craven Road, Reading, RG1 5AN, UK
| | - Sara di Carlo
- Oxford OesophagoGastric Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LJ, UK
- Queen's Medical Centre, Derby Road, Nottingham, NG7 2UH, UK
| | - Fergus V Gleeson
- Department of Nuclear Medicine, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Nicholas D Maynard
- Oxford OesophagoGastric Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LJ, UK
| | - Kevin M Bradley
- Department of Nuclear Medicine, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Mark R Middleton
- NIHR Oxford Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| |
Collapse
|
24
|
Griffin Y. Esophageal Cancer: Role of Imaging in Primary Staging and Response Assessment Post Neoadjuvant Therapy. Semin Ultrasound CT MR 2016; 37:339-51. [PMID: 27342898 DOI: 10.1053/j.sult.2016.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Advances in the early detection and treatment of esophageal cancer have meant improved survival rates for patients with esophageal cancer. Accurate pretreatment and post-neoadjuvant treatment staging of esophageal cancer is essential for assessing operability and determining the optimum treatment plan. This article reviews the multimodality imaging approach in the diagnosis, staging, and assessment of treatment response in esophageal cancer.
Collapse
Affiliation(s)
- Yvette Griffin
- Department of Radiology, Leicester Royal Infirmary, Leicester, UK.
| |
Collapse
|
25
|
Djuric-Stefanovic A, Micev M, Stojanovic-Rundic S, Pesko P, Saranovic D. Absolute CT perfusion parameter values after the neoadjuvant chemoradiotherapy of the squamous cell esophageal carcinoma correlate with the histopathologic tumor regression grade. Eur J Radiol 2015; 84:2477-84. [PMID: 26467704 DOI: 10.1016/j.ejrad.2015.09.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/21/2015] [Accepted: 09/27/2015] [Indexed: 12/19/2022]
Abstract
PURPOSE To analyze value of the computed tomography (CT) perfusion imaging in response evaluation of the esophageal carcinoma to neoadjuvant chemoradiotherapy (nCRT) using the histopathology as reference standard. METHODS Forty patients with the squamous cell esophageal carcinoma were re-evaluated after the nCRT by CT examination, which included low-dose CT perfusion study that was analyzed using the deconvolution-based CT perfusion software (Perfusion 3.0, GE). Histopathologic assessment of tumor regression grade (TRG) according to Mandard's criteria served as reference standard of response evaluation. Statistical analysis was performed using Spearman's rank correlation coefficient (r(S)) and Kruskal-Wallis's test. RESULTS The perfusion CT parameter values, measured after the nCRT in the segment of the esophagus that had been affected by neoplasm prior to therapy, significantly correlated with the TRG: blood flow (BF) (r(S)=0.851; p<0.001), blood volume (BV) (r(S)=0.732; p<0.001) and mean transit time (MTT) (r(S)=-0.386; p=0.014). Median values of BF and BV significantly differed among TRG 1-4 groups (p<0.001), while maximal esophageal wall thickness did not (p=0.102). Median BF and BV were gradually rose and MTT decreased as TRG increased, from 21.4 ml/min/100 g (BF), 1.6 ml/100 g (BV) and 8.6 s (MTT) in TRG 1 group, to 37.3 ml/min/100 g, 3.5 ml/100 g and 7.5 s in TRG 2 group, 81.4 ml/min/100 g, 4.1 ml/100 g and 3.8 s in TRG 3 group, and 121.1 ml/min/100 g, 4.9 ml/100 g and 3.7 s in TRG 4 group. In all 15 patients who achieved complete histopathologic regression (TRG 1), BF was <30.0 ml/min/100 g. CONCLUSIONS CT perfusion could improve the accuracy in response evaluation of the esophageal carcinoma to nCRT.
Collapse
Affiliation(s)
- A Djuric-Stefanovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia; Department of Digestive Radiology (First Surgery University Clinic), Center of Radiology and MR, Clinical Center of Serbia, Belgrade, Serbia.
| | - M Micev
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia; Department of Pathology, First Surgery University Clinic, Clinical Center of Serbia, Belgrade, Serbia
| | - S Stojanovic-Rundic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia; Clinic for Radiation Oncology and Diagnostics, Department of Radiation Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - P Pesko
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia; First Surgery University Clinic, Clinical Center of Serbia, Belgrade, Serbia
| | - Dj Saranovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia; Department of Digestive Radiology (First Surgery University Clinic), Center of Radiology and MR, Clinical Center of Serbia, Belgrade, Serbia
| |
Collapse
|
26
|
Abstract
The value of 18F-FDG PET/CT in the initial diagnosis and in the locoregional staging of esophagogastric junction adenocarcinoma is not fully established. However, 18F-FDG PET/CT is widely accepted as the best modality for identification of suspected metastases in staging of the disease. Results published in the literature suggest that 18F-FDG PET/CT may provide useful information for response assessment to neoadjuvancy and to differentiate responding and nonresponding tumors. We review the potential role of 18F-FDG PET/CT imaging in staging, restaging, and prognostic value after chemoradiation therapy in esophagogastric junction adenocarcinoma.
Collapse
|
27
|
Bollschweiler E, Hölscher AH, Schmidt M, Warnecke-Eberz U. Neoadjuvant treatment for advanced esophageal cancer: response assessment before surgery and how to predict response to chemoradiation before starting treatment. Chin J Cancer Res 2015; 27:221-30. [PMID: 26157318 DOI: 10.3978/j.issn.1000-9604.2015.04.04] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/20/2015] [Indexed: 12/22/2022] Open
Abstract
Patients with advanced esophageal cancer (T3-4, N) have a poor prognosis. Chemoradiation or chemotherapy before esophagectomy with adequate lymphadenectomy is the standard treatment for patients with resectable advanced esophageal carcinoma. However, only patients with major histopathologic response (regression to less than 10% of the primary tumor) after preoperative treatment will have a prognostic benefit of preoperative chemoradiation. Using current therapy regimens about 40% to 50% of the patients show major histopathological response. The remaining cohort does not benefit from this neoadjuvant approach but might benefit from earlier surgical resection. Therefore, it is an aim to develop tools for response prediction before starting the treatment and for early response assessment identifying responders. The current review discusses the different imaging techniques and the most recent studies about molecular markers for early response prediction. The results show that [(18)F]-fluorodeoxyglucose-positron emission tomography (FDG-PET) has a good sensitivity but the specificity is not robust enough for routine clinical use. Newer positron emission tomography detector technology, the combination of FDG-PET with computed tomography, additional evaluation criteria and standardization of evaluation may improve the predictive value. There exist a great number of retrospective studies using molecular markers for prediction of response. Until now the clinical use is missing. But the results of first prospective studies are promising. A future perspective may be the combination of imaging technics and special molecular markers for individualized therapy. Another aspect is the response assessment after finishing neoadjuvant treatment protocol. The different clinical methods are discussed. The results show that until now no non-invasive method is valid enough to assess complete histopathological response.
Collapse
Affiliation(s)
- Elfriede Bollschweiler
- 1 Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany ; 2 Institute of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - Arnulf H Hölscher
- 1 Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany ; 2 Institute of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - Matthias Schmidt
- 1 Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany ; 2 Institute of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - Ute Warnecke-Eberz
- 1 Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany ; 2 Institute of Nuclear Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
28
|
Zhang XY, Yan WP, Sun Y, Li XT, Chen Y, Fan MY, Wu Y, Liang Z, Xiong HC, Wang ZL, Sun YS, Chen KN. CT Signs Can Predict Treatment Response and Long-Term Survival: A Study in Locally Advanced Esophageal Cancer with Preoperative Chemotherapy. Ann Surg Oncol 2015; 22 Suppl 3:S1380-7. [DOI: 10.1245/s10434-015-4531-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Indexed: 12/26/2022]
|
29
|
Yip C, Cook GJR, Landau DB, Davies A, Goh V. Performance of different imaging modalities in assessment of response to neoadjuvant therapy in primary esophageal cancer. Dis Esophagus 2015; 29:116-30. [PMID: 25604614 DOI: 10.1111/dote.12315] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- C Yip
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK.,Department of Radiation Oncology, National Cancer Center, Singapore
| | - G J R Cook
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| | - D B Landau
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK.,Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - A Davies
- Department of General Surgery, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - V Goh
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK.,Department of Radiology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
30
|
Ringe KI, Meyer S, Ringe BP, Winkler M, Wacker F, Raatschen HJ. Value of oral effervescent powder administration for multidetector CT evaluation of esophageal cancer. Eur J Radiol 2014; 84:215-20. [PMID: 25497235 DOI: 10.1016/j.ejrad.2014.11.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 10/20/2014] [Accepted: 11/09/2014] [Indexed: 12/19/2022]
Abstract
PURPOSE To assess the value of oral effervescent powder (EP) for evaluation of esophageal distension, and for detection and staging of esophageal cancer with contrast-enhanced CT. MATERIALS AND METHODS 84 patients without esophageal pathology and 52 patients with histological confirmed diagnosis of esophageal cancer were included in this prospective IRB-approved study. Half of the patients in both groups received EP prior to CT. Esophageal distension was assessed by planimetry of the inner (IA) and outer area (OA). Two blinded readers evaluated the datasets separately with regard to diagnosis of esophageal cancer (yes/no) and staging (T0-T4), if applicable. Distension results were compared (t-Test). In patients with cancer sensitivity, specificity, NPV and PPV were calculated. CT staging results were compared to histopathology (Cohen-k). RESULTS IA and IA/OA were significantly larger after EP as compared to the group without EP (p<0.05). Sensitivity, specificity, NPV and PPV for cancer detection cancer were as follows: 78%/78%, 98%/98%, 95%/95%, 87%/87% with EP; 60%/68%, 98%/98%, 94%/94%, 80%/83% without EP. Staging with EP was good (k=0.84/0.67) and moderate without EP (k=0.58/0.59). CONCLUSIONS Administration of EP prior to CT results in good distension of the esophagus, and improves detection and staging of esophageal cancer, as compared to control studies without EP.
Collapse
Affiliation(s)
- Kristina I Ringe
- Department of Diagnostic and Interventional Radiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany.
| | - Simone Meyer
- Department of Diagnostic and Interventional Radiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany.
| | - Bastian P Ringe
- Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany.
| | - Michael Winkler
- Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany.
| | - Frank Wacker
- Department of Diagnostic and Interventional Radiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany.
| | - Hans-Juergen Raatschen
- Department of Diagnostic and Interventional Radiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany.
| |
Collapse
|