1
|
Zhang Q, Zhang T, Gu J, Zhang X, Mao Y, Zhu Y, Zhang J, Wang J, Chen S, Cao Y, Wang M, Wang C. Survival benefits of postoperative radiotherapy in esophageal cancer during the immunotherapy era:a retrospective cohort study based on the SEER database and a single-center registry in China. Front Immunol 2025; 16:1548520. [PMID: 40066458 PMCID: PMC11891367 DOI: 10.3389/fimmu.2025.1548520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/04/2025] [Indexed: 05/13/2025] Open
Abstract
Purpose The aim of this study was to investigate the survival benefits of postoperative radiotherapy (PORT) in patients with resectable esophageal cancer (EC) after neoadjuvant therapy in the Immunotherapy era. Methods The study was designed as a retrospective cohort study, which included a total of 733 patients with EC from the SEER database and a single-center cohort. We used propensity score matching (PSM) to equilibrate patient characteristics. The investigation incorporated Kaplan-Meier survival analysis and the Cox proportional risk regression model to assess outcomes. Results PORT did not significantly improve survival in the overall cohort, with a median overall survival of 38 months (p=0.56) in the SEER cohort and 39 months (p=0.75) in the Chinese cohort. However, in the immunotherapy subgroup, the Chinese cohort demonstrated that immunotherapy combined with PORT significantly improved survival (p=0.044).Multivariate Cox regression analysis demonstrated that patients aged 50-59 years (HR=5.93, 95% CI: 1.67-21.06) and those aged ≥70 years (HR=10.96, 95% CI:3.04-39.56) had increased survival risks compared to patients aged <50 years. Additionally, ypT3-4 stage patients exhibited a higher risk than those with ypT1-2 stage (HR=2.12, 95% CI: 1.14-3.93, p=0.017).Similar trends were observed in cT3-4 staging, R1/R2 and no immunotherapy. Lymph node metastasis also showed a progressive relationship with survival risk, with patients categorized as ypN1 (HR=1.90), ypN2 (HR=4.24), and ypN3 (HR=6.68) experiencing increasingly higher risks (p<0.05). Conclusions The collaborative effect of immunotherapy and PORT potentially enhances survival outcomes for patients with EC. However, further prospective research is essential to confirm our results.
Collapse
Affiliation(s)
- Qian Zhang
- Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tao Zhang
- Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiaqi Gu
- Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xuemei Zhang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Yuxin Mao
- Department of Oncology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Yingying Zhu
- Department of Oncology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Jin Zhang
- Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jingyi Wang
- Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuyang Chen
- Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Cao
- Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Muhong Wang
- Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chunbo Wang
- Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
2
|
Liu D, Liu A, Guo L, Li Y, Li Y, Chi Y, Lin H, Yu J, Li M. Postoperative Adjuvant Therapy Benefits Non-pCR Patients Rather Than pCR Patients for Locally Advanced ESCC: A Multicenter Real-World Study. Thorac Cancer 2025; 16:e70021. [PMID: 39988453 PMCID: PMC11847616 DOI: 10.1111/1759-7714.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/04/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND There is no unified standard in adjuvant therapy (AT) for patients with esophageal squamous cell carcinoma (ESCC) after neoadjuvant therapy and surgery. We evaluated the significance of AT for these patients and explored its influencing factors. METHODS ESCC patients who underwent neoadjuvant therapy and surgery from 2019 to 2022 at three centers were divided into AT (n = 227) and non-AT groups (n = 435). Baseline characteristics were balanced using propensity score matching (PSM). Primary endpoints were disease-free survival (DFS) and overall survival (OS), assessed using the Kaplan-Meier method. Subgroup analyses and univariate and multivariate Cox regression analyses were conducted to identify the prognostic factors. RESULTS The median follow-up period is 36 (2-72) months. After PSM, the total population had 1-, 2-, and 3-year OS rates of 71.3%, 66.0%, and 64.1%, respectively. There were no statistically significant differences in DFS (HR: 0.79; 95% CI: 0.55-1.14, p = 0.21) or OS (HR: 0.75; 95% CI: 0.49-1.13, p = 0.17) between AT and non-AT groups. Subgroup analysis revealed that non-pCR patients benefited from AT in DFS (p = 0.042) and OS (p = 0.033). Moreover, in non-pCR patients who received AT, BMI ≥ 21.5 kg/m2 and ypN0 were independent protective factors of DFS. ypN0 was an independent protective factor of OS. In terms of AT regimens, the Kaplan-Meier analysis revealed that adjuvant immunochemotherapy (AICT) provided superior survival benefits than adjuvant radiotherapy and adjuvant chemotherapy. CONCLUSIONS Postoperative AT benefited ESCC patients with non-pCR, while AICT may be a relatively better AT regimen in real-world data, which deserves further exploration.
Collapse
Affiliation(s)
- Defeng Liu
- Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Ao Liu
- Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Longxiang Guo
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
- Department of OncologyDongying People's HospitalDongyingChina
| | - Yi Li
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Yuanlin Li
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Yuxiang Chi
- Cheeloo College of MedicineShandong UniversityJinanChina
- Institute of Oncology, Shandong Provincial HospitalShandong UniversityJinanChina
| | - Haiqun Lin
- Department of Radiation OncologyThe Second Hospital of Shandong UniversityJinanChina
| | - Jinming Yu
- Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Minghuan Li
- Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| |
Collapse
|
3
|
Xia L, Shi W, Cai Y, Liao Z, Huang Z, Qiu H, Wang J, Chen Y. Comparison of long-term survival of neoadjuvant therapy plus surgery versus upfront surgery and the role of adjuvant therapy for T1b-2N0-1 esophageal cancer: a population study of the SEER database and Chinese cohort. Int J Surg 2025; 111:70-79. [PMID: 38920327 PMCID: PMC11745756 DOI: 10.1097/js9.0000000000001886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND For stage T1b-2N0-1 esophageal cancer, the impact of neoadjuvant therapy plus surgery (NS), surgery alone (SA), and surgery plus adjuvant therapy (ST) on cancer-specific survival (CSS) and overall survival (OS) is uncertain. METHODS Stage T1b-2N0-1 esophageal cancer patients from the SEER database and two Chinese cancer centers were included in this study. The Kaplan-Meier method was used to plot survival curves, which were compared using the log-rank test. Propensity score matching was used to equalize differences between the groups. Cox proportional hazards regression models were used to analyze prognostic factors. A nomogram for OS was developed after screening the variables using the Cox proportional hazards regression model and the least absolute shrinkage and selection operator. The performance of the nomogram was assessed by the Harrell concordance index (C-index), the area under the curve (AUC) of the receiver operating characteristic curve, calibration plots, and decision curve analysis. RESULTS After propensity score matching analysis, the 3-year CSS and OS rates in the NS group compared to the SA group were 80.3% versus 62.1% ( P =0.016) and 75.8% versus 55.5% ( P =0.006), the 3-year CSS and OS rates in the NS group compared to the ST group were 71.3% versus 68.3% ( P =0.560) and 69.8% versus 62.9% ( P =0.330), the 3-year CSS and OS rates in the SA group compared to the ST group were 54.6% versus 66.7% ( P =0.220) and 50.2% versus 57.9% ( P =0.290), respectively. The predictive nomogram for OS in T1b-2N0-1 patients ultimately incorporated five clinicopathological variables: T stage, N stage, age, examined lymph nodes, and therapy modality. The nomogram C-index for predicting OS was 0.648, 0.663, and 0.666 in the training group, external validation group-1, and external validation group-2, respectively. The 1-year, 3-year, and 5-year predicted AUC values of the OS prediction model were 0.659, 0.639, and 0.612 for the training group, and 0.786, 0.758, and 0.692 for validation group-1, and 0.805, 0.760, and 0.693 for validation group-2, respectively. CONCLUSION For patients with stage T1b-2N0-1 esophageal cancer, neoadjuvant therapy significantly improves prognosis compared to surgery alone, those presenting with positive lymph nodes after upfront surgery can achieve survival benefits from adjuvant therapy.
Collapse
Affiliation(s)
- Lingling Xia
- Cancer Center, Renmin Hospital of Wuhan University
| | - Wei Shi
- Cancer Center, Renmin Hospital of Wuhan University
| | - Yuxin Cai
- Cancer Center, Renmin Hospital of Wuhan University
| | - Zhengkai Liao
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Zhen Huang
- Cancer Center, Renmin Hospital of Wuhan University
| | - Hu Qiu
- Cancer Center, Renmin Hospital of Wuhan University
| | - Jing Wang
- Cancer Center, Renmin Hospital of Wuhan University
| | | |
Collapse
|
4
|
Xie X, Zhang H, He H, Wu B, Chen Y, Lin W, Feng Q, Chen Q. Postoperative adjuvant immunotherapy for pathological stage II-IVa esophageal squamous cell carcinoma after radical surgery does not improve disease-free recurrence rates. Front Med (Lausanne) 2024; 11:1517001. [PMID: 39735702 PMCID: PMC11671493 DOI: 10.3389/fmed.2024.1517001] [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: 10/25/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Background/objectives Postoperative adjuvant therapy for esophageal squamous cell carcinoma (ESCC) primarily includes chemotherapy and chemoradiotherapy. The survival benefits of postoperative adjuvant therapy for R0-resected ESCC remain controversial. Immunotherapy is being gradually applied perioperatively for esophageal cancer, but the efficacy of postoperative immunotherapy in ESCC is unclear. This study aimed to evaluate the effectiveness of postoperative immunotherapy for esophageal cancer. Toward this goal, we explored the differences between postoperative immunotherapy combined with chemotherapy and postoperative adjuvant chemotherapy alone. Methods This retrospective study evaluated patients who underwent radical surgery for esophageal cancer at Gaozhou People's Hospital between January 2020 and August 2022 and received postoperative adjuvant therapy. Patients were divided into two groups according to the adjuvant treatment regimens: postoperative adjuvant chemotherapy (aCT) and postoperative adjuvant immunotherapy combined with chemotherapy (aICT) groups. Data on baseline characteristics, surgical-related indicators, adverse event rates during adjuvant therapy, and 2-year postoperative follow-up were collected for both groups. Results A total of 76 patients were included: 36 and 40 patients in the aICT and aCT groups, respectively. There were no significant differences in baseline data between the two groups. During the adjuvant treatment period, the incidence of hypothyroidism was significantly higher in the aICT group than in the aCT group (25.0% vs. 2.5%, p = 0.007). During the 2-year follow-up, local and recurrence rates were 17.5 and 12.5% in the aCT group and 13.9 and 5.6% in the aICT group, respectively, showing no significant difference between the two groups (p = 0.489). Conclusion For patients with pathologically confirmed locally advanced ESCC after surgery, postoperative immunotherapy did not confer better disease-free recurrence rates compared to postoperative adjuvant therapy. Nonetheless, with research advancements, the role of immunotherapy in the treatment of ESCC is likely to expand, offering new hope for these patients.
Collapse
Affiliation(s)
- Xihao Xie
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Thoracic Surgery, Gaozhou People’s Hospital Affiliated to Guangdong Medical University, Gaozhou, China
| | - Hai Zhang
- Department of Thoracic Surgery, Gaozhou People’s Hospital Affiliated to Guangdong Medical University, Gaozhou, China
| | - Haiquan He
- Department of Thoracic Surgery, Gaozhou People’s Hospital Affiliated to Guangdong Medical University, Gaozhou, China
| | - Bomeng Wu
- Department of Thoracic Surgery, Gaozhou People’s Hospital Affiliated to Guangdong Medical University, Gaozhou, China
| | - Ying Chen
- Department of Thoracic Surgery, Gaozhou People’s Hospital Affiliated to Guangdong Medical University, Gaozhou, China
| | - Wanli Lin
- Department of Thoracic Surgery, Gaozhou People’s Hospital Affiliated to Guangdong Medical University, Gaozhou, China
| | - Qingyi Feng
- Department of Thoracic Surgery, Gaozhou People’s Hospital Affiliated to Guangdong Medical University, Gaozhou, China
| | - Qunqing Chen
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Zeng Y, Su X, Zhou T, Jia J, Liu J, Yu W, Zhang Q, Song X, Fu X, Cai X. Propensity-matched study on locally advanced esophageal cancer: surgery versus post-operative radiotherapy. Radiat Oncol 2024; 19:130. [PMID: 39334405 PMCID: PMC11428459 DOI: 10.1186/s13014-024-02528-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND This study aims to delineate the long-term outcomes and recurrence patterns of locally advanced thoracic esophageal squamous cell carcinoma (TESCC) patients managed with or without postoperative radiotherapy (PORT). METHODS A retrospective cohort from two academic centers, encompassing patients who initially underwent esophagectomy and were pathologically staged T3-4, was analyzed. Survival outcomes were constructed using Kaplan-Meier method, with survival significance was evaluated using the log-rank test. Propensity score matching (PSM) was utilized to balance potential selection bias. RESULTS Among the 506 patients, 251 underwent surgery alone and 255 received radiotherapy following radical surgery. With a median follow-up of 49.1 months, PORT significantly improved 5-year overall survival (53.8% vs. 25.3%; p < 0.001) and 5-year disease-free survival rates (45.3% vs. 8.5%; p < 0.001) compared to surgery alone. These differences in survival outcomes persisted even after PSM (p < 0.001 for both). Treatment failure was significantly less frequent in the PORT group (46.7%) compared to the surgery-only group (90.0%; p < 0.001), with corresponding reductions in locoregional recurrence (9.4% vs. 54.1%; p < 0.001). This underscores the significant association between PORT and disease control. CONCLUSION The absence of neoadjuvant chemoradiotherapy highlights the importance of PORT in improving survival and reducing recurrence in advanced T3-4 TESCC patients. This study underscores the importance of PORT as a salvage treatment for locally advanced TESCC patients without neoadjuvant chemoradiotherapy.
Collapse
Affiliation(s)
- Ya Zeng
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China
| | - Xi Su
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China
| | - Tongfang Zhou
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China
| | - Jingyi Jia
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China
| | - Jun Liu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China
| | - Wen Yu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China
| | - Xinyun Song
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China
| | - Xiaolong Fu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China.
| | - Xuwei Cai
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai, 200030, China.
| |
Collapse
|
6
|
Chen J, Wang Y, Wu M, Yu K, Liu J, Chang J. Vasculogenic mimicry triggers early recidivation and resistance to adjuvant therapy in esophageal cancer. BMC Cancer 2024; 24:1132. [PMID: 39261780 PMCID: PMC11389244 DOI: 10.1186/s12885-024-12903-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
OBJECTIVE To investigate the impact of vasculogenic mimicry (VM) and postoperative adjuvant therapy on the prognosis and survival of patients with esophageal squamous cell carcinoma (ESCC), as well as to assess whether VM affects the clinical benefit of postoperative adjuvant therapy. METHODS This single-center retrospective analysis included patients who underwent radical surgery for ESCC, which was documented in the medical record system. The presence or absence of VM in surgical specimens was determined using double staining with PAS/CD31. Stratification was applied based on adjuvant therapy and VM status. Survival curves and COX modeling were used to analyze the impact of the presence or absence of VM on the benefit of adjuvant therapy and the survival prognosis of patients. RESULTS VM-positive patients were more prone to postoperative recurrence and metastasis. VM was identified as an independent risk factor for progression-free survival (PFS) (p < 0.001, 95% CI:1.809-3.852) and overall survival (OS) (p < 0.001, 95% CI:1.603-2.786) in postoperative ESCC. Postoperative adjuvant therapy significantly prolonged PFS (p = 0.008) and OS time (p < 0.001) in patients with stage II and III ESCC, with concurrent chemoradiotherapy being the most effective. However, the presence of VM significantly reduced the benefits of postoperative adjuvant therapy (p < 0.001). CONCLUSION VM negatively impacts the prognosis of postoperative ESCC patients and reduces the efficacy of postoperative adjuvant therapy.
Collapse
Affiliation(s)
- Jue Chen
- Department of Oncology, Affiliated Hospital of Yangzhou University, No. 368 Hanjiang Middle Road, Hanjiang District, 225001, Yangzhou, China.
- Medical College of Yangzhou University, 225001, Yangzhou, China.
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the National Administration of Traditional Chinese Medicine, 225001, Yangzhou, China.
| | - Yu Wang
- Medical College of Yangzhou University, 225001, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the National Administration of Traditional Chinese Medicine, 225001, Yangzhou, China
| | - Mengke Wu
- Medical College of Yangzhou University, 225001, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the National Administration of Traditional Chinese Medicine, 225001, Yangzhou, China
| | - Keke Yu
- Medical College of Yangzhou University, 225001, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the National Administration of Traditional Chinese Medicine, 225001, Yangzhou, China
| | - Junchi Liu
- Medical College of Yangzhou University, 225001, Yangzhou, China
| | - Jiayu Chang
- Medical College of Yangzhou University, 225001, Yangzhou, China
| |
Collapse
|
7
|
Guo XW, Ji L, Xi XX, Zhao WW, Liu YC, Zhou SB, Ji SJ. Predictive potential of preoperative Naples prognostic score-based nomogram model for the prognosis in surgical resected thoracic esophageal squamous cell carcinoma patients: A retrospective cohort study. Medicine (Baltimore) 2024; 103:e38038. [PMID: 38701277 PMCID: PMC11062709 DOI: 10.1097/md.0000000000038038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
The present study aimed to establish an effective prognostic nomogram model based on the Naples prognostic score (NPS) for resectable thoracic esophageal squamous cell carcinoma (ESCC). A total of 277 patients with ESCC, who underwent standard curative esophagectomy and designated as study cohort, were retrospectively analyzed. The patients were divided into different groups, including NPS 0, NPS 1, NPS 2, and NPS 3 or 4 groups, for further analysis, and the results were validated in an external cohort of 122 ESCC patients, who underwent surgery at another cancer center. In our multivariate analysis of the study cohort showed that the tumor-node-metastasis (TNM) stage, systemic inflammation score, and NPS were the independent prognostic factors for the overall survival (OS) and progression-free survival (PFS) durations. In addition, the differential grade was also an independent prognostic factor for the OS in the patients with ESCC after surgery (all P < .05). The area under the curve of receiver operator characteristics for the PFS and OS prediction with systemic inflammation score and NPS were 0.735 (95% confidence interval [CI] 0.676-0.795, P < .001) and 0.835 (95% CI 0.786-0.884, P < .001), and 0.734 (95% CI 0.675-0.793, P < .001) and 0.851 (95% CI 0.805-0.896, P < .001), respectively. The above independent predictors for OS or PFS were all selected in the nomogram model. The concordance indices (C-indices) of the nomogram models for predicting OS and PFS were 0.718 (95% CI 0.681-0.755) and 0.669 (95% CI 0.633-0.705), respectively, which were higher than that of the 7th edition of American Joint Committee on Cancer TNM staging system [C-index 0.598 (95% CI 0.558-0.638) for OS and 0.586 (95% CI 0.546-0.626) for PFS]. The calibration curves for predicting the 5-year OS or PFS showed a good agreement between the prediction by nomogram and actual observation. In the external validation cohort, the nomogram discrimination for OS was better than that of the 7th edition of TNM staging systems [C-index: 0.697 (95% CI 0.639-0.755) vs 0.644 (95% CI 0.589-0.699)]. The calibration curves showed good consistency in predicting the 5-year survival between the actual observation and nomogram predictions. The decision curve also showed a higher potential of the clinical application of predicting the 5-years OS of the proposed nomogram model as compared to that of the 7th edition of TNM staging systems. The preoperative NPS-based nomogram model had a certain potential role for predicting the prognosis of ESCC patients.
Collapse
Affiliation(s)
- Xin-Wei Guo
- Department of Radiation Oncology, Affiliated Taixing People’s Hospital of Nanjing Medical University, Kangda College, Taixing, People’s Republic of China
| | - Lei Ji
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Xiao-Xiang Xi
- Department of Thoracic Surgery, Affiliated Taixing People’s Hospital of Nanjing Medical University, Kangda College, Taixing, People’s Republic of China
| | - Wei-Wei Zhao
- Department of Radiation Oncology, Affiliated Taixing People’s Hospital of Nanjing Medical University, Kangda College, Taixing, People’s Republic of China
| | - Yang-Chen Liu
- Department of Radiation Oncology, Affiliated Taixing People’s Hospital of Nanjing Medical University, Kangda College, Taixing, People’s Republic of China
| | - Shao-Bing Zhou
- Department of Radiation Oncology, Affiliated Taixing People’s Hospital of Nanjing Medical University, Kangda College, Taixing, People’s Republic of China
| | - Sheng-Jun Ji
- Department of Radiotherapy and Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, People’s Republic of China
| |
Collapse
|
8
|
Veziant J, Bouché O, Aparicio T, Barret M, El Hajbi F, Lepilliez V, Lesueur P, Maingon P, Pannier D, Quero L, Raoul JL, Renaud F, Seitz JF, Serre AA, Vaillant E, Vermersch M, Voron T, Tougeron D, Piessen G. Esophageal cancer - French intergroup clinical practice guidelines for diagnosis, treatments and follow-up (TNCD, SNFGE, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO, ACHBT, SFP, RENAPE, SNFCP, AFEF, SFR). Dig Liver Dis 2023; 55:1583-1601. [PMID: 37635055 DOI: 10.1016/j.dld.2023.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION This document is a summary of the French intergroup guidelines regarding the management of esophageal cancer (EC) published in July 2022, available on the website of the French Society of Gastroenterology (SNFGE) (www.tncd.org). METHODS This collaborative work was conducted under the auspices of several French medical and surgical societies involved in the management of EC. Recommendations were graded in three categories (A, B and C), according to the level of evidence found in the literature until April 2022. RESULTS EC diagnosis and staging evaluation are mainly based on patient's general condition assessment, endoscopy plus biopsies, TAP CT-scan and 18F FDG-PET. Surgery alone is recommended for early-stage EC, while locally advanced disease (N+ and/or T3-4) is treated with perioperative chemotherapy (FLOT) or preoperative chemoradiation (CROSS regimen) followed by immunotherapy for adenocarcinoma. Preoperative chemoradiation (CROSS regimen) followed by immunotherapy or definitive chemoradiation with the possibility of organ preservation are the two options for squamous cell carcinoma. Salvage surgery is recommended for incomplete response or recurrence after definitive chemoradiation and should be performed in an expert center. Treatment for metastatic disease is based on systemic therapy including chemotherapy, immunotherapy or combined targeted therapy according to biomarkers testing such as HER2 status, MMR status and PD-L1 expression. CONCLUSION These guidelines are intended to provide a personalised therapeutic strategy for daily clinical practice and are subject to ongoing optimization. Each individual case should be discussed by a multidisciplinary team.
Collapse
Affiliation(s)
- Julie Veziant
- Department of Digestive and Oncological Surgery, Claude Huriez Hospital, CHU Lille, University of Lille, Lille F-59000, France.
| | - Olivier Bouché
- Department of Digestive Oncology, CHU Reims, Reims, France
| | - T Aparicio
- Department of Gastroenterology and Digestive Oncology, AP-HP, Saint-Louis Hospital, Paris, France
| | - M Barret
- Gastroenterology Department, Cochin Hospital, APHP, Paris, France
| | - F El Hajbi
- Department of Oncology, Centre Oscar Lambret, Lille, France
| | - V Lepilliez
- Gastroenterology Department, Jean Mermoz Private Hospital, Ramsay Santé, Lyon, France
| | - P Lesueur
- Department of Radiation Oncology, Centre Guillaume le Conquérant, Le Havre, France
| | - P Maingon
- Department of Radiation Oncology, La Pitié-Salpêtrière, APHP, Sorbonne University, Paris, France
| | - D Pannier
- Department of Oncology, Centre Oscar Lambret, Lille, France
| | - L Quero
- Department of Radiation Oncology, Saint-Louis Hospital, APHP, Paris, France
| | - J L Raoul
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - F Renaud
- Department of Pathology, La Pitié-Salpêtrière, APHP, Sorbonne University, Paris, France
| | - J F Seitz
- Department of Digestive Oncology, La Timone, Aix Marseille Université, Marseille, France
| | - A A Serre
- Department of Radiotherapy, Centre Léon Bérard, Lyon, France
| | | | - M Vermersch
- Medical Imaging Department, Valencienne Hospital Centre, Valencienne 59300, France
| | - T Voron
- Department of General and Digestive Surgery, Sorbonne Université, AP-HP, Hôpital Saint Antoine, 184 rue du faubourg Saint-Antoine, Paris 75012, France
| | - D Tougeron
- Department of Gastro-Enterology and Hepatology, Poitiers University Hospital, Poitiers, France
| | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, Claude Huriez Hospital, CHU Lille, University of Lille, Lille F-59000, France
| |
Collapse
|
9
|
Yao Q, Zheng H, Huang S, Lin M, Yang J, Li J. Low versus high dose of postoperative radiotherapy for locally advanced esophageal squamous cell carcinoma: a propensity score-matched analysis. PRECISION RADIATION ONCOLOGY 2023; 7:101-110. [PMID: 40337268 PMCID: PMC11935151 DOI: 10.1002/pro6.1192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 05/09/2025] Open
Abstract
Objective This study aimed to investigate the effect of different postoperative radiotherapy doses on the prognosis of patients with esophageal squamous cell carcinoma (ESCC). Methods A total of 199 patients (aged 18-75 years) with locally advanced ESCC who underwent esophagectomy and postoperative radiotherapy/chemoradiotherapy at the Fujian Cancer Hospital between July 2008 and January 2018 were included. Based on the postoperative radiotherapy dose, the patients were divided into a low-dose group (50-50.4 Gy; median dose 50 Gy) and a high-dose group (>50.4 Gy; median dose 60 Gy). Neoadjuvant and adjuvant chemotherapy regimens included PF (fluorouracil and cisplatin) and TP (paclitaxel and cisplatin) regimens. Patients were followed-up every 3 months in the first 2 years after surgery, every 6 months for the next 3 years, and then subsequently once a year. The primary endpoints were overall survival (OS) and progression-free survival (PFS) rates. The propensity-score matching (PSM) method was applied to identify a 1:1, well-balanced matched cohort with 33 patients in each group for survival comparison. Results Among the 199 patients enrolled in this study, 144 and 55 were in the low-dose and high-dose groups, respectively. Univariate and multivariate analyses showed that pathological N classification, vascular tumor emboli, and postoperative radiotherapy dose were independent prognostic factors for both OS and PFS, all p < 0.05. Before PSM, the OS and the PFS of the low-dose group were significantly longer than those of the high-dose group, both p < 0.05. After PSM, better OS and PFS rates were observed in the low-dose group, both p < 0.05. The results showed that patients with pathological stages N0-2 or N3, negative surgical margins, and no vascular tumor emboli could obtain a significant benefit in both OS and PFS after treatment with a low dose of postoperative radiotherapy (50-50.4 Gy). In the subgroup with positive surgical margins, treatment with a low dose of postoperative radiotherapy offered a non-significant survival benefit compared to treatment with a high dose of postoperative radiotherapy. Conclusions Our study revealed that for patients with ESCC, the low-dose group (50-50.4 Gy) had a significantly higher OS and PFS than the high-dose group (>50.4 Gy). It was suggested that 50-50.4 Gy might be the recommended postoperative radiotherapy dose for ESCC patients.
Collapse
Affiliation(s)
- Qiwei Yao
- Department of Radiation OncologyClinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhouChina
| | - Hongying Zheng
- Department of Radiation OncologyClinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhouChina
| | - Shuyun Huang
- Department of Radiation OncologyClinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhouChina
| | - Mingqiang Lin
- Department of Radiation OncologyClinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhouChina
| | - Jun Yang
- Department of Radiation OncologyClinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhouChina
| | - Jiancheng Li
- Department of Radiation OncologyClinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhouChina
| |
Collapse
|
10
|
Tondolo V, Casà C, Rizzo G, Leone M, Quero G, Alfieri V, Boldrini L, Bulajic M, Corsi D, Micciché F. Management of Esophago-Gastric Junction Carcinoma: A Narrative Multidisciplinary Review. Cancers (Basel) 2023; 15:2597. [PMID: 37174063 PMCID: PMC10177387 DOI: 10.3390/cancers15092597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Esophagogastric junction (EGJ) carcinoma represents a specific site of disease, given the opportunities for multimodal clinical care and management and the possibilities of combined treatments. It encompasses various clinical subgroups of disease that are heterogeneous and deserve different treatments; therefore, the guidelines have progressively evolved over time, considering the evidence provided by clinical trials. The aim of this narrative review was to summarize the main evidence, which orientates the current guidelines, and to collect the main ongoing studies to address existing gray areas.
Collapse
Affiliation(s)
- Vincenzo Tondolo
- U.O.C. di Chirurgia Digestiva e del Colon-Retto, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy; (V.T.); (V.A.)
| | - Calogero Casà
- U.O.C. di Radioterapia Oncologica, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy; (C.C.); (M.L.); (F.M.)
| | - Gianluca Rizzo
- U.O.C. di Chirurgia Digestiva e del Colon-Retto, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy; (V.T.); (V.A.)
| | - Mariavittoria Leone
- U.O.C. di Radioterapia Oncologica, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy; (C.C.); (M.L.); (F.M.)
| | - Giuseppe Quero
- U.O.C. di Chirurgia Digestiva, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Virginia Alfieri
- U.O.C. di Chirurgia Digestiva e del Colon-Retto, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy; (V.T.); (V.A.)
- Università Campus Bio-Medico College, 00128 Rome, Italy
| | - Luca Boldrini
- U.O.C. di Radioterapia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Milutin Bulajic
- U.O.C. di Endoscopia Digestiva, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy;
| | - Domenico Corsi
- U.O.C. di Oncologia Medica, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy;
| | - Francesco Micciché
- U.O.C. di Radioterapia Oncologica, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy; (C.C.); (M.L.); (F.M.)
| |
Collapse
|
11
|
Yang W, Yang Y, Ma X, Ma M, Han B. Surgery and postoperative radiotherapy affect the prognosis of esophageal cancer: A SEER analysis. Medicine (Baltimore) 2023; 102:e32925. [PMID: 36862870 PMCID: PMC9981363 DOI: 10.1097/md.0000000000032925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
The principal treatment modalities for esophageal cancer are radiation, chemotherapy and surgery or a combination of them. In some sense, technological advances have tremendously heightened patients' survival rates. Nevertheless, the debate on the prognostic value of postoperative radiotherapy (PORT) has never ceased. On that account, this study made an effort to probe deep into the effects of PORT and surgery on the prognosis of stage III esophageal cancer. Our study included patients diagnosed with stage III esophageal cancer between 2004 and 2015 through the Surveillance, Epidemiology, and End Results (SEER) program. We performed propensity score matching (PSM) on the basis of whether surgery was carried out and whether PORT conducted. We identified the independent risk factors by multivariate Cox regression and constructed a nomogram model. In this research, we included 3940 patients, and the median follow-up is 14 months: 1932 cases without surgery; 2008 cases with surgery, and 322 cases of them underwent PORT. In the postPSM patient cohort, patients who underwent surgery had a median overall survival rate (OS) of 19.0 (95% confidence interval [CI] 17.2-20.8) and a median cancer-specific survival rate (CSS) of 23.0 (95% CI 20.6-25.3) months, which were remarkably higher than those without surgery (P < .001). The OS(P < .05)and CSS(P < .05)of the patients who underwent PORT were lower than those who did not. Similar results were obtained in the groups of N0 and N1. This study revealed surgery can heighten patients' survival rate, while PORT could not elevate patients' survival rate in stage III esophageal cancer patients.
Collapse
Affiliation(s)
- Wenwen Yang
- The First Clinical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yanjiang Yang
- Qilu hospital of Shandong University, Shandong University, Jinan, Shandong Province, China
| | - Xiang Ma
- The First Clinical College of Lanzhou University, Lanzhou, Gansu Province, China
| | - Minjie Ma
- Department of Thoracic Surgery, the First Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- Gansu Province International Cooperation Base for Research and Application of Key Technology of Thoracic Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Biao Han
- Department of Thoracic Surgery, the First Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- Gansu Province International Cooperation Base for Research and Application of Key Technology of Thoracic Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- *Correspondence: Biao Han, Department of Thoracic Surgery, the First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China (e-mail: )
| |
Collapse
|
12
|
Pu J, Teng Z, Zhang T, Wang B, Zhang D, Yang Q, Yang Q, Sun X, Long W. Expression of Polyadenylate-binding Protein Cytoplasmic 1 (PABPC1) in Combination With RAD51 as Prognostic Biomarker in Patients Who Underwent Postoperative Chemotherapy for Esophageal Squamous Cell Carcinoma. Appl Immunohistochem Mol Morphol 2023; 31:189-195. [PMID: 36735495 DOI: 10.1097/pai.0000000000001100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 12/30/2022] [Indexed: 02/04/2023]
Abstract
Molecular markers in the prognosis of esophageal squamous cell carcinoma (ESCC) patients who received postoperative treatments are lacking. This research aims to evaluate the prognostic value of polyadenylate-binding protein cytoplasmic 1 (PABPC1) alone and in combination with RAD51 in ESCC patients who underwent postoperative chemotherapy (CT). A total of 103 ESCC patients who underwent postoperative CT and 103 matched ones who received surgery alone were analyzed in this study. PABPC1 and RAD51 expression was assessed in cancer samples by immunohistochemistry. PABPC1 high expression (PABPC1-HE) but not that of RAD51 was associated with poor patients' survival, regardless of the postoperative treatment or node status. Patients with PABPC1 low expression and RAD51 negative expression [RAD51- (PABPC1-LE/RAD51-)] tumor had good overall survival (OS) in both the CT treated and untreated groups. Patients with PABPC1-LE/RAD51+ and PABPC1-HE/RAD51+ tumors had longer OS in the CT treated group than in the untreated group. However, PABPC1-HE/RAD51- was associated with a poor outcome in both groups and the patients with PABPC1-HE/RAD51- tumor had hardly any benefit from CT in N+ status. PABPC1 alone and in combination with RAD51 was a prognostic biomarker for OS in ESCC patients who received postoperative CT.
Collapse
Affiliation(s)
| | | | | | | | | | - Qin Yang
- Pathology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Qiao Yang
- Pathology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Xingwang Sun
- Pathology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Wenbo Long
- Pathology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| |
Collapse
|
13
|
He X, Zhou Y, Chen W, Zhao X, Duan L, Zhou H, Li M, Yu Y, Zhao J, Guo Y, Gu H, Jiang Y, Dong Z, Liu K. Repurposed pizotifen malate targeting NRF2 exhibits anti-tumor activity through inducing ferroptosis in esophageal squamous cell carcinoma. Oncogene 2023; 42:1209-1223. [PMID: 36841865 DOI: 10.1038/s41388-023-02636-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 02/27/2023]
Abstract
Targeted therapy attempts are needed to enhance esophageal squamous cell carcinoma (ESCC) patients' overall survival and satisfaction of life. Nuclear factor erythroid 2-related factor 2 (NRF2), as a high-confidence cancer driver gene, controls the antioxidant response, metabolic balance and redox homeostasis in cancer and is regarded as a potent molecular target for cancer treatment. Here, we attempted to find a new NRF2 inhibitor and study the underlying molecular mechanism in ESCC. We found that up-regulated NRF2 protein was negatively correlated with patient prognosis and promoted tumor proliferation in ESCC. Moreover, Pizotifen malate (PZM), a FDA-approved medication, bound to the Neh1 domain of NRF2 and prevented NRF2 protein binding to the ARE motif of target genes, suppressing transcription activity of NRF2. PZM treatment suppressed tumor development in ESCC PDX model by inducing ferroptosis via down-regulating the transcription of GPX4, GCLC, ME1 and G6PD. Our study illustrates that the over expression of NRF2 indicates poor prognosis and promotes tumor proliferation in ESCC. PZM, as a novel NRF2 inhibitor, inhibits the tumor growth by inducing ferroptosis and elucidates a potent NRF2-based therapy strategy for patients with ESCC.
Collapse
Affiliation(s)
- Xinyu He
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yubing Zhou
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Wenjing Chen
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Xiaokun Zhao
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Lina Duan
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Hao Zhou
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mingzhu Li
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Yin Yu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China
| | - Jimin Zhao
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China.,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China
| | - Yaping Guo
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Huihui Gu
- Basic Medicine Sciences Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yanan Jiang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Zigang Dong
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China.
| | - Kangdong Liu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, Henan, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China. .,Basic Medicine Sciences Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
14
|
Yang W, Liu F, Xu R, Yang W, He Y, Liu Z, Zhou F, Heng F, Hou B, Zhang L, Chen L, Zhang F, Cai F, Xu H, Lin M, Liu M, Pan Y, Liu Y, Hu Z, Chen H, He Z, Ke Y. Is Adjuvant Therapy a Better Option for Esophageal Squamous Cell Carcinoma Patients Treated With Esophagectomy? A Prognosis Prediction Model Based on Multicenter Real-World Data. Ann Surg 2023; 277:e61-e69. [PMID: 34091512 DOI: 10.1097/sla.0000000000004958] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To construct a prediction model for more precise evaluation of prognosis which will allow personalized treatment recommendations for adjuvant therapy in patients following resection of ESCC. BACKGROUND Marked heterogeneity of patient prognosis and limited evidence regarding survival benefit of various adjuvant therapy regimens pose challenges in the clinical treatment of ESCC. METHODS Based on comprehensive clinical data obtained from 4129 consecutive patients with resected ESCC in a high-risk region in China, we identified predictors for overall survival through a 2-phase selection based on Cox proportional hazard regression and minimization of Akaike information criterion. The model was internally validated using bootstrapping and externally validated in 1815 patients from a non-high-risk region in China. RESULTS The final model incorporates 9 variables: age, sex, primary site, T stage, N stage, number of lymph nodes harvested, tumor size, adjuvant treatment, and hemoglobin level. A significant interaction was also observed between N stage and adjuvant treatment. N1+ stage patients were likely to benefit from addition of adjuvant therapy as opposed to surgery alone, but adjuvant therapy did not improve overall survival for N0 stage patients. The C -index of the model was 0.729 in the training cohort, 0.723 after bootstrapping, and 0.695 in the external validation cohort. This model outperformed the seventh edition American Joint Committee on Cancer staging system in prognostic prediction and risk stratification. CONCLUSIONS The prediction model constructed in this study may facilitate precise prediction of survival and inform decision-making about adjuvant therapy according to N stage.
Collapse
Affiliation(s)
- Wenlei Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Fangfang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ruiping Xu
- Anyang Cancer Hospital, Anyang, Henan Province, People's Republic of China
| | - Wei Yang
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Yu He
- Department of Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Zhen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Fuyou Zhou
- Anyang Cancer Hospital, Anyang, Henan Province, People's Republic of China
| | - Fanxiu Heng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of information Technology Service, Peking University Cancer Hospital & institute, Beijing, People's Republic of China
| | - Bolin Hou
- Linkdoc AI Research (LAIR), Beijing, People's Republic of China
| | - Lixin Zhang
- Anyang Cancer Hospital, Anyang, Henan Province, People's Republic of China
| | - Lei Chen
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Fan Zhang
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Fen Cai
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Huawen Xu
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Miaoping Lin
- Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Mengfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yaqi Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zhe Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Huanyu Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zhonghu He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yang Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| |
Collapse
|
15
|
Han W, Chang X, Zhang W, Yang J, Yu S, Deng W, Ni W, Zhou Z, Chen D, Feng Q, Liang J, Hui Z, Wang L, Gao S, Lin Y, Chen X, Chen J, Xiao Z. Effect of Adjuvant Radiation Dose on Survival in Patients with Esophageal Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:5879. [PMID: 36497360 PMCID: PMC9736548 DOI: 10.3390/cancers14235879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Background: For patients with esophageal squamous cell carcinoma (ESCC) treated with surgery alone, the incidence of local-regional recurrence remains unfavorable. Postoperative radiotherapy (PORT) has been associated with increased local-regional recurrence-free survival (LRFS), although its application is limited by concerns of PORT-related toxicities. Methods: Among 3591 patients with ESCC analyzed in this study, 2765 patients with T3-4N0 and T1-4N1-3 lesions and specific local-regional status information were analyzed in a subsequent analysis of adjuvant radiation dose (aRTD) effect. Application of the restricted cubic spline regression model revealed a non-linear relationship between aRTD and survival/radiotoxicity. Linear regression analysis (LRA) was performed to evaluate correlations between LRFS and overall survival (OS)/ disease-free survival (DFS). Results: For patients staged T1−2N0, T1−2N1−3, T3−4N0, and T3−4N1−3, 5-year OS in PORT and non-PORT groups were 77.38% vs. 72.91%, p = 0.919, 52.35% vs. 46.60%, p = 0.032, 73.41% vs. 61.19%, p = 0.005 and 38.30% vs. 25.97%, p < 0.001. With aRTD escalation, hazard ratios (HRs) of OS/DFS declined until aRTD exceeded 50Gy, then increased, whereas that of LRFS declined until aRTD exceeded 50 Gy, then remained steady. HR of treatment-related mortality was stable until aRTD exceeded 50 Gy, then increased. LRA revealed strong correlations between LRFS and OS/DFS (r = 0.984 and r = 0.952, respectively). An absolute 1% advancement in LRFS resulted in 0.32% and 0.34% improvements in OS and DFS. Conclusions: An aRTD of 50Gy was well-tolerated, with favorable survival resulting from PORT-related LRFS improvement in patients staged T3−4N0 or T1-4N1−3. Further stratification analyses based on tumor burden would help determine potential PORT-beneficiaries.
Collapse
Affiliation(s)
- Weiming Han
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiao Chang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jingsong Yang
- Department of Radiation Oncology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shufei Yu
- Department of Radiation Oncology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Wei Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wenjie Ni
- Department of Radiation Oncology, Beijing Shijitan Hospital, Capital Medical University, Ninth School of Clinical Medicine, Beijing 100038, China
| | - Zongmei Zhou
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dongfu Chen
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qinfu Feng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jun Liang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhouguang Hui
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lvhua Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu Lin
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Xiaohui Chen
- Department of Thoracic Surgery, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Junqiang Chen
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Zefen Xiao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
16
|
Feng J, Wang L, Yang X, Chen Q, Cheng X. The usefulness of pretreatment controlling nutritional status score for predicting recurrence in patients with esophageal squamous cell carcinoma undergoing neoadjuvant immunochemotherapy: A real-world study. Front Immunol 2022; 13:1015365. [PMID: 36505443 PMCID: PMC9729701 DOI: 10.3389/fimmu.2022.1015365] [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: 08/09/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Background The controlling nutritional status (CONUT) score, as an immune-nutritional index, has been reported to be related to prognosis in several cancers. Neoadjuvant immunochemotherapy (nICT) is an emerging pattern for cancer treatment in recent years. However, the usefulness of CONUT in esophageal squamous cell carcinoma (ESCC) with nICT has not been reported so far. This study attempted to clarify the usefulness of CONUT in predicting disease-free survival (DFS) in ESCC with nICT. Methods Two hundred sixteen ESCC patients receiving nICT between 2019 and 2021 were retrospectively enrolled. Based on CONUT, the patients were divided into two groups: low groups (score ≤ 2) and high (score ≥ 3) groups. The relationships between CONUT and clinical characteristics were estimated. Cox regression analyses with hazard ratios (HRs) and 95% confidence intervals (CIs) were also performed to evaluate the prognostic factors of DFS. Results Fifty-nine (27.3%) patients achieved pathologic complete response (pCR), and 30 (13.9%) cases had a recurrence. There were 150 cases (69.4%) in low CONUT group and 66 cases (30.6%) in high CONUT group, respectively. The results revealed that vessel invasion (P = 0.037), postoperative pneumonia (P = 0.001), advanced ypT stage (P = 0.011), cTNM stage (P = 0.007), and ypTNM stage (P < 0.001) were significantly related to patients with a high CONUT score. A high pCR rate was found in patients with a low CONUT score (33.3% vs. 13.6%, P = 0.003), and a high recurrence rate was found in patients with a high CONUT score (24.2% vs. 9.3%, P = 0.004), respectively. Patients with a low CONUT score had a better 1-year DFS than those with a high CONUT score (90.7% vs. 75.8%, P = 0.004). Multivariate analyses indicated that the pretreatment CONUT score was an independent predictor regarding DFS (HR = 2.221, 95% CI: 1.067-4.625, P = 0.033). Conclusion A better response and a lower recurrence were found in ESCC patients with a lower pretreatment CONUT. As a useful index for immune-nutritional status, the CONUT might be a reliable prognostic indicator in ESCC patients with nICT.
Collapse
Affiliation(s)
- Jifeng Feng
- Department of Thoracic Oncological Surgery, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Science, Hangzhou, China,Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Science, Hangzhou, China,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Liang Wang
- Department of Thoracic Oncological Surgery, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Science, Hangzhou, China
| | - Xun Yang
- Department of Thoracic Oncological Surgery, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Science, Hangzhou, China
| | - Qixun Chen
- Department of Thoracic Oncological Surgery, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Science, Hangzhou, China,*Correspondence: Qixun Chen, ; Xiangdong Cheng,
| | - Xiangdong Cheng
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Science, Hangzhou, China,*Correspondence: Qixun Chen, ; Xiangdong Cheng,
| |
Collapse
|
17
|
Abdelhakeem A, Blum Murphy M. Adjuvant Therapies for Esophageal Cancer. Thorac Surg Clin 2022; 32:457-465. [DOI: 10.1016/j.thorsurg.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Chen H, Sun G, Han Z, Wang H, Li J, Ye H, Song C, Zhang J, Wang P. Anti-CXCL8 Autoantibody: A Potential Diagnostic Biomarker for Esophageal Squamous Cell Carcinoma. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58101480. [PMID: 36295640 PMCID: PMC9607113 DOI: 10.3390/medicina58101480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/15/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022]
Abstract
Background and Objectives: Esophageal squamous cell carcinoma (ESCC) is one of the most common malignancies. Anti-tumor associated antigen autoantibodies (TAAbs) can be used as biomarkers for tumor detection. The aim of this study was to identify a reliable TAAb as the diagnostic marker for ESCC. Materials and Methods: The Cancer Genome Atlas (TCGA) database was used to screen candidate genes. The mRNA expression of the key gene was then verified by micro array dataset GSE44021 from the Gene Expression Omnibus (GEO) database and the diag nostic value of the corresponding autoantibody to the key gene in ESCC was detected by enzyme-linked im muno sorbent assay (ELISA). Results: CXCL8 was identified as the key gene. The dataset GSE44021 showed that CXCL8 mRNA expression was prominently over-expressed in ESCC tissues compared with normal tissues. ELISA results showed that the level of anti-CXCL8 autoantibody in ESCC patients was significantly higher than in normal controls and the receiver operating char ac teristic (ROC) curve indicated that anti-CXCL8 autoantibody could discriminate ESCC patients from normal controls, with the area under the ROC curve (AUC) for the verification cohort, and the validation cohort were 0.713 and 0.751, respectively. Conclusions: Our study illustrated that anti-CXCL8 autoantibody had good diagnostic value, and may become a candidate biomarker for ESCC.
Collapse
Affiliation(s)
- Huili Chen
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Guiying Sun
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Zhuo Han
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Huimin Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Jiaxin Li
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Hua Ye
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Chunhua Song
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Jianying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Peng Wang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Correspondence: ; Tel.: +86-0371-67781453
| |
Collapse
|
19
|
Zheng SY, Qi WX, Zhao SG, Chen JY. No survival benefit could be obtained from adjuvant radiotherapy in esophageal cancer treated with neoadjuvant chemotherapy followed by surgery: A SEER-based analysis. Front Oncol 2022; 12:897476. [PMID: 36185174 PMCID: PMC9516333 DOI: 10.3389/fonc.2022.897476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe aim of this study is to assess the clinical benefit of postoperative radiotherapy (PORT) in patients with esophageal cancer (EC) who treated with neoadjuvant chemotherapy (NAC) and surgery via a national population-based database.MethodsPatients diagnosed with EC between 2004 and 2015 were identified from the Surveillance, Epidemiology, and End Results (SEER) database. Kaplan–Meier survival analysis was used to compare the overall survival (OS) and cause-specific survival (CSS) difference between PORT vs. no-radiotherapy (RT) groups before and after propensity score matching (PSM). After PSM for baseline characteristics, Cox proportional hazard regression was performed to investigate the factors associated with OS.ResultsA total of 321 patients were included in the analysis. Of them, 91 patients (28%) received PORT. In the unmatched population, the no-RT group had improved OS compared with PORT (44 vs. 25 months, p = 0.002), and CSS was similar in patients undergoing NAC with or without PORT (42 vs. 71 months, p = 0.17). After PSM for baseline characteristics, the OS benefit of the no-RT group over the PORT group remained significant with a median OS of 46 vs. 27 months (p = 0.02), and CSS remained comparable between groups (83 vs. 81 months, p = 0.49). In subgroup analyses, PORT did not improve the OS among patients with adenocarcinoma in the subgroups of cN0, cN1, and cN2–3 (all p > 0.05). In Cox regression, aged ≥71 years old, cT3–4, cN2–3, and receiving PORT were independent predictors of worse OS, whereas cT4 and cN2–3 were independent predictors of worse CSS (all p < 0.05).ConclusionsThe present study demonstrated that no survival benefit could be obtained from the additional use of PORT after NAC and surgery in patients with EC. Well-designed prospective trials are needed to confirm our findings.
Collapse
Affiliation(s)
| | | | | | - Jia-Yi Chen
- *Correspondence: Sheng-Guang Zhao, ; Jia-Yi Chen,
| |
Collapse
|
20
|
Machine Learning and Novel Biomarkers Associated with Immune Infiltration for the Diagnosis of Esophageal Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:6732780. [PMID: 36081670 PMCID: PMC9448540 DOI: 10.1155/2022/6732780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/08/2022] [Accepted: 07/13/2022] [Indexed: 11/18/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) accounts for the main esophageal cancer type, which is related to advanced stage and poor survivals. Therefore, novel diagnostic biomarkers are critically needed. In the current research, we aimed to screen novel diagnostic biomarkers based on machine learning. The expression profiles were obtained from GEO datasets (GSE20347, GSE38129, and GSE75241) and TCGA datasets. Differentially expressed genes (DEGs) were screened between 47 ESCC and 47 nontumor samples. The LASSO regression model and SVM-RFE analysis were carried out for the identification of potential markers. ROC analysis was carried out to assess discriminatory abilities. The expressions and diagnostic values of the candidates in ESCC were demonstrated in the GSE75241 datasets and TCGA datasets. We also explore the correlations between the critical genes and cancer immune infiltrates using CIBERSORT. In this study, we identified 27 DEGs in ESCC: 5 genes were significantly elevated, and 22 genes were significantly decreased. Based on the results of the SVM-RFE and LASSO regression model, we identified five potential diagnostic biomarkers for ESCC, including GPX3, COL11A1, EREG, MMP1, and MMP12. However, the diagnostic values of only GPX3, MMP1, and MMP12 were confirmed in GSE75241 datasets. Moreover, in TCGA datasets, we further confirmed that GPX3 expression was distinctly decreased in ESCC specimens, while the expression of MMP1 and MMP12 was noticeably increased in ESCC specimens. Immune cell infiltration analysis revealed that the expression of GPX3, MMP1, and MMP12 was associated with several immune, such as T cells CD8, macrophages M2, macrophages M0, and dendritic cells activated. Overall, our findings suggested GPX3, MMP1, and MMP12 as novel diagnostic marker and correlated with immune infiltrates in ESCC patients.
Collapse
|
21
|
Tang B, Zhang Q, Liu K, Huang Y. Exosomal circRNA FNDC3B promotes the progression of esophageal squamous cell carcinoma by sponging miR-490-5p and regulating thioredoxin reductase 1 expression. Bioengineered 2022; 13:13829-13848. [PMID: 35703190 PMCID: PMC9275986 DOI: 10.1080/21655979.2022.2084484] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Exosomal circular RNAs (circRNAs) have been reported to play critical roles in esophageal squamous cell carcinoma (ESCC). We aimed to investigate the function of exosomal circRNA FNDC3B (circFNDC3B). The RNA levels and protein levels were examined using RT-qPCR and western blot (WB) assays. Colony formation and EdU assays were used to assess cell proliferative ability. Cell migratory and invasive abilities were detected by wound healing and transwell assays. Cell apoptosis was measured by flow cytometry. Glycolysis was measured using commercial kits. Transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) were applied to examine the morphology and size of exosomes. Dual-luciferase reporter, RIP and RNA pull-down assays assessed the interaction of miR-490-5p with circFNDC3B or thioredoxin reductase 1 (TXNRD1). Xenograft tumor model determined the role of exosomal circFNDC3B in vivo. We observed that circFNDC3B was upregulated in ESCC samples and cells, as well as ESCC-derived exosomes. CircFNDC3B could be delivered via exosomes in tumor cells, and the colony formation, proliferation, migration, invasion, glycolysis, and in vivo growth ability of recipient cells were weakened after co-incubation with exosomal circFNDC3B-knockdown donor cells. CircFNDC3B was a miR-490-5p sponge, and miR-490-5p inhibition reversed the role of exosomal circFNDC3B-downregulating in ESCC cells. TXNRD1 was a miR-490-5p target, and TXNRD1 elevation weakened the anti-cancer function of miR-490-5p upregulation in ESCC cells. CircFNDC3B mediated TXNRD1 expression by interacting with miR-490-5p. In conclusion, exosomal circFNDC3B drove ESCC progression via regulating the miR-490-5p/TXNRD1 axis.AbbreviationsEC: esophageal cancer; ESCC: esophageal squamous cell carcinoma; circRNA: circular RNA; WB: western blot; TEM: transmission electron microscopy; NTA: nanoparticle tracking analysis; TXNRD1: thioredoxin reductase 1; IHC: immunohistochemistry; RT-qPCR: reverse transcription-polymerase quantitative chain reaction; GLUT1: glucose transport protein type 1; LDHA: lactate dehydrogenase A.
Collapse
Affiliation(s)
- Bo Tang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong City, China
| | - Qingfeng Zhang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong City, China
| | - Kui Liu
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong City, China
| | - Yun Huang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong City, China
| |
Collapse
|
22
|
Mei LX, Mo JX, Chen Y, Dai L, Wang YY, Chen MW. Esophagectomy versus definitive chemoradiotherapy as initial treatment for clinical stage I esophageal cancer: a systematic review and meta-analysis. Dis Esophagus 2022; 35:6329176. [PMID: 34318324 DOI: 10.1093/dote/doab049] [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: 01/16/2021] [Revised: 05/19/2021] [Accepted: 06/27/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Esophagectomy and definitive chemoradiotherapy are commonly used in the treatment of stage I esophageal cancer (EC). The present study aims to compare the efficacy and safety of esophagectomy and definitive chemoradiotherapy as the initial treatment for clinical stage I EC. METHODS This study was registered with the International Prospective Register of Systematic Reviews (CRD42020197203). Relevant studies were identified through PubMed, Web of Science, EMBASE, and Cochrane Library from database inception to June 30, 2020. Hazard ratio (HR) with 95% confidence intervals (CI) was employed to compare overall survival (OS) and progression-free survival (PFS). Odds ratio (OR) with 95% CI was employed to compare treatment-related death, complications, and tumor recurrence. RESULTS A total of 13 non-randomized controlled studies involving 3,346 patients were included. Compared with definitive chemoradiotherapy, esophagectomy showed an improved OS (HR 0.69, 95% CI 0.55-0.86; P < 0.001), PFS (HR 0.47, 95% CI 0.33-0.67; P < 0.001), and a lower risk of tumor recurrence (OR 0.43, 95% CI 0.30-0.61; P < 0.001). There was no significant difference in the incidence of complications (OR 1.11, 95% CI 0.75-1.65; P = 0.60) and treatment-related death (OR 1.15, 95% CI 0.31-4.30; P = 0.84) between the two treatments. CONCLUSIONS Current evidence shows esophagectomy has superior survival benefits as the initial treatment for clinical stage I EC. It is still the preferred choice for patients with clinical stage I EC. However, future high-quality randomized controlled trials are needed to validate this conclusion.
Collapse
Affiliation(s)
- Li-Xiang Mei
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun-Xian Mo
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yong Chen
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lei Dai
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yong-Yong Wang
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ming-Wu Chen
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
23
|
Cellini F, Manfrida S, Casà C, Romano A, Arcelli A, Zamagni A, De Luca V, Colloca GF, D’Aviero A, Fuccio L, Lancellotta V, Tagliaferri L, Boldrini L, Mattiucci GC, Gambacorta MA, Morganti AG, Valentini V. Modern Management of Esophageal Cancer: Radio-Oncology in Neoadjuvancy, Adjuvancy and Palliation. Cancers (Basel) 2022; 14:431. [PMID: 35053594 PMCID: PMC8773768 DOI: 10.3390/cancers14020431&n974851=v901586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The modern management of esophageal cancer is crucially based on a multidisciplinary and multimodal approach. Radiotherapy is involved in neoadjuvant and adjuvant settings; moreover, it includes radical and palliative treatment intention (with a focus on the use of a stent and its potential integration with radiotherapy). In this review, the above-mentioned settings and approaches will be described. Referring to available international guidelines, the background evidence bases will be reviewed, and the ongoing, more relevant trials will be outlined. Target definitions and radiotherapy doses to administer will be mentioned. Peculiar applications such as brachytherapy (interventional radiation oncology), and data regarding innovative approaches including MRI-guided-RT and radiomic analysis will be reported. A focus on the avoidance of surgery for major clinical responses (particularly for SCC) is detailed.
Collapse
Affiliation(s)
- Francesco Cellini
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Stefania Manfrida
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Calogero Casà
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Angela Romano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
- Correspondence:
| | - Alessandra Arcelli
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.A.); (A.Z.); (A.G.M.)
| | - Alice Zamagni
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.A.); (A.Z.); (A.G.M.)
| | - Viola De Luca
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Giuseppe Ferdinando Colloca
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Andrea D’Aviero
- Radiation Oncology, Mater Olbia Hospital, 07026 Olbia, Italy;
| | - Lorenzo Fuccio
- Department of Medical and Surgical Sciences, IRCSS—S. Orsola-Malpighi Hospital, 40138 Bologna, Italy;
| | - Valentina Lancellotta
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Luca Tagliaferri
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Gian Carlo Mattiucci
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Radiation Oncology, Mater Olbia Hospital, 07026 Olbia, Italy;
| | - Maria Antonietta Gambacorta
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Alessio Giuseppe Morganti
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.A.); (A.Z.); (A.G.M.)
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES), Alma Mater Studiorum, Bologna University, 40126 Bologna, Italy
| | - Vincenzo Valentini
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| |
Collapse
|
24
|
Cellini F, Manfrida S, Casà C, Romano A, Arcelli A, Zamagni A, De Luca V, Colloca GF, D’Aviero A, Fuccio L, Lancellotta V, Tagliaferri L, Boldrini L, Mattiucci GC, Gambacorta MA, Morganti AG, Valentini V. Modern Management of Esophageal Cancer: Radio-Oncology in Neoadjuvancy, Adjuvancy and Palliation. Cancers (Basel) 2022; 14:431. [PMID: 35053594 PMCID: PMC8773768 DOI: 10.3390/cancers14020431&n923648=v907986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The modern management of esophageal cancer is crucially based on a multidisciplinary and multimodal approach. Radiotherapy is involved in neoadjuvant and adjuvant settings; moreover, it includes radical and palliative treatment intention (with a focus on the use of a stent and its potential integration with radiotherapy). In this review, the above-mentioned settings and approaches will be described. Referring to available international guidelines, the background evidence bases will be reviewed, and the ongoing, more relevant trials will be outlined. Target definitions and radiotherapy doses to administer will be mentioned. Peculiar applications such as brachytherapy (interventional radiation oncology), and data regarding innovative approaches including MRI-guided-RT and radiomic analysis will be reported. A focus on the avoidance of surgery for major clinical responses (particularly for SCC) is detailed.
Collapse
Affiliation(s)
- Francesco Cellini
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Stefania Manfrida
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Calogero Casà
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Angela Romano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
- Correspondence:
| | - Alessandra Arcelli
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.A.); (A.Z.); (A.G.M.)
| | - Alice Zamagni
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.A.); (A.Z.); (A.G.M.)
| | - Viola De Luca
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Giuseppe Ferdinando Colloca
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Andrea D’Aviero
- Radiation Oncology, Mater Olbia Hospital, 07026 Olbia, Italy;
| | - Lorenzo Fuccio
- Department of Medical and Surgical Sciences, IRCSS—S. Orsola-Malpighi Hospital, 40138 Bologna, Italy;
| | - Valentina Lancellotta
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Luca Tagliaferri
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Gian Carlo Mattiucci
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Radiation Oncology, Mater Olbia Hospital, 07026 Olbia, Italy;
| | - Maria Antonietta Gambacorta
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Alessio Giuseppe Morganti
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.A.); (A.Z.); (A.G.M.)
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES), Alma Mater Studiorum, Bologna University, 40126 Bologna, Italy
| | - Vincenzo Valentini
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| |
Collapse
|
25
|
Cellini F, Manfrida S, Casà C, Romano A, Arcelli A, Zamagni A, De Luca V, Colloca GF, D’Aviero A, Fuccio L, Lancellotta V, Tagliaferri L, Boldrini L, Mattiucci GC, Gambacorta MA, Morganti AG, Valentini V. Modern Management of Esophageal Cancer: Radio-Oncology in Neoadjuvancy, Adjuvancy and Palliation. Cancers (Basel) 2022; 14:431. [PMID: 35053594 PMCID: PMC8773768 DOI: 10.3390/cancers14020431] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
The modern management of esophageal cancer is crucially based on a multidisciplinary and multimodal approach. Radiotherapy is involved in neoadjuvant and adjuvant settings; moreover, it includes radical and palliative treatment intention (with a focus on the use of a stent and its potential integration with radiotherapy). In this review, the above-mentioned settings and approaches will be described. Referring to available international guidelines, the background evidence bases will be reviewed, and the ongoing, more relevant trials will be outlined. Target definitions and radiotherapy doses to administer will be mentioned. Peculiar applications such as brachytherapy (interventional radiation oncology), and data regarding innovative approaches including MRI-guided-RT and radiomic analysis will be reported. A focus on the avoidance of surgery for major clinical responses (particularly for SCC) is detailed.
Collapse
Affiliation(s)
- Francesco Cellini
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Stefania Manfrida
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Calogero Casà
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Angela Romano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Alessandra Arcelli
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.A.); (A.Z.); (A.G.M.)
| | - Alice Zamagni
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.A.); (A.Z.); (A.G.M.)
| | - Viola De Luca
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Giuseppe Ferdinando Colloca
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Andrea D’Aviero
- Radiation Oncology, Mater Olbia Hospital, 07026 Olbia, Italy;
| | - Lorenzo Fuccio
- Department of Medical and Surgical Sciences, IRCSS—S. Orsola-Malpighi Hospital, 40138 Bologna, Italy;
| | - Valentina Lancellotta
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Luca Tagliaferri
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Gian Carlo Mattiucci
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Radiation Oncology, Mater Olbia Hospital, 07026 Olbia, Italy;
| | - Maria Antonietta Gambacorta
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| | - Alessio Giuseppe Morganti
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.A.); (A.Z.); (A.G.M.)
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES), Alma Mater Studiorum, Bologna University, 40126 Bologna, Italy
| | - Vincenzo Valentini
- Dipartimento Universitario Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (F.C.); (G.C.M.); (M.A.G.); (V.V.)
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.M.); (C.C.); (V.D.L.); (G.F.C.); (V.L.); (L.T.); (L.B.)
| |
Collapse
|
26
|
Wang S, Song M, Zhang B. Trichostatin A enhances radiosensitivity and radiation-induced DNA damage of esophageal cancer cells. J Gastrointest Oncol 2021; 12:1985-1995. [PMID: 34790366 DOI: 10.21037/jgo-21-560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/29/2021] [Indexed: 01/02/2023] Open
Abstract
Background Trichostatin A (TSA) is emerging as a potential component of anticancer therapy. In this study, we aimed to identify the radiosensitizing effects of TSA in esophageal squamous carcinoma cell lines and identify the genomic alteration of histone acetylation associated with TSA treatment. Methods EC109 and KYSE450 cells were pretreated with TSA (0.1 µM) for 12 hours prior to irradiation, and the cell viability, flow cytometry, and comet assays were performed to analyze cell growth, cell apoptosis, and DNA damage, respectively. Chromatin immunoprecipitation sequencing (ChIP-Seq) was performed to identify the acetylation sites of histone H3 lysine 9 (H3K9), which was altered by TSA. Results Our data showed that TSA could sensitize esophageal cancer cells to radiation by inducing cell cycle arrest and increasing cell apoptosis. DNA damage induced by radiation was enhanced by TSA treatment. In addition, a total of 105 differential peak-related genes were found to be associated with TSA treatment, which was identified using ChIP-Seq with specific antibodies against acetylated histone H3K9. Conclusions Our data suggest that pretreatment with TSA can enhance ionizing radiation-induced DNA damage of esophageal cancer cells, which was associated with the altered histone modification of whole genome. TSA has potential implications for clinical use in increasing the anticancer efficacy of radiation.
Collapse
Affiliation(s)
- Shaobo Wang
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Song
- Department of Neurology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Zhang
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
27
|
Kou J, Wang X, Wei Y, Zhao R, Wang X, He J, Li X, Wang X. Aurora kinase inhibitor VX-680 enhances sensitivity of esophageal squamous cell carcinoma cells to cisplatin chemotherapy. Anticancer Drugs 2021; 32:969-977. [PMID: 34016831 DOI: 10.1097/cad.0000000000001089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is malignant cancer with a high mortality rate. Cisplatin is one of the most potent chemotherapy agents used in the treatment of ESCC. However, chemoresistance and severe adverse effects of cisplatin become major obstacles to clinical utility. The combination treatment with molecule-targeted drugs and chemotherapy agents is a promising treatment strategy for cancer to improve antineoplastic responses. VX-680 is a potent inhibitor of Aurora kinases. This study was performed to investigate if VX-680 and cisplatin can synergistically inhibit the malignant behavior of ESCC cells. The results obtained from 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di- phenytetrazoliumromide assay and combination index analysis demonstrated that the combination of VX-680 and cisplatin synergistically enhanced cytotoxic effects in ESCC cells. 2-(4-Amidinophenyl)-6-indolecarbamidine dihydrochloride staining and western blot analysis suggested that VX-680 increased cisplatin-mediated cell apoptosis. Further analysis revealed that VX-680 combined with cisplatin could attenuate cell migration and angiogenesis confirmed by wound-healing assay and tube formation assay. Subsequently, VX-680 and cisplatin combined treatment significantly promoted cell-cell cohesion, and reduced cell-extracellular matrix interaction, as analyzed by the cell dissociation assay and cell-matrix attachment assay. In addition, the combination of VX-680 and cisplatin markedly decreased the expressions of matrix metalloproteinases-2 (MMP-2), vascular endothelial growth factor (VEGF), p-extracellular signal-regulated protein kinase and p-RAC-α serine/threonine-protein kinase compared to VX-680 or cisplatin only treatment. Altogether, these findings strongly suggest that the combination of VX-680 and cisplatin could exert a synergistic antitumor effect in ESCC cells and this combination might represent a promising therapeutic strategy against ESCC.
Collapse
Affiliation(s)
- Junting Kou
- Department of Biochemistry and Molecular Biology, Shanxi Medical University
| | - Xuewei Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University
| | - Yuan Wei
- Department of Biochemistry and Molecular Biology, Shanxi Medical University
| | - Rong Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University
| | - Xiuli Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University
| | - Jiefeng He
- Department of General Surgery, Shanxi Academy of Medical Sciences and Shanxi Bethune Hospital
| | - Xiaozhong Li
- Department of Infectious Diseases, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Xiaoxia Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University
| |
Collapse
|
28
|
Yang X, Han B, He Z, Zhang Y, Lin K, Su H, Hosseini DK, Sun H, Yang M, Chen X. RNA-Binding Proteins CLK1 and POP7 as Biomarkers for Diagnosis and Prognosis of Esophageal Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:715027. [PMID: 34568328 PMCID: PMC8458940 DOI: 10.3389/fcell.2021.715027] [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] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/09/2021] [Indexed: 12/24/2022] Open
Abstract
The abnormality of RNA-binding proteins (RBPs) is closely related to the tumorigenesis and development of esophageal squamous cell carcinoma (ESCC), and has been an area of interest for research recently. In this study, 162 tumors and 11 normal samples are obtained from The Cancer Genome Atlas database, among which 218 differentially expressed RBPs are screened. Finally, a prognostic model including seven RBPs (CLK1, DDX39A, EEF2, ELAC1, NKRF, POP7, and SMN1) is established. Further analysis reveals that the overall survival (OS) rate of the high-risk group is lower than that of the low-risk group. The area under the receiver operating characteristic (ROC) curve (AUC) of the training group and testing group is significant (AUCs of 3 years are 0.815 and 0.694, respectively, AUCs of 5 years are 0.737 and 0.725, respectively). In addition, a comprehensive analysis of seven identified RBPs shows that most RBPs are related to OS in patients with ESCC, among which EEF2 and ELCA1 are differentially expressed at the protein level of ESCC and control tissues. CLK1 and POP7 expressions in esophageal cancer tumor samples are undertaken using the tissue microarray, and show that CLK1 mRNA levels are relatively lower, and POP7 mRNA levels are higher compared with non-cancerous esophageal tissues. Survival analysis reveals that a higher expression of CLK1 predicts a significant worse prognosis, and a lower expression of POP7 predicts a worse prognosis in esophageal cancer. These results suggest that CLK1 may promote tumor progression, and POP7 may hinder the development of esophageal cancer. In addition, gene set enrichment analysis reveals that abnormal biological processes related to ribosomes and abnormalities in classic tumor signaling pathways such as TGF-β are important driving forces for the occurrence and development of ESCC. Our results provide new insights into the pathogenesis of ESCC, and seven RBPs have potential application value in the clinical prognosis prediction of ESCC.
Collapse
Affiliation(s)
- Xiuping Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Baoai Han
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zuhong He
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ya Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kun Lin
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongguo Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Davood K Hosseini
- Department of Internal Medicine, Hackensack University Medical Center, Hackensack, NJ, United States
| | - Haiying Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minlan Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiong Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
29
|
A commentary on "A meta-analysis on surgery with or without postoperative radiotherapy to treat squamous cell esophageal carcinoma" (Int J Surg 2020;80:184 -191). Int J Surg 2021; 88:105925. [PMID: 33746057 DOI: 10.1016/j.ijsu.2021.105925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 11/21/2022]
|
30
|
Han N, Zhang Y. A commentary on "A meta-analysis on surgery with or without postoperative radiotherapy to treat squamous cell esophageal carcinoma" [Int. J. Surg. 80 (2020) 184-191]. Int J Surg 2021; 88:105908. [PMID: 33676030 DOI: 10.1016/j.ijsu.2021.105908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Ning Han
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Liaoning, 110042, China
| | - Yaotian Zhang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Liaoning, 110042, China.
| |
Collapse
|
31
|
Abbasi AN, Qureshi BM. Commentary on '' A meta-analysis on surgery with or without postoperative radiotherapy to treat squamous cell esophageal carcinoma". Int J Surg 2020; 82:5. [PMID: 32822836 DOI: 10.1016/j.ijsu.2020.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 08/01/2020] [Indexed: 11/19/2022]
Affiliation(s)
- Ahmed Nadeem Abbasi
- Radiation Oncology Section, Department of Oncology, The Aga Khan University, Pakistan.
| | - Bilal Mazhar Qureshi
- Radiation Oncology Section, Department of Oncology, The Aga Khan University, Pakistan
| |
Collapse
|