1
|
Cai M, Song B, He D, Xu C, Luo R, Qian Y, Kam S, Huo X, Wang J, Vieth M, Zhong Y. Endoscopic resection after downstaging of oesophageal carcinoma by neoadjuvant chemoimmunotherapy: - a new multimodal concept? Gut 2025:gutjnl-2024-333337. [PMID: 40139748 DOI: 10.1136/gutjnl-2024-333337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/23/2025] [Indexed: 03/29/2025]
Affiliation(s)
- Mingyan Cai
- Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Baohui Song
- Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Dongli He
- Zhongshan Hospital Xuhui Branch, Fudan University, Shanghai, China
| | - Chen Xu
- Pathology, Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Rongkui Luo
- Pathology, Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Yang Qian
- Department of Radiation Oncology, Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Sikei Kam
- Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Xucheng Huo
- Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Jian Wang
- Department of Radiotherapy, Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany
| | - Yunshi Zhong
- Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| |
Collapse
|
2
|
Yang Y, Liu Z, Wong I, Gao X, Zhang H, Liu J, Eyck BM, Shao J, Han Y, van der Wilk BJ, Chao Y, Law S, Wijnhoven BPL, van Lanschot JJB, Li Z. Detecting residual disease after neoadjuvant chemoradiotherapy for oesophageal squamous cell carcinoma: The prospective multicentre preSINO trial. Br J Surg 2025; 112:znaf004. [PMID: 39937490 PMCID: PMC11816269 DOI: 10.1093/bjs/znaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (nCRT) in patients with oesophageal squamous cell carcinoma (OSCC) may lead to clinical complete response (cCR). It is important to know the accuracy of clinical response evaluations (CREs) before advocating active surveillance instead of oesophagectomy. METHODS This was a prospective, multicentre study of patients with locally advanced OSCC. They received the first CRE (bite-on-bite biopsies) 4-6 weeks after nCRT. Patients with residual tumour underwent surgery. Patients with a cCR at CRE-1 underwent a second CRE 10-12 weeks after nCRT using PET-CT, bite-on-bite biopsies and endoscopic ultrasound fine-needle aspiration (EUS-FNA). All patients without distant metastases underwent surgery. Primary endpoint was the accuracy of CREs for detecting Tumour Regression Grade (TRG)3-4 or TRG1-2 with ypN+ residual tumour with a prespecified false-negative rate (FNR) of 19.5%. Circulating-tumour DNA (ctDNA) at CREs was performed for exploratory analysis. RESULTS In total 309 patients were included. Eighteen of 133 patients with TRG3-4 or TRG1-2 with ypN+ residual tumours were not detected by bite-on-bite biopsies and EUS-FNA (FNR: 13.5%). Sensitivity, specificity, negative predictive value and positive predictive value of detecting any residual tumour were 81.7%, 93.2%, 68.7% and 96.5% respectively. PET-CT detected interval distant metastases in 13 (4.9%) of 268 patients presurgically. After a minimum 12-month follow-up, systemic recurrence rates were 28.0% in patients with positive ctDNA at CREs and 5.3% in those with negative ctDNA. CONCLUSIONS Bite-on-bite biopsies and EUS-FNA were accurate in detecting residual disease after nCRT in OSCC. Positive ctDNA at CREs may indicate an increased risk of systemic metastases.
Collapse
Affiliation(s)
- Yang Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhichao Liu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ian Wong
- Department of Surgery, LKS Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Xing Gao
- Division of Thoracic Surgery, Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan, Taiwan, China
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Hong Zhang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ben M Eyck
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Jinchen Shao
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Berend J van der Wilk
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Yinkai Chao
- Division of Thoracic Surgery, Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan, Taiwan, China
| | - Simon Law
- Department of Surgery, LKS Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Bas P L Wijnhoven
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - J Jan B van Lanschot
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Li Y, Su X, Shang Y, Liu H, Wang W, Zhang A, Shi G. Comparative evaluation of imaging methods for prognosis assessment in esophageal squamous cell carcinoma: focus on diffusion-weighted magnetic resonance imaging, computed tomography and esophagography. Front Oncol 2024; 14:1397266. [PMID: 39026975 PMCID: PMC11256006 DOI: 10.3389/fonc.2024.1397266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Objective To identify the most sensitive imaging examination method to evaluate the prognosis of esophageal squamous cell carcinoma (ESCC). Materials and methods Thirty patients with esophageal squamous cell carcinoma (ESCC) participated in the study and underwent chemoradiotherapy (CRT). They were divided into two groups based on their survival status: the survival group and non-survival group. The diagnostic tests were utilized to determine the most effective imaging examination method for assessing the prognosis. Results 1. There were no significant differences in tumor length shown on esophagography or computed tomography (CT) or the maximal esophageal wall thickness shown on CT at the specified time points between the two groups. 2. The tumor length on diffusion-weighted imaging (DWI) in the survival group was significantly lower than in the non-survival group at the end of the sixth week of treatment (P=0.001). The area under the ROC curve was 0.840 (P=0.002), and the diagnostic efficiency was moderately accurate. 3. The apparent diffusion coefficient (ADC) values of the survival group were significantly higher than those in the non-survival group at the end of the fourth week and sixth week of treatment (both P<0.001). Areas under the curve were 0.866 and 0.970, with P values of 0.001 and <0.001 and good diagnostic accuracy. Cox regression analyses indicated the ADC at the end of the sixth week of treatment was an independent risk factor. Conclusions Compared with esophagography and CT, DW-MRI has certain advantages in predicting the prognosis of ESCC.
Collapse
Affiliation(s)
- Yang Li
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaohua Su
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Yuguang Shang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hui Liu
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Weishuai Wang
- CS Service AP, Siemens Healthineers Digital Health Technology (Shanghai) Co., Ltd. Beijing Branch, Beijing, China
| | - Andu Zhang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gaofeng Shi
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
4
|
Fan L, Yang Z, Chang M, Chen Z, Wen Q. CT-based delta-radiomics nomogram to predict pathological complete response after neoadjuvant chemoradiotherapy in esophageal squamous cell carcinoma patients. J Transl Med 2024; 22:579. [PMID: 38890720 PMCID: PMC11186275 DOI: 10.1186/s12967-024-05392-4] [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: 07/21/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND This study developed a nomogram model using CT-based delta-radiomics features and clinical factors to predict pathological complete response (pCR) in esophageal squamous cell carcinoma (ESCC) patients receiving neoadjuvant chemoradiotherapy (nCRT). METHODS The study retrospectively analyzed 232 ESCC patients who underwent pretreatment and post-treatment CT scans. Patients were divided into training (n = 186) and validation (n = 46) sets through fivefold cross-validation. 837 radiomics features were extracted from regions of interest (ROIs) delineations on CT images before and after nCRT to calculate delta values. The LASSO algorithm selected delta-radiomics features (DRF) based on classification performance. Logistic regression constructed a nomogram incorporating DRFs and clinical factors. Receiver operating characteristic (ROC) and area under the curve (AUC) analyses evaluated nomogram performance for predicting pCR. RESULTS No significant differences existed between the training and validation datasets. The 4-feature delta-radiomics signature (DRS) demonstrated good predictive accuracy for pCR, with α-binormal-based and empirical AUCs of 0.871 and 0.869. T-stage (p = 0.001) and differentiation degree (p = 0.018) were independent predictors of pCR. The nomogram combined the DRS and clinical factors improved the classification performance in the training dataset (AUCαbin = 0.933 and AUCemp = 0.941). The validation set showed similar performance with AUCs of 0.958 and 0.962. CONCLUSIONS The CT-based delta-radiomics nomogram model with clinical factors provided high predictive accuracy for pCR in ESCC patients after nCRT.
Collapse
Affiliation(s)
- Liyuan Fan
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhe Yang
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Minghui Chang
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Zheng Chen
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Qiang Wen
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
5
|
He Q, Shi X, Yan J, Wu M, Gu C, Yu X. Circulating tumor DNA serial monitoring of relapse and responses to tislelizumab immunotherapy as second‑line monotherapy for metastatic esophageal squamous cell carcinoma: A prospective study. Mol Clin Oncol 2024; 20:29. [PMID: 38414510 PMCID: PMC10895470 DOI: 10.3892/mco.2024.2727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/08/2024] [Indexed: 02/29/2024] Open
Abstract
Anti-programmed cell death 1 immuno-monotherapy has become the second-line standard treatment for advanced esophageal squamous cell carcinoma (ESCC) after the failure of first-line chemotherapy. However, new biomarkers are still needed to identify patients at risk of tumor progression and to select patients with advanced ESCC who are likely to benefit from immunotherapy. A total of 12 patients with advanced ESCC treated with tislelizumab were prospectively enrolled and endoscopic biopsy samples were collected. Plasma was obtained prior to and after every 2-3 treatment cycles with tislelizumab and when disease progression occurred. Targeted sequencing of 425 genes from plasma cell-free DNA, DNA from leukocytes and fixed esophageal tumor biopsies was performed. The patients underwent imaging analyses every 6-8 weeks until disease progression. The association between status of circulating tumor DNA (ctDNA) or changes in ctDNA following tislelizumab immunotherapy and response, tumor progression and survival was determined. All patients had evaluable next-generation sequencing results at the time of analysis. The results showed that patients with ESCC with liver metastasis had a significantly shorter median progression-free survival (mPFS: 1.4 vs. 11.7 months; P=0.037). TSC complex subunit 2 [11.7 months vs. not reached (NR); P=0.004] and zinc finger protein 217 (11.7 months vs. NR; P=0.022) gene mutations were the independent and negative prognostic factors for median overall survival (OS), respectively. Of note, ctDNA dynamic changes expressed as ∆ mutant molecules per milliliter of plasma (∆MMPM; MMPM detected at the first monitoring time-point after the first infusion of tislelizumab as baseline MMPM) predicted progression-free survival (PFS) and OS more accurately compared to the ctDNA change of an individual gene. ∆MMPM <20% was an independent predictor of PFS (2.8 vs. 14.6 months; P=0.029), although there was no significant difference for OS (16.7 vs. 17.6 months; P=0.830). In conclusion, changes in ctDNA levels were associated with anti-tumor effects, progression and disease-specific survival. ctDNA sequencing is promising for predicting response and progression after tislelizumab immunotherapy as second-line monotherapy for advanced ESCC [the present study was part of the RATIONALE-302 study (ClinicalTrials.gov identifier no. NCT03430843; 29.01.2018)].
Collapse
Affiliation(s)
- Qiong He
- Department of Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| | - Xun Shi
- Department of Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| | - Junrong Yan
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210032, P.R. China
| | - Mengmeng Wu
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210032, P.R. China
| | - Cuiping Gu
- Department of Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| | - Xinmin Yu
- Department of Oncology, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
6
|
Chi CL, Gao X, Hsieh HY, Cheng YH, Yang ZH, Chao YK. Survival Outcomes of Patients with Esophageal Cancer Who Did Not Proceed to Surgery after Neoadjuvant Treatment. Cancers (Basel) 2023; 15:4049. [PMID: 37627076 PMCID: PMC10452185 DOI: 10.3390/cancers15164049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND This retrospective study examined outcomes in esophageal squamous cell carcinoma (ESCC) patients who did not undergo surgical resection after neoadjuvant chemoradiotherapy (nCRT). METHODS Patients receiving nCRT between 2012 and 2020 were divided into two groups: group 1 (scheduled surgery) and group 2 (no surgery). Group 2 was further categorized into subgroups based on reasons for not proceeding to surgery: group 2a (disease progression), group 2b (poor general conditions), and group 2c (patient refusal). Overall survival (OS) was the primary outcome. RESULTS Group 1 comprised 145 patients, while subgroups 2a, 2b, and 2c comprised 24, 16, and 31 patients, respectively. The 3-year OS rate was significantly lower in group 2 compared with group 1 (34% versus 56%, p < 0.001). A subgroup analysis showed varying 3-year OS rates: 13% for group 2a, 25% for group 2b, and 58% for group 2c (p < 0.001). Propensity score matching for group 2c and group 1 revealed no significant difference in 3-year OS rates (p = 0.91). CONCLUSION One-third of ESCC patients receiving nCRT did not undergo surgical resection. Overall survival in this group was generally poorer, except for those who refused surgery (group 2c).
Collapse
Affiliation(s)
- Chun-Ling Chi
- Division of Thoracic Surgery, Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan 333, Taiwan; (C.-L.C.); (X.G.); (H.-Y.H.); (Y.-H.C.); (Z.-H.Y.)
| | - Xing Gao
- Division of Thoracic Surgery, Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan 333, Taiwan; (C.-L.C.); (X.G.); (H.-Y.H.); (Y.-H.C.); (Z.-H.Y.)
- Department of Surgery, Erasmus Medical Center, 3015GD Rotterdam, The Netherlands
| | - Hsiang-Yu Hsieh
- Division of Thoracic Surgery, Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan 333, Taiwan; (C.-L.C.); (X.G.); (H.-Y.H.); (Y.-H.C.); (Z.-H.Y.)
| | - Yi-Hsuan Cheng
- Division of Thoracic Surgery, Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan 333, Taiwan; (C.-L.C.); (X.G.); (H.-Y.H.); (Y.-H.C.); (Z.-H.Y.)
| | - Zhi-Hao Yang
- Division of Thoracic Surgery, Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan 333, Taiwan; (C.-L.C.); (X.G.); (H.-Y.H.); (Y.-H.C.); (Z.-H.Y.)
| | - Yin-Kai Chao
- Division of Thoracic Surgery, Chang Gung Memorial Hospital-Linkou, Chang Gung University, Taoyuan 333, Taiwan; (C.-L.C.); (X.G.); (H.-Y.H.); (Y.-H.C.); (Z.-H.Y.)
| |
Collapse
|
7
|
Albin J, Fahrig L, Siemanowski J, Rehkaemper J, Gebauer F, Zander T, Buettner R, Bruns CJ, Schroeder W, Alakus H, Hieggelke L, Quaas A. FGFR2-amplified tumor clones are markedly heterogeneously distributed in carcinomas of the upper gastrointestinal tract. J Cancer Res Clin Oncol 2023; 149:5289-5300. [PMID: 36416959 PMCID: PMC10349760 DOI: 10.1007/s00432-022-04460-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND FGFR2 is a therapy-relevant target in tumors of the upper gastrointestinal tract (GIT), and clinical trials are currently underway to test the efficacy of FGFR2 inhibitors. Tumor heterogeneity is one of the relevant causes of treatment failure. Almost nothing is known about the heterogeneous distribution of FGFR2-amplified clones in adenocarcinomas of the upper GIT. PATIENTS AND METHODS To assess FGFR2 gene copy number alteration and intratumoral heterogeneity of upper GIT adenocarcinomas, we analyzed 893 patient-derived formalin-fixed paraffin-embedded tumor specimens, including primary operated and neoadjuvant-treated tumors (462 gastric carcinomas and 429 esophageal adenocarcinomas) as well as complementary lymph node and distant metastasis by fluorescence in situ hybridization. RESULTS Twenty-six gastric tumors (5.6%) and 21 esophageal adenocarcinomas (4.9%) showed FGFR2 amplification. Overall, 93% of gastric carcinomas and 83% of esophageal carcinomas showed heterogeneous amplification. FGFR2 amplification was found in different histological growth patterns, including intestinal and diffuse type according to the Lauren classification. In the primary gastric carcinoma group, FGFR2 amplification was associated with poor prognosis (p = 0.005). CONCLUSION Homogeneous FGFR2 amplification in tumors of the upper GIT is the exception. This has highly relevant implications in the nature of FGFR2 diagnostics (sufficient tumor cell number, determination of amplification at metastasis versus primary tumor, etc.) and on the response probability of appropriate inhibitors. It is relevant that the often poorly treatable and aggressive subtype of diffuse carcinomas (poorly cohesive carcinomas) also shows FGFR2 amplification and that an individualized therapy option with FGFR2 inhibitors could be an option in this group.
Collapse
Affiliation(s)
- Jan Albin
- Institute of Pathology, University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | - Luca Fahrig
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Cologne, Germany
| | - Janna Siemanowski
- Institute of Pathology, University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | - Jan Rehkaemper
- Institute of Pathology, University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Cologne, Germany
| | - Thomas Zander
- Department of Internal Medicine I, University Hospital Cologne, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | | | - Wolfgang Schroeder
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Cologne, Germany
| | - Lena Hieggelke
- Institute of Pathology, University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany.
| |
Collapse
|
8
|
Zhang X, Lu M, Zhu J, Zhang C, Wang M. Altered genome‑wide hydroxymethylation analysis for neoadjuvant chemoradiotherapy followed by surgery in esophageal cancer. Exp Ther Med 2022; 25:29. [PMID: 36561617 PMCID: PMC9748644 DOI: 10.3892/etm.2022.11728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/14/2022] [Indexed: 11/26/2022] Open
Abstract
Esophageal cancer has high incidence rate in China. Neoadjuvant chemoradiotherapy (nCRT) has become the standard treatment for esophageal squamous cell carcinoma (ESCC). However, there are few reliable epigenetic parameters for patients with ESCC undergoing neoadjuvant therapy. Genomic extract from tumor tissue was amplified and sequenced using the Illumina HiSeq4000 to quantify genes associated methylation or hydromethylation in 12 patients with ESCC undergoing nCRT. The genome-wide hydroxymethylation were analyzed by methylated and hydroxymethylated DNA immunoprecipitation sequencing by MACS2 software and UCSC RefSeq database. Abnormal DNA methylation was statistically different between nCRT-well (showed a pathological complete response to nCRT) and nCRT-poor (showed incomplete pathological response to nCRT) patients. Levels of ten-eleven translocation 1, 2 and 3 mRNA and protein were higher in tumor tissue in nCRT-well group patients than in nCRT-poor group patients. Illumina HiSeq 4000 sequencing identified 2925 hypo-differentially hydroxymethylated region (DhMRs) and 292 hyper-DhMRs in promoter between nCRT-well and nCRT-poor patients. Biological processes associated with hyper-DhMRs included 'snRNA processing', 'hormone-mediated signaling pathway' and 'cellular response'. Metabolic processes were associated with hypo-DhMRs. These data may explain the functional response to nCRT in patients with abnormal promoter of methylation gene-associated mRNA expression. The present results implied that hyper-DhMRs and hypo-DhMRs affect molecular pathways, such as hippo and Notch signaling pathways, highlighting epigenetic modifications associated with clinical response to nCRT in patients with esophageal cancer.
Collapse
Affiliation(s)
- Xianjing Zhang
- The Second Clinical Department, Medical School of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Mingzhu Lu
- Department of Pathology, Changzhou Cancer Hospital, Soochow University, Changzhou, Jiangsu 213032, P.R. China
| | - Jing Zhu
- Department of Laboratory Medicine, Suzhou Science and Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China
| | - Changsong Zhang
- Department of Laboratory Medicine, Suzhou Science and Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215153, P.R. China,Correspondence to: Dr Changsong Zhang, Department of Laboratory Medicine, Suzhou Science and Technology Town Hospital, Gusu School, Nanjing Medical University, 1 Lijiang Road, Suzhou, Jiangsu 215153, P.R. China
| | - Meihua Wang
- Department of Pathology, Changzhou Cancer Hospital, Soochow University, Changzhou, Jiangsu 213032, P.R. China,Correspondence to: Dr Changsong Zhang, Department of Laboratory Medicine, Suzhou Science and Technology Town Hospital, Gusu School, Nanjing Medical University, 1 Lijiang Road, Suzhou, Jiangsu 215153, P.R. China
| |
Collapse
|
9
|
Hoppe S, Meder L, Gebauer F, Ullrich RT, Zander T, Hillmer AM, Buettner R, Plum P, Puppe J, Malter W, Quaas A. Trophoblast Cell Surface Antigen 2 (TROP2) as a Predictive Bio-Marker for the Therapeutic Efficacy of Sacituzumab Govitecan in Adenocarcinoma of the Esophagus. Cancers (Basel) 2022; 14:cancers14194789. [PMID: 36230712 PMCID: PMC9562858 DOI: 10.3390/cancers14194789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION The Trophoblast cell surface antigen 2 (TROP2) is expressed in many carcinomas and may represent a target for treatment. Sacituzumab govitecan (SG) is a TROP2-directed antibody-drug conjugate (ADC). Nearly nothing is known about the biological effectiveness of SG in esophageal adenocarcinoma (EAC). MATERIAL AND METHODS We determined the TROP2 expression in nearly 600 human EAC. In addition, we used the EAC cell lines (ESO-26, OACM5.1C, and FLO-1) and a xenograft mouse model to investigate this relationship. RESULTS Of 598 human EACs analyzed, 88% showed varying degrees of TROP2 positivity. High TROP2 positive ESO-26 and low TROP2 positive OACM5.1C showed high sensitivity to SG in contrast to negative FLO-1. In vivo, the ESO-26 tumor shows a significantly better response to SG than the TROP2-negative FLO-1 tumor. ESO-26 vital tumor cells show similar TROP2 expression on all carcinoma cells as before therapy initiation, FLO-1 is persistently negative. DISCUSSION Our data suggest that sacituzumab govitecan is a new therapy option in esophageal adenocarcinoma and the TROP2 expression in irinotecan-naïve EAC correlates with the extent of treatment response by sacituzumab govitecan. TROP2 is emerging as a predictive biomarker in completely TROP2-negative tumors. This should be considered in future clinical trials.
Collapse
Affiliation(s)
- Sascha Hoppe
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Lydia Meder
- Internal Medicine, Oncology Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
- Mildred Scheel School of Oncology Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Florian Gebauer
- Department of Gynecology and Obstetrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Roland T. Ullrich
- Internal Medicine, Oncology Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
- Mildred Scheel School of Oncology Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Thomas Zander
- Internal Medicine, Oncology Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Axel M. Hillmer
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Reinhard Buettner
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Patrick Plum
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Julian Puppe
- Department of Gynecology and Obstetrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Wolfram Malter
- Department of Gynecology and Obstetrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
| | - Alexander Quaas
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Köln, Germany
- Correspondence: ; Tel.: +49-0221-478-5257; Fax: +49-0221-478-6360
| |
Collapse
|
10
|
Taghizadeh Kermani A, Ghanbarzadeh R, Joudi Mashhad M, Javadinia SA, Emadi Torghabeh A. Predictive Value of Endoscopic Observations and Biopsy After Neoadjuvant Chemoradiotherapy in Assessing the Pathologic Complete Response of Patients With Esophageal Squamous Cell Carcinoma. Front Oncol 2022; 12:859079. [PMID: 35646696 PMCID: PMC9132043 DOI: 10.3389/fonc.2022.859079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction No standard method has been defined to evaluate the therapeutic response of esophageal cancer to neoadjuvant chemoradiotherapy (CRT). This study aimed to determine the predictive value of endoscopic evaluation and biopsy after CRT in predicting the complete pathological response to neoadjuvant CRT in patients with esophageal squamous cell carcinoma (SCC). Materials and Method This prospective, descriptive study was conducted on patients with stage II and III esophageal SCC who could undergo esophagectomy. Patients underwent neoadjuvant CRT. Four to six weeks after the end of treatment, re-endoscopy was performed and a biopsy was taken in the presence of a tumor lesion. In the absence of a tumor lesion, the marked site of the esophagus was removed as a blind biopsy. Gastrologist observations during endoscopy and the result of the pathological examination of an endoscopic biopsy were recorded. The patient underwent esophagectomy. The pathology obtained from endoscopic biopsy was compared with the pathology response obtained from esophagectomy. Results Sixty-nine patients were included in the study, of which 32 underwent esophagectomy. In an endoscopic examination after CRT, 28 patients had macroscopic tumor remnants and 4 patients did not. Pathological examination of the samples obtained from endoscopy showed no tumor remnants in 10 patients (31.3%), and in 22 patients (68.7%), living tumor remnants were seen in the biopsy specimen. Pathologic evaluation of the samples obtained by surgical resection showed that in 13 patients, there were no viable carcinomas in the esophagus or lymph nodes removed, and the rate of pathologic complete response was 40.6. Sensitivity, specificity, positive predictive, and negative predictive values of endoscopic observations were 94.7, 23, 64.2, and 75%, respectively. Preoperative biopsy sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were 68.4, 30.7, 59, and 40%, respectively. Conclusion Considering the negative and positive predictive values of endoscopic observations and biopsy after neoadjuvant CRT, it seems that these two methods alone are not suitable for assessing the pathologic complete response after neoadjuvant treatment.
Collapse
Affiliation(s)
| | - Raha Ghanbarzadeh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Joudi Mashhad
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Alireza Javadinia
- Non-Communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Ali Emadi Torghabeh
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Damanakis A, Plum PS, Gebauer F, Schröder W, Büttner R, Zander T, Bruns CJ, Quaas A. Fructose-1,6-bisphosphatase 1 (FBP1) is an independent biomarker associated with a favorable prognosis in esophageal adenocarcinoma. J Cancer Res Clin Oncol 2022; 148:2287-2293. [PMID: 35477823 PMCID: PMC9349078 DOI: 10.1007/s00432-022-04025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022]
Abstract
Introduction Despite modern multimodal therapeutic regimens, the prognosis of esophageal adenocarcinoma (EAC) is still poor and there is a lack of biological markers estimating the patients’ prognosis. Fructose-1,6-biphosphatase (FBP1) is a key enzyme in gluconeogenesis and is associated with tumor initiation in several cancers. Therefore, this study aims to characterize its implication for EAC patients. Methods and materials A total of 571 EAC patients who underwent multimodal treatment between 1999 and 2017 were analyzed for FBP1 expression using immunohistochemistry. Results 82.5% of the EACs show FBP1 expression in the tumor albeit with different intensities categorizing specimens accordingly into score 0 (no expression), score 1 (weak expression), score 2 (moderate expression) and score 3 (strong expression) (score 1 = 25.0%, score 2 = 35.9%, score 3 = 21.5%). Intratumoral FBP1 expression was significantly associated with a better prognosis (p = 0.024). This observation was particularly relevant among patients who received primary surgery without neoadjuvant treatment (p = 0.004). In multivariate analysis, elevated FBP1 expression was an independent biomarker associated with a favorable prognosis. Discussion Despite being associated with a favorable prognosis, the majority of patients with high FBP1 expression also require individualized therapy options to ensure long-term survival. Recently, it has been shown that the presence of the FBP1 protein increases the sensitivity of pancreatic cancer cells to the bromodomain and extraterminal domain (BET) inhibitor JQ1. Conclusion We described for the first time the prognostic and possibly therapeutic relevance of FBP1 in EAC. The efficiency of the BET inhibitor in EAC should be verified in clinical studies and special attention should be paid to the effects of neoadjuvant therapy on FBP1 expression. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-022-04025-x.
Collapse
Affiliation(s)
- Alexander Damanakis
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Patrick Sven Plum
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, Cologne, Germany. .,Gastrointestinal Cancer Group Cologne (GCGC), Cologne, Germany.
| | - Florian Gebauer
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Wolfgang Schröder
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Thomas Zander
- Department I of Internal Medicine, Center for Integrated Oncology (CIO), Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Gastrointestinal Cancer Group Cologne (GCGC), Cologne, Germany
| | - Christiane Josephine Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Gastrointestinal Cancer Group Cologne (GCGC), Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Gastrointestinal Cancer Group Cologne (GCGC), Cologne, Germany
| |
Collapse
|
12
|
Kütting F, Gebauer F, Zweerink S, Krämer L, Schramm C, Quaas A, Bruns C, Goeser T, Nierhoff D. Expression of Neighbor of Punc E11 (NOPE) in early stage esophageal adenocarcinoma is associated with reduced survival. Sci Rep 2022; 12:3584. [PMID: 35246597 PMCID: PMC8897453 DOI: 10.1038/s41598-022-07580-y] [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: 06/11/2021] [Accepted: 12/02/2021] [Indexed: 11/10/2022] Open
Abstract
Current recommendations suggest neoadjuvant treatment in node-positive esophageal cancer or tumors staged T3 and upwards but some T2 N0 patients might benefit from neoadjuvant therapy. It is of clinical relevance to identify this subgroup. Loss of epithelial apicobasal polarity is a key factor in the development of invasive capabilities of carcinoma. The oncofetal stem/progenitor cell marker NOPE is expressed in adult depolarized murine hepatocytes and in murine/human hepatocellular carcinoma. We analyzed NOPE expression in 363 patients with esophageal adenocarcinoma using an RNA Scope Assay on a tissue microarray and correlated results with clinical data. Median follow-up was 57.7 months with a 5-year survival rate of 26.6%. NOPE was detectable in 32 patients (8.8%). In pT1/2 stages, NOPE expression was associated with a significantly reduced median OS of 6.3 months (95% CI 1.2-19.4 months), the median OS is not reached in the NOPE-negative group (calculated mean OS 117.1 months) (P = 0.012). In advanced tumor stages, a NOPE dependent survival difference was not detected. This is the first report of NOPE expression demonstrating a prognostic value in esophageal cancer. Early stage, NOPE positive patients are at a high risk of tumor progression and may benefit from neoadjuvant treatment analogous to advanced stage cancer.
Collapse
Affiliation(s)
- Fabian Kütting
- Department of Gastroenterology and Hepatology, University Hospital of Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Florian Gebauer
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Susanne Zweerink
- Department of Gastroenterology and Hepatology, University Hospital of Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Laurenz Krämer
- Department of Gastroenterology and Hepatology, University Hospital of Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Christoph Schramm
- Department of Gastroenterology and Hepatology, University Hospital of Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Tobias Goeser
- Department of Gastroenterology and Hepatology, University Hospital of Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Dirk Nierhoff
- Department of Gastroenterology and Hepatology, University Hospital of Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
13
|
Chidambaram S, Markar SR. Clinical utility and applicability of circulating tumor DNA testing in esophageal cancer: a systematic review and meta-analysis. Dis Esophagus 2022; 35:doab046. [PMID: 34286823 PMCID: PMC8832526 DOI: 10.1093/dote/doab046] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022]
Abstract
Esophageal cancer is an aggressive malignancy with a relatively poor prognosis even after multimodality therapy. Currently, patients undergo a series of investigations that can be invasive and costly or pose secondary risks to their health. In other malignancies, liquid biopsies of circulating tumor DNA (ctDNA) are used in clinical practice for diagnostic and surveillance purposes. This systematic review summarizes the latest evidence for the clinical applicability of ctDNA technology in esophageal cancer. A systematic review of the literature was performed using MEDLINE, EMBASE, the Cochrane Review and Scopus databases. Articles were evaluated for the use of ctDNA for diagnosis and monitoring of patients with esophageal cancer. Quality assessment of studies was performed using the QUADAS-2 tool. A meta-analysis was performed to assess the diagnostic accuracy of sequencing methodologies. We included 15 studies that described the use of ctDNA technology in the qualitative synthesis and eight studies involving 414 patients in the quantitative analysis. Of these, four studies assessed its utility in cancer diagnosis, while four studies evaluated its use for prognosis and monitoring. The pooled sensitivity and specificity for diagnostic studies were 71.0% (55.7-82.6%) and 98.6% (33.9-99.9%), while the pooled sensitivity and specificity for surveillance purposes were 48.9% (29.4-68.8%) and 95.5% (90.6-97.9%). ctDNA technology is an acceptable method for diagnosis and monitoring with a moderate sensitivity and high specificity that is enhanced in combination with current imaging methods. Further work should demonstrate the practical integration of ctDNA in the diagnostic and surveillance clinical pathway.
Collapse
Affiliation(s)
| | - Sheraz R Markar
- Department of Surgery and Cancer, Imperial College London, London, UK
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
14
|
Shi Y, Ge X, Ju M, Zhang Y, Di X, Liang L. Circulating Tumor Cells in Esophageal Squamous Cell Carcinoma - Mini Review. Cancer Manag Res 2021; 13:8355-8365. [PMID: 34764697 PMCID: PMC8577339 DOI: 10.2147/cmar.s337489] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/24/2021] [Indexed: 12/30/2022] Open
Abstract
Esophageal cancer has high incidence and mortality rates and a low five-year survival rate of <15% owing to its strong capabilities of invasion, relapse and metastasis. The classic view holds that metastasis and diffusion is an advanced event during cancer progression, but recent studies show that distant diffusion of primary cancer cells may actually be an early event. Detection of circulating tumor cells (CTCs) in the circulation may indicate tumor spread, so CTCs are considered to be the key factor of metastatic cascade. In recent years, despite research progress on CTCs, there is a lack of systematic and important evidence to confirm the diagnostic, monitoring and prognostic values of CTCs in esophageal squamous cell carcinoma (ESCC). In this review, we clarify the relationship between CTC values and ESCC and provide more reliable evidence to improve the management and treatment of ESCC.
Collapse
Affiliation(s)
- Yujing Shi
- Jurong People's Hospital, Zhenjiang, 212400, People's Republic of China
| | - Xiaolin Ge
- Jiangsu Provincial People's Hospital, Nanjing, 212000, People's Republic of China
| | - Mengyang Ju
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, 5650871, Japan
| | - Yumeng Zhang
- Nanjing Medical University, Nanjing, 212000, People's Republic of China
| | - Xiaoke Di
- Jiangsu Provincial People's Hospital, Nanjing, 212000, People's Republic of China
| | - Liang Liang
- Jurong People's Hospital, Zhenjiang, 212400, People's Republic of China
| |
Collapse
|
15
|
Zhou N, Mitchell KG, Corsini EM, Truong VTT, Antonoff MB, Mehran RJ, Rajaram R, Rice DC, Roth JA, Sepesi B, Swisher SG, Vaporciyan AA, Walsh GL, Ajani JA, Hofstetter WL. Analysis of trimodal and bimodal therapy in a selective-surgery paradigm for locally advanced oesophageal squamous cell carcinoma. Br J Surg 2021; 108:1207-1215. [PMID: 34095952 DOI: 10.1093/bjs/znab162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/14/2021] [Indexed: 11/14/2022]
Abstract
BACKGROUND Long-term survival outcomes of trimodal therapy (TMT; chemoradiation plus surgery) and bimodal therapy (BMT; chemoradiation) have seldom been analysed. In a selective-surgery paradigm, the benefit of TMT in patients with a complete clinical response is controversial. Factors associated with survival in patients with a clinical complete response to chemoradiation were evaluated. METHODS Patients with stage II-III oesophageal squamous cell carcinoma treated with TMT or BMT from 2002 to 2017 were evaluated. The BMT group consisted of patients who were otherwise eligible for surgery but underwent chemoradiation alone followed by observation. This group included patients who later had salvage oesophagectomy. Survival was evaluated and compared between TMT and BMT groups. Elastic net regularization was performed to select co-variables for Cox multivariable survival analysis in patients with a clinical complete response. RESULTS Of 143 patients, 60 (41.9 per cent) underwent TMT and 83 (58.0 per cent) BMT. Patients who underwent TMT had longer median overall survival than those who had BMT (77 versus 33 months; P = 0.019). For patients with a clinical complete response, TMT achieved longer median overall survival than BMT (123 versus 55 months; P = 0.04). BMT had a high locoregional recurrence rate (48 versus 6 per cent; P < 0.001); 26 of 29 patients with locoregional recurrence in the BMT groupunderwent salvage resection. Cox multivariable analysis demonstrated that upper-mid oesophageal tumour location (hazard ratio (HR) 2.04; P = 0.024) and tumour length (HR 1.18; P = 0.046) were associated with worse survival. Although TMT was not associated with survival, it was a predictor of reduced recurrence (HR 0.28; P = 0.028). The maximum standardized uptake value after chemoradiation also predicted recurrence (HR 1.33; P < 0.001). CONCLUSION In patients who achieve a clinical complete response, TMT reduces locoregional recurrence but may not prolong survival. The differences in survival outcomes may be due to patient selection; therefore, a selective-surgery strategy in oesophageal squamous cell carcinoma is a reasonable approach.
Collapse
Affiliation(s)
- N Zhou
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - K G Mitchell
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - E M Corsini
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - V T T Truong
- Center for Clinical Research and Evidence-Based Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
| | - M B Antonoff
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - R J Mehran
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - R Rajaram
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - D C Rice
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - J A Roth
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - B Sepesi
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - S G Swisher
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - A A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - G L Walsh
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - J A Ajani
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - W L Hofstetter
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
16
|
Mori T, Kumagai K, Nasu K, Yoshizawa T, Kuwano K, Hamada Y, Kanazawa H, Suzuki R. Clonal Expansion of Tumor-Infiltrating T Cells and Analysis of the Tumor Microenvironment within Esophageal Squamous Cell Carcinoma Relapsed after Definitive Chemoradiation Therapy. Int J Mol Sci 2021; 22:ijms22031098. [PMID: 33499345 PMCID: PMC7865796 DOI: 10.3390/ijms22031098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 12/18/2022] Open
Abstract
(1) Background: Comparable prognoses after definitive chemoradiation therapy (CRT) to surgery alone for esophageal squamous cell carcinoma (ESCC) have been previously reported; however, no robust prognostic markers have been established. The clonality of tumor-infiltrating lymphocytes (TILs) and tumor microenvironments (TMEs) in ESCC relapsed after CRT were examined to explore prognostic markers. (2) Methods: Clonality of TIL and TME were examined in ESCC with and without preceding CRT, as well as oral squamous cell carcinoma (OSCC) and healthy volunteers as controls. The clonality of TIL was assessed by T-cell receptor (TCR) α and β repertoire analyses and evaluated by diversity indices. The TME was assessed by quantitative polymerase chain reaction evaluating PD-L1 and CD8B. (3) Results: The clonal expansion of TIL was significantly induced within ESCCs and OSCCs, when compared to healthy volunteers, and was mostly induced within ESCCs after definitive CRT. Diversity indices of TIL were not associated with the prognosis, but the ratio of PD-L1 mRNA to CD8B mRNA in TME was significantly associated with a poor prognosis after salvage surgery (p = 0.007). (4) Conclusions: The clonal expansion of TIL is induced after definitive CRT for ESCC, and the ratio of PD-L1 mRNA to CD8B mRNA within tumor tissues is a prognostic marker candidate for salvage esophagectomy after CRT.
Collapse
Affiliation(s)
- Takahiro Mori
- Departments of Clinical Oncology and Gastroenterological Surgery, National Hospital Organization Sagamihara National Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara 252-0392, Japan
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Rheumatology and Allergy, National Hospital Organization Sagamihara National Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara 252-0385, Japan; (K.K.); (K.N.); (T.Y.); (K.K.); (R.S.)
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan;
- Correspondence:
| | - Kenichi Kumagai
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Rheumatology and Allergy, National Hospital Organization Sagamihara National Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara 252-0385, Japan; (K.K.); (K.N.); (T.Y.); (K.K.); (R.S.)
- Department of Surgery, National Hospital Organization Sagamihara National Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara 252-0392, Japan;
- Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aobaku, Sendai 980-8574, Japan
| | - Keisuke Nasu
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Rheumatology and Allergy, National Hospital Organization Sagamihara National Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara 252-0385, Japan; (K.K.); (K.N.); (T.Y.); (K.K.); (R.S.)
- Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aobaku, Sendai 980-8574, Japan
| | - Takamasa Yoshizawa
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Rheumatology and Allergy, National Hospital Organization Sagamihara National Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara 252-0385, Japan; (K.K.); (K.N.); (T.Y.); (K.K.); (R.S.)
- Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aobaku, Sendai 980-8574, Japan
| | - Koji Kuwano
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Rheumatology and Allergy, National Hospital Organization Sagamihara National Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara 252-0385, Japan; (K.K.); (K.N.); (T.Y.); (K.K.); (R.S.)
- Department of Oral-Maxillofacial Surgery and Orthodontics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yoshiki Hamada
- Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aobaku, Sendai 980-8574, Japan
| | - Hideki Kanazawa
- Department of Oral-Maxillofacial Surgery and Orthodontics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ryuji Suzuki
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Rheumatology and Allergy, National Hospital Organization Sagamihara National Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara 252-0385, Japan; (K.K.); (K.N.); (T.Y.); (K.K.); (R.S.)
| |
Collapse
|
17
|
Quaas A, Pamuk A, Klein S, Quantius J, Rehkaemper J, Barutcu AG, Rueschoff J, Zander T, Gebauer F, Hillmer A, Buettner R, Schroeder W, Bruns CJ, Löser H, Schoemig-Markiefka B, Alakus H. Sex-specific prognostic effect of CD66b-positive tumor-infiltrating neutrophils (TANs) in gastric and esophageal adenocarcinoma. Gastric Cancer 2021; 24:1213-1226. [PMID: 34009535 PMCID: PMC8502159 DOI: 10.1007/s10120-021-01197-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Tumor-associated neutrophils (TANs) have recently been identified as a relevant component of the tumor microenvironment (TME) in solid tumors. Within the TME TANs mediate either tumor-promoting or tumor-inhibiting activities. So far, their prognostic relevance remains to be determined. This study aims to evaluate the prognostic relevance of TANs in different molecular subtypes of gastric and esophageal adenocarcinoma. METHODS We analyzed a total of 1118 Caucasian patients divided into gastric adenocarcinoma (n = 458) and esophageal adenocarcinoma cohort (n = 660) of primarily resected and neoadjuvant-treated individuals. The amount of CD66b + TANs in the tumor stroma was determined using quantitative image analysis and correlated to both molecular, as well as clinical data. RESULTS An accumulation of TANs in the tumor stroma of gastric carcinomas was associated to a significant favorable prognosis (p = 0.026). A subgroup analysis showed that this effect was primarily related to the molecular chromosomal instable subtype (CIN) of gastric carcinomas (p = 0.010). This was only observed in female patients (p = 0.014) but not in male patients (p = 0.315). The same sex-specific effect could be confirmed in adenocarcinomas of the esophagus (p = 0.027), as well as in female individuals after receiving neoadjuvant therapy (p = 0.034). CONCLUSIONS Together, we show a sex-specific prognostic effect of TANs in gastric cancer within a Caucasian cohort. For the first time, we showed that this sex-specific prognostic effect of TANs can also be seen in esophageal cancer.
Collapse
Affiliation(s)
- Alexander Quaas
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Aylin Pamuk
- grid.411097.a0000 0000 8852 305XDepartment of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Sebastian Klein
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jennifer Quantius
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jan Rehkaemper
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Atakan G. Barutcu
- grid.411097.a0000 0000 8852 305XDepartment of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Josef Rueschoff
- Institute of Pathology, Nordhessen and Targos Molecular Pathology GmbH, Kassel, Germany
| | - Thomas Zander
- grid.6190.e0000 0000 8580 3777Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| | - Florian Gebauer
- grid.411097.a0000 0000 8852 305XDepartment of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Axel Hillmer
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Reinhard Buettner
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Wolfgang Schroeder
- grid.411097.a0000 0000 8852 305XDepartment of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Christiane J. Bruns
- grid.411097.a0000 0000 8852 305XDepartment of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Heike Löser
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Birgid Schoemig-Markiefka
- grid.411097.a0000 0000 8852 305XInstitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Hakan Alakus
- grid.411097.a0000 0000 8852 305XDepartment of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
18
|
Loeser H, Kraemer M, Gebauer F, Bruns C, Schröder W, Zander T, Alakus H, Hoelscher A, Buettner R, Lohneis P, Quaas A. Indoleamine 2,3-Dioxygenase (IDO) Expression Is an Independent Prognostic Marker in Esophageal Adenocarcinoma. J Immunol Res 2020; 2020:2862647. [PMID: 33029538 PMCID: PMC7527882 DOI: 10.1155/2020/2862647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase (IDO) is an interferon-inducible immune checkpoint expressed on tumor-infiltrating lymphocytes (TILs). IDO is known as a poor prognostic marker in esophageal squamous cell cancer, while a positive effect was shown for breast cancer. A comprehensive analysis of IDO expression in a well-defined cohort of esophageal adenocarcinoma (EAC) is missing. METHODS We analyzed 551 patients with EAC using single-protein and multiplex immunohistochemistry as well as mRNA in situ technology for the expression and distribution of IDO on subtypes of TILs (INF-γ mRNA and CD4- and CD8-positive T lymphocytes). RESULTS IDO expression on TILs was seen in up to 59.6% of tumors, and expression on tumor cells was seen in 9.2%. We found a strong positive correlation of IDO-positive TILs, CD3-positive T lymphocytes, and INF-γ mRNA-producing TILs in the tumor microenvironment of EACs showing significantly better overall survival (47.7 vs. 22.7 months, p < 0.001) with emphasis on early tumor stages (pT1/2: 142.1 vs. 37.1 months, p < 0.001). In multivariate analysis, IDO is identified as an independent prognostic marker. CONCLUSIONS Our study emphasizes the importance of immunomodulation in EAC marking IDO as a potential biomarker. Beyond this, IDO might indicate a subgroup of EAC with an explicit survival benefit.
Collapse
Affiliation(s)
- Heike Loeser
- Institute of Pathology, University Hospital Cologne, Germany
- Gastrointestinal Cancer Group Cologne, Department I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| | - Max Kraemer
- Institute of Pathology, University Hospital Cologne, Germany
| | - Florian Gebauer
- Gastrointestinal Cancer Group Cologne, Department I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Germany
| | - Wolfgang Schröder
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Germany
| | - Thomas Zander
- Gastrointestinal Cancer Group Cologne, Department I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
- Department I of Internal Medicine, Center for Integrated Oncology (CIO), University Hospital Cologne, Germany
| | - Hakan Alakus
- Gastrointestinal Cancer Group Cologne, Department I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Germany
| | - Arnulf Hoelscher
- Center for Esophageal and Gastric Surgery, AGAPLESION Markus Krankenhaus, Frankfurt, Germany
| | | | - Philipp Lohneis
- Institute of Pathology, University Hospital Cologne, Germany
- Gastrointestinal Cancer Group Cologne, Department I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, Germany
- Gastrointestinal Cancer Group Cologne, Department I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
19
|
Narumiya K, Bollschweiler E, Hölscher AH, Yamamoto M, Drebber U, Alakus H, Metzger R, Warnecke-Eberz U. Different response rates to chemotherapy between Japanese and German esophageal squamous cell carcinoma: patients may be influenced by ERCC1 or ABCB1. Future Oncol 2020; 16:2075-2087. [PMID: 32611208 DOI: 10.2217/fon-2020-0489] [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] [Indexed: 12/13/2022] Open
Abstract
Aim: To find out differences in biomarkers between Japanese and German patients responsible for response after neoadjuvant radio/chemotherapy and survival for esophageal squamous cell carcinoma. Materials & methods: A total of 60 patients from Japan and 127 patients from Germany with esophageal squamous cell carcinoma were analyzed according to three SNPs by real-time PCR. Results: The distribution of the genotypes of ERCC1 rs16115 and ABCB1 C3435T rs1045642 was significantly different between both patients' groups. Japanese patients had significantly less good response to 5-fluorouracil/cisplatin chemotherapy. The influence of the three SNPs on response varied between patients from Japan and Germany. Conclusion: Different expressions of ERCC1 and ABCB1 SNPs of Japanese patients compared with the German patients partially explain the different response.
Collapse
Affiliation(s)
- Kosuke Narumiya
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Kawada 8-1, Shinjyuku, Tokyo, 1628666, Japan
| | | | - Arnulf H Hölscher
- Medical Faculty, University of Cologne, Kerpener Str. 62, Cologne, 50924, Germany.,Head Contilia Center for Esophageal Diseases, Elisabeth Hospital, Klara-Kopp-Weg 1, Essen, 45138, Germany
| | - Masakazu Yamamoto
- Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Kawada 8-1, Shinjyuku, Tokyo, 1628666, Japan
| | - Uta Drebber
- Institute of Pathology, University of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| | - Hakan Alakus
- Department of General, Visceral & Cancer Surgery, University of Cologne, Kerpner Str. 62, Cologne, 50924, Germany
| | - Ralf Metzger
- Department of General, Visceral, Thoracic & Cancer Surgery, Caritas Klinikum, Rhine Str. 2, Saarbrücken, 66113, Germany
| | - Ute Warnecke-Eberz
- Department of General, Visceral & Cancer Surgery, University of Cologne, Kerpner Str. 62, Cologne, 50924, Germany
| |
Collapse
|
20
|
Maffazzioli L, Zilio MB, Klamt AL, Duarte JA, Mazzini GS, Campos VJ, Chedid MF, Gurski RR. ADC as a predictor of pathologic response to neoadjuvant therapy in esophageal cancer: a systematic review and meta-analysis. Eur Radiol 2020; 30:3934-3942. [PMID: 32157409 DOI: 10.1007/s00330-020-06723-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/31/2020] [Accepted: 02/05/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Diffusion-weighted magnetic resonance imaging (DWI) is part of clinical practice. The aim of this study was to evaluate the role of apparent diffusion coefficient (ADC) as a predictor of pathologic response to neoadjuvant therapy (nCRT) in patients with esophageal cancer (EC). METHODS The MEDLINE, Embase, and Google Scholar databases were systematically searched for studies using ADC to evaluate response to neoadjuvant therapy in patients with EC. Methodological quality of the studies was evaluated with the QUADAS tool. Data from eligible studies were extracted and evaluated by two independent reviewers. Meta-analyses were performed comparing mean ADC values between responders and non-responders to nCRT in three different scenarios: baseline (BL) absolute values; percent change between intermediate (IM) values and BL; and percent change between final follow-up (FU) value and baseline BL. RESULTS Seven studies (n = 158 patients) were included. Responders exhibited a statistically significant percent increase in ADC during nCRT (mean difference [MD] 21.06%, 95%CI = 13.04-29.09; I2 = 49%; p = 0.12). A similar increase was identified in the complete pathologic response (pCR) versus non-complete pathologic response (npCR) subgroup (MD = 25.68%, 95%CI = 18.87-32.48; I2 = 0%; p = 0.60). At the end of treatment, responders also exhibited a statistically significant percent increase in ADC (MD = 22.49%, 95%CI = 9.94-35.05; I2 = 0%; p = 0.46). BL ADC was not associated with any definition of pathologic response (MD = 0.11%, 95%CI = - 0.21-0.42; I2 = 85%; p < 0.01). CONCLUSION These results suggest that ADC can be used as a predictor of pathologic response, with a statistically significant association between percent ADC increase during and after treatment and pCR. ADC may serve as a tool to help in guiding clinical decisions. KEY POINTS • DWI is routinely included in MRI oncological protocols. • ADC can be used as a predictor of pathologic response, with a statistically significant association between percent ADC increase during and after treatment and pCR.
Collapse
Affiliation(s)
- Leticia Maffazzioli
- Division of Radiology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2350, 2nd Floor, Porto Alegre, 90035-903, Brazil.
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil.
| | - Mariana B Zilio
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| | - Alexandre L Klamt
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
- Division of Gastroenterology, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| | - Juliana A Duarte
- Division of Radiology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2350, 2nd Floor, Porto Alegre, 90035-903, Brazil
| | - Guilherme S Mazzini
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| | - Vinicius J Campos
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
| | - Marcio F Chedid
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| | - Richard R Gurski
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| |
Collapse
|
21
|
Triantafyllou T, Wijnhoven B. Multidisciplinary treatment of esophageal cancer: The role of active surveillance after neoadjuvant chemoradiation. Ann Gastroenterol Surg 2020; 4:352-359. [PMID: 32724878 PMCID: PMC7382442 DOI: 10.1002/ags3.12350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/29/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022] Open
Abstract
The optimal treatment of esophageal cancer is still controversial. Neoadjuvant chemoradiotherapy followed by radical esophagectomy is a standard treatment. Morbidity after esophagectomy however is still considerable and has an impact on patients' quality of life. Given a pathologic complete response rate of approximately 30% in patients after neoadjuvant chemoradiation followed by surgery, active surveillance has been introduced as a new alternative approach. Active surveillance involves regular clinical response evaluations in patients after neoadjuvant therapy to detect residual or recurrent disease. As long as there is no suspicion of disease activity, surgery is withheld. Esophagectomy is reserved for patients presenting with an incomplete response or resectable recurrent disease. Active surveillance after neoadjuvant treatment has been previously applied in other types of malignancy with encouraging results. This paper discusses its role in esophageal cancer.
Collapse
Affiliation(s)
- Tania Triantafyllou
- Department of SurgeryHippocration General Hospital of AthensNational and Kapodistrian University of AthensAthensGreece
| | - Bas Wijnhoven
- Department of SurgeryErasmus University Medical CenterRotterdamthe Netherlands
| |
Collapse
|
22
|
Y Chromosome Loss is a Frequent Event in Barrett's Adenocarcinoma and Associated with Poor Outcome. Cancers (Basel) 2020; 12:cancers12071743. [PMID: 32629877 PMCID: PMC7408596 DOI: 10.3390/cancers12071743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 01/06/2023] Open
Abstract
Background: The loss of the Y chromosome in various malignant diseases has been described previously. There are no reliable information on the actual frequency, significance and homogeneity of Y chromosome loss (LoY) in esophageal adenocarcinoma (EAC). Methods: 400 male EAC including lymph-node metastases were analyzed with commercially available Y chromosome specific fluorescence in-situ probes. The results were correlated with molecular and immunohistochemical markers and clinicopathological aspects. Results: The entire cohort (n = 400) showed a singular LoY of one chromosome arm in 1.0% (q-arm) and 2.8% (p-arm), complete LoY in 52.5%. LoY was strongly associated with shortened overall-survival (OS). Patients with preserved Y chromosome had a median OS of 58.8 months, patients with LoY an OS of 19.4 months (p < 0.001). Multivariate analysis showed LoY as an independent prognostic marker with a hazard ratio of 1.835 (95% CI 1.233–2.725). LoY correlated with TP53 mutations (p = 0.003), KRAS amplification (p = 0.004), loss of ARID1a (p = 0.045) and presence of LAG3 (p = 0.018). Conclusions: Loss of the Y chromosome is a very common phenomenon in EAC. The LoY is heterogeneously distributed within the tumor, but corresponding lymph node metastases frequently show homogeneous LoY, indicating a selection and metastasizing advantage with poor prognosis. To date, the male predominance of EAC (7–9:1) is unclear, so genetic explanatory models are favored. The LoY in EAC may be biologically and functionally relevant and additional genomic or functional analyses are needed.
Collapse
|
23
|
Lymphocyte activation gene-3 (LAG3) mRNA and protein expression on tumour infiltrating lymphocytes (TILs) in oesophageal adenocarcinoma. J Cancer Res Clin Oncol 2020; 146:2319-2327. [PMID: 32592066 PMCID: PMC7382658 DOI: 10.1007/s00432-020-03295-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/20/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Lymphocyte activation gene-3 (LAG3) is an immunosuppressive checkpoint molecule expressed on T cells. The frequency and distribution of LAG3 expression in oesophageal adenocarcinoma (EAC) is unknown. Aim of the study was the evaluation and distribution of LAG3 on tumour infiltrating lymphocytes (TILs) and correlation with clinico-pathological and molecular data. METHODS We analysed tumor tissue samples using immunohistochemistry, multi-colour immunofluorescence and mRNA in-situ technology. The analyses were performed on a multi-spot tissue microarray (TMA) with 165 samples, followed by an evaluation on a single-spot TMA with 477 samples. These results were correlated with clinical and molecular tumour data. RESULTS LAG3 expression on TILs was detectable in 10.5% on the multi-spot TMA and 11.4% on the single-spot TMA. There was a strong correlation between protein expression and mRNA expression (p < 0.001) in TILs. LAG 3 expression was correlated with CD4+ and CD8+ T-cells within the tumor (p < 0.001). LAG3 expression showed an improved overall survival (OS) compared to patients without LAG3 expression (median OS 70.2 vs. 26.9 months; p = 0.046). The effect was even clearer in the group of patients with tumour stages > pT2 (70.2 vs 25.0 months; p = 0.037). CONCLUSION This is the first description of LAG3 expression on TILs in EAC, underscoring the importance of immunomodulation in EAC. Our data suggest an impact of LAG3 in a relevant subset of EAC. Therapeutic studies investigating the efficacy of LAG3 inhibition in EAC will also provide predictive evidence and relevance of the immunohistochemical determination of LAG3 expression.
Collapse
|
24
|
Moentenich V, Comut E, Gebauer F, Tuchscherer A, Bruns C, Schroeder W, Buettner R, Alakus H, Loeser H, Zander T, Quaas A. Mesothelin expression in esophageal adenocarcinoma and squamous cell carcinoma and its possible impact on future treatment strategies. Ther Adv Med Oncol 2020; 12:1758835920917571. [PMID: 32547645 PMCID: PMC7249595 DOI: 10.1177/1758835920917571] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/06/2020] [Indexed: 12/29/2022] Open
Abstract
Background Mesothelin is expressed at very low levels by normal mesothelial cells but is overexpressed in several human cancers. This makes mesothelin a promising target for immunotherapy. Limited data exist about mesothelin expression in esophageal carcinoma. In a current clinical trial, the highly potent anti-mesothelin antibody anetumab ravtansine is used in patients with mesothelin-positive tumors. Response rates are correlated with mesothelin expression (using the Ventana antibody) in tumor cells. No data are available on expression levels using the Ventana antibody. Most data have been generated using the Novocastra antibody. As patients are selected for clinical trials based on antibody staining of tumor samples, a comparison of these two available antibodies is crucial. Methods We analyzed 481 esophageal carcinomas [373 esophageal adenocarcinomas (EACs), 108 esophageal squamous cell carcinomas (ESCCs)] using two different monoclonal antibodies (Novocastra and Ventana) for mesothelin expression (low-mid and high-level expression, as used in one clinical trial). We also checked for the correlation of these results with clinical and molecular data. Results We revealed different staining results for both antibodies in EACs: Ventana: 53.6% (low expression: 25.3%; high expression: 28.3%) and Novocastra: 35.7% (low expression: 21.2%; high expression 14.5%). In ESCC we found comparable staining results: Ventana: 13.3% (low expression: 9.5%; high expression: 3.8%) and Novocastra: 13% (low expression: 11.1%; high expression: 1.9%). ARID1a-deficient EAC patients demonstrated significantly higher rates of mesothelin-positive tumors than ARID1a intact EAC patients. No correlations were found with other molecular alterations (TP53 mutation, ERBB2 amplification) or survival rates. Conclusion To the best of our knowledge, this is the largest study analyzing the importance of mesothelin expression in esophageal carcinoma. This study revealed a significant number of mesothelin-positive esophageal carcinomas, especially adenocarcinomas. New therapeutic targets are urgently required to improve the outcome of patients with locally advanced or metastasized esophageal carcinoma. The inhibition of mesothelin can be a new attractive target.
Collapse
Affiliation(s)
- Valeska Moentenich
- Department of Oncology and Hematology, University of Cologne, Kerpener Strasse 62, Cologne 50937, Germany
| | - Erdem Comut
- Institute of Pathology, Pammukale University, Turkey
| | - Florian Gebauer
- Department of Visceral Surgery, University of Cologne, Germany
| | - Armin Tuchscherer
- Department of Oncology and Hematology, University of Cologne, Germany
| | | | | | | | - Hakan Alakus
- Department of Visceral Surgery, University of Cologne, Germany
| | - Heike Loeser
- Institute of Pathology, University of Cologne, Germany
| | - Thomas Zander
- Department of Oncology and Hematology, University of Cologne, Germany
| | | |
Collapse
|
25
|
Hamai Y, Emi M, Ibuki Y, Murakami Y, Nishibuchi I, Nagata Y, Furukawa T, Kurokawa T, Ohsawa M, Yoshikawa T, Okada M. Predictions of Pathological Features and Recurrence Based on FDG-PET Findings of Esophageal Squamous Cell Carcinoma after Trimodal Therapy. Ann Surg Oncol 2020; 27:4422-4430. [PMID: 32405978 DOI: 10.1245/s10434-020-08609-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND The degree of metabolic activity in tumor cells can be determined by 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET). Associations between FDG uptake by primary tumors of locally advanced esophageal squamous cell carcinoma (ESCC) under trimodal therapy and the pathological features of such tumors have not been fully investigated. PATIENTS AND METHODS We evaluated relationships between the maximal standardized uptake (SUVmax) in primary tumors on preoperative PET images and pathological features as well as cancer recurrence in 143 patients with ESCC who underwent neoadjuvant chemoradiotherapy (NCRT) followed by surgery. RESULTS The post-SUVmax significantly differed after NCRT for ypT and ypN status, lymphatic invasion (LI), venous invasion (VI), and recurrence. Furthermore, the %ΔSUVmax (rate of decrease between before and after NCRT) for LI, VI, and recurrence significantly differed. Univariate and multivariate analyses selected post-SUVmax and %ΔSUVmax as independent preoperative predictors of recurrence-free survival [hazard ratio (HR) 1.46; 95% confidence interval (CI) 1.24-1.72 and HR 0.97; 95% CI 0.96-0.99, respectively; p < 0.001 for both]. Recurrence-free and overall survival were significantly stratified according to optimal SUVmax cutoffs for predicting recurrence (post- and %ΔSUVmax: 2.8 and 70, respectively). CONCLUSIONS The post- and %ΔSUVmax of primary tumors were significantly associated with the pathological features and recurrence of ESCC under trimodal therapy. Therefore, FDG-PET can preoperatively predict the degree of aggressive tumor behavior in ESCC under trimodal therapy.
Collapse
Affiliation(s)
- Yoichi Hamai
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan.
| | - Manabu Emi
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Yuta Ibuki
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Yuji Murakami
- Department of Radiation Oncology, Hiroshima University, Hiroshima, Japan
| | - Ikuno Nishibuchi
- Department of Radiation Oncology, Hiroshima University, Hiroshima, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Hiroshima University, Hiroshima, Japan
| | - Takaoki Furukawa
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Tomoaki Kurokawa
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Manato Ohsawa
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Toru Yoshikawa
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
26
|
Accuracy of Detecting Residual Disease After Neoadjuvant Chemoradiotherapy for Esophageal Cancer: A Systematic Review and Meta-analysis. Ann Surg 2020; 271:245-256. [PMID: 31188203 DOI: 10.1097/sla.0000000000003397] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The aim of this study was to perform a meta-analysis on the accuracy of endoscopic biopsies, EUS, and 18F-FDG PET(-CT) for detecting residual disease after neoadjuvant chemoradiotherapy (nCRT) for esophageal cancer. SUMMARY OF BACKGROUND DATA After nCRT, one-third of patients have a pathologically complete response in the resection specimen. Before an active surveillance strategy could be offered to these patients, clinically complete responders should be accurately identified. METHODS Embase, Medline, Cochrane, and Web-of-Science were searched until February 2018 for studies on accuracy of endoscopic biopsies, EUS, or PET(-CT) for detecting locoregional residual disease after nCRT for squamous cell- or adenocarcinoma. Pooled sensitivities and specificities were calculated using random-effect meta-analyses. RESULTS Forty-four studies were included for meta-analyses. For detecting residual disease at the primary tumor site, 12 studies evaluated endoscopic biopsies, 11 qualitative EUS, 14 qualitative PET, 8 quantitative PET using maximum standardized uptake value (SUVmax), and 7 quantitative PET using percentage reduction of SUVmax (%ΔSUVmax). Pooled sensitivities and specificities were 33% and 95% for endoscopic biopsies, 96% and 8% for qualitative EUS, 74% and 52% for qualitative PET, 69% and 72% for PET-SUVmax, and 73% and 63% for PET-%ΔSUVmax. For detecting residual nodal disease, 11 studies evaluated qualitative EUS with a pooled sensitivity and specificity of 68% and 57%, respectively. In subgroup analyses, sensitivity of PET-%ΔSUVmax and EUS for nodal disease was higher in squamous cell carcinoma than adenocarcinoma. CONCLUSIONS Current literature suggests insufficient accuracy of endoscopic biopsies, EUS, and 18F-FDG PET(-CT) as single modalities for detecting residual disease after nCRT for esophageal cancer.
Collapse
|
27
|
Moentenich V, Gebauer F, Comut E, Tuchscherer A, Bruns C, Schroeder W, Buettner R, Alakus H, Loeser H, Zander T, Quaas A. Claudin 18.2 expression in esophageal adenocarcinoma and its potential impact on future treatment strategies. Oncol Lett 2020; 19:3665-3670. [PMID: 32391091 PMCID: PMC7204493 DOI: 10.3892/ol.2020.11520] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 11/12/2019] [Indexed: 01/04/2023] Open
Abstract
The incidence of esophageal adenocarcinoma (EAC) has rapidly increased, particularly in the Western world. Despite improvements in perioperative treatments, the overall survival of patients remains low. Claudin 18.2 is a tight junction protein that is exclusively expressed in the gastric epithelia. However, following malignant transformation, gastric cancer metastases maintain this expression. Therefore, claudin 18.2 is a promising target for immunotherapy. Previous clinical trials have revealed improved anti-tumor activity in patients treated with an anti-claudin antibody by investigating the expression of claudin 18.2 in tumor cells. However, there is currently very limited data on the importance of claudin 18.2 expression in EAC. The present study analyzed the distribution of claudin 18.2 using immunohistochemistry in 485 patients with EAC, including their lymph node metastases. Additionally, these results were associated with clinical and molecular data. Claudin 18.2 was detected in 89/485 patients (18.4%). No correlations between expression and clinicopathological data (sex, age, pT stage, lymph node metastasis and grading) were observed. However, significantly decreased claudin 18.2 expression was observed in tumor types with upregulated human epidermal growth factor receptor 2 expression (P=0.036). Additionally, neoadjuvant treatment did not have any significant impact on claudin 18.2 expression (P=0.331). To the best of our knowledge, the present study is the largest systematic investigation of claudin 18.2 protein expression in EAC. The results obtained suggested that claudin 18.2 may serve as a promising therapeutic target in a substantial number of patients with EAC.
Collapse
Affiliation(s)
- Valeska Moentenich
- Department of Oncology and Hematology, University of Cologne, D-50937 Cologne, Germany
| | - Florian Gebauer
- Department of Visceral Surgery, University of Cologne, D-50937 Cologne, Germany
| | - Erdem Comut
- Institute of Pathology, Pammukale University, Denizli 20160, Turkey
| | - Armin Tuchscherer
- Department of Oncology and Hematology, University of Cologne, D-50937 Cologne, Germany
| | - Christiane Bruns
- Department of Visceral Surgery, University of Cologne, D-50937 Cologne, Germany
| | - Wolfgang Schroeder
- Department of Visceral Surgery, University of Cologne, D-50937 Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University of Cologne, D-50937 Cologne, Germany
| | - Hakan Alakus
- Department of Visceral Surgery, University of Cologne, D-50937 Cologne, Germany
| | - Heike Loeser
- Institute of Pathology, University of Cologne, D-50937 Cologne, Germany
| | - Thomas Zander
- Department of Oncology and Hematology, University of Cologne, D-50937 Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, D-50937 Cologne, Germany
| |
Collapse
|
28
|
Azad TD, Chaudhuri AA, Fang P, Qiao Y, Esfahani MS, Chabon JJ, Hamilton EG, Yang YD, Lovejoy A, Newman AM, Kurtz DM, Jin M, Schroers-Martin J, Stehr H, Liu CL, Hui ABY, Patel V, Maru D, Lin SH, Alizadeh AA, Diehn M. Circulating Tumor DNA Analysis for Detection of Minimal Residual Disease After Chemoradiotherapy for Localized Esophageal Cancer. Gastroenterology 2020; 158:494-505.e6. [PMID: 31711920 PMCID: PMC7010551 DOI: 10.1053/j.gastro.2019.10.039] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Biomarkers are needed to risk stratify after chemoradiotherapy for localized esophageal cancer. These could improve identification of patients at risk for cancer progression and selection of additional therapy. METHODS We performed deep sequencing (CAncer Personalized Profiling by deep Sequencing, [CAPP-Seq]) analyses of plasma cell-free DNA collected from 45 patients before and after chemoradiotherapy for esophageal cancer, as well as DNA from leukocytes and fixed esophageal tumor biopsy samples collected during esophagogastroduodenoscopy. Patients were treated from May 2010 through October 2015; 23 patients subsequently underwent esophagectomy, and 22 did not undergo surgery. We also sequenced DNA from blood samples from 40 healthy control individuals. We analyzed 802 regions of 607 genes for single-nucleotide variants previously associated with esophageal adenocarcinoma or squamous cell carcinoma. Patients underwent imaging analyses 6-8 weeks after chemoradiotherapy and were followed for 5 years. Our primary aim was to determine whether detection of circulating tumor DNA (ctDNA) after chemoradiotherapy is associated with risk of tumor progression (growth of local, regional, or distant tumors, detected by imaging or biopsy). RESULTS The median proportion of tumor-derived DNA in total cell-free DNA before treatment was 0.07%, indicating that ultrasensitive assays are needed for quantification and analysis of ctDNA from localized esophageal tumors. Detection of ctDNA after chemoradiotherapy was associated with tumor progression (hazard ratio, 18.7; P < .0001), formation of distant metastases (hazard ratio, 32.1; P < .0001), and shorter disease-specific survival times (hazard ratio, 23.1; P < .0001). A higher proportion of patients with tumor progression had new mutations detected in plasma samples collected after chemoradiotherapy than patients without progression (P = .03). Detection of ctDNA after chemoradiotherapy preceded radiographic evidence of tumor progression by an average of 2.8 months. Among patients who received chemoradiotherapy without surgery, combined ctDNA and metabolic imaging analysis predicted progression in 100% of patients with tumor progression, compared with 71% for only ctDNA detection and 57% for only metabolic imaging analysis (P < .001 for comparison of either technique to combined analysis). CONCLUSIONS In an analysis of cell-free DNA in blood samples from patients who underwent chemoradiotherapy for esophageal cancer, detection of ctDNA was associated with tumor progression, metastasis, and disease-specific survival. Analysis of ctDNA might be used to identify patients at highest risk for tumor progression.
Collapse
Affiliation(s)
- Tej D. Azad
- Department of Radiation Oncology, Stanford University, Stanford, California, USA,Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Aadel A. Chaudhuri
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | - Penny Fang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yawei Qiao
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mohammad S. Esfahani
- Department of Radiation Oncology, Stanford University, Stanford, California, USA,Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Jacob J. Chabon
- Department of Radiation Oncology, Stanford University, Stanford, California, USA,Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Emily G. Hamilton
- Department of Radiation Oncology, Stanford University, Stanford, California, USA,Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Yi D. Yang
- Department of Radiation Oncology, Stanford University, Stanford, California, USA,Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Alex Lovejoy
- Department of Radiation Oncology, Stanford University, Stanford, California, USA,Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Aaron M. Newman
- Stanford Cancer Institute, Stanford University, Stanford, California, USA,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - David M. Kurtz
- Stanford Cancer Institute, Stanford University, Stanford, California, USA,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Michael Jin
- Stanford Cancer Institute, Stanford University, Stanford, California, USA,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Joseph Schroers-Martin
- Stanford Cancer Institute, Stanford University, Stanford, California, USA,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Henning Stehr
- Department of Radiation Oncology, Stanford University, Stanford, California, USA,Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Chih Long Liu
- Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Angela Bik-Yu Hui
- Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Viren Patel
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dipen Maru
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ash A. Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, California, USA,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California, USA
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University, Stanford, California; Stanford Cancer Institute, Stanford University, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California.
| |
Collapse
|
29
|
Schallenberg S, Bork J, Essakly A, Alakus H, Buettner R, Hillmer AM, Bruns C, Schroeder W, Zander T, Loeser H, Gebauer F, Quaas A. Loss of the SWI/SNF-ATPase subunit members SMARCF1 (ARID1A), SMARCA2 (BRM), SMARCA4 (BRG1) and SMARCB1 (INI1) in oesophageal adenocarcinoma. BMC Cancer 2020; 20:12. [PMID: 31906887 PMCID: PMC6945480 DOI: 10.1186/s12885-019-6425-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022] Open
Abstract
Background The SWI/SNF complex is an important chromatin remodeler, commonly dysregulated in cancer, with an estimated mutation frequency of 20%. ARID1A is the most frequently mutated subunit gene. Almost nothing is known about the other familiar members of the SWI/SNF complexes, SMARCA2 (BRM), SMARCA4 (BRG1) and SMARCB1 (INI1), in oesophageal adenocarcinoma (EAC). Methods We analysed a large cohort of 685 patients with EAC. We used four different antibodies to detect a loss-of-protein of ARID1A BRM, BRG1 and INI1 by immunohistochemistry and correlated these findings with molecular and clinical data. Results Loss of ARID1A, BRG1, BRM and INI1 was observed in 10.4, 3.4, 9.9 and 2% of EAC. We found a co-existing protein loss of ARID1A and BRM in 9.9% and of ARID1A and BRG1 in 2.2%. Patients with loss of ARID1A and TP53 wildtype EACs showed a shortened overall survival compared with AIRDA1A-positive tumours [median overall survival was 60.1 months (95%CI 1.2–139.9 months)] in patients with ARIDA-1A expression and 26.2 months (95%CI 3.7–19.1 months) in cases of ARIDA-1A loss (p = 0.044). Tumours with loss or expression of ARID1A and TP53 loss were not associated with a difference in survival. Only one tumour revealed high microsatellite instability (MSI-H) with concomitant ARID1A loss. All other ARID1A loss-EACs were microsatellite-stable (MSS). No predictive relevance was seen for SWI/SNF-complex alterations and simultaneous amplification of different genes (PIK3CA, KRAS, c-MYC, MET, GATA6, ERBB2). Conclusion Our work describes, for the first time, loss of one of the SWI/SNF ATPase subunit proteins in a large number of adenocarcinomas of the oesophagus. Several papers discuss possible therapeutic interventions for tumours showing a loss of function of the SWI/SNF complex, such as PARP inhibitors or PI3K and AKT inhibitors. Future studies will be needed to show whether SWI/SNF complex-deficient EACs may benefit from personalized therapy.
Collapse
Affiliation(s)
- Simon Schallenberg
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Julian Bork
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Ahlem Essakly
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Axel M Hillmer
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Wolfgang Schroeder
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Department I of Internal Medicine, Center for Integrated Oncology (CIO)University of Cologne, Cologne, Germany
| | - Heike Loeser
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
30
|
Nagai Y, Yoshida N, Baba Y, Harada K, Imai K, Iwatsuki M, Karashima R, Koga Y, Nomoto D, Okadome K, Ishimoto T, Imamura Y, Watanabe M, Baba H. Clinical significance of evaluating endoscopic response to neoadjuvant chemotherapy in esophageal squamous cell carcinoma. Dig Endosc 2020; 32:39-48. [PMID: 31120560 DOI: 10.1111/den.13449] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/20/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIM Clinical significance of endoscopic response (ER) after neoadjuvant chemotherapy (NAC) for esophageal cancer has not been fully understood. Thus, the present study aimed to investigate the association between ER to NAC and its clinicopathological outcomes in patients with esophageal squamous cell carcinoma (ESCC). METHODS In total, 141 patients who underwent NAC and subsequent esophagectomy for ESCC were included. ER to NAC was retrospectively evaluated based on macroscopic findings of the primary tumor, which was classified into three categories: endoscopic no response (eNR), endoscopic partial response (ePR), and endoscopic good response (eGR). An endoscopic responder was defined as patients with eGR/ePR. RESULTS Approximately 89.4% of patients had cStage II-III disease, and 7.1% had pathological complete response. Upon ER evaluation, eNR, ePR, and eGR were observed in 46 (32.6%), 54 (38.3%), and 41 (29.1%) patients, respectively. Pathological responders significantly increased as the ER grade became better. Among preoperative clinical factors, only ER significantly correlated with pathological response in univariate and multivariate analysis. Endoscopic responders showed a significantly better prognosis than did eNR patients (P < 0.001), although the overall survival (OS) of the patients with eGR and ePR was equivalent. Endoscopic responder, ypT, ypN, and pathological responder were significant predictors of OS in the univariate analysis, and endoscopic responder, ypN, and pathological responder were independent predictors in the multivariate analysis. CONCLUSION This study suggests that ER can be a simple and important tool to predict the pathological response and survival of patients who undergo NAC for ESCC.
Collapse
Affiliation(s)
- Yohei Nagai
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuto Harada
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ryuichi Karashima
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Koga
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daichi Nomoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuo Okadome
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yu Imamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
31
|
PIK3CA and KRAS Amplification in Esophageal Adenocarcinoma and their Impact on the Inflammatory Tumor Microenvironment and Prognosis. Transl Oncol 2019; 13:157-164. [PMID: 31865178 PMCID: PMC6931191 DOI: 10.1016/j.tranon.2019.10.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/25/2022] Open
Abstract
Gene amplifications of PIK3CA or KRAS induce a downstream activation of the AKT-mTOR or RAF-ERK-pathways. Interactions of the active AKT pathway have been implicated in the inflammatory tumor microenvironment. Nothing is known about these interactions or prognostic power in esophageal adenocarcinoma (EAC). We retrospectively analyzed a large cohort of 685 EAC considering KRAS and PIK3CA gene amplification using fluorescence in situ hybridization (FISH) and immunohistochemistry. These results were correlated with clinical and molecular data as well as the inflammatory tumor microenvironment. Amplifications of KRAS were seen in 94 patients (17.1%), PIK3CA amplifications in 23 patients (5.0%). KRAS amplifications significantly correlated with nodal positive patients and poorer overall survival (OS) in the subgroup without neoadjuvant treatment (p = 0.004), coamplifications of Her2 (p = 0.027), and TP53 mutations (p = 0.016). PIK3CA amplifications significantly correlated with a high amount of tumor infiltrating T cells (p = 0.003) and showed a tendency to better OS (p = 0.068). A correlation with checkpoint makers (PD-L1, LAG3, VISTA, TIM3, IDO) could not be revealed. Our findings are the first to link the KRAS amplified genotype with lymphonodal positivity and poor prognosis and the PIK3CA-amplified genotype with a T cell–rich microenvironment in EAC. Future studies must show whether these two genotype subgroups can be therapeutically influenced. A dual inhibition of MEK and SHP2T could be effective in the subgroup of KRAS amplified EACs and an immune checkpoint blockade may prove to be particularly promising in the subgroup of PIK3CA-amplified EACs.
Collapse
|
32
|
Brinkmann S, Noordman BJ, Hölscher AH, Biermann K, van Klaveren D, Bollschweiler E, Pütz K, van Lanschot JJB, Drebber U. External Validation of Pretreatment Pathological Tumor Extent in Patients with Neoadjuvant Chemoradiotherapy Plus Surgery for Esophageal Cancer. Ann Surg Oncol 2019; 27:1250-1258. [PMID: 31691114 PMCID: PMC7060166 DOI: 10.1245/s10434-019-08024-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Indexed: 11/26/2022]
Abstract
Background This study was conducted to validate a pretreatment (i.e. prior to neoadjuvant chemoradiotherapy) pathological staging system in the resection specimen after neoadjuvant chemoradiotherapy for esophageal cancer. The study investigated the prognostic value of pretreatment pathological T and N categories (prepT and prepN categories) in both an independent and a combined patient cohort. Methods Patients with esophageal cancer treated with neoadjuvant chemotherapy and esophagectomy between 2012 and 2015 were included. PrepT and prepN categories were estimated based on the extent of tumor regression and regressional changes of lymph nodes in the resection specimen. The difference in Akaike’s information criterion (ΔAIC) was used to assess prognostic performance. PrepN and ypN categories were combined to determine the effect of nodal sterilization on prognosis. A multivariable Cox regression model was used to identify combined prepN and ypN categories as independent prognostic factors. Results The prognostic strength of the prepT category was better than the cT and ypT categories (ΔAIC 7.7 vs. 3.0 and 2.9, respectively), and the prognostic strength of the prepN category was better than the cN category and similar to the ypN category (ΔAIC 29.2 vs. − 1.0 and 27.9, respectively). PrepN + patients who became ypN0 had significantly worse survival than prepN0 patients (2-year overall survival 69% vs. 86% in 137 patients; p = 0.044). Similar results were found in a combined cohort of 317 patients (2-year overall survival 62% vs. 85%; p = 0.002). Combined prepN/ypN stage was independently associated with overall survival. Conclusions These results independently confirm the prognostic value of prepTNM staging. PrepTNM staging is of additional prognostic value to cTNM and ypTNM. PrepN0/ypN0 patients have a better survival than prepN +/ypN0 patients. Electronic supplementary material The online version of this article (10.1245/s10434-019-08024-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sebastian Brinkmann
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Bo J Noordman
- Department of Surgery, Erasmus MC - University Medical Centre, Rotterdam, The Netherlands.
| | - Arnulf H Hölscher
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany.,Centre for Esophageal and Gastric Surgery, AGAPLESION Markus Krankenhaus, Frankfurt, Germany
| | - Katharina Biermann
- Department of Pathology, Erasmus MC - University Medical Centre, Rotterdam, The Netherlands
| | - David van Klaveren
- Department of Public Health, Erasmus MC - University Medical Centre, Rotterdam, The Netherlands
| | - Elfriede Bollschweiler
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Katharina Pütz
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - J Jan B van Lanschot
- Department of Surgery, Erasmus MC - University Medical Centre, Rotterdam, The Netherlands
| | - Uta Drebber
- Institute of Pathology, University of Cologne, Cologne, Germany
| |
Collapse
|
33
|
Shah T, Kushnir V, Mutha P, Majhail M, Patel B, Schutzer M, Mogahanaki D, Smallfield G, Patel M, Zfass A. Neoadjuvant cryotherapy improves dysphagia and may impact remission rates in advanced esophageal cancer. Endosc Int Open 2019; 7:E1522-E1527. [PMID: 31681831 PMCID: PMC6823095 DOI: 10.1055/a-0957-2798] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/25/2019] [Indexed: 12/21/2022] Open
Abstract
Background and study aims Liquid nitrogen spray cryotherapy (LNSC) can provide rapid dysphagia relief, and is postulated to stimulate a local antitumor immune response. The aim of this prospective pilot clinical trial was to evaluate the safety and efficacy of LNSC when administered prior to chemoradiotherapy. Patients and methods Treatment-naïve adult patients with dysphagia at the time of biopsy-proven squamous carcinoma or adenocarcinoma of the esophagus were prospectively enrolled at two tertiary medical centers. Patients underwent a single session of LNSC. The primary outcome measure was change in dysphagia at 1 and 2 weeks post-cryotherapy. A secondary outcome measure was clinical complete response rate (CR) following chemoradiotherapy. Results Twenty-five patients were screened, of whom 21 patients were eligible and enrolled. There were seven with metastatic and 14 with locally advanced cancer. The primary outcome of dysphagia improvement of ≥ 1 point occurred in 15/21 patients (71 %) at 1 week, and 10/20 patients (50 %) at 2 weeks. The median dysphagia score improved by 1 at 1 week ( P = 0.0003), and 0.5 at 2 weeks ( P = 0.02). Six of nine patients (67 %) with locally advanced cancer who completed chemoradiation did not have residual tumor cells on mucosal biopsy, and five of nine patients (56 %) had a clinical CR. There were no serious cryotherapy-related complications. Conclusions LNSC provided safe and effective palliation for esophageal cancer patients who presented with dysphagia at index diagnosis. Its combination with chemoradiotherapy did not lead to any serious toxicity. Our study provides a scientific rationale for pursuing larger clinical trials addressing synergistic effects of combining LNSC with chemoradiation.
Collapse
Affiliation(s)
- Tilak Shah
- Division of Gastroenterology, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia, United States,Division of Gastroenterology, Virginia Commonwealth University Health System,Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States,Corresponding author Tilak Shah, MD, MHS McGuire VAMCGastroenterology; 111N1201 Broad Rock BlvdRichmond, VA 23224+1-804-675-5816
| | - Vladimir Kushnir
- Section of Gastroenterology, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Pritesh Mutha
- Division of Gastroenterology, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia, United States,Division of Gastroenterology, Virginia Commonwealth University Health System
| | - Mankanchan Majhail
- Division of Gastroenterology, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia, United States
| | - Bhaumik Patel
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States,Division of Hematology-Oncology, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia, United States
| | - Matthew Schutzer
- Radiation oncology Service, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia, United States
| | - Drew Mogahanaki
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States,Radiation oncology Service, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia, United States
| | - George Smallfield
- Division of Gastroenterology, Virginia Commonwealth University Health System
| | - Milan Patel
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Alvin Zfass
- Division of Gastroenterology, Hunter Holmes McGuire VA Medical Center, Richmond, Virginia, United States,Division of Gastroenterology, Virginia Commonwealth University Health System
| |
Collapse
|
34
|
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
|
35
|
Neoadjuvant therapy for locally advanced gastric cancer patients. A population pharmacodynamic modeling. PLoS One 2019; 14:e0215970. [PMID: 31071108 PMCID: PMC6508715 DOI: 10.1371/journal.pone.0215970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/05/2019] [Indexed: 01/27/2023] Open
Abstract
Background Perioperative chemotherapy (CT) or neoadjuvant chemoradiotherapy (CRT) in patients with locally advanced gastric (GC) or gastroesophageal junction cancer (GEJC) has been shown to improve survival compared to an exclusive surgical approach. However, most patients retain a poor prognosis due to important relapse rates. Population pharmacokinetic-pharmacodynamic (PK/PD) modeling may allow identifying at risk-patients. We aimed to develop a mechanistic PK/PD model to characterize the relationship between the type of neoadjuvant therapy, histopathologic response and survival times in locally advanced GC and GEJC patients. Methods Patients with locally advanced GC and GEJC treated with neoadjuvant CT with or without preoperative CRT were analyzed. Clinical response was assessed by CT-scan and EUS. Pathologic response was defined as a reduction on pTNM stage compared to baseline cTNM. Metastasis development risk and overall survival (OS) were described using the population approach with NONMEM 7.3. Model evaluation was performed through predictive checks. Results A low correlation was observed between clinical and pathologic TNM stage for both T (R = 0.32) and N (R = 0.19) categories. A low correlation between clinical and pathologic response was noticed (R = -0.29). The OS model adequately described the observed survival rates. Disease recurrence, cTNM stage ≥3 and linitis plastica absence, were correlated to a higher risk of death. Conclusion Our model adequately described clinical response profiles, though pathologic response could not be predicted. Although the risk of disease recurrence and survival were linked, the identification of alternative approaches aimed to tailor therapeutic strategies to the individual patient risk warrants further research.
Collapse
|
36
|
Loeser H, Kraemer M, Gebauer F, Bruns C, Schröder W, Zander T, Persa OD, Alakus H, Hoelscher A, Buettner R, Lohneis P, Quaas A. The expression of the immune checkpoint regulator VISTA correlates with improved overall survival in pT1/2 tumor stages in esophageal adenocarcinoma. Oncoimmunology 2019; 8:e1581546. [PMID: 31069143 PMCID: PMC6492979 DOI: 10.1080/2162402x.2019.1581546] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/25/2019] [Accepted: 02/06/2019] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint modulation in cancer has been demonstrated as a high-value therapeutic strategy in many tumor entities. VISTA is an immune checkpoint receptor regulating T-cell function. To the best of our knowledge, nothing is known about the expression and prognostic impact of VISTA on tumor infiltrating lymphocytes (TILs) in the tumor microenvironment of esophageal adenocarcinoma (EAC). We analyzed in total 393 EACs within a test-cohort (n = 165) and a validation-cohort (n = 228) using a monoclonal antibody (clone D1L2G). These data were statistically correlated with clinical as well as molecular data. 22.2% of the tumor cohort presented with a VISTA expression on TILs. These patients demonstrated an improved median overall survival compared to patients without VISTA expression (202.2 months vs. 21.6 months; p < 0.0001). The favorable outcome of VISTA positive tumors is significant in the entire cohort but mainly driven by the general better prognosis of T1/T2 tumors. However, in the pT1/2 group, VISTA positive tumors show a tremendous survival benefit compared to VISTA negative tumors revealing real long-term survivors in this particular subgroup. The survival difference is independent of the T-stage. This unique characteristic could influence neoadjuvant therapy concepts for EAC, since a profit of therapy could be reduced in the already favorable subgroup of VISTA positive tumors. VISTA emerges as a prognostic biomarker for long-term survival especially in the group of early TNM-stages. Future studies have to show the relevance of VISTA positive TILs within a tumor concerning response to specific immune checkpoint inhibition.
Collapse
Affiliation(s)
- Heike Loeser
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Max Kraemer
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Wolfgang Schröder
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Department I of Internal Medicine, Centre for Integrated Oncology (CIO), University of Cologne, Cologne, Germany
| | - Oana-Diana Persa
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Arnulf Hoelscher
- Centre for Esophageal and Gastric Surgery, AGAPLESION Markus Krankenhaus, Frankfurt, Germany
| | | | - Philipp Lohneis
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, Cologne, Germany
| |
Collapse
|
37
|
Eyck BM, van der Wilk BJ, Lagarde SM, Wijnhoven BPL, Valkema R, Spaander MCW, Nuyttens JJME, van der Gaast A, van Lanschot JJB. Neoadjuvant chemoradiotherapy for resectable oesophageal cancer. Best Pract Res Clin Gastroenterol 2018; 36-37:37-44. [PMID: 30551855 DOI: 10.1016/j.bpg.2018.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/19/2018] [Indexed: 01/31/2023]
Abstract
At present, treatment of potentially curable oesophageal cancer includes neoadjuvant chemoradiotherapy followed by oesophagectomy. Alternatively, neoadjuvant chemotherapy is used. To date, strong evidence on the superiority of one modality over the other has not been provided. Currently, up to one-third of patients show a pathologically complete response after neoadjuvant chemoradiotherapy. To optimise the efficacy of neoadjuvant treatment for individual patients, prediction of response to neoadjuvant treatment is highly desired. Therefore, several clinical diagnostic modalities have been investigated for early response evaluation, of which positron emission tomography (PET) has been studied most extensively. To identify patients who might benefit from postponing or even omitting surgery, recent advances have been made in evaluating response after completion of neoadjuvant chemoradiotherapy. This review provides an overview of current evidence and recent advances in neoadjuvant chemoradiotherapy for oesophageal cancer and discusses the use of neoadjuvant chemotherapy compared to chemoradiotherapy. Moreover, clinical response evaluation to neoadjuvant chemoradiotherapy is reviewed.
Collapse
Affiliation(s)
- B M Eyck
- Department of Surgery, Erasmus MC - University Medical Centre Rotterdam, Dr. Molenwaterplein 40, 3000 CA, Rotterdam, the Netherlands.
| | - B J van der Wilk
- Department of Surgery, Erasmus MC - University Medical Centre Rotterdam, Dr. Molenwaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - S M Lagarde
- Department of Surgery, Erasmus MC - University Medical Centre Rotterdam, Dr. Molenwaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - B P L Wijnhoven
- Department of Surgery, Erasmus MC - University Medical Centre Rotterdam, Dr. Molenwaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - R Valkema
- Department of Radiology and Nuclear Medicine, Erasmus MC - University Medical Centre Rotterdam, Dr. Molenwaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - M C W Spaander
- Department of Gastroenterology & Hepatology, Erasmus MC - University Medical Centre Rotterdam, Dr. Molenwaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - J J M E Nuyttens
- Department of Radiotherapy, Erasmus MC - University Medical Centre Rotterdam, Dr. Molenwaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - A van der Gaast
- Department of Medical Oncology, Erasmus MC - University Medical Centre Rotterdam, Dr. Molenwaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - J J B van Lanschot
- Department of Surgery, Erasmus MC - University Medical Centre Rotterdam, Dr. Molenwaterplein 40, 3000 CA, Rotterdam, the Netherlands
| |
Collapse
|
38
|
Fang P, Musall BC, Son JB, Moreno AC, Hobbs BP, Carter BW, Fellman BM, Mawlawi O, Ma J, Lin SH. Multimodal Imaging of Pathologic Response to Chemoradiation in Esophageal Cancer. Int J Radiat Oncol Biol Phys 2018; 102:996-1001. [PMID: 29685377 PMCID: PMC6119639 DOI: 10.1016/j.ijrobp.2018.02.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 02/07/2018] [Accepted: 02/20/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE To examine the value of early changes in quantitative diffusion-weighted imaging and 18F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) for discriminating complete pathologic response (pCR) to chemoradiation in esophageal cancer. METHODS AND MATERIALS Twenty esophageal cancer patients treated with chemoradiation followed by surgery were prospectively enrolled. Patients underwent magnetic resonance imaging and FDG-PET/CT scans at baseline, interim (2 weeks after chemoradiation start), and first follow-up. On the basis of pathologic findings at surgery, patients were categorized into tumor regression groups (TRG1, TRG2, and TRG3+). Distributions of summary statistics in apparent diffusion coefficient (ADC) and FDG-PET at baseline and relative changes at interim and follow-up scans were compared between pCR/TRG1 and non-pCR/TRG2+ groups and across readers. Receiver operating characteristics were evaluated for summary measures to characterize discrimination of pCR from non-pCR. RESULTS Relative changes in tumor volume ADC (ΔADC) mean and 25th and 10th percentiles from baseline to interim were able to completely discriminate (area under the curve = 1, P < .0011) between pCR and non-pCR (thresholds = 27.7%, 29.2%, and 32.1%, respectively) and were found to have high interreader reliability (95% limits of agreement of 1.001, 0.944, and 0.940, respectively). Relative change in total lesion glycolysis (TLG) from baseline to interim was significantly different among pCR and non-pCR groups (P=.0117) and yielded an area under the curve of 0.947 (95% confidence interval 0.8505-1.043). An optimal threshold of 59% decrease in TLG provided optimal sensitivity (specificity) of 1.000 (0.867). Changes in ADC summary measures were negatively correlated with that of TLG (Spearman, -0.495, P=.027). CONCLUSIONS Quantitative volume ΔADC and TLG during treatment may serve as early imaging biomarkers for discriminating pathologic response to chemoradiation in esophageal cancer. Validation of these data in larger, prospective, multicenter studies is essential.
Collapse
Affiliation(s)
- Penny Fang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Benjamin C Musall
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jong Bum Son
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amy C Moreno
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Brian P Hobbs
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Brett W Carter
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bryan M Fellman
- Department of Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Osama Mawlawi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
39
|
Quaas A, Heydt C, Gebauer F, Alakus H, Loeser H, Buettner R, Hillmer A, Bruns C, Merkelbach-Bruse S, Zander T, Frommolt P. Genomic Characterization of TP53-Wild-Type Esophageal Carcinoma. Transl Oncol 2018; 12:154-161. [PMID: 30317074 PMCID: PMC6187090 DOI: 10.1016/j.tranon.2018.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 12/28/2022] Open
Affiliation(s)
| | - Carina Heydt
- Institute of Pathology, University of Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, University of Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University of Cologne, Germany
| | - Heike Loeser
- Institute of Pathology, University of Cologne, Germany
| | | | - Axel Hillmer
- Institute of Pathology, University of Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, University of Cologne, Germany
| | | | - Thomas Zander
- Department I of Internal Medicine, Center for Integrated Oncology (CIO), University of Cologne, Germany
| | | |
Collapse
|
40
|
Zhang W, Luo Y, Wang X, Han G, Wang P, Yuan W, Dai SB. Dose-escalated radiotherapy improved survival for esophageal cancer patients with a clinical complete response after standard-dose radiotherapy with concurrent chemotherapy. Cancer Manag Res 2018; 10:2675-2682. [PMID: 30147366 PMCID: PMC6097517 DOI: 10.2147/cmar.s160909] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose For esophageal cancer patients with a clinical complete response (cCR) after standard-dose radiotherapy with concurrent chemotherapy, data on the survival outcomes and recurrence patterns remain scarce. To evaluate the impact of dose escalation on overall survival for this subset of patients, we carried out the current investigation. Materials and methods Medical records of 80 esophageal cancer patients with a cCR after standard-dose radiotherapy with concurrent chemotherapy at our center from 2010 to 2014 were allocated into the standard-dose group (50.4 Gy, observation group) or the radiation dose-escalation group (59.4 Gy, control group). In this cohort study, we compared the outcomes between the 2 groups. Results There were no differences in patient characteristics between the 2 groups. The median recurrence-free survival and overall survival times for all patients were 38 and 54 months, respectively. Patients in the control group had significantly better 5-year recurrence-free survival rate (12% vs 0%, p=0.019) and 5-year overall survival rate (42.8% vs 21.0%, p=0.028) than the observation group. Additionally, local control rate was significantly higher in the control group (p=0.04), and ~60% of treatment failures were local failures even for patients achieving cCR after chemoradiotherapy. There were no significant differences in treatment-related toxicities between the groups. Conclusion The results of the current study suggest that for esophageal cancer patients with a cCR after standard-dose radiotherapy with concurrent chemotherapy, those with dose-escalated radiotherapy showed significantly better local control, recurrence-free survival, and overall survival than patients receiving 50.4 Gy radiotherapy.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China,
| | - Yijun Luo
- Department of Oncology, The People's Hospital of Jiangxi Province, Nanchang, Jiangxi, People's Republic of China
| | - Xiaoli Wang
- Department of Oncology, The People's Hospital of Jiangxi Province, Nanchang, Jiangxi, People's Republic of China
| | - Gaohua Han
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China,
| | - Peng Wang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China,
| | - Wei Yuan
- Department of Cardiology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China,
| | - Sheng-Bin Dai
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China,
| |
Collapse
|
41
|
Cosgrove ND, Mullady DK. Endoscopic evaluation of the esophageal cancer patient after chemoradiotherapy for persistent/recurrent cancer. Dis Esophagus 2018; 31:5040371. [PMID: 29931309 DOI: 10.1093/dote/doy023] [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] [Indexed: 12/11/2022]
Abstract
Endoscopy has an important role in the pre- and post-treatment staging of esophageal cancer. Complete pathologic response following neoadjuvant chemoradiation therapy occurs in approximately 25% of patients. However, the ability to accurately detect this preoperatively with currently available endoscopic modalities is limited such that the default pathway is for fit patients to proceed with surgical resection. This article discusses the available endoscopic modalities (primarily Esophagogastroduodenoscopy [EGD] with mucosal biopsies and endoscopic ultrasonography with or without fine needle aspiration) used for post-treatment staging of esophageal cancer. We present data regarding the benefits and limitations of endoscopic methods in assessing for residual disease. Unfortunately, endoscopic modalities are not accurate enough to identify complete pathological responsers who may avoid surgical resection.
Collapse
Affiliation(s)
- N D Cosgrove
- Division of Gastroenterology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - D K Mullady
- Division of Gastroenterology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
42
|
Noordman BJ, Spaander MCW, Valkema R, Wijnhoven BPL, van Berge Henegouwen MI, Shapiro J, Biermann K, van der Gaast A, van Hillegersberg R, Hulshof MCCM, Krishnadath KK, Lagarde SM, Nieuwenhuijzen GAP, Oostenbrug LE, Siersema PD, Schoon EJ, Sosef MN, Steyerberg EW, van Lanschot JJB. Detection of residual disease after neoadjuvant chemoradiotherapy for oesophageal cancer (preSANO): a prospective multicentre, diagnostic cohort study. Lancet Oncol 2018; 19:965-974. [PMID: 29861116 DOI: 10.1016/s1470-2045(18)30201-8] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/04/2018] [Accepted: 03/06/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND After neoadjuvant chemoradiotherapy for oesophageal cancer, roughly half of the patients with squamous cell carcinoma and a quarter of those with adenocarcinoma have a pathological complete response of the primary tumour before surgery. Thus, the necessity of standard oesophagectomy after neoadjuvant chemoradiotherapy should be reconsidered for patients who respond sufficiently to neoadjuvant treatment. In this study, we aimed to establish the accuracy of detection of residual disease after neoadjuvant chemoradiotherapy with different diagnostic approaches, and the optimal combination of diagnostic techniques for clinical response evaluations. METHODS The preSANO trial was a prospective, multicentre, diagnostic cohort study at six centres in the Netherlands. Eligible patients were aged 18 years or older, had histologically proven, resectable, squamous cell carcinoma or adenocarcinoma of the oesophagus or oesophagogastric junction, and were eligible for potential curative therapy with neoadjuvant chemoradiotherapy (five weekly cycles of carboplatin [area under the curve 2 mg/mL per min] plus paclitaxel [50 mg/m2 of body-surface area] combined with 41·4 Gy radiotherapy in 23 fractions) followed by oesophagectomy. 4-6 weeks after completion of neoadjuvant chemoradiotherapy, patients had oesophagogastroduodenoscopy with biopsies and endoscopic ultrasonography with measurement of maximum tumour thickness. Patients with histologically proven locoregional residual disease or no-pass during endoscopy and without distant metastases underwent immediate surgical resection. In the remaining patients a second clinical response evaluation was done (PET-CT, oesophagogastroduodenoscopy with biopsies, endoscopic ultrasonography with measurement of maximum tumour thickness, and fine-needle aspiration of suspicious lymph nodes), followed by surgery 12-14 weeks after completion of neoadjuvant chemoradiotherapy. The primary endpoint was the correlation between clinical response during clinical response evaluations and the final pathological response in resection specimens, as shown by the proportion of tumour regression grade (TRG) 3 or 4 (>10% residual carcinoma in the resection specimen) residual tumours that was missed during clinical response evaluations. This study was registered with the Netherlands Trial Register (NTR4834), and has been completed. FINDINGS Between July 22, 2013, and Dec 28, 2016, 219 patients were included, 207 of whom were included in the analyses. Eight of 26 TRG3 or TRG4 tumours (31% [95% CI 17-50]) were missed by endoscopy with regular biopsies and fine-needle aspiration. Four of 41 TRG3 or TRG4 tumours (10% [95% CI 4-23]) were missed with bite-on-bite biopsies and fine-needle aspiration. Endoscopic ultrasonography with maximum tumour thickness measurement missed TRG3 or TRG4 residual tumours in 11 of 39 patients (28% [95% CI 17-44]). PET-CT missed six of 41 TRG3 or TRG4 tumours (15% [95% CI 7-28]). PET-CT detected interval distant histologically proven metastases in 18 (9%) of 190 patients (one squamous cell carcinoma, 17 adenocarcinomas). INTERPRETATION After neoadjuvant chemoradiotherapy for oesophageal cancer, clinical response evaluation with endoscopic ultrasonography, bite-on-bite biopsies, and fine-needle aspiration of suspicious lymph nodes was adequate for detection of locoregional residual disease, with PET-CT for detection of interval metastases. Active surveillance with this combination of diagnostic modalities is now being assessed in a phase 3 randomised controlled trial (SANO trial; Netherlands Trial Register NTR6803). FUNDING Dutch Cancer Society.
Collapse
Affiliation(s)
- Bo Jan Noordman
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands.
| | - Manon C W Spaander
- Department of Gastroenterology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Roelf Valkema
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Bas P L Wijnhoven
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | | | - Joël Shapiro
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Katharina Biermann
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Ate van der Gaast
- Department of Medical Oncology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | | | | | | | - Sjoerd M Lagarde
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | | | | | - Peter D Siersema
- Department of Gastroenterology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands; Department of Gastroenterology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Erik J Schoon
- Department of Gastroenterology, Catharina Hospital, Eindhoven, Netherlands
| | - Meindert N Sosef
- Department of Surgery, Zuyderland Medical Centre, Heerlen, Netherlands
| | - Ewout W Steyerberg
- Department of Public Health, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands; Department of Medical Statistics and Bioinformatics, Leiden University Medical Centre, Leiden, Netherlands
| | - J Jan B van Lanschot
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
43
|
Cho CJ, Kang HJ, Park SJ, Kim GH, Park SH, Eo SH, Kim MJ, Kim YH, Park SR, Kim JH, Ahn JY, Kim DH, Choi KD, Song HJ, Jung HY, Park YS, Lee GH. Novel endoscopic categorization for prediction of chemoradiotherapy response in locally advanced esophageal cancer. J Gastroenterol Hepatol 2018; 33:1213-1219. [PMID: 29193280 DOI: 10.1111/jgh.14055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/26/2017] [Accepted: 11/21/2017] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Preoperative chemoradiotherapy (CRT) followed by esophagectomy is a well-known treatment modality for patients with locally advanced esophageal cancer (EC). This study developed an algorithm to predict pathological complete response (CR) in these patients using post-CRT endoscopic category with biopsy and validated the proposed algorithm. METHODS A retrospective review of 141 consecutive patients who completed preoperative CRT and underwent surgical resection for locally advanced EC was performed. The post-CRT endoscopic findings of each patient were stratified into five categories. RESULTS The distribution of post-CRT endoscopic categories was significantly different between the pathological CR and non-pathological CR groups (P < 0.001). About 76.8% (73/95) of patients in category 0, 1, or 2 achieved pathological CR. In contrast, 91.3% (42/46) of endoscopic categories 3 and 4 patients did not achieve pathological CR. Sensitivity of post-CRT biopsy was 11.1%. Therefore, an algorithm combining biopsy results and dichotomized post-CRT endoscopic category (category 0, 1, or 2 vs category 3 or 4) was developed. The sensitivity, specificity, and accuracy in predicting pathological CR by the proposed algorithm were 64.8%, 95.9%, and 82.8%, respectively. In the multivariate analysis, the proposed algorithm remained a significant negative factor of survival (P < 0.001). CONCLUSIONS Algorithm using post-CRT endoscopic category with biopsy may help identify locally advanced EC patients who achieved pathological CR after preoperative CRT. Modalities to accurately detect subepithelial remnant EC may further aid in predicting pathological CR.
Collapse
Affiliation(s)
- Charles J Cho
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyo Jeong Kang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Se Jeong Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ga Hee Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seong Hwan Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo-Heang Eo
- Department of Statistics, Korea University, Seoul, Korea
| | - Min-Ju Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yong-Hee Kim
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sook Ryun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Hoon Kim
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji Yong Ahn
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Do Hoon Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kee Don Choi
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho June Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hwoon-Yong Jung
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gin Hyug Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
44
|
Hamai Y, Hihara J, Emi M, Furukawa T, Murakami Y, Nishibuchi I, Nagata Y, Ibuki Y, Yamakita I, Kurokawa T, Okada M. Preoperative prediction of a pathologic complete response of esophageal squamous cell carcinoma to neoadjuvant chemoradiotherapy. Surgery 2018; 164:S0039-6060(18)30016-3. [PMID: 29519558 DOI: 10.1016/j.surg.2018.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/05/2018] [Accepted: 01/10/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND The accurate prediction of a pathologic complete response (ypT0N0M [LYM] 0 ypStage 0) before operation is essential for selecting appropriate strategies for treating esophageal cancer after neoadjuvant chemoradiotherapy. METHODS We reviewed 130 consecutive patients with esophageal squamous cell carcinoma who were evaluated preoperatively using upper gastrointestinal endoscopy, computed tomography, and 18F-fluorodeoxyglucose-positron emission tomography after neoadjuvant chemoradiotherapy and subsequently underwent esophagectomy. Our aim was to determine the diagnostic abilities of computed tomography, 18F-fluorodeoxyglucose-positron emission tomography, and endoscopy to predict preoperatively a pathologic complete response of the primary site of the locally advanced esophageal squamous cell carcinoma and associated lymph nodes to trimodal neoadjuvant chemoradiotherapy. Associations between clinical complete response (ycT0N0M [LYM] 0 ycStage 0) and pathologic complete response were investigated preoperatively. RESULTS Twenty-nine (22.3%) and 43 (33.1%) patients, respectively, achieved clinical complete response and pathologic complete response, which were associated (P=.001). The sensitivity and specificity, as well as the positive and negative predictive values of clinical complete response to define pathologic complete response were 39.5%, 86.2%, 58.6%, and 74.3%, respectively. Univariate and multivariate analyses selected clinical complete response as the sole independent preoperative predictor of pathologic complete response (clinical complete responses versus non-clinical complete responses: odds ratio: 0.26, 95% confidence interval, 0.10-0.65, P=.004). Recurrence-free and overall survival (OS) rates were better in patients with than in those without clinical complete response (5-year recurrence-free and overall survival: 69.0% vs 41.4% and 75.9% vs 45.0%, respectively, both P=.02). Furthermore, clinical complete response was an independent preoperative predictor of recurrence-free survival (clinical complete response versus nonclinical complete response: hazard ratio: 2.20, 95% confidence interval, 1.08-4.45, P=.03). CONCLUSION Although pathologic complete response was predictable preoperatively to some extent, the accuracy was somewhat low. Considerable caution should be exercised when selecting the watch-and-wait approach with operation as needed and omitting planned operative intervention even for patients who achieve clinical complete response after neoadjuvant chemoradiotherapy.
Collapse
Affiliation(s)
- Yoichi Hamai
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| | - Jun Hihara
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Manabu Emi
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Takaoki Furukawa
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yuji Murakami
- Department of Radiation Oncology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ikuno Nishibuchi
- Department of Radiation Oncology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuta Ibuki
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Ichiko Yamakita
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Tomoaki Kurokawa
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
45
|
Semenkovich TR, Meyers BF. Surveillance versus esophagectomy in esophageal cancer patients with a clinical complete response after induction chemoradiation. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:81. [PMID: 29666804 DOI: 10.21037/atm.2018.01.31] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There currently exists an area of controversy in treatment of esophageal cancer for patients who have an apparent clinical complete response (cCR) after induction chemoradiation. A standard treatment is to offer these patients an esophagectomy, but increasingly there is interest from both the patient and provider for active surveillance with so-called "salvage" esophagectomies for local recurrence as an alternative treatment paradigm. In this article, we review the existing evidence that stakeholders should consider for clinical decision-making in this specific patient population, including: the accuracy of post-induction clinical restaging, the reliability of operative risk assessment, the feasibility and adherence to surveillance strategies, and the observed outcomes in these patients after salvage esophagectomy or continued active surveillance. We also briefly discuss quality of life and future directions for this field.
Collapse
Affiliation(s)
- Tara R Semenkovich
- Department of Cardiothoracic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Bryan F Meyers
- Department of Cardiothoracic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
46
|
Li QW, Qiu B, Wang B, Wang DL, Yin SH, Yang H, Liu JL, Fu JH, Liu MZ, Xie CM, Liu H. Prediction of pathologic responders to neoadjuvant chemoradiotherapy by diffusion-weighted magnetic resonance imaging in locally advanced esophageal squamous cell carcinoma: a prospective study. Dis Esophagus 2018; 31:4259168. [PMID: 29036528 DOI: 10.1093/dote/dox121] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 09/06/2017] [Indexed: 12/11/2022]
Abstract
This study aims to investigate the role of diffusion-weighted magnetic resonance imaging (DW-MRI) in ESCC patients receiving neoadjuvant concurrent chemoradiotherapy (CCRT), and the efficacy of apparent diffusion coefficient (ADC) values in predicting pathologic response to neoadjuvant CCRT. Twenty-eight locally advanced ESCC patients treated with neoadjuvant CCRT followed by radical resection were prospectively enrolled. DW-MRI was recommended to be performed within 2 weeks before and 4-6 weeks after neoadjuvant CCRT. The calculated ADCs pre- (ADC1) and post- (ADC2) neoadjuvant CCRT, the definite (ΔADC) and percentage changes (ΔADC%) were analyzed for the efficacy of predicting pathologic response to neoadjuvant CCRT. Twenty patients had been identified as responders (tumor regression grade 1-2). Among them, ADC2 (3.02 ± 0.84 vs. 2.12 ± 0.44 × 10-3 mm2/s, P = 0.001) and ΔADC (1.22 ± 0.78 vs 0.64 ± 0.26 × 10-3 mm2/s, P = 0.007) were significantly higher than those of nonresponders (tumor regression grade: 3-5). Receiver operating characteristic analysis revealed that ADC2 exhibited an overall accuracy of in 71.4% in predicting pathologic response, with a sensitivity of 60.0%, a specificity of 100%, a positive predictive value of 100%, and a negative predictive value of 50.0%, when 3.04 × 10-3 mm2/s was used as the cutoff value. ADC value could be useful in predicting pathologic response to neoadjuvant CCRT in ESCC patients. High postneoadjuvant CCRT ADC is a predictive indicator for good response.
Collapse
Affiliation(s)
- Q-W Li
- Departments of Radiation Oncology
| | - B Qiu
- Departments of Radiation Oncology
| | - B Wang
- Departments of Radiation Oncology
| | | | - S-H Yin
- Departments of Medical Imaging
| | | | - J-L Liu
- Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center Guangzhou, Guangdong, China
| | | | - M-Z Liu
- Departments of Radiation Oncology
| | - C-M Xie
- Departments of Medical Imaging
| | - H Liu
- Departments of Radiation Oncology
| |
Collapse
|
47
|
Loeser H, Schallenberg S, von Winterfeld M, Tharun L, Alakus H, Hölscher A, Bollschweiler E, Buettner R, Zander T, Quaas A. High protein and mRNA expression levels of TUBB3 (class III ß-tubulin) are associated with aggressive tumor features in esophageal adenocarcinomas. Oncotarget 2017; 8:115179-115189. [PMID: 29383151 PMCID: PMC5777763 DOI: 10.18632/oncotarget.23112] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022] Open
Abstract
Background Esophageal adenocarcinomas show an increasing incidence in the Western world and their overall survival remains low. Microtubules are multifunctional cytoskeletal proteins involved in crucial cellular roles, including maintenance of cell shape, intracellular transport, meiosis, and mitosis. Microtubulus-TUBB3 was found overexpressed in several carcinomas suggesting a significant role in cancer development. High levels of TUBB3 expression were also described to be associated with poor clinical outcome in various cancers. It was shown that overexpression of TUBB3 could be related to reduced efficiency of taxane-based targeting anticancer drugs in several cancer types. Results There is a statistically significant association between high TUBB3 protein and TUBB3 mRNA expression and shortened survival (p<0,0001). Prognostic impact of TUBB3 expression is seen in patients with and without multimodal treatment. Multivariate analysis revealed a strong TUBB3 expression to be an independent prognosis factor. Validation of protein expression by mRNA in situ hybridization underlines the credibility of the immunohistochemical results. Discussion Our study emphasized the significant importance of TUBB3 in esophageal adenocarcinoma. TUBB3 serves as an independent prognostic marker and may be a valuable biomarker for routine application in esophageal adenocarcinoma especially to address the need for adjuvant treatment in individuals following neoadjuvant therapy and surgery. Future prospective studies are needed which include the results of TUBB3 in preoperative biopsy material to proof the prognostic impact of TUBB3. Materials and Methods 280 esophageal adenocarcinomas that underwent primary surgical resection or resection after neoadjuvant therapy were analyzed by mRNA-in-situ-hybridization (RNAscope®) and by immunohistochemistry (TUBB3 rabbit monoclonal antibody; Epitomics).
Collapse
Affiliation(s)
- Heike Loeser
- Institute of Pathology, University of Cologne, Cologne, Germany
| | | | | | - Lars Tharun
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Arnulf Hölscher
- Department of Thorax and Oesophageal Surgery, Agaplesion Markus Krankenhaus, Frankfurt/Main, Germany
| | - Elfriede Bollschweiler
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | | | - Thomas Zander
- Department I of Internal Medicine, Center for Integrated Oncology (CIO), University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, Cologne, Germany
| |
Collapse
|
48
|
Noordman BJ, Wijnhoven BPL, Lagarde SM, Biermann K, van der Gaast A, Spaander MCW, Valkema R, van Lanschot JJB. Active surveillance in clinically complete responders after neoadjuvant chemoradiotherapy for esophageal or junctional cancer. Dis Esophagus 2017; 30:1-8. [PMID: 28881890 DOI: 10.1093/dote/dox100] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Indexed: 12/11/2022]
Abstract
Neoadjuvant chemoradiotherapy (nCRT) followed by surgery is standard of care for locally advanced esophageal cancer in many countries. After nCRT up to one third of all patients have a pathologically complete response in the resection specimen, posing an ethical imperative to reconsider the necessity of standard surgery in all operable patients after nCRT. An active surveillance strategy following nCRT, in which patients are subjected to frequent clinical investigations after the completion of neoadjuvant therapy, has been evaluated in other types of cancer with promising results. In esophageal cancer, both patients who are cured by neoadjuvant therapy alone as well as patients with subclinical disseminated disease at the time of completion of neoadjuvant therapy may benefit from such an organ sparing approach. Active surveillance is currently applied in selected patients with esophageal cancer who refuse surgery or are medically unfit for major surgery after completion of nCRT, but this strategy is not (yet) adopted as an alternative to standard surgery or definitive chemoradiation. The available literature is scarce, but suggests that long-term oncological outcomes after active surveillance are noninferior compared to standard surgical resection, providing justification for comparison of both treatments in a phase III trial. This review gives an overview of the current knowledge regarding active surveillance after completion of nCRT in esophageal cancer and outlines future research perspectives.
Collapse
Affiliation(s)
| | | | | | | | | | | | - R Valkema
- Department of Radiology and Nuclear Medicine, Erasmus MC - University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | |
Collapse
|
49
|
Uberoi GS, Uberoi AS, Bhutani MS. Endoscopic and Imaging Predictors of Complete Pathologic Response After Chemoradiation for Esophageal Cancer. Curr Gastroenterol Rep 2017; 19:57. [PMID: 28983771 DOI: 10.1007/s11894-017-0594-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
PURPOSE OF REVIEW Locally advanced esophageal cancer is frequently treated preoperatively with chemotherapy and radiation. The degree of response to this preoperative therapy varies in different patients, and a complete pathological response (pCR) has important implications in the management and prognosis of these patients. This is a review of the different modalities currently available to detect pCR and the clinical context of their use. RECENT FINDINGS While research is still ongoing, no single technique has emerged as the modality of choice to reliably predict pCR in all patients. Studies investigating the sensitivity, specificity, and accuracy of these modalities have had promising results, but no single modality has been firmly validated as the modality if choice. The emergence of functional imaging techniques and the use of biomarkers are newer developments which need further evaluation before adoption in routine clinical practice. While no single technique reliably predicts pCR, a combination of imaging and diagnostic modalities (endoscopic appearance, biopsy, EUS, and PET/CT) may provide a better diagnostic yield rather than any of these modalities taken alone.
Collapse
Affiliation(s)
- Guneesh S Uberoi
- Department of Gastroenterology, Hepatology and Nutrition-Unit 1466, UT MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030-4009, USA
| | - Angad S Uberoi
- Department of Gastroenterology, Hepatology and Nutrition-Unit 1466, UT MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030-4009, USA
| | - Manoop S Bhutani
- Department of Gastroenterology, Hepatology and Nutrition-Unit 1466, UT MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030-4009, USA.
| |
Collapse
|
50
|
Chao YK, Chuang WY, Yeh CJ, Chang HK, Tseng CK. Anatomical distribution of residual cancer in patients with oesophageal squamous cell carcinoma who achieved clinically complete response after neoadjuvant chemoradiotherapy. Eur J Cardiothorac Surg 2017; 53:201-208. [DOI: 10.1093/ejcts/ezx261] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/09/2017] [Indexed: 02/04/2023] Open
|