501
|
Yoshikawa T, Muro K, Shitara K, Oh DY, Kang YK, Chung HC, Kudo T, Chin K, Kadowaki S, Hamamoto Y, Hironaka S, Yoshida K, Yen CJ, Omuro Y, Bai LY, Maeda K, Ozeki A, Yoshikawa R, Kitagawa Y. Effect of First-line S-1 Plus Oxaliplatin With or Without Ramucirumab Followed by Paclitaxel Plus Ramucirumab on Advanced Gastric Cancer in East Asia: The Phase 2 RAINSTORM Randomized Clinical Trial. JAMA Netw Open 2019; 2:e198243. [PMID: 31373648 PMCID: PMC6681552 DOI: 10.1001/jamanetworkopen.2019.8243] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/08/2019] [Indexed: 12/24/2022] Open
Abstract
Importance Ramucirumab, a human IgG 1 antibody against vascular endothelial growth factor receptor 2, has been shown to improve progression-free survival and overall survival in patients with advanced gastric cancer in the second-line setting. Objective To compare progression-free survival for S-1 and oxaliplatin plus ramucirumab with that for S-1 and oxaliplatin plus placebo in patients with advanced gastric cancer. Design, Setting, and Participants This phase 2, double-blind randomized clinical trial (RAINSTORM [First-line S-1 Plus Oxaliplatin With or Without Ramucirumab Followed by Paclitaxel Plus Ramucirumab in Patients With Advanced Gastric Cancer]) was conducted from October 12, 2015, to April 11, 2018, at 36 sites in Japan, South Korea, and Taiwan. Participants were chemotherapy-naive patients (n = 189) with metastatic gastric or gastroesophageal adenocarcinoma. Analyses of the full analysis set and safety population were conducted between November 27, 2017, and June 4, 2018. Interventions Patients randomized to the ramucirumab plus S-1 and oxaliplatin arm received S-1, 80 to 120 mg/d twice daily, on days 1 to 14 and oxaliplatin, 100 mg/m2, on day 1 with ramucirumab, 8 mg/kg, on days 1 and 8 in part A (21-day cycle). Patients randomized to the placebo plus S-1 and oxaliplatin arm received the same S-1 and oxaliplatin dosage as well as placebo on days 1 and 8 in part A. Eligible patients received second-line paclitaxel, 80 mg/m2, on days 1, 8, and 15 and ramucirumab, 8 mg/kg, on days 1 and 15 in part B (28-day cycle). Main Outcomes and Measures The primary end point was progression-free survival, analyzed using the stratified log-rank test; the hazard ratio (HR) was estimated using the stratified Cox proportional hazards regression model. Secondary end points included overall survival and adverse events. Results In total, 189 patients were randomized and received treatment: 96 to the ramucirumab plus S-1 and oxaliplatin arm and 93 to the placebo plus S-1 and oxaliplatin arm. Among the 189 patients, 121 (64.0%) were male, and the median (range) age was 62.0 (26-84) years. Median progression-free survival was not prolonged in the ramucirumab plus S-1 and oxaliplatin arm compared with the placebo plus S-1 and oxaliplatin arm (6.34 [80% CI, 5.65-6.93] vs 6.74 [80% CI, 5.75-7.13] months; HR, 1.07; 80% CI, 0.86-1.33; P = .70). Median overall survival was 14.65 (80% CI, 12.39-15.67) months in the ramucirumab plus S-1 and oxaliplatin arm and 14.26 (80% CI, 13.83-17.31) months in the placebo plus S-1 and oxaliplatin arm (HR, 1.11; 80% CI, 0.89-1.40; P = .55). The most commonly reported grade 3 or higher treatment-emergent adverse events in the ramucirumab plus S-1 and oxaliplatin arm in part A were decreased neutrophil count (14 patients [14.6%]), hypertension (10 patients [10.4%]), and anemia (10 patients [10.4%]). Conclusions and Relevance In this randomized clinical trial, the addition of ramucirumab to first-line S-1 and oxaliplatin treatment did not prolong progression-free survival or overall survival compared with S-1 and oxaliplatin alone among East Asian patients with advanced gastric cancer; no new safety signals for ramucirumab were identified. Trial Registration ClinicalTrials.gov identifier: NCT02539225.
Collapse
Affiliation(s)
- Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center Hospital, Yokohama, Japan
- currently affiliated with Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Hyun Cheol Chung
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Toshihiro Kudo
- Osaka University Graduate School of Medicine, Osaka, Japan
- currently affiliated with Osaka International Cancer Institute, Osaka, Japan
| | - Keisho Chin
- The Cancer Institute Hospital of the Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | | | - Shuichi Hironaka
- Chiba Cancer Center, Chiba, Japan
- currently affiliated with Oita University Faculty of Medicine, Oita, Japan
| | | | - Chia-Jui Yen
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yasushi Omuro
- Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Li-Yuan Bai
- Division of Hematology and Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | | | | | | | | |
Collapse
|
502
|
Koh J, Lee KW, Nam SK, Seo AN, Kim JW, Kim JW, Park DJ, Kim HH, Kim WH, Lee HS. Development and Validation of an Easy-to-Implement, Practical Algorithm for the Identification of Molecular Subtypes of Gastric Cancer: Prognostic and Therapeutic Implications. Oncologist 2019; 24:e1321-e1330. [PMID: 31371521 DOI: 10.1634/theoncologist.2019-0058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/10/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is a heterogeneous disease, and substantial efforts have been made to develop a molecular biology-based classification system for GC. Analysis of the genomic signature is not always feasible, and thus, we aimed to (i) develop and validate a practical immunohistochemistry (IHC)- and polymerase chain reaction (PCR)-based molecular classification of GC and (ii) to assess HER2 status according to this classification. MATERIALS AND METHODS A total of 894 consecutive patients with GC from two individual cohorts (training, n = 507; validation, n = 387) were classified using Epstein-Barr virus (EBV) in situ hybridization, microsatellite instability (MSI) testing, and IHC for E-cadherin and p53. RESULTS We were able to classify patients into five groups in the training cohort: group 1 (MSI+), group 2 (EBV-, MSI-, non-epithelial-mesenchymal transition [non-EMT]-like, p53-), group 3 (EBV+), group 4 (EBV-, MSI-, non-EMT-like, p53+), and group 5 (EBV-, MSI-, EMT-like). In the training cohort, each group showed different overall survival (OS) after gastrectomy (p < .001); group 1 had the best prognosis, and group 5 showed the worst survival outcome. The significant impact of the classification system on OS was also verified in the validation cohort (p = .004). HER2 positivity was observed in 6.5% of total population, and most of HER2-positive cases (93.1%) were included in groups 2 and 4. CONCLUSION We developed and validated a modified IHC- and PCR-based molecular classification system in GC, which showed significant impact on survival, irrespective of stage or other clinical variables. We also found close association between HER2 status and non-EMT phenotype in our classification system. IMPLICATIONS FOR PRACTICE Molecular classification of gastric cancer suggested by previous studies mostly relies on extensive genomic data analysis, which is not always available in daily practice. The authors developed a simplified immunohistochemistry- and polymerase chain reaction-based molecular classification of gastric cancer and proved the prognostic significance of this classification, as well as the close association between HER2 status and certain groups of the classification, in the largest consecutive cohort of gastric cancer. Results of this study suggest that this scheme is a cost-effective, easy-to-implement, and feasible way of classifying gastric cancer in daily clinical practice, also serving as a practical tool for aiding therapeutic decisions and predicting prognosis.
Collapse
Affiliation(s)
- Jiwon Koh
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soo Kyung Nam
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - An Na Seo
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ji-Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
503
|
GLI1 activation by non-classical pathway integrin α vβ 3/ERK1/2 maintains stem cell-like phenotype of multicellular aggregates in gastric cancer peritoneal metastasis. Cell Death Dis 2019; 10:574. [PMID: 31366904 PMCID: PMC6668446 DOI: 10.1038/s41419-019-1776-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022]
Abstract
Peritoneal metastasis is one of the most important causes of postoperative death in patients with gastric cancer, and the exact mechanism remains unclear. The proliferation of multicellular aggregates of exfoliated malignant gastric cells in the abdominal cavity is the focus of current research. However, the mechanism how gastric cancer multicellular aggregates survive remains unclear. In this study, we demonstrated that multicellular aggregates of exfoliated gastric cancer cells in the abdominal cavity expressed a stem cell-Like phenotype. We found that Integrin αvβ3 not only mediated adhesion of gastric cancer multicellular aggregates to form independent functional units, but also maintained their stem cell-like phenotype by the non-classical pathway Integrin αvβ3/ERK1/2/GLI1. In addition, ERK1/2 directly regulates the transcriptional activity of GLI1. GLI1 is a key effector of the Integrin αvβ3 pathway in regulating stem cell-like phenotype in multicellular aggregates. Our data indicates that although there is a crosstalk between the non-classical Integrin αvβ3 pathway and the classical Hedgehog pathway, the activation of GLI1 is almost independent of the Hedgehog pathway in multicellular aggregates of gastric cancer cells. Our study provides a basis for blocking GLI1 activity in the prevention and treatment of peritoneal metastases of gastric cancer.
Collapse
|
504
|
Li J, Xi H, Guo X, Gao Y, Xie T, Qiao Z, Chen L. Surgical outcomes and learning curve analysis of robotic gastrectomy for gastric cancer: Multidimensional analysis compared with three‑dimensional high‑definition laparoscopic gastrectomy. Int J Oncol 2019; 55:733-744. [PMID: 31364736 DOI: 10.3892/ijo.2019.4851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/04/2019] [Indexed: 11/06/2022] Open
Abstract
The present ambispective cohort study was performed to compare the short‑term surgical outcomes, including financial cost and surgeons' acceptance, of robotic versus three‑dimensional high‑definition (3D‑HD) laparoscopic gastrectomy for patients with gastric cancer (GC). Between 2011 and 2017, 517 patients with GC were enrolled for treatment with either robotic gastrectomy [408 patients, including 73 treated by one of the authors (LC)] or 3D‑HD laparoscopic gastrectomy (109 patients, including 71 treated by LC). The cumulative summation method was developed to analyze the learning curves of robotic and 3D‑HD laparoscopic gastrectomy performed by LC. In the analysis of all 517 patients, there were no significant differences in the clinicopathological characteristics between the two treatment groups, with the exception of smoking status (P<0.001). The robotic group had a shorter operative time (OT; 209 vs. 228 min, P=0.004), fewer postoperative days (PODs) to first flatus (3 vs. 4 days, P=0.025), more PODs to removal of the drainage and nasogastric tubes (12 vs. 9 days, P=0.001; 6 vs. 4 days, P=0.001, respectively), and more postoperative complications (21.3 vs. 9.2%, P=0.003). Comparison of these short‑term outcomes of robotic and 3D‑HD laparoscopic gastrectomy performed by LC (144 patients) revealed that only the number of retrieved lymph nodes (27 in the robotic group vs. 33 in the 3D‑HD group; P=0.038) and PODs to removal of the nasogastric tube (5 days in the robotic group vs. 3 days in the 3D‑HD group; P<0.001) were significantly different. The OT stabilized after around 21 robotic gastrectomy procedures and 19 3D‑HD laparoscopic gastrectomy procedures. The cost‑effectiveness analysis revealed that robotic gastrectomy had a significantly higher total cost than 3D‑HD laparoscopic gastrectomy (124,907 vs. 94,395 RMB, P<0.001). With comparable surgical outcomes, lower financial cost and higher surgeons' acceptance, 3D‑HD laparoscopic gastrectomy is highly recommended as a minimally invasive surgical method for patients with GC prior to the popularization of robotic surgery.
Collapse
Affiliation(s)
- Jiyang Li
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Hongqing Xi
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Xin Guo
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Yunhe Gao
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Tianyu Xie
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Zhi Qiao
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Lin Chen
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
505
|
Wang ZK, Lin JX, Li P, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Tu RH, Huang CM, Zheng CH. Higher Risk of Lymph Node Metastasis in Young Patients with Early Gastric Cancer. J Cancer 2019; 10:4389-4396. [PMID: 31413759 PMCID: PMC6691700 DOI: 10.7150/jca.30260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/14/2019] [Indexed: 12/11/2022] Open
Abstract
Objective: Whether age affects lymph node metastasis (LNM) in patients with gastric cancer (GC) is currently inconclusive. This study investigates the effect of age on LNM in patients with GC. Methods: From January 1988 to December 2013, 22,808 GC patients underwent gastrectomy at the Surveillance, Epidemiology, and End Results database were included. The relationship between age and LNM was analyzed. Results: The median number of examined lymph nodes (ELNs) was 12 (interquartile range [IQR], 7-20) among the 22,808 patients with GC, and the median numbers of ELNs were 10 (IQR, 5-18), 12 (IQR, 6-19), 13 (IQR, 7-21) and 13 (IQR, 7-21) in patients with T1 to T4 disease, respectively. A total of 13,780 (60.4%) patients presented with LNM. The LNM rates were 69.6%, 66.1%, 64.7%, 61.8%, 57.8% and 55.6% for patients in the 20-39, 40-49, 50-59, 60-69, 70-79 and ≥ 80 age groups, respectively (P < 0.001). The LNM rates and the number of positive lymph nodes were correlated with age among patients whose diseases were of the same T stage (all P < 0.01). Multivariate analysis showed that age was an independent predictor for LNM in patients with early gastric cancer (EGC) (P < 0.05), and linear regression analysis showed that the LNM rate was higher in young patients with EGC (P < 0.05). Conclusions: Age is an independent predictor for LNM in EGC. Moreover, LNM is more common in young patients with EGC than in other age groups, which indicates that limited lymph node dissection may not be appropriate for young patients with EGC.
Collapse
Affiliation(s)
- Zu-Kai Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
506
|
Yu C, Zhang Y. Characterization of the prognostic values of CXCR family in gastric cancer. Cytokine 2019; 123:154785. [PMID: 31344595 DOI: 10.1016/j.cyto.2019.154785] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/07/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The role of CXC chemokine receptors (CXCRs) in gastric cancer (GC) has been an increasing focus. However, comprehensive prognostic values of CXCR members in GC are yet to be clearly defined. METHODS Multiple public available datasets, including Kaplan-Meier (KM) plotter, oncomine, the cancer genome atlas (TCGA), SurvExpress platform and the tumor immune estimation resource (TIMER), were used for mRNA expression and prognostic characterization. Nomogram method was used for clinical model prediction. RESULTS CXCR3, CXCR4 and CXCR5 displayed significantly up-regulated expression in tumor compared to normal. High mRNA expression of CXCR2 (HR = 0.77, 95%CI: 0.62-0.95, p = 0.014), CXCR3 (HR = 0.74, 95%CI: 0.61-0.90, p = 0.0024), CXCR4 (HR = 0.7, 95%CI: 0.58-0.86, p = 0.00048), CXCR5 (HR = 0.72, 95%CI: 0.59-0.87, p = 0.00093) and CXCR6 (HR = 0.66, 95%CI: 0.54-0.81, p = 4.9e-05) was significantly associated with favorable overall survival (OS). The prognostic values of CXCR members were also explored in subtypes, including HER2 status, Lauren classification, pathological stages. The low risk group of CXCR signature displayed a significantly favorable OS compared to the high risk group (HR = 3.22, 95% CI = 2.21-4.69, p = 1.057e-09). Nomogram clinical models were established for both OS (C-index: 0.692; 95%CI: 0.648-0.736) and recurrence free survival (C-index: 0.731; 95%CI: 0.675-0.786). In addition, CXCR6 and CD8+T cells featured the highest correlation (partial-cor = 0.781, p = 4.17e-77). CONCLUSION This study identified distinct expression and prognostic values of CXCR members in GC using public databases.
Collapse
Affiliation(s)
- Chaoran Yu
- Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200025, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200025, PR China; Department of Gastrointestinal Surgery, Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 20025, PR China.
| | - Yujie Zhang
- Department of Gastrointestinal Surgery, Tongji Hospital, Tongji Medical College in Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
507
|
Yu C, Hao X, Zhang S, Hu W, Li J, Sun J, Zheng M. Characterization of the prognostic values of the NDRG family in gastric cancer. Therap Adv Gastroenterol 2019; 12:1756284819858507. [PMID: 31384305 PMCID: PMC6647212 DOI: 10.1177/1756284819858507] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 05/07/2019] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The N-myc downstream-regulated gene (NDRG) family, NDRG1-4, has been involved in a wide spectrum of biological functions in multiple cancers. However, their prognostic values remain sparse in gastric cancer (GC). Therefore, it is crucial to systematically investigate the prognostic values of the NDRG family in GC. METHODS The prognostic values of the NDRG family were evaluated by Kaplan-Meier Plotter and SurvExpress. The mRNA of the NDRG family was investigated in The Cancer Genome Atlas (TCGA). Transcription factors (TFs) and miRNAs associated with the NDRG family were predicted by NetworkAnalysis. The prognostic values of DNA methylation levels were analyzed by MethSurv. The correlation between immune cells and the NDRG family was evaluated by the Tumor Immune Estimation Resource (TIMER) database. RESULTS High levels of mRNA expression of NDRG2 and NDRG3 were associated with a favorable prognosis in all GCs. In HER2 - GC, NDRG1 was significantly associated with a poor prognosis of GC [hazard ratio (HR) = 1.65, 95% confidence interval (CI) = 1.16-2.33, p = 0.0046]. In HER2 + GC, NDRG4 showed a poor prognosis (HR = 1.4, 95% CI: 1.06-1.85, p = 0.017). NDRG4 was an independent prognostic factor in recurrence-free survival by TCGA cohort. The low-risk NDRG-signature group displayed a significantly favorable survival outcome than the high-risk group (HR = 1.76, 95% CI: 1.2-2.59, p = 0.00385). The phosphorylated protein NDRG1 (NDRG1_pT346) displayed a favorable overall survival and was significantly associated with HER2 and phosphorylated HER2. Epidermis development was the top biological process (BP) for coexpressed genes associated with NDRG1 and NDRG4, while mitotic nuclear division and mitotic cell processes were the top BPs for NDRG2 and NDRG3, respectively. Overall, 6 CpGs of NDRG1, 4 CpGs of NDRG2, 3 CpGs of NDRG3 and 24 CpGs of NDRG4 were associated with significant prognosis. CD4+ T-cells showed the highest correlation with NDRG4 (correlation = 0.341, p = 2.14e-11). Furthermore, BCL6 in follicular helper T-cells (Tfh) cells showed the highest association with NDRG4 (correlation = 0.438, p = 00e+00). CONCLUSIONS This study analyzed the multilevel prognostic values and biological roles of the NDRG family in GC.
Collapse
Affiliation(s)
- Chaoran Yu
- Department of Gastrointestinal Surgery, Ruijin
Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai,
China
- Shanghai Minimally Invasive Surgery Center,
Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine,
Shanghai, China
| | - Xiaohui Hao
- Department of Gastrointestinal Surgery, Ruijin
Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai,
China
- Shanghai Minimally Invasive Surgery Center,
Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine,
Shanghai, China
| | - Sen Zhang
- Department of Gastrointestinal Surgery, Ruijin
Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai,
China
- Shanghai Minimally Invasive Surgery Center,
Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine,
Shanghai, China
| | - Wenjun Hu
- Department of Gastrointestinal Surgery, Ruijin
Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai,
China
- Shanghai Minimally Invasive Surgery Center,
Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine,
Shanghai, China
| | - Jianwen Li
- Department of Gastrointestinal Surgery, Ruijin
Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai,
China
- Shanghai Minimally Invasive Surgery Center,
Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine,
Shanghai, China
| | - Jing Sun
- Department of Gastrointestinal Surgery, Ruijin
Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai,
China
- Shanghai Minimally Invasive Surgery Center,
Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine,
Shanghai, China
| | | |
Collapse
|
508
|
Wang SB, Qi WX, Chen JY, Xu C, Kirova YM, Cao WG, Cai R, Cao L, Yan M, Cai G. Competing risk nomogram predicting initial loco-regional recurrence in gastric cancer patients after D2 gastrectomy. Radiat Oncol 2019; 14:128. [PMID: 31315683 PMCID: PMC6637492 DOI: 10.1186/s13014-019-1332-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
Background Lacking quantitative evaluations of clinicopathological features and the risk factors for loco-regional recurrence (LRR) in gastric cancer after D2 gastrectomy, we aimed to develop a competing risk nomogram to identify the risk predictors for initial LRR. Methods We retrospectively analysed 1105 patients who underwent radical gastrectomy with D2 resection for stage I-III gastric cancer. A nomogram predicting initial LRR of gastric cancer was conducted based on Fine and Grey’s competing risk analysis. The predictive accuracy and discriminative ability of the model were determined using the concordance index (C-index) and calibration curve. Decision tree analysis was performed for patient grouping. Results At a median follow-up of 28.4 months, 274 patients developed 373 first recurrence events (local, regional, and distant disease). The median recurrence-free survival (RFS) was 16.7 months. Multivariate competing risk analysis showed that age (SHR, 1.72; 95% CI, 1.10–2.83, p = 0.031), CEA (SHR, 1.94; 95% CI, 1.09–3.46, p = 0.024), pT4 (SHR, 2.77; 95% CI, 1.01–7.57, p = 0.047), lymph node metastasis (SHR 1.92, 95% CI: 1.09–3.38, p = 0.024) and LVI (SHR, 1.84; 95% CI, 1.06–3.20, p = 0.028) were independent risk factors for LRR (all p < 0.05). The nomogram incorporating these factors achieved good agreement between prediction and actual observation with a concordance index of 0.738 (95% CI, 0.767 to 0.709). In a subgroup analysis of node-positive patients, pN3b was associated with increased peritoneal and distant metastasis (p = 0.048). The para-aortic lymph nodes were the most frequent sites (n = 71) of LRR, and among them, the 16a2 and 16b1 nodes exhibited even more prevalence (90.1 and 81.7%). Conclusions Adjuvant radiotherapy might be recommended in gastric cancer patients ≥65 years old or those with pN+, pT4, LVI, or increased CEA levels, particularly in high-risk or pN1-3a patients. The competing risk nomograms may be considered as convenient and individualized predictive tools for LRR in gastric cancer after D2 gastrectomy. It is also recommended that the clinical target volume (CTV) include 16a2 and 16b1 regions of para-aortic lymph nodes.
Collapse
Affiliation(s)
- Shu-Bei Wang
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Wei-Xiang Qi
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Jia-Yi Chen
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Cheng Xu
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Youlia M Kirova
- Department of Radiation Oncology, Institute Curie, Paris, France
| | - Wei-Guo Cao
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Rong Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Lu Cao
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Min Yan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Gang Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Rui Jin Er Road, Shanghai, 200025, China.
| |
Collapse
|
509
|
Zhang D, Wu JR, Duan XJ, Wang KH, Zhao Y, Ni MW, Liu SY, Zhang XM, Zhang B. A Bayesian Network Meta-Analysis for Identifying the Optimal Taxane-Based Chemotherapy Regimens for Treating Gastric Cancer. Front Pharmacol 2019; 10:717. [PMID: 31333452 PMCID: PMC6624233 DOI: 10.3389/fphar.2019.00717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 06/05/2019] [Indexed: 01/30/2023] Open
Abstract
Background: Several taxane-based chemotherapy regimens are effective in the treatment of gastric cancer; nevertheless, their comparative efficacy and safety remain disputed. This network meta-analysis (NMA) was designed to compare the efficacy and safety of different taxane-based chemotherapy regimens against gastric cancer. Methods: A comprehensive search was conducted to identify all relevant randomized controlled trials (RCTs) in multiple electronic databases. A Bayesian NMA was performed to combine the direct and indirect evidence and estimate the comparative efficacy and safety of different taxane-based chemotherapy regimens simultaneously by utilizing WinBUGS 1.4.3 and Stata 13.1 software. The efficacy outcomes included overall survival rate (OS), progression-free survival (PFS), and overall response rate (ORR), and the safety outcomes were adverse reactions (ADRs), namely, neutropenia, leucopenia, vomiting, and fatigue. Results: A total of 37 RCTs were identified involving 7,178 patients with gastric cancer, and 10 taxane-based chemotherapy regimens (RT, T, TC, TCF, TF, TO, TOF, mTCF, mTF, and mTOF) were collected in gastric cancer therapy. According to the results of cluster analysis, compared with other taxane-based chemotherapy regimens, the regimens of TOF, mTCF, and TF were associated with the most favorable clinical efficacy in improving OS, PFS, and ORR. On the other hand, the regimens of T and mTF had the potential to be the most tolerable and acceptable therapeutic alternative in terms of ADRs. Conclusions: The current NMA provides the evidence that the combination of taxanes (paclitaxel or docetaxel) and fluorouracil is associated with the most preferable and beneficial option for patients with gastric cancer, although additional results from multicenter trials and high-quality studies will be pivotal for supporting our findings.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jia-Rui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Jiao Duan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Kai-Huan Wang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhao
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Meng-Wei Ni
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shu-Yu Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Meng Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Bing Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
510
|
Courtois S, Lehours P, Bessède E. The therapeutic potential of metformin in gastric cancer. Gastric Cancer 2019; 22:653-662. [PMID: 30900101 DOI: 10.1007/s10120-019-00952-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/11/2019] [Indexed: 02/07/2023]
Abstract
Metformin is a biguanide molecule used since 1957 to treat type 2 diabetes patients. In addition to its hypoglycemic effects, epidemiological studies have shown that metformin can be associated with a decrease in cancer development risk in diabetic populations. Thus, since 2005 this molecule is largely studied for its antitumoural properties in different types of cancer. The potential antitumoural effect of metformin in gastric cancer has been poorly studied. Here, we detailed the different described mechanisms implicated in the antitumoural effect of metformin in gastric cancer, from the signalling pathways to the functional effects on gastric cancer cell lines and gastric cancer stem cells.
Collapse
Affiliation(s)
- Sarah Courtois
- INSERM, Univ. Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, 33000, Bordeaux, France.
| | - Philippe Lehours
- INSERM, Univ. Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, 33000, Bordeaux, France.,French National Reference Center for Campylobacters and Helicobacters in Bordeaux (CNRCH), University Hospital of Bordeaux, Bordeaux, France
| | - Emilie Bessède
- INSERM, Univ. Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, 33000, Bordeaux, France.,French National Reference Center for Campylobacters and Helicobacters in Bordeaux (CNRCH), University Hospital of Bordeaux, Bordeaux, France
| |
Collapse
|
511
|
Li L, Guo LY, Mao J. A network meta-analysis protocol of adjuvant chemotherapy for unresectable patients with advanced gastric cancer. Medicine (Baltimore) 2019; 98:e16108. [PMID: 31305394 PMCID: PMC6641784 DOI: 10.1097/md.0000000000016108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The treatment methods about surgery, chemotherapy, and radiation therapy were recommended for gastric cancer (GC) patients by National Comprehensive Cancer Network (NCCN) guidelines. However, for the advanced gastric cancer patients or with metastatic lesions who have lost their chance of surgery, the current adjuvant chemotherapy treatments are still controversial. Therefore, this network meta-analysis is mainly to assess the relative efficacy of different adjuvant chemotherapy regimens for advanced gastric cancer (AGC). METHODS In order to compare the relative efficacy among different adjuvant chemotherapy regimens for AGC patients, randomized controlled trials (RCTs) and non-RCTs were systematic searched in PubMed, Web of Science, Clinical Trials, Cochrane Library and Embase database. R-3.4.1 software will be used for data analysis. The risk of bias in RCTs and non-RCTs will be evaluated through the risk of bias tool from the Cochrane Handbook version 5.1.0 and non-randomized studies of interventions (ROBINS-I), respectively. RESULTS AND CONCLUSION The results of this network meta-analysis will evaluate the relative effectiveness and rank the interventions among all chemotherapy methods for unresectable AGC patients, and provide more evidence-based guidance in clinical practice. PROTOCOL REGISTRATION NUMBER CRD42018111835.
Collapse
|
512
|
Huang L, Liu H, Yu J, Lin T, Hu YF, Li TJ, Li GX. Long-Term Outcomes in Laparoscopic D2 Gastrectomy for Gastric Cancer: a Large Comprehensive Study Proposing Novel Hypotheses. J Gastrointest Surg 2019; 23:1349-1361. [PMID: 30478532 DOI: 10.1007/s11605-018-4008-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/09/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND The long-term outcomes of laparoscopic gastrectomy (LG) versus open gastrectomy (OG) for gastric cancer (GC) remain obscure, especially for advanced cancer and disease affecting the upper stomach and in older patients. This study aimed to comprehensively assess the long-term efficacy of LG for GC using a large prospective database. METHODS Totally, 1877 consecutive patients (1186 receiving LG and 691 OG) operated in 2004-2016 were analyzed, with a median follow-up of 63 months. Association of LG versus OG with disease-specific survival (DSS) and disease-free survival (DFS) overall and in various subgroups were investigated using multivariable Cox regression. Propensity score matching (PSM) was performed for sensitivity analysis. RESULTS Before PSM, overall, there was no significant association of LG versus OG with survival after multivariable adjustment; however, in subgroup analyses, LG was associated with superior DSS in patients aged ≥ 70 years and those with upper GC. No significant associations regarding DFS were observed overall or in stratifications. PSM analyses revealed that LG was associated with better DSS also in patients aged ≥ 70 years (hazard ratio (HR) = 0.33, 95% confidence interval (CI) = 0.15-0.72) and in those with upper GC (HR = 0.51, 95% CI = 0.29-0.91), and with better DFS in those with upper GC (HR = 0.60, 95% CI = 0.37-0.99). Multivariable analysis showed that age, hepatitis B, performance status, tumor histology, stage, and vascular invasion were significantly associated with post-LG survival. LG-specific nomograms were then constructed with concordance indexes of 0.814 (DSS) and 0.809 (DFS) and excellent calibration. CONCLUSIONS In this large institutional analysis, while LG for GC was associated with DSS and DFS similar to those for OG overall, non-inferior LG-associated survival especially DSS was observed in some subgroups rarely investigated in prospective or randomized settings. There could still be biases even after PSM due to confounders not accounted for in this observational study. However, these findings offer novel hypotheses for further validation.
Collapse
Affiliation(s)
- Lei Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Tian Lin
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Yan-Feng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Tuan-Jie Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| | - Guo-Xin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
513
|
Cleary JM, Horick NK, McCleary NJ, Abrams TA, Yurgelun MB, Azzoli CG, Rubinson DA, Brooks GA, Chan JA, Blaszkowsky LS, Clark JW, Goyal L, Meyerhardt JA, Ng K, Schrag D, Savarese DM, Graham C, Fitzpatrick B, Gibb KA, Boucher Y, Duda DG, Jain RK, Fuchs CS, Enzinger PC. FOLFOX plus ziv-aflibercept or placebo in first-line metastatic esophagogastric adenocarcinoma: A double-blind, randomized, multicenter phase 2 trial. Cancer 2019; 125:2213-2221. [PMID: 30913304 PMCID: PMC6763367 DOI: 10.1002/cncr.32029] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/06/2018] [Accepted: 01/10/2019] [Indexed: 02/04/2023]
Abstract
BACKGROUND Antiangiogenic therapy is a proven therapeutic modality for refractory gastric and gastroesophageal junction adenocarcinoma. This trial assessed whether the addition of a high affinity angiogenesis inhibitor, ziv-aflibercept, could improve the efficacy of first-line mFOLFOX6 (oxaliplatin, leucovorin, and bolus plus infusional 5- fluorouracil) in metastatic esophagogastric adenocarcinoma. METHODS Patients with treatment-naive metastatic esophagogastric adenocarcinoma were randomly assigned (in a 2:1 ratio) in a multicenter, placebo-controlled, double-blind trial to receive first-line mFOLFOX6 with or without ziv-aflibercept (4 mg/kg) every 2 weeks. The primary endpoint was 6-month progression-free survival (PFS). RESULTS Sixty-four patients were randomized to receive mFOLFOX6 and ziv-aflibercept (43 patients) or mFOLFOX6 and a placebo (21 patients). There was no difference in the PFS, overall survival, or response rate. Patients treated with mFOLFOX6/ziv-aflibercept tended to be more likely to discontinue study treatment for reasons other than progressive disease (P = .06). The relative dose intensity of oxaliplatin and 5-fluorouracil was lower in the mFOLFOX6/ziv-aflibercept arm during the first 12 and 24 weeks of the trial. There were 2 treatment-related deaths due to cerebral hemorrhage and bowel perforation in the mFOLFOX6/ziv-aflibercept cohort. CONCLUSIONS Ziv-aflibercept did not increase the anti-tumor activity of first-line mFOLFOX6 in metastatic esophagogastric cancer, potentially because of decreased dose intensity of FOLFOX. Further evaluation of ziv-aflibercept in unselected, chemotherapy-naive patients with metastatic esophagogastric adenocarcinoma is not warranted.
Collapse
Affiliation(s)
- James M. Cleary
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Nora K. Horick
- Biostatistics Center, Massachusetts General Hospital, Boston, Massachusetts
| | - Nadine Jackson McCleary
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Thomas A. Abrams
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Matthew B. Yurgelun
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Christopher G. Azzoli
- Steele Laboratories for Tumor Biology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Douglas A. Rubinson
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Gabriel A. Brooks
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Jennifer A. Chan
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | | | - Jeffrey W. Clark
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Lipika Goyal
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey A. Meyerhardt
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Kimmie Ng
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Deborah Schrag
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Diane M.F. Savarese
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Christopher Graham
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Bridget Fitzpatrick
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Kathryn A. Gibb
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Yves Boucher
- Steele Laboratories for Tumor Biology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Dan G. Duda
- Steele Laboratories for Tumor Biology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Rakesh K. Jain
- Steele Laboratories for Tumor Biology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | | | - Peter C. Enzinger
- Center for Esophageal and Gastric Cancer, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Division of Gastrointestinal Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
514
|
Chen AH, Chan WH, Lee YH, Tseng JH, Yeh TS, Chiu CT, Chen JS, Chen CM. Routine chest CT for staging of gastric cancer. Br J Surg 2019; 106:1197-1203. [PMID: 31210362 PMCID: PMC6771689 DOI: 10.1002/bjs.11186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/18/2019] [Accepted: 03/04/2019] [Indexed: 12/21/2022]
Abstract
Background International guidelines on clinical staging of gastric cancer recommend the use of chest CT for the detection of pulmonary metastases. This study assessed the clinical value of routine chest CT in the staging of gastric cancer. Methods This retrospective study included patients identified from the gastric cancer registry of Chang Gung Memorial Hospital, Linkou, Taiwan. All patients who underwent clinical staging between 2008 and 2014 were included. The pattern, site and number of metastases at initial presentation and after surgery with curative intent were evaluated. Pulmonary metastases were defined as multiple small round pulmonary nodules with a random distribution or of variable size. Results Some 1669 patients were included, of whom 478 (28·6 per cent) had metastatic disease at clinical presentation. The majority of metastases were to the peritoneum (75·7 per cent of patients) or liver (30·5 per cent), and only 27 patients (5·6 per cent) had pulmonary metastases at presentation, none of which were isolated to the lung. Of these 27 patients, 11 had primary lesions located at the cardia/fundus. In 19 patients the lung metastases were also detected on the staging chest X‐ray. After surgery there were 196 cancer recurrences. Some 15 patients (7·6 per cent) had lung metastasis and this was not the only site of metastases in any patient. The prevalence of lung metastasis at presentation of the disease and after surgery was 1·6 and 1·5 per cent respectively. Conclusion This study does not support the routine use of chest CT for staging of gastric cancer as isolated pulmonary metastasis in the absence of other metastatic sites could not be detected.
Collapse
Affiliation(s)
- A-H Chen
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - W-H Chan
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Y-H Lee
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - J-H Tseng
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - T-S Yeh
- Department of Surgery, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - C-T Chiu
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - J-S Chen
- Division of Haematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - C-M Chen
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
515
|
Rawicz-Pruszyński K, Mielko J, Ciseł B, Skórzewska M, Pikuła A, Gęca K, Skoczylas T, Kubiatowski T, Kurylcio A, Polkowski WP. Blast from the past: Perioperative use of the Maruyama computer program for prediction of lymph node involvement in the surgical treatment of gastric cancer following neoadjuvant chemotherapy. Eur J Surg Oncol 2019; 45:1957-1963. [PMID: 31178298 DOI: 10.1016/j.ejso.2019.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/13/2019] [Accepted: 06/01/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Surgical quality assurance is a key element of gastric cancer treatment. The Maruyama Computer Program (MCP) allows to predict lymph node involvement in stations no. 1-16. The aim of the current study was to evaluate the accuracy of the MCP predictions in GC patients treated with neoadjuvant chemotherapy (nCTH) followed by gastrectomy with adequate lymphadenectomy. METHODS 101 patients who underwent preoperative nCTH followed by D2 gastrectomy with curative intent were analysed. The response to nCTH was measured using the tumour regression grade system. RESULTS Test sensitivity, specificity, PPV, NPV and accuracy of the MCP were 92%, 33%, 41%, 89%, and 53%, respectively. In patients with response to nCTH, number of false positive (FP) results was significantly higher than in patients who did not respond to nCTH both in the N1 (56.3% vs 28.9%, p < 0.0001) and in the N2 (59% vs 41%, p < 0.0001) trier. The risk for FP results was 6 times higher in N1 (OR = 6.50, 95%CI: 3.91-10.82,; p < 0.0001) and N2 (OR = 5.84, 95%CI: 2.85-11.96; p < 0.0001) triers. In patients with intestinal type GC, the risk for FP results was 4 times higher than in other histologic types of GC in both N1 (OR = 4.23, 95%CI: 2.58-6.95; p < 0.0001) and N2 (OR = 4.23, 95%CI: 2.02-9.62; p = 0.0002) triers. CONCLUSIONS MCP predictions in the GC patients treated with nCTH have low specificity due to significantly high number of FP results. Noticeably low accuracy level of predictions indicate a need for new prediction models, based on Laurén classification, since it may provide some information on expected regression grade.
Collapse
Affiliation(s)
- Karol Rawicz-Pruszyński
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłowska 13 St., 20-080, Lublin, Poland.
| | - Jerzy Mielko
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłowska 13 St., 20-080, Lublin, Poland.
| | - Bogumiła Ciseł
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłowska 13 St., 20-080, Lublin, Poland.
| | - Magdalena Skórzewska
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłowska 13 St., 20-080, Lublin, Poland.
| | - Agnieszka Pikuła
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłowska 13 St., 20-080, Lublin, Poland.
| | - Katarzyna Gęca
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłowska 13 St., 20-080, Lublin, Poland.
| | - Tomasz Skoczylas
- 2nd Department and Clinic of General, Gastroenterological and Gastrointestinal Cancer Surgery, Medical University of Lublin, Staszica 16 St., 20-081, Lublin, Poland.
| | - Tomasz Kubiatowski
- Department of Clinical Oncology, St. John of Dukla Lublin Region Cancer Center, Jaczewskiego 7 St., 20-090, Lublin, Poland.
| | - Andrzej Kurylcio
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłowska 13 St., 20-080, Lublin, Poland.
| | - Wojciech Piotr Polkowski
- Department of Surgical Oncology, Medical University of Lublin, Radziwiłowska 13 St., 20-080, Lublin, Poland.
| |
Collapse
|
516
|
Ham IH, Lee D, Hur H. Role of Cancer-Associated Fibroblast in Gastric Cancer Progression and Resistance to Treatments. JOURNAL OF ONCOLOGY 2019; 2019:6270784. [PMID: 31281359 PMCID: PMC6590541 DOI: 10.1155/2019/6270784] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/23/2019] [Indexed: 12/21/2022]
Abstract
Although the survival of gastric cancer (GC) patients has gradually improved, the outcomes of advanced GC patients remain unsatisfactory despite standard treatment with conventional chemotherapy or targeted agents. Several studies have shown that cancer-associated fibroblasts (CAFs), a major component of tumor stroma in GC, may have significant roles in GC progression and resistance to treatments. CAFs are a major source of various secreted molecules in the tumor microenvironment, which stimulate cancer cells and other noncancerous components of GC. Surprisingly, these factors could be involved in gastric carcinogenesis. Cytokines, including interleukin-6 and interleukin-11, or growth factors, such as fibroblast growth factor produced from CAFs, can directly activate GC cells and consequently lead to the development of an aggressive phenotype. Galectin-1 or hepatocyte growth factor can be involved in CAF-derived neovascularization in GC. In addition, recent studies showed that CAFs can affect tumor immunity through M2 polarization of tumor-associated macrophages. Finally, the current study aimed to introduce several inhibitory agents and evaluate their suppressive effects on CAFs in patients with GC progression. However, further studies are required to evaluate their safety and select appropriate patients for application in clinical settings.
Collapse
Affiliation(s)
- In-Hye Ham
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dagyeong Lee
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Brain Korea 21 Plus Research Center for Biomedical Sciences, Ajou University, Suwon, Republic of Korea
- Department of Biomedical Science, Graduated School of Ajou University, Suwon, Republic of Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Brain Korea 21 Plus Research Center for Biomedical Sciences, Ajou University, Suwon, Republic of Korea
- Department of Biomedical Science, Graduated School of Ajou University, Suwon, Republic of Korea
| |
Collapse
|
517
|
Sun Z, Zheng H, Yu J, Huang W, Li T, Chen H, Hu Y, Zhao M, Liu H, Jiang Y, Li G. Liver Metastases in Newly Diagnosed Gastric Cancer: A Population-Based Study from SEER. J Cancer 2019; 10:2991-3005. [PMID: 31281476 PMCID: PMC6590027 DOI: 10.7150/jca.30821] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Purpose: Population-based data on the proportion and prognosis of liver metastases at diagnosis of gastric cancer are currently lacking. Besides, the treatment of gastric cancer with liver metastases is still controversial now. Methods: Patients with gastric cancer and liver metastases (GCLM) at the time of diagnosis in advanced gastric cancer were identified using the Surveillance, Epidemiology, and End Result (SEER) database of the National Cancer Institute. Multivariable logistic and Cox regression were performed to identify predictors of the presence of GCLM at diagnosis and factors associated with all-cause mortality. Results: We identified 3507 patients with gastric cancer and liver metastases at the time of diagnosis, representing 16.89% of the entire cohort and 44.12% of the subset with metastatic disease to any distant site. Among entire cohort, multivariable logistic regression identified thirteen factors (age, race, sex, original, tumor location, pathology grade, Lauren classification, T staging, N staging, tumor size, number of extrahepatic metastatic sites to bone, lung, and brain, insurance situation and smoking) as predictors of the presence of liver metastases at diagnosis. Median survival among the entire cohort with GCLM was 4.0 months (interquartile range: 1.0-10.0 mo). Patients receiving comprehensive therapy had longer median overall survival, of which the median survival was 12.0 months (interquartile range: 6.0-31.0 mo). Multivariable Cox model in SEER cohort confirmed nine factors (age, tumor location, Lauren classification, T staging, number of extrahepatic metastatic sites to bone, lung, and brain, surgery, chemotherapy, RSC and marital status) as independent predictors for overall survival. Conclusions: The findings of this study provided population-based estimates of the proportion and prognosis for LM at time of GC diagnosis. These findings provide preventive guidelines for screening and treatment of LM in GC patients.
Collapse
Affiliation(s)
- Zepang Sun
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Huan Zheng
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, 510515 Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Weicai Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Tuanjie Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Hao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Mingli Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Yuming Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| |
Collapse
|
518
|
Chen QY, Zhong Q, Wang W, Chen S, Li P, Xie JW, Wang JB, Lin JX, Lu J, Cao LL, Lin M, Tu RH, Huang ZN, Lin JL, Zheng HL, Liu ZY, Zheng CH, Peng JS, Zhou ZW, Huang CM. Prognosis of Young Survivors of Gastric Cancer in China and the U.S.: Determining Long-Term Outcomes Based on Conditional Survival. Oncologist 2019; 24:e260-e274. [PMID: 30470692 PMCID: PMC6656502 DOI: 10.1634/theoncologist.2018-0220] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Young survivors of gastric cancer (GC) have better prognoses than elderly patients, yet their disease-specific survival (DSS) has received little attention. PATIENTS AND METHODS Data on young patients (aged ≤40 years) with GC undergoing resections at three Chinese institutions (n = 542) and from the SEER database (n = 533) were retrospectively analyzed. Three-year conditional disease-specific survival (CS3) was assessed. The effects of well-known prognostic factors over time were analyzed by time-dependent Cox regression. RESULTS Overall, young Chinese patients with GC had a better 5-year DSS than U.S. patients (62.8% vs. 54.1%; p < .05). The disease-specific mortality likelihood of the entire cohort was not constant over time, with most deaths occurring during the first 3 years after surgery but peaking at 1 and 2 years in China and the U.S., respectively. Based on 5-year survivorship, the CS3 rates of both groups were similar (90.9% [U.S.] vs. 91.5% [China]; p > .05). Cox regression showed that for Chinese patients, site, size, T stage, and N stage were independent prognostic factors at baseline (p < .05). For U.S. patients, grade, T stage. and N stage significantly affected DSS at baseline (p < .05). In both groups, only T stage continuously affected DSS within 3 years after gastrectomy. However, for both groups, the initial well-known prognostic factors lost prognostic significance after 5 years of survival (all p > .05). Although the 5-year DSS rates of young Chinese patients with T3 and T4a disease were significantly better than those of young U.S. patients, in each T stage, the CS3 of both regions trended toward consistency over time. CONCLUSION For young patients with GC, the factors that predict survival at baseline vary over time. Although the initial 5-year DSS is heterogeneous, insight into conditional survival will help clinicians evaluate the long-term prognoses of survivors while ignoring population differences. IMPLICATIONS FOR PRACTICE With the increasing number of young survivors of gastric cancer (GC), it is essential for clinicians to understand the dynamic prognosis of these patients. Based on large data sets from China and the U.S., this study found that the prognostic factors that predict survival for young patients with GC at baseline vary over time. Although the initial 5-year disease-specific survival is heterogeneous, insight into conditional survival will help clinicians evaluate the long-term prognoses of survivors while ignoring population differences. This knowledge may be more effective in helping young patients with GC to manage future uncertainties, especially when they need to make important life plans.
Collapse
Affiliation(s)
- Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangdong, People's Republic of China
| | - Shi Chen
- Department of Esophageal and Gastrointestinal Surgery, The Sixth Hospital Affiliated to Sun Yat-Sen University, Sun Yat-Sen University Research Center of Diagnosis and Treatment of Gastric Cancer, Guangzhou, People's Republic of China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jia-Bing Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ju-Li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jun-Sheng Peng
- Department of Esophageal and Gastrointestinal Surgery, The Sixth Hospital Affiliated to Sun Yat-Sen University, Sun Yat-Sen University Research Center of Diagnosis and Treatment of Gastric Cancer, Guangzhou, People's Republic of China
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangdong, People's Republic of China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| |
Collapse
|
519
|
Cherukuri AR, Lubner MG, Zea R, Hinshaw JL, Lubner SJ, Matkowskyj KA, Foltz ML, Pickhardt PJ. Tissue sampling in the era of precision medicine: comparison of percutaneous biopsies performed for clinical trials or tumor genomics versus routine clinical care. Abdom Radiol (NY) 2019; 44:2074-2080. [PMID: 30032384 DOI: 10.1007/s00261-018-1702-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE The purpose of the study was to determine if patients undergoing percutaneous biopsy for genetic profiling are undergoing more biopsies (procedures, passes per procedure), or experiencing more procedure-related complications. METHODS 60 patients undergoing biopsy procedures for genetic profiling were retrospectively compared with 60 consecutive control patients undergoing routine biopsies. Procedural details and related complications were collected. Results were analyzed using t-tests and logistic regression. RESULTS Biopsied organs included mainly lung (n = 31), liver (n = 50), and lymph nodes (n = 18). The average number of core biopsy passes was 3.45 in the study group and 2.18 in the control group (0.73, 1.81; p = 0.0001). The average study patient underwent 1.44 biopsy procedures by radiology from 2016 to 2017, whereas the average control patient underwent 1.08 (0.1657, 0.5010, p = 0.0002). Results were similar when looking at the subset of patients undergoing liver biopsies. In our cohort of 120 patients total, only 6 complications were noted. There were 4 complications in the control patients and 2 complications in the study patients, all of which were pneumothoraces in patients undergoing lung biopsy; only 2 of these required treatment. The odds ratio for a complication occurring from an increase in one core biopsy is 1.07 (0.601, 1.573; p = 0.775), suggesting no significant relationship among the number of biopsies taken and the probability of complication in this cohort. CONCLUSIONS Patients being biopsied for genetic profiling or clinical study enrollment are undergoing more biopsy procedures and more biopsy passes per procedure, but are not experiencing a detectable increased rate of complications in this small cohort, single-center study.
Collapse
Affiliation(s)
- Anjuli R Cherukuri
- Departments of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA
| | - Meghan G Lubner
- Departments of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA.
| | - Ryan Zea
- Biostatistics, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA
| | - J Louis Hinshaw
- Departments of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA
| | - Sam J Lubner
- Internal Medicine - Division of Human Oncology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA
| | - Kristina A Matkowskyj
- Pathology and Lab Medicine, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA
| | - Marcia L Foltz
- Departments of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA
| | - Perry J Pickhardt
- Departments of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Sciences Center, 600 Highland Ave, Madison, WI, 53792, USA
| |
Collapse
|
520
|
Kim JH, Lee HH, Seo HS, Jung YJ, Park CH. Stage-specific difference in timing and pattern of initial recurrence after curative surgery for gastric cancer. Surg Oncol 2019; 30:81-86. [PMID: 31500791 DOI: 10.1016/j.suronc.2019.05.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/13/2019] [Accepted: 05/27/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Gastric cancer (GC) follow-up schedule after curative surgery is universally recommended based on the pathologic stage, but their details, including intervals and modalities of surveillance have not yet been standardised. The aim of this study was to investigate the characteristics of GC recurrence by stage to establish optimal postoperative surveillance strategies. METHODS Medical information on 5095 patients with GC who underwent curative intent gastrectomy in our institution between January 1989 and December 2013 was reviewed retrospectively. Moreover, 656 patients who had recurrences after radical surgery were identified. Clinicopathologic characteristics, timing and pattern of recurrence, and survival data of these patients were analysed. RESULTS Among the 656 patients, 50 (7.6%), 123 (18.8%), and 483 (73.6%) had stages I, II, and III GC, respectively. The median times to initial recurrence in patients with stages I, II, and III GC were 23.5 months (interquartile range [IQR], 13.0-33.0 months), 13.0 months (IQR, 9.0-25.0 months), and 12.0 months (IQR, 7.0-21.0 months), respectively. In patients with stage I GC, more than half (58%) of them had distant organ metastasis; otherwise, peritoneal dissemination (39%) was the most common pattern in patients with stage III GC. CONCLUSIONS Despite the low incidence, the time of initial recurrence in stage I GC was longer than those in stage II and III GC. Moreover, the pattern of initial recurrence was also different according to the pathologic stage. Therefore, clinicians should consider stage-specific differences of recurrence in setting up surveillance strategies after curative surgery for GC patients.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Han Hong Lee
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| | - Ho Seok Seo
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yoon Ju Jung
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Cho Hyun Park
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
521
|
Aoki M, Shoji H, Nagashima K, Imazeki H, Miyamoto T, Hirano H, Honma Y, Iwasa S, Okita N, Takashima A, Kato K, Higuchi K, Boku N. Hyperprogressive disease during nivolumab or irinotecan treatment in patients with advanced gastric cancer. ESMO Open 2019; 4:e000488. [PMID: 31231567 PMCID: PMC6555603 DOI: 10.1136/esmoopen-2019-000488] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 12/24/2022] Open
Abstract
Background Nivolumab showed a survival benefit for advanced gastric cancer (AGC). However, an acceleration of tumour growth during immunotherapy, (hyperprogressive disease, HPD) has been reported in various cancers. This study reviewed the HPD in patients with AGC treated with nivolumab or irinotecan. Methods The subjects of this retrospective study were patients with AGC with measurable lesions, and their tumour growth rates (TGR) during nivolumab or irinotecan were compared with those during prior therapy. HPD was defined as an increase in TGR more than twofold. Results 34 and 66 patients received nivolumab and irinotecan in third or later line between June 2009 and September 2018 at our hospital; 22 patients receiving nivolumab had prior treatment with irinotecan, and one patient received irinotecan after nivolumab. Nivolumab and irinotecan showed no differences in disease control rates (38.2% and 34.8%) and in progression-free survival (PFS) (HR 1.1, 95% CI 0.7 to 1.6, p=0.802). The incidence of HPD was slightly higher after nivolumab (29.4%) than after irinotecan (13.5%) (p=0.0656), showing no differences in background between the patients with and without HPD. Compared between HPD and PD other than HPD after nivolumab, the HRs for PFS and overall survival (OS) were 1.1 (95% CI 0.5 to 2.7; p=0.756), and 2.1 (95% CI 0.7 to 5.8; p=0.168), but such clear difference in OS was not observed after irinotecan. Conclusions HPD was observed more frequently after nivolumab compared with irinotecan, which was associated with a poor prognosis after nivolumab but not so clearly after irinotecan.
Collapse
Affiliation(s)
- Masahiko Aoki
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
- Internal Medicine (II), Osaka Medical College, Osaka, Japan
| | - Hirokazu Shoji
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Kengo Nagashima
- Research Center for Medical and Health Data Science, The Institute of Statistical Mathematics, Tokyo, Japan
- Clinical and Translational Research Center, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Imazeki
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiro Miyamoto
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
- Internal Medicine (II), Osaka Medical College, Osaka, Japan
| | - Hidekazu Hirano
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshitaka Honma
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Satoru Iwasa
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Natsuko Okita
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Atsuo Takashima
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | | | - Narikazu Boku
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
522
|
Efficacy and Safety of Xiao Ai Ping Injection Combined with Chemotherapy in Advanced Gastric Cancer: A Systematic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3821053. [PMID: 31236124 PMCID: PMC6545757 DOI: 10.1155/2019/3821053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 04/16/2019] [Indexed: 12/24/2022]
Abstract
Xiao Ai Ping injection (XAPI), extracted from the Chinese herbal medicine Marsdenia tenacissima, is widely used in the adjuvant treatment of tumors in China. The present study aimed to evaluate the efficacy and safety of XAPI combined with chemotherapy for treating patients with advanced gastric cancer. Seven databases were searched for relevant studies published up to October 1, 2018, and Review Manager 5.3 software and Stata 12.0 software were used for meta-analysis. Fourteen studies, representing 1097 enrolled patients, were included in our analysis. Compared with chemotherapy alone, combination treatment with XAPI and the XELOX regimen (capecitabine plus oxaliplatin) was found to improve the objective response rate (ORR) [RR=1.36; 95%CI (1.10, 1.70); P=0.006], disease control rate (DCR) [RR=1.15; 95% CI (1.04, 1.28); P=0.010], and Karnofsky Performance Status (KPS) improvement rate [RR=1.51; 95%CI (1.14, 2.00); P=0.004] and to reduce the incidence of leukopenia [RR=0.68; 95%CI (0.55,0.84); P=0.0005], liver damage [RR=0.59; 95% CI (0.37, 0.92); P=0.02], renal impairment [RR=0.39; 95% CI (0.18, 0.85); P=0.02], and hand-foot syndrome [RR=0.56; 95%CI (0.35,0.90); P=0.02]. However, median progression-free survival (PFS), 1-year survival rate, and median overall survival (OS) were not extended by XAPI plus XELOX. Combination treatment with XAPI and the SOX regimen (tegafur plus oxaliplatin) did not improve ORR or DCR, but it did enhance the KPS improvement rate [RR=1.73; 95%CI (1.23,2.43); P=0.002] and reduce the incidence of nausea and vomiting [RR=0.66; 95% CI (0.50, 0.88); P=0.004]. XAPI in combination with the FOLFOX regimen (fluorouracil/calcium folinate/oxaliplatin) enhanced only the KPS improvement rate [RR=1.68; 95%CI (1.18,2.39); P=0.004] and had no significant effect on ORR or DCR or the incidence of adverse events. A single study reported that XAPI combined with the CPT-11 regimen (irinotecan) was superior to chemotherapy alone with respect to DCR and also reduced the incidence of leukopenia, liver damage, and hand-foot syndrome during chemotherapy, while prolonging PFS. Finally, one study reported that XAPI combined with the TP regimen (palitaxel plus cisplatin) improved ORR and KPS improvement rate to a greater extent than TP alone. Although the present review has some limitations, the findings suggest that XAPI combined with chemotherapy may represent a beneficial treatment strategy, particularly the combination of XAPI and XELOX.
Collapse
|
523
|
Ulanja MB, Beutler BD, Rishi M, Konam KG, Zell SC, Patterson DR, Ambika S, Gullapalli N. Influence of race and geographic setting on the management of gastric adenocarcinoma. J Surg Oncol 2019; 120:270-279. [PMID: 31102468 DOI: 10.1002/jso.25503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/07/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVES Conflicting evidence indicates that both race and geographic setting may influence the management of malignancies such as gastric adenocarcinoma (GAC). METHODS We designed a retrospective cohort study utilizing data from the Surveillance, Epidemiology, and End Results program to identify patients with resectable GAC (N = 15 991). Exposures of interest were race and geographic region of diagnosis (West [WE], Midwest [MW], South [SO], or Northeast [NE]). Endpoints included: (1) recommendation against surgery and (2) gastric adenocarcinoma-specific survival (GACSS). Multivariable logistic and Cox regression models were used to identify pertinent associations. RESULTS A total of 15 991 patients were included (2007-2015). In adjusted analysis, African American individuals more frequently received a recommendation against surgical resection than White (adjusted odds ratio [aOR] = 0.86; 95% confidence interval [CI], 0.76-0.98), Asian American (aOR = 0.55; 95% CI, 0.46-0.65), and American Indian (aOR = 0.50; 95% CI, 0.31-0.82) individuals. In addition to race-based discrepancies, there was a significant association between geography and management: individuals diagnosed with GAC in the SO were more likely to receive a recommendation against surgery (odds ratio = 1.35; 95% CI, 1.23-1.49) and exhibited poorer GACSS as compared with those in the WE, MW, or NE regions. CONCLUSIONS Race and geographic region of diagnosis affect treatment recommendations and GACSS among individuals with resectable tumors. African Americans with resectable cancers are more likely to receive a recommendation against surgery than individuals of other racial groups.
Collapse
Affiliation(s)
- Mark B Ulanja
- Department of Internal Medicine, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Bryce D Beutler
- Department of Internal Medicine, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Mohit Rishi
- Department of Internal Medicine, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Kenneth G Konam
- Department of Medicine, Health and Society, College of Arts and Sciences, Vanderbilt University, Nashville, Tennessee
| | - Steven C Zell
- Department of Internal Medicine, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Darryll R Patterson
- Department of Internal Medicine, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Santhosh Ambika
- Department of Internal Medicine, Reno School of Medicine, University of Nevada, Reno, Nevada.,Renown Institute for Cancer, Department of Hematology/Oncology, Reno, Nevada
| | - Nageshwara Gullapalli
- Department of Internal Medicine, Reno School of Medicine, University of Nevada, Reno, Nevada
| |
Collapse
|
524
|
Notch signaling pathway regulates CD4 +CD25 +CD127 dim/- regulatory T cells and T helper 17 cells function in gastric cancer patients. Biosci Rep 2019; 39:BSR20182044. [PMID: 30988066 PMCID: PMC6522723 DOI: 10.1042/bsr20182044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 04/02/2019] [Accepted: 04/06/2019] [Indexed: 02/06/2023] Open
Abstract
Regulatory T cells (Tregs) and T helper 17 (Th17) cells contribute to cancer progression and prognosis. However, regulatory factors associated with Tregs-Th17 balance were not completely understood. We previously demonstrated an immune-modulatory capacity by Notch signaling inactivation to reverse Tregs-Th17 disequilibrium in chronic hepatitis C. Thus, the aim of current study was to assess the role of Notch signaling in modulation Tregs and Th17 cells function in gastric cancer (GC) patients. A total of 51 GC patients and 18 normal controls (NCs) were enrolled. Notch1 and Notch2 mRNA expressions were semiquantified by real-time polymerase chain reaction. Tregs/Th17 percentages, transcriptional factors, and cytokines production were investigated in response to the stimulation of Notch signaling inhibitor DAPT. Both Notch1 and Notch2 mRNA expressions were elevated in GC tissues and peripheral bloods in GC patients. CD4+CD25+CD127dim/- Tregs and Th17 cells percentage was also elevated in GC patients compared with in NCs. DAPT treatment did not affect frequency of either circulating Tregs or Th17 cells, however, reduced FoxP3/RORγt mRNA expression and interleukin (IL)-35/IL-17 production in purified CD4+ T cells from GC patients. Moreover, blockade of Notch signaling also inhibited the suppressive function of purified CD4+CD25+CD127dim/- Tregs from GC patients, which presented as elevation of cellular proliferation and IL-35 secretion. The current data further provided mechanism underlying Tregs-Th17 balance in GC patients. The link between Notch signaling and Th cells might lead to a new therapeutic target for GC patients.
Collapse
|
525
|
Xiao FK, Guo S, Yang F, Zhao LS, Wang LD. MDM2 and its functional polymorphism SNP309 contribute to the development of esophageal carcinoma. J Gene Med 2019; 21:e3086. [PMID: 30861607 DOI: 10.1002/jgm.3086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/11/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Overexpression of the murine double minute 2 (MDM2) has been explored in many tumors with high proliferation and anti-apoptosis ability. However, the role of MDM2 and its functional single nucleotide polymorphism (SNP) rs2279744 (also known as SNP309) in esophageal squamous cell carcinoma (ESCC) remains unclear. METHODS We performed a genotype study of blood samples in 360 ESCC patients and 360 healthy control individuals to determine the risk of various rs2279744 in ESCC. To further evaluate the role of rs2279744 in regulating MDM2 expression, we performed an allele-specific reporter assay and investigated whether the SNP-containing sequences functioned as an active enhancer. To examine the functional role of MDM2 on esophageal cancer cell lines, we carried out an MTS assay and flow cytometry analysis. RESULTS From the genotyping study, we found that GG genotype of SNP309 significantly increased the risk of ESCC in an additive model [odds ratio (OR) = 2.55, 95% confidence interval (CI) = 1.66-3.89, p = 1.50 × 10-5 ) and in a recessive model (OR = 2.44, 95% CI = 1.69-3.51, p = 1.60 × 10-8 ). Furthermore, the G allele was significantly associated with a higher risk of ESCC (OR = 1.56, 95% CI = 1.26-1.92, p = 2.81 × 10-5 ). In multivariate logistic regression analysis, the GG genotype had increased the occurrence of ESCC by 2.39-fold (95% CI = 1.48-3.8). Compared to the T allele, the variant G allele had significantly higher luciferase activity on the promoter of MDM2 in both cell lines. By transfecting the gene to ESCC lines, we showed that overexpression of MDM2 significantly promote cell proliferation and anti-apoptosis. CONCLUSIONS The MDM2 promoter SNP309 is a risk factor for esophageal cancer. MDM2 promotes the proliferation and anti-apoptosis of ESCC.
Collapse
Affiliation(s)
- Fan-Kai Xiao
- Henan Key Laboratory for Esophageal Cancer Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sen Guo
- Department of Cardiology and Hypertension, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fan Yang
- Department of Cardiology and Hypertension, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Luo-Sha Zhao
- Department of Cardiology and Hypertension, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li-Dong Wang
- Henan Key Laboratory for Esophageal Cancer Research, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
526
|
Choi MH, Jung SE, Lee YJ, Yoon SB. More Frequent Follow-up CT Scans in Postsurgical Resection Patients Than in Postendoscopic Resection Patients of Early Gastric Cancers: Impracticality of CTs for Mucosal Cancer. Acad Radiol 2019; 26:651-657. [PMID: 30268723 DOI: 10.1016/j.acra.2018.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 07/25/2018] [Accepted: 07/25/2018] [Indexed: 02/07/2023]
Abstract
RATIONALE AND OBJECTIVES The aim of this study was to compare the utility and timing of computed tomography (CT) in the detection of recurrent tumors between patients after surgical and endoscopic resections for early gastric cancer (EGC). MATERIALS AND METHODS A total of 670 patients after surgical (n = 535) and endoscopic (n = 135) resections for EGC between 2007 and 2009 were enrolled. The mean numbers of CT and endoscopy between both treatment groups were compared. The mean and cumulative dose length products of CT examinations were calculated. The modality that detected recurrence was compared between the two groups using the Pearson chi-square test. RESULTS The mean interval of CT was significantly shorter and the mean number of CTs was significantly larger in the surgical resection group than in the endoscopic resection group. All 34 gastric recurrences were diagnosed by endoscopy. All seven extragastric recurrences occurred in patients treated for EGC with submucosal invasion. Six extragastric recurrences were detected by CT out of a total of 5417 CT scans. The average cumulative dose length product was significantly higher in the surgical group than in the endoscopic resection group (P = 0.004). CONCLUSION Follow-up CTs were performed more frequently in patients after surgical resection than in patients after endoscopic resection of EGCs. However, CT scans were not effective in detecting recurrent tumors after either treatment methods, especially for patients treated for mucosal gastric cancer.
Collapse
Affiliation(s)
- Moon Hyung Choi
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seung Eun Jung
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea.
| | - Young Joon Lee
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seung Bae Yoon
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
527
|
Newhook TE, Agnes A, Blum M, Estrella JS, Das P, Ho L, Ajani JA, Minsky BD, Mansfield P, Badgwell BD. Laparoscopic Hyperthermic Intraperitoneal Chemotherapy is Safe for Patients with Peritoneal Metastases from Gastric Cancer and May Lead to Gastrectomy. Ann Surg Oncol 2019; 26:1394-1400. [PMID: 30680477 DOI: 10.1245/s10434-018-07140-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Laparoscopic hyperthermic intraperitoneal chemotherapy (LS-HIPEC) is a novel strategy for patients with gastric adenocarcinoma (GA) metastatic to the peritoneum. We evaluated the safety profile of LS-HIPEC for patients with positive peritoneal cytology (PPC) or carcinomatosis from GA. METHODS Outcomes were reviewed of patients with stage IV GA with peritoneal involvement who received LS-HIPEC from June 2014 to January 2017. LS-HIPEC included a 60-minute perfusion of mitomycin-C (30 mg) and cisplatin (200 mg) with inflow temperatures of 41-42 °C and outflow temperatures of 39-40 °C. RESULTS A total of 71 LS-HIPEC procedures were performed in 44 patients. At diagnosis, 68% (n = 30) had carcinomatosis and 32% (n = 14) had isolated PPC. Three patients (7%) underwent LS-HIPEC for intractable ascites. All patients initially received systemic chemotherapy, and 20 patients (45%) received pre-procedural chemoradiotherapy. The median number of LS-HIPEC procedures performed per patient was one (range 1-5 procedures). There were no conversions to laparotomy, two outflow catheter obstructions, and one major (Clavien-Dindo grade III) surgical complication within 30 days. A total of seven postoperative adverse hematologic events (> CTCAE 2) were observed in five patients (11%), without any major renal or gastrointestinal adverse events within 30 days. The median overall length of hospital stay after LS-HIPEC was 2 (range 2-11) days. Eleven patients (25%) underwent secondary gastrectomy following resolution of peritoneal cytology. CONCLUSIONS Laparoscopic HIPEC is a safe procedure and may be repeated in patients with peritoneal metastases from gastric cancer. Future studies are required to determine the optimal HIPEC regimen and timing relative to systemic therapy to best minimize morbidity.
Collapse
Affiliation(s)
- Timothy E Newhook
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Annamaria Agnes
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mariela Blum
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeannelyn S Estrella
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linus Ho
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bruce D Minsky
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Mansfield
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian D Badgwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
528
|
Liu K, Chen XZ, Zhang WH, Zhang DY, Luo Y, Yu Y, Yang K, Yang SJ, Chen XL, Sun LF, Zhao LY, Zhou ZG, Hu JK. "Four-Step Procedure" of laparoscopic exploration for gastric cancer in West China Hospital: a retrospective observational analysis from a high-volume institution in China. Surg Endosc 2019; 33:1674-1682. [PMID: 30478700 PMCID: PMC6484818 DOI: 10.1007/s00464-018-6605-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 11/19/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND The preoperative work-up has limitations on finding peritoneal dissemination (PD) in gastric cancer patients. Laparoscopic exploration (LE) can discover radiographically occult PD, obtain accurate stage and avert futile laparotomy. The aim of our study was to introduce "Four-Step Procedure" LE in West China Hospital and further evaluate its safety and feasibility. METHODS We conducted a retrospective analysis on 165 patients from July 2016 to December 2017 who underwent "Four-Step Procedure" LE in gastrointestinal surgery department of West China Hospital. All the patients were diagnosed with gastric adenocarcinoma without explicit distant metastasis through Computed Tomography and/or Gastrointestinal Ultrasonography. Peritoneal lavage cytological examination (CY) was routinely performed during LE in our research. The "Four-Step" technical process of LE was introduced comprehensively. The clinicopathologic features and the presence of PD or CY at LE were analyzed, and the stratified analysis by cT and cN stages on the proportion of P1 and/or CY1 was also reported in this study. RESULTS Total of 165 patients accepted LE in our study, among these patients: 27 (16.4%) patients with P1 and/or CY1: 19 (11.5%) patients were found PD (P1), 17 (10.3%) patients with positive cytological examination (CY1) and 9 (3.6%) patients with P1Cy1. The stratified analysis by cT stage indicated that there was no P1 and/or Cy1 in cT1-cT2 stages, 1 (2.7%) patient with P1 and 1 (2.7%) with Cy1 in cT3 stage, 18 (20.0%) patients with P1 and 16 (17.8%) with Cy1 in cT4 stage. After LE, there were 74 (44.8%) patients underwent laparoscopic assistant gastrectomy, 25 (15.2%) patients with open gastrectomy, 50 (30.3%) patients with neoadjuvant chemotherapy and 16 (9.7%) patients with palliative chemotherapy and/or conversion therapy. CONCLUSION "Four-Step Procedure" LE is reliable and feasible for gastric cancer. From our study, LE has unique superiority on ascertaining PD and cytological examination and LE should be recommended in cT4 stage gastric cancer before resection.
Collapse
Affiliation(s)
- Kai Liu
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xin-Zu Chen
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Wei-Han Zhang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Dong-Yang Zhang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Yi Luo
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yue Yu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Kun Yang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Shi-Jie Yang
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xiao-Long Chen
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Li-Fei Sun
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Lin-Yong Zhao
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Zong-Guang Zhou
- Department of Gastrointestinal Surgery and Laboratory of Digestive Surgery, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Jian-Kun Hu
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China.
| |
Collapse
|
529
|
Shitara K, Iwata H, Takahashi S, Tamura K, Park H, Modi S, Tsurutani J, Kadowaki S, Yamaguchi K, Iwasa S, Saito K, Fujisaki Y, Sugihara M, Shahidi J, Doi T. Trastuzumab deruxtecan (DS-8201a) in patients with advanced HER2-positive gastric cancer: a dose-expansion, phase 1 study. Lancet Oncol 2019; 20:827-836. [PMID: 31047804 DOI: 10.1016/s1470-2045(19)30088-9] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Trastuzumab deruxtecan (DS-8201a) is a novel HER2-targeted antibody-drug conjugate with a humanised anti-HER2 antibody, cleavable peptide-based linker, and topoisomerase I inhibitor payload. A phase 1, non-randomised, open-label, multiple-dose study was done to assess the safety, tolerability, and activity of trastuzumab deruxtecan in HER2-expressing advanced solid tumours. The dose escalation (part 1) has previously been reported and the recommended doses for expansion of 5·4 mg/kg or 6·4 mg/kg were established. In this Article, we report the safety and preliminary activity results from this phase 1 trial in all patients with HER2-positive gastric or gastro-oesophageal junction cancer who received trastuzumab deruxtecan at the recommended doses for expansion. METHODS This was an open-label, dose-escalation and dose-expansion phase 1 trial done at eight hospitals and clinics in the USA and six in Japan. Eligible patients were at least 18 years old in the USA and at least 20 years old in Japan and had advanced solid tumours (regardless of HER2 expression in dose escalation or HER2 expression or mutation in dose expansion). The recommended doses for expansion of 5·4 mg/kg or 6·4 mg/kg trastuzumab deruxtecan were administered intravenously to patients once every 3 weeks until withdrawal of consent, unacceptable toxicity, or progressive disease. In this Article, all patients with HER2-positive gastric or gastro-oesophageal junction cancer with previous trastuzumab treatment who received trastuzumab deruxtecan were analysed together. The primary endpoints of the study were safety and preliminary activity (proportion of patients who achieved an objective response as assessed by the investigators). The activity evaluable set included all patients who received at least one dose of trastuzumab deruxtecan at the recommended doses for expansion, and for whom both baseline and post-treatment activity data were available. The safety analysis set included all patients who received at least one dose of trastuzumab deruxtecan at the recommended doses for expansion. Enrolment for patients with gastric or gastro-oesophageal junction cancer has completed. This trial is registered at ClinicalTrials.gov, number NCT02564900, and ClinicalTrials.jp, number JapicCTI-152978. FINDINGS Between Aug 28, 2015, and Aug 10, 2018, 44 patients with HER2-positive gastric or gastro-oesophageal junction cancer received at least one dose of trastuzumab deruxtecan at the recommended doses for expansion. All patients had at least one treatment-emergent adverse event. The most frequent grade 3 or worse treatment-emergent adverse events included anaemia (13 [30%]) and decreases in neutrophil (nine [20%]), platelet (eight [18%]), and white blood cell (seven [16%]) counts. Serious treatment-emergent adverse events occurred in 11 (25%) patients. There were four pneumonitis cases (three grade 2 and one grade 3). There were no drug-related deaths due to treatment-emergent adverse events. 19 (43·2%; 95% CI 28·3-59·0) of 44 patients had a confirmed objective response. INTERPRETATION Trastuzumab deruxtecan had a manageable safety profile and showed preliminary activity in heavily pretreated patients with HER2-positive gastric or gastro-oesophageal junction cancer. These results support further investigation of trastuzumab deruxtecan for HER2-positive gastric or gastro-oesophageal junction cancer post-trastuzumab. FUNDING Daiichi Sankyo Co, Ltd.
Collapse
Affiliation(s)
- Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Shunji Takahashi
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kenji Tamura
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Haeseong Park
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Shanu Modi
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Junji Tsurutani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan; Department of Medical Oncology, Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan
| | - Shigenori Kadowaki
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Kensei Yamaguchi
- Department of Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoru Iwasa
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kaku Saito
- Research and Development, Daiichi Sankyo Co, Ltd, Tokyo, Japan
| | | | - Masahiro Sugihara
- Biostatistics and Data Management, Daiichi Sankyo Co, Ltd, Tokyo, Japan
| | - Javad Shahidi
- Research and Development, Daiichi Sankyo, Inc, Basking Ridge, NJ, USA
| | - Toshihiko Doi
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| |
Collapse
|
530
|
Li Q, Ge Y, Chen X, Wang L, Xia Y, Xu Z, Li Z, Wang W, Yang L, Zhang D, Xu Z. LEM domain containing 1 promotes proliferation via activating the PI3K/Akt signaling pathway in gastric cancer. J Cell Biochem 2019; 120:15190-15201. [PMID: 31021450 DOI: 10.1002/jcb.28783] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/26/2018] [Accepted: 01/10/2019] [Indexed: 01/31/2023]
Affiliation(s)
- Qiang Li
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Yugang Ge
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Xiaofeng Chen
- Department of Oncology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Lu Wang
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Yiwen Xia
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Zhipeng Xu
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Zheng Li
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Weizhi Wang
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Li Yang
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Diancai Zhang
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Zekuan Xu
- Department of General Surgery The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| |
Collapse
|
531
|
van Putten M, Lemmens VEPP, van Laarhoven HWM, Pruijt HFM, Nieuwenhuijzen GAP, Verhoeven RHA. Poor compliance with perioperative chemotherapy for resectable gastric cancer and its impact on survival. Eur J Surg Oncol 2019; 45:1926-1933. [PMID: 30982656 DOI: 10.1016/j.ejso.2019.03.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 01/27/2019] [Accepted: 03/28/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND In several Western European countries it is recommended to treat gastric cancer patients with perioperative chemotherapy if they are eligible for surgery. However, little is known about its use in daily clinical practice. This study examines the use of perioperative treatment and its impact on survival in the Netherlands. METHODS Patients diagnosed with potentially resectable gastric cancer (cT1N+/cT2-T3,X any cN, cM0,X) between 2006 and 2014 were selected from the Netherlands Cancer Registry (N = 5824). Treatment trends were examined. Propensity score matching was used to create a subsample to reduce selection bias. Cox regression analysis was used to assess differences in overall survival. RESULTS The percentage of patients treated with perioperative treatment increased from 3% in 2006 to 26% in 2014 and the use of only surgery decreased from 60% to 26%. 35% of all patients did not undergo surgery. Of the patients who underwent preoperative chemotherapy and surgery, 43% did not commence postoperative treatment. Cox regression analysis showed a better overall survival for patients who underwent perioperative treatment compared to patients who underwent preoperative treatment only (HR = 0.80 95%CI 0.70-0.93; propensity matched sample: HR = 0.84 95%CI 0.71-0.99), whereas survival was comparable for patients who underwent preoperative chemotherapy versus surgery alone (HR = 0.89 95%CI 0.77-1.02, propensity matched sample: HR = 0.85 95%CI 0.72-1.01). CONCLUSION This population-based study highlights that a significant proportion of the patients did not receive perioperative treatment. More research is necessary to elucidate the importance of the individual components of perioperative treatment.
Collapse
Affiliation(s)
- Margreet van Putten
- Department of Research, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, the Netherlands.
| | - Valery E P P Lemmens
- Department of Research, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, the Netherlands; Department of Public Health, Erasmus MC - University Medical Centre Rotterdam, the Netherlands
| | | | - Hans F M Pruijt
- Department of Internal Medicine, Jeroen Bosch Hospital, 's Hertogenbosch, the Netherlands
| | | | - Rob H A Verhoeven
- Department of Research, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, the Netherlands
| |
Collapse
|
532
|
Chen QY, Zhong Q, Zhou JF, Qiu XT, Dang XY, Cai LS, Su GQ, Xu DB, Liu ZY, Li P, Guo KQ, Xie JW, Chen QX, Wang JB, Li TW, Lin JX, Lin SM, Lu J, Cao LL, Lin M, Tu RH, Huang ZN, Lin JL, Lin W, He QL, Zheng CH, Huang CM. Development and External Validation of Web-Based Models to Predict the Prognosis of Remnant Gastric Cancer after Surgery: A Multicenter Study. JOURNAL OF ONCOLOGY 2019; 2019:6012826. [PMID: 31093283 PMCID: PMC6481035 DOI: 10.1155/2019/6012826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Remnant gastric cancer (RGC) is a rare malignant tumor with poor prognosis. There is no universally accepted prognostic model for RGC. METHODS We analyzed data for 253 RGC patients who underwent radical gastrectomy from 6 centers. The prognosis prediction performances of the AJCC7th and AJCC8th TNM staging systems and the TRM staging system for RGC patients were evaluated. Web-based prediction models based on independent prognostic factors were developed to predict the survival of the RGC patients. External validation was performed using a cohort of 49 Chinese patients. RESULTS The predictive abilities of the AJCC8th and TRM staging systems were no better than those of the AJCC7th staging system (c-index: AJCC7th vs. AJCC8th vs. TRM, 0.743 vs. 0.732 vs. 0.744; P>0.05). Within each staging system, the survival of the two adjacent stages was not well discriminated (P>0.05). Multivariate analysis showed that age, tumor size, T stage, and N stage were independent prognostic factors. Based on the above variables, we developed 3 web-based prediction models, which were superior to the AJCC7th staging system in their discriminatory ability (c-index), predictive homogeneity (likelihood ratio chi-square), predictive accuracy (AIC, BIC), and model stability (time-dependent ROC curves). External validation showed predictable accuracies of 0.780, 0.822, and 0.700, respectively, in predicting overall survival, disease-specific survival, and disease-free survival. CONCLUSIONS The AJCC TNM staging system and the TRM staging system did not enable good distinction among the RGC patients. We have developed and validated visual web-based prediction models that are superior to these staging systems.
Collapse
Affiliation(s)
- Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jun-Feng Zhou
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xian-Tu Qiu
- Department of Gastrointestinal Surgery and Gastrointestinal Surgery Research Institute, The Affiliated Hospital of Putian University, Putian, China
| | - Xue-Yi Dang
- Department of General Surgery, Shanxi Provincial Cancer Hospital, Shanxi, China
| | - Li-Sheng Cai
- Department of General Surgery Unit 4, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Guo-Qiang Su
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Dong-Bo Xu
- Department of Gastrointestinal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Kai-Qing Guo
- Department of General Surgery, Shanxi Provincial Cancer Hospital, Shanxi, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qiu-Xian Chen
- Department of General Surgery Unit 4, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Teng-Wen Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Shuang-Ming Lin
- Department of Gastrointestinal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ju-Li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Wei Lin
- Department of Gastrointestinal Surgery and Gastrointestinal Surgery Research Institute, The Affiliated Hospital of Putian University, Putian, China
| | - Qing-Liang He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
533
|
Abstract
Apatinib [Aitan® (brand name in China)], also known as rivoceranib, is a novel, small molecule, selective vascular endothelial growth factor receptor-2 (VEGFR-2) tyrosine kinase inhibitor and is the second anti-angiogenic drug to be approved in China for the treatment of advanced or metastatic gastric cancer. This article summarizes the pharmacological properties of apatinib and reviews its clinical use in chemotherapy-experienced patients with advanced gastric adenocarcinoma, including gastroesophageal adenocarcinoma (GEA), or with other advanced cancers such as non-small cell lung cancer (NSCLC), breast cancer, gynaecological cancers, hepatocellular carcinoma (HCC), thyroid cancer and sarcomas. As third- or subsequent-line therapy, oral apatinib significantly prolonged median progression-free survival (PFS) and overall survival (OS) compared with placebo and had a manageable safety profile in Chinese patients with advanced or metastatic gastric cancer or GEA participating in randomized, double-blind, multicentre, phase 2 and 3 trials. More limited evidence also supports it use as subsequent-line treatment in Chinese patients with other advanced or metastatic solid tumours, including NSCLC, breast cancer and HCC. Further clinical experience and long-term pharmacovigilance data are required to more definitively establish the efficacy and safety profile of apatinib, including its use in combination with other chemotherapy agents and its role in the management of other types of advanced or metastatic solid tumours. In the meantime, given its convenient administration regimen and the limited treatment options and poor prognosis for patients with advanced or metastatic solid tumours, apatinib is an important, emerging treatment option for adult patients with advanced gastric adenocarcinoma or GEA who have progressed or relapsed after chemotherapy.
Collapse
Affiliation(s)
- Lesley J Scott
- Springer, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
534
|
Wang Y, Zhang J, Guo S, Dong Z, Meng X, Zheng G, Yang D, Zheng Z, Zhao Y. Implication of lymph node staging in migration and different treatment strategies for stage T2N0M0 and T1N1M0 resected gastric cancer: a SEER population analysis. Clin Transl Oncol 2019; 21:1499-1509. [PMID: 30903518 DOI: 10.1007/s12094-019-02078-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/28/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE The purpose of this study was to explore the differences between stage T2N0M0 and stage T1N1M0 gastric cancer (GC) and to identify the necessity of adjuvant treatment (AT) for these stages. METHODS Between years 2004 and 2015, 1971 stage IB GC patients who underwent radical surgery were recruited using the Surveillance, Epidemiology and End Results database. We conducted univariate/multivariate analyses, the propensity score matching and evaluated gastric cancer-specific survival (GCSS) and overall survival (OS) with the log-rank test. RESULTS T1N1M0 had a significantly worse survival than T2N0M0 in both GCSS and OS before and after the propensity score matching. Examined lymph nodes (ELN) ≤ 15 and T1N1M0 were independent risk factors for worse GCSS and OS in stage IB GC. The absence of adjuvant chemotherapy (CT) was an independent risk factor for worse GCSS and OS in T1N1M0 but not in T2N0M0. AT demonstrated similar GCSS and OS with surgery alone (SA) for T2N0M0 but better survival for T1N1M0. Compared to CT and adjuvant chemoradiotherapy (CRT) group, SA demonstrated significantly worse GCSS and OS for T1N1M0. There was no significant difference between CT and CRT in both T2N0M0 and T1N1M0 stages. T2N0M0 had a better survival than T1N1M0 in ELN ≤ 15 subgroup. However, similar survival was demonstrated in ELN > 15 subgroup. CONCLUSIONS T2N0M0 GC has a better survival rate than T1N1M0 GC when ELN are ≤ 15. Moreover, T2N0M0 GC may not benefit from AT. T1N1M0 GC requires CT but not adjuvant radiotherapy.
Collapse
Affiliation(s)
- Y Wang
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang City, 110042, Liaoning Province, China
| | - J Zhang
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang City, 110042, Liaoning Province, China
| | - S Guo
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang City, 110042, Liaoning Province, China
| | - Z Dong
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang City, 110042, Liaoning Province, China
| | - X Meng
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang City, 110042, Liaoning Province, China
| | - G Zheng
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang City, 110042, Liaoning Province, China
| | - D Yang
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang City, 110042, Liaoning Province, China
| | - Z Zheng
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang City, 110042, Liaoning Province, China
| | - Y Zhao
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang City, 110042, Liaoning Province, China.
| |
Collapse
|
535
|
Muro K, Jen MH, Cheng R. Is ramucirumab and paclitaxel therapy beneficial for second-line treatment of metastatic gastric or junctional adenocarcinoma for patients with ascites? Analysis of RAINBOW phase 3 trial data. Cancer Manag Res 2019; 11:2261-2267. [PMID: 30962715 PMCID: PMC6433106 DOI: 10.2147/cmar.s193739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Purpose Second-line treatment with ramucirumab-paclitaxel has demonstrated statistically significant and clinically meaningful survival outcomes compared to paclitaxel-alone in patients with advanced gastric cancer (HR=0.807, 95% CI 0.678–0.962; P=0.017). Post hoc, exploratory analyses of RAINBOW patient data were performed to examine whether ascites impacted the efficacy and safety of ramucirumab-paclitaxel. Patients and methods Patients were placed in with- or without-ascites subgroups based on baseline information collected on case report forms. The Kaplan–Meier method was used to estimate median progression-free survival (PFS) and overall survival (OS) of the ascites subgroups. HR and 95% CI were calculated using the Cox proportional hazards model. Survival distributions within the two arms in each ascites subgroup were compared using the log-rank test. Results There were 36% of RAINBOW trial patients (237/665) that had ascites at baseline (with-ascites subgroup); 64% of patients (428/665) had no baseline ascites (without-ascites subgroup). Most baseline characteristics were balanced. The with-ascites subgroup had a higher percentage of patients with peritoneal metastases (91% vs 23%) as expected. Overall survival for the with-ascites subgroup was worse than for the without-ascites subgroup (median OS for placebo-treated patients: 5.2 vs 8.5 months, respectively). However, OS treatment effects did not seem to differ significantly among patients with ascites (OS stratified HR=0.864, 95% CI=0.644–1.161; P=0.3362) vs those without ascites (OS stratified HR=0.745, 95% CI=0.593–0.936; P=0.0115). Similar results were observed for PFS. Ramucirumab treatment was associated with a greater incidence of all-grade vomiting for the with-ascites subgroup vs the without-ascites subgroup (ramucirumab arm: 39.2% vs 18.8%; placebo arm: 23.3% vs 19.5%, respectively). The incidence of adverse events of special interest was not elevated among the ramucirumab-treated ascites subgroup over the without-ascites subgroup. Conclusion The benefit/risk profile of ramucirumab-paclitaxel remains favorable in patients with ascites and is consistent with the findings of the RAINBOW trial.
Collapse
Affiliation(s)
- Kei Muro
- Department of Clinical Oncology, Outpatient Treatment Center, Aichi Cancer Center Hospital, Nagoya, Japan,
| | - Min-Hua Jen
- Eli Lilly and Company Ltd, Windlesham, Surrey, UK
| | | |
Collapse
|
536
|
Chen D, Chen G, Jiang W, Fu M, Liu W, Sui J, Xu S, Liu Z, Zheng X, Chi L, Lin D, Li K, Chen W, Zuo N, Lu J, Chen J, Li G, Zhuo S, Yan J. Association of the Collagen Signature in the Tumor Microenvironment With Lymph Node Metastasis in Early Gastric Cancer. JAMA Surg 2019; 154:e185249. [PMID: 30698615 DOI: 10.1001/jamasurg.2018.5249] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Importance Lymph node status is the primary determinant in treatment decision making in early gastric cancer (EGC). Current evaluation methods are not adequate for estimating lymph node metastasis (LNM) in EGC. Objective To develop and validate a prediction model based on a fully quantitative collagen signature in the tumor microenvironment to estimate the individual risk of LNM in EGC. Design, Setting, and Participants This retrospective study was conducted from August 1, 2016, to May 10, 2018, at 2 medical centers in China (Nanfang Hospital and Fujian Provincial Hospital). Participants included a primary cohort (n = 232) of consecutive patients with histologically confirmed gastric cancer who underwent radical gastrectomy and received a T1 gastric cancer diagnosis from January 1, 2008, to December 31, 2012. Patients with neoadjuvant radiotherapy, chemotherapy, or chemoradiotherapy were excluded. An additional consecutive cohort (n = 143) who received the same diagnosis from January 1, 2011, to December 31, 2013, was enrolled to provide validation. Baseline clinicopathologic data of each patient were collected. Collagen features were extracted in specimens using multiphoton imaging, and the collagen signature was constructed. An LNM prediction model based on the collagen signature was developed and was internally and externally validated. Main Outcomes and Measures The area under the receiver operating characteristic curve (AUROC) of the prediction model and decision curve were analyzed for estimating LNM. Results In total, 375 patients were included. The primary cohort comprised 232 consecutive patients, in whom the LNM rate was 16.4% (n = 38; 25 men [65.8%] with a mean [SD] age of 57.82 [10.17] years). The validation cohort consisted of 143 consecutive patients, in whom the LNM rate was 20.9% (n = 30; 20 men [66.7%] with a mean [SD] age of 54.10 [13.19] years). The collagen signature was statistically significantly associated with LNM (odds ratio, 5.470; 95% CI, 3.315-9.026; P < .001). Multivariate analysis revealed that the depth of tumor invasion, tumor differentiation, and the collagen signature were independent predictors of LNM. These 3 predictors were incorporated into the new prediction model, and a nomogram was established. The model showed good discrimination in the primary cohort (AUROC, 0.955; 95% CI, 0.919-0.991) and validation cohort (AUROC, 0.938; 95% CI, 0.897-0.981). An optimal cutoff value was selected in the primary cohort, which had a sensitivity of 86.8%, a specificity of 93.3%, an accuracy of 92.2%, a positive predictive value of 71.7%, and a negative predictive value of 97.3%. The validation cohort had a sensitivity of 90.0%, a specificity of 90.3%, an accuracy of 90.2%, a positive predictive value of 71.1%, and a negative predictive value of 97.1%. Among the 375 patients, a sensitivity of 87.3%, a specificity of 92.1%, an accuracy of 91.2%, a positive predictive value of 72.1%, and a negative predictive value of 96.9% were found. Conclusions and Relevance This study's findings suggest that the collagen signature in the tumor microenvironment is an independent indicator of LNM in EGC, and the prediction model based on this collagen signature may be useful in treatment decision making for patients with EGC.
Collapse
Affiliation(s)
- Dexin Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Gang Chen
- Department of Pathology, Fujian Provincial Cancer Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Wei Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Meiting Fu
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Wenju Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,Department of Pathology, Fujian Provincial Cancer Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Jian Sui
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Shuoyu Xu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Zhangyuanzhu Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoling Zheng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,Department of Endoscopy Center, Fujian Provincial Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Liangjie Chi
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Dajia Lin
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.,Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Kai Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Weisheng Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ning Zuo
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Jianping Lu
- Department of Pathology, Fujian Provincial Cancer Hospital, Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Shuangmu Zhuo
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, Fujian, People's Republic of China
| | - Jun Yan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
537
|
Wang FH, Shen L, Li J, Zhou ZW, Liang H, Zhang XT, Tang L, Xin Y, Jin J, Zhang YJ, Yuan XL, Liu TS, Li GX, Wu Q, Xu HM, Ji JF, Li YF, Wang X, Yu S, Liu H, Guan WL, Xu RH. The Chinese Society of Clinical Oncology (CSCO): clinical guidelines for the diagnosis and treatment of gastric cancer. Cancer Commun (Lond) 2019; 39:10. [PMID: 30885279 PMCID: PMC6423835 DOI: 10.1186/s40880-019-0349-9] [Citation(s) in RCA: 306] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/01/2019] [Indexed: 02/08/2023] Open
Abstract
China is one of the countries with the highest incidence of gastric cancer. There are differences in epidemiological characteristics, clinicopathological features, tumor biological characteristics, treatment patterns, and drug selection between gastric cancer patients from the Eastern and Western countries. Non-Chinese guidelines cannot specifically reflect the diagnosis and treatment characteristics for the Chinese gastric cancer patients. The Chinese Society of Clinical Oncology (CSCO) arranged for a panel of senior experts specializing in all sub-specialties of gastric cancer to compile, discuss, and revise the guidelines on the diagnosis and treatment of gastric cancer based on the findings of evidence-based medicine in China and abroad. By referring to the opinions of industry experts, taking into account of regional differences, giving full consideration to the accessibility of diagnosis and treatment resources, these experts have conducted experts' consensus judgement on relevant evidence and made various grades of recommendations for the clinical diagnosis and treatment of gastric cancer to reflect the value of cancer treatment and meeting health economic indexes. This guideline uses tables and is complemented by explanatory and descriptive notes covering the diagnosis, comprehensive treatment, and follow-up visits for gastric cancer.
Collapse
Affiliation(s)
- Feng-Hua Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Jin Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120 P. R. China
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Han Liang
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Tianjin’s Clinical Research Cancer for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 P. R. China
| | - Xiao-Tian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Lei Tang
- Medical Imaging Department, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Yan Xin
- Pathology Laboratory of Gastrointestinal Tumor, The First Hospital of China Medical University, Shenyang, 110001 Liaoning P. R. China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center, China and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 P. R. China
| | - Yu-Jing Zhang
- Department of Radiotherapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Xiang-Lin Yuan
- Department of Medical Oncology, Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430030 Hubei P. R. China
| | - Tian-Shu Liu
- Department of Medical Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032 P. R. China
| | - Guo-Xin Li
- Department of General Surgery, Nanfang Hospital Affiliated to Southern Medical University, Guangzhou, 510515 Guangdong P. R. China
| | - Qi Wu
- Department of Endoscopy Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Hui-Mian Xu
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, 110001 Liaoning P. R. China
| | - Jia-Fu Ji
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142 P. R. China
| | - Yuan-Fang Li
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Xin Wang
- Department of Radiation Oncology, National Cancer Center, China and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 P. R. China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032 P. R. China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital Affiliated to Southern Medical University, Guangzhou, 510515 Guangdong P. R. China
| | - Wen-Long Guan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 Guangdong P. R. China
| |
Collapse
|
538
|
Ding H, Sun J, Li R, Wang G. Retracted Article: Long non-coding RNA GACAT1 alleviates doxorubicin and vincristine resistance through a PTEN/AKT/mTOR/S6K1 regulatory pathway in gastric cancer. RSC Adv 2019; 9:8048-8055. [PMID: 35521206 PMCID: PMC9061239 DOI: 10.1039/c8ra10030f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/22/2019] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is a major global health problem. Chemotherapy is a common therapeutic strategy for cancers including GC. However, chemoresistance strikingly limits the clinical applications of chemotherapeutic drugs. Long non-coding RNAs (lncRNAs) have been widely reported to be implicated in the pathogenesis and chemoresistance of cancers including GC. Our work aims to investigate the roles and molecular mechanisms of lncRNA gastric cancer-associated transcript 1 (GACAT1) in regulating doxorubicin (ADR) and vincristine (VCR) resistance in GC. In this text, RT-qPCR assay showed that GACAT1 expression was markedly reduced in ADR- or VCR-resistant GC (SGC7901/ADR or SGC7901/VCR) cells and GC tissues. CCK-8 assay and flow cytometry analysis revealed that GACAT1 overexpression alleviated the resistance of GC cells to ADR and VCR. RT-qPCR and western blot assay disclosed that GACAT1 deactivated the AKT/mTOR/S6K1 signaling pathway and promoted PTEN expression in SGC7901/ADR or SGC7901/VCR cells. Restoration experiments demonstrated that GACAT1 attenuated ADR or VCR resistance by regulating the PTEN/AKT/mTOR/S6K1 pathway in SGC7901/ADR or SGC7901/VCR cells. In vivo experiments demonstrated that GACAT1 overexpression inhibited tumor growth and enhanced ADR- or VCR-mediated anti-tumor effects in GC xenograft tumor models. Taken together, these data revealed that GACAT1 weakened the resistance of GC cells to ADR and VCR by the PTEN/AKT/mTOR/S6K1 regulatory pathway in vitro and in vivo, shedding new light on GACAT1 upregulation as a potential strategy to alleviate chemoresistance in GC.
Collapse
Affiliation(s)
- Hengxuan Ding
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University Jianshe Road 1 Zhengzhou 450052 P. R. China +86-0371-67967137
| | - Junfeng Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University Jianshe Road 1 Zhengzhou 450052 P. R. China +86-0371-67967137
| | - Ruixin Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University Jianshe Road 1 Zhengzhou 450052 P. R. China +86-0371-67967137
| | - Guojun Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University Jianshe Road 1 Zhengzhou 450052 P. R. China +86-0371-67967137
| |
Collapse
|
539
|
Sakata T, Takahata T, Kimura T, Yasuhara I, Kojima T, Akazai Y, Mimura T, Lefor AK. A single-institution retrospective analysis of gastric carcinoma with positive peritoneal lavage cytology and without serosal invasion: A case series. Ann Med Surg (Lond) 2019; 39:10-15. [PMID: 30792857 PMCID: PMC6370578 DOI: 10.1016/j.amsu.2019.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/21/2018] [Accepted: 01/23/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Peritoneal dissemination of gastric cancer is often associated with serosal infiltration. The aim of this study was to evaluate the clinical importance of peritoneal lavage cytology in patients with gastric carcinoma without serosal invasion. The incidence and impact on prognosis of positive cytology were analyzed. METHODS Of 2768 patients with gastric cancer, outcomes and pathological characteristics of 973 patients were reviewed retrospectively. All patients underwent peritoneal lavage at laparotomy for curative or palliative resection of gastric cancer between 1999 and 2017. Among these, 479 who underwent surgery from January 1999 to March 2012 were also reviewed to analyze 5-year survival. RESULTS Of 973 patients enrolled, 338 (35%) did not have serosal invasion, and peritoneal cytology was positive in 4/338 (1.2%). Of these four patients, one had submucosal invasion and three had muscularis propria invasion. Of 635 patients with serosal invasion, peritoneal cytology was positive in 74/635 (12%). Of 479 patients reviewed for survival, cytology was positive in 32/479, with 3/32 (9%) surviving for five years, and cytology was negative in 447 patients with 266/447 (60%) surviving for five years. CONCLUSIONS Cytologic evaluation should be routinely performed in patients with early-stage gastric cancer.
Collapse
Affiliation(s)
- Taizo Sakata
- Department of Surgery, Okayama Saiseikai General Hospital, Okayama, Japan
| | - Takaomi Takahata
- Department of Surgery, Okayama Saiseikai General Hospital, Okayama, Japan
| | - Toshikazu Kimura
- Department of Surgery, Okayama Saiseikai General Hospital, Okayama, Japan
| | - Isao Yasuhara
- Department of Surgery, Okayama Saiseikai General Hospital, Okayama, Japan
| | - Toru Kojima
- Department of Surgery, Okayama Saiseikai General Hospital, Okayama, Japan
| | - Yoshihiro Akazai
- Department of Surgery, Okayama Saiseikai General Hospital, Okayama, Japan
| | - Tetsushige Mimura
- Department of Surgery, Okayama Saiseikai General Hospital, Okayama, Japan
| | | |
Collapse
|
540
|
Garg PK, Jara M, Alberto M, Rau B. The role of Pressurized IntraPeritoneal Aerosol Chemotherapy in the management of gastric cancer: A systematic review. Pleura Peritoneum 2019; 4:20180127. [PMID: 31198852 PMCID: PMC6545873 DOI: 10.1515/pp-2018-0127] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/13/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The quest to cure or to contain the disease in cancer patients leads to new strategies and techniques being added to the armamentarium of oncologists. Pressurized IntraPeritoneal Aerosol Chemotherapy (PIPAC) is a recently described surgical technique which is being evaluated at many centers for the management of peritoneal metastasis (PM). The present study is a systematic review to evaluate the current role of PIPAC in the management of gastric cancer associated PM. METHODS A systematic search was conducted in Pubmed and EMBASE database using relevant keywords and confirming to the PRISMA guidelines to identify the articles describing the role of PIPAC in gastric cancer associated PM. All the studies which were published prior to July 1, 2018 in English literature and reported the role of PIPAC in gastric cancer associated PM were included in the systematic review. RESULTS The search yielded 79 articles; there were ten published studies which have reported the use of PIPAC in gastric cancer associated PM. A total of 129 patients with gastric cancer associated PM were treated in the studies. Only two studies had an exclusive cohort of gastric cancer patients while eight other studies had a heterogeneous population with a small proportion of gastric cancer patients. There was only one study highlighting the role of PIPAC in neoadjuvant setting to downgrade the peritoneal carcinomatosis index. All the studies revealed that PIPAC is feasible and has minimal perioperative morbidity, even after repeated applications. CONCLUSION There is a scarcity of English literature related to the role of PIPAC in gastric cancer associated PM. PIPAC is a safe and well-tolerated procedure which has the potential to contain spreading PM. Further studies are warranted to better define the role of PIPAC in gastric cancer associated PM.
Collapse
Affiliation(s)
- Pankaj Kumar Garg
- Department of Surgical Oncology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Maximilian Jara
- Department of General Surgery, Campus Virchow Klinikum, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Miguel Alberto
- Department of General Surgery, Campus Virchow Klinikum, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Beate Rau
- Department of General Surgery, Campus Virchow Klinikum, Charité, Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
541
|
Fuchs CS, Shitara K, Di Bartolomeo M, Lonardi S, Al-Batran SE, Van Cutsem E, Ilson DH, Alsina M, Chau I, Lacy J, Ducreux M, Mendez GA, Alavez AM, Takahari D, Mansoor W, Enzinger PC, Gorbounova V, Wainberg ZA, Hegewisch-Becker S, Ferry D, Lin J, Carlesi R, Das M, Shah MA, Karaseva NA, Kowalyszyn RD, Hernandez CA, Csoszi T, De Vita F, Pfeiffer P, Sugimoto N, Kocsis J, Csilla A, Bodoky G, Garnica Jaliffe G, Protsenko S, Madi A, Wojcik E, Brenner B, Folprecht G, Sarosiek T, Peltola KJ, Bono P, Ayala H, Aprile G, Gerardo CG, Huitzil Melendez FD, Falcone A, Di Costanzo F, Tehfe M, Mineur L, García Alfonso P, Obermannova R, Senellart H, Petty R, Samuel L, Acs PI, Hussein MA, Nechaeva MN, Erdkamp F, Won E, Bendell JC, Gallego Plazas J, Lorenzen S, Melichar B, Escudero MA, Pezet D, Phelip JM, Kaen DL, Reeves JAJ, Longo Muñoz F, Madhusudan S, Barone C, Fein LE, Gomez Villanueva A, Hebbar M, Prausova J, Visa Turmo L, Vidal Barrull J, Yilmaz MKN, Beny A, Van Laarhoven H, DiCarlo BA, Esaki T, Fujitani K, Geboes K, Geva R, Kadowaki S, Leong S, Machida N, Raj MS, Ramirez Godinez FJ, Ruzsa A, Ford H, Lawler WE, Maisey NR, Petera J, Shacham-Shmueli E, Sinapi I, et alFuchs CS, Shitara K, Di Bartolomeo M, Lonardi S, Al-Batran SE, Van Cutsem E, Ilson DH, Alsina M, Chau I, Lacy J, Ducreux M, Mendez GA, Alavez AM, Takahari D, Mansoor W, Enzinger PC, Gorbounova V, Wainberg ZA, Hegewisch-Becker S, Ferry D, Lin J, Carlesi R, Das M, Shah MA, Karaseva NA, Kowalyszyn RD, Hernandez CA, Csoszi T, De Vita F, Pfeiffer P, Sugimoto N, Kocsis J, Csilla A, Bodoky G, Garnica Jaliffe G, Protsenko S, Madi A, Wojcik E, Brenner B, Folprecht G, Sarosiek T, Peltola KJ, Bono P, Ayala H, Aprile G, Gerardo CG, Huitzil Melendez FD, Falcone A, Di Costanzo F, Tehfe M, Mineur L, García Alfonso P, Obermannova R, Senellart H, Petty R, Samuel L, Acs PI, Hussein MA, Nechaeva MN, Erdkamp F, Won E, Bendell JC, Gallego Plazas J, Lorenzen S, Melichar B, Escudero MA, Pezet D, Phelip JM, Kaen DL, Reeves JAJ, Longo Muñoz F, Madhusudan S, Barone C, Fein LE, Gomez Villanueva A, Hebbar M, Prausova J, Visa Turmo L, Vidal Barrull J, Yilmaz MKN, Beny A, Van Laarhoven H, DiCarlo BA, Esaki T, Fujitani K, Geboes K, Geva R, Kadowaki S, Leong S, Machida N, Raj MS, Ramirez Godinez FJ, Ruzsa A, Ford H, Lawler WE, Maisey NR, Petera J, Shacham-Shmueli E, Sinapi I, Yamaguchi K, Hara H, Beck JT, Błasińska-Morawiec M, Villalobos Valencia R, Alcindor T, Bajaj M, Berry S, Gomez CM, Dammrich D, Patel R, Taieb J, Ten Tije A, Burkes RL, Cabanillas F, Firdaus I, Chua CC, Hironaka S, Hofheinz RD, Lim HJ, Nordsmark M, Piko B, Verma U, Wadsley J, Yukisawa S, Gutiérrez Delgado F, Denlinger CS, Kallio R, Pikiel J, Wojcik-Tomaszewska J, Brezden-Masley C, Jang RWJ, Pribylova J, Sakai D, Bartoli MA, Cats A, Grootscholten M, Dichmann RA, Hool H, Shaib W, Tsuji A, Van den Eynde M, Velez-Cortez H, Asmis TR. Ramucirumab with cisplatin and fluoropyrimidine as first-line therapy in patients with metastatic gastric or junctional adenocarcinoma (RAINFALL): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2019; 20:420-435. [PMID: 30718072 DOI: 10.1016/s1470-2045(18)30791-5] [Show More Authors] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/03/2018] [Accepted: 10/16/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND VEGF and VEGF receptor 2 (VEGFR-2)-mediated signalling and angiogenesis can contribute to the pathogenesis and progression of gastric cancer. We aimed to assess whether the addition of ramucirumab, a VEGFR-2 antagonist monoclonal antibody, to first-line chemotherapy improves outcomes in patients with metastatic gastric or gastro-oesophageal junction adenocarcinoma. METHODS For this double-blind, randomised, placebo-controlled, phase 3 trial done at 126 centres in 20 countries, we recruited patients aged 18 years or older with metastatic, HER2-negative gastric or gastro-oesophageal junction adenocarcinoma, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, and adequate organ function. Eligible patients were randomly assigned (1:1) with an interactive web response system to receive cisplatin (80 mg/m2, on the first day) plus capecitabine (1000 mg/m2, twice daily for 14 days), every 21 days, and either ramucirumab (8 mg/kg) or placebo on days 1 and 8, every 21 days. 5-Fluorouracil (800 mg/m2 intravenous infusion on days 1-5) was permitted in patients unable to take capecitabine. The primary endpoint was investigator-assessed progression-free survival, analysed by intention to treat in the first 508 patients. We did a sensitivity analysis of the primary endpoint, including a central review of CT scans. Overall survival was a key secondary endpoint. This study is registered with ClinicalTrials.gov, number NCT02314117. FINDINGS Between Jan 28, 2015, and Sept 16, 2016, 645 patients were randomly assigned to receive ramucirumab plus fluoropyrimidine and cisplatin (n=326) or placebo plus fluoropyrimidine and cisplatin (n=319). Investigator-assessed progression-free survival was significantly longer in the ramucirumab group than the placebo group (hazard ratio [HR] 0·753, 95% CI 0·607-0·935, p=0·0106; median progression-free survival 5·7 months [5·5-6·5] vs 5·4 months [4·5-5·7]). A sensitivity analysis based on central independent review of the radiological images did not corroborate the investigator-assessed difference in progression-free survival (HR 0·961, 95% CI 0·768-1·203, p=0·74). There was no difference in overall survival between groups (0·962, 0·801-1·156, p=0·6757; median overall survival 11·2 months [9·9-11·9] in the ramucirumab group vs 10·7 months [9·5-11·9] in the placebo group). The most common grade 3-4 adverse events were neutropenia (85 [26%] of 323 patients in the ramucirumab group vs 85 [27%] of 315 in the placebo group), anaemia (39 [12%] vs 44 [14%]), and hypertension (32 [10%] vs 5 [2%]). The incidence of any-grade serious adverse events was 160 (50%) of 323 patients in the ramucirumab group and 149 (47%) of 315 patients in the placebo group. The most common serious adverse events were vomiting (14 [4%] in the ramucirumab group vs 21 [7%] in the placebo group) and diarrhoea (11 [3%] vs 19 [6%]). There were seven deaths in each group, either during study treatment or within 30 days of discontinuing study treatment, which were the result of treatment-related adverse events. In the ramucirumab group, these adverse events were acute kidney injury, cardiac arrest, gastric haemorrhage, peritonitis, pneumothorax, septic shock, and sudden death (n=1 of each). In the placebo group, these adverse events were cerebrovascular accident (n=1), multiple organ dysfunction syndrome (n=2), pulmonary embolism (n=2), sepsis (n=1), and small intestine perforation (n=1). INTERPRETATION Although the primary analysis for progression-free survival was statistically significant, this outcome was not confirmed in a sensitivity analysis of progression-free survival by central independent review, and did not improve overall survival. Therefore, the addition of ramucirumab to cisplatin plus fluoropyrimidine chemotherapy is not recommended as first-line treatment for this patient population. FUNDING Eli Lilly and Company.
Collapse
Affiliation(s)
- Charles S Fuchs
- Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT, USA.
| | - Kohei Shitara
- National Cancer Center Hospital East, Kashiwa, Japan
| | | | | | - Salah-Eddin Al-Batran
- Institute of Clinical Cancer Research at Krankenhaus Nordwest, UCT-University Cancer Center, Frankfurt, Germany
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg, Leuven and KULeuven, Belgium
| | - David H Ilson
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Alsina
- Vall d'Hebron University Hospital and Institute of Oncology, Barcelona, Spain
| | - Ian Chau
- Royal Marsden Hospital, Sutton, Surrey, United Kingdom
| | - Jill Lacy
- Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT, USA
| | - Michel Ducreux
- Gustave Roussy Cancer Centre, Grand Paris, Villejuif, France; Université Paris-Saclay, France
| | | | | | | | | | | | | | - Zev A Wainberg
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - David Ferry
- Eli Lilly and Company, New York City, NY, USA
| | - Ji Lin
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Mayukh Das
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Manish A Shah
- Weill Cornell Medical College, NY, USA; New York Presbyterian Hospital, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
542
|
Tu RH, Lin JX, Li P, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Zheng CH, Huang CM. The prognostic value of a Surgical Outcome Risk Tool in patients after radical gastrectomy for gastric cancer and its guiding significance for postoperative chemotherapy. Surg Oncol 2019; 28:128-134. [PMID: 30851886 DOI: 10.1016/j.suronc.2018.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 10/18/2018] [Accepted: 11/08/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To provide individualized survival predictors for patients after radical gastrectomy and to guide adjuvant treatment. METHODS The data of patients with gastric cancer treated in our department from January 1996 to December 2014 were analyzed retrospectively. A Surgical Outcome Risk Tool (SORT) was used to indicate the physical condition of the patients. RESULTS A total of 5327 patients in the study. The 5-year overall survival rates in the low-, moderate- and high-risk group were 61.6%, 54.7% and 44.4%, respectively (p < 0.001). A multivariate analysis showed that SORT was an independent prognostic factor for gastric cancer. There was significant chemotherapy benefit after surgery in stage II-III patients with a SORT<3 and in stage III patients with a SORT≥3 (p < 0.05). However, chemotherapy did not improve the 5-year overall survival in stage II patients with a SORT≥3 (62.4% vs. 48.3%, p = 0.196). The calculated difference between two nomogram models was the potential benefit of adjuvant chemotherapy for patients with gastric cancer. CONCLUSION A SORT is an independent risk factor for predicting the prognosis of gastric cancer, and postoperative adjuvant chemotherapy should be carefully adopted in stage II patients with a SORT≥3.
Collapse
Affiliation(s)
- Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
543
|
Liao G, Wang Z, Li H, Min J, Zhong J, Mariella S, Qian K, Zhang W. Learning curve and short-term outcomes of modularized LADG for advanced gastric cancer: A retrospective study. Medicine (Baltimore) 2019; 98:e14670. [PMID: 30855456 PMCID: PMC6417604 DOI: 10.1097/md.0000000000014670] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Laparoscopy-assisted distal gastrectomy (LADG) is a complicated procedure. To reduce the difficulty of the operation and standardize the surgical procedure, we explored a new operation mode, which we termed modularized LADG (MLADG). To further extend the new operation mode, we conducted this study to evaluate the short-term outcomes of MLADG for advanced gastric cancer, and determine the learning curve.Data from 100 consecutive patients who received LADG between October 2016 and October 2017 were retrospectively analyzed. Short-term outcomes, such as operation time and intraoperative blood loss, were evaluated, and the learning curve was calculated.For MLADG, the mean operation time was 168.2 ± 13.0 minutes, the mean intraoperative blood loss was 93.6 ± 29.1 ml, the mean number of harvested lymph nodes was 28.6 ± 4.2, and conversion to open surgery occurred in only 1 case. In addition, MLADG had an acceptable postoperative complication incidence and fast postoperative recovery. After the first 20 cases, the operation skill reached a mature and stable level.Our results indicate that MLADG is an oncologically feasible and technically safe surgical procedure. For the trainees with rich experience in open distal gastrectomy, the learning curve is considered to be completed after 20 MLADG cases.
Collapse
|
544
|
Lin H, Han D, Fu G, Liu C, Wang L, Han S, Liu B, Yu J. Concurrent apatinib and docetaxel vs apatinib monotherapy as third- or subsequent-line therapy for advanced gastric adenocarcinoma: a retrospective study. Onco Targets Ther 2019; 12:1681-1689. [PMID: 30881023 PMCID: PMC6400117 DOI: 10.2147/ott.s193801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose The aim of this study was to assess the efficacy and safety of concurrent apatinib and docetaxel therapy vs apatinib monotherapy as third- or subsequent-line treatment for advanced gastric adenocarcinoma (GAC). Methods Patients, who had received apatinib with or without docetaxel as third or more line therapy for advanced GAC, were retrospectively reviewed. Propensity score matching (PSM) analysis was performed to minimize the potential confounding bias. Kaplan–Meier curve and log-rank test were used to analyze the survival. Prognostic factors were estimated by Cox regression. Adverse events (AEs) were evaluated using CTCAE 4.0. Results Thirty-four patients received concurrent therapy, whereas 31 received monotherapy. The median progression-free survival (PFS) and overall survival (OS) in monotherapy and con-therapy groups were 2.5 and 4 months (P=0.002), 3.3 and 6 months (P=0.004), respectively. After PSM, the median PFS and OS in the con-therapy group were also superior to the monotherapy group (P=0.004 and P=0.017). Cox regression suggested that Eastern Cooperative Oncology Group performance status (ECOG PS; HR =2.437, 95% CI: 1.349–4.404, P=0.003), CA199 (HR =1.001, 95% CI: 1.000–1.002, P=0.016), and treatment options (HR =0.388, 95% CI: 0.222–0.679, P=0.001) had significant effects on OS. Grade 3/4 toxicities in the monotherapy and con-therapy groups were as follows: leukopenia (0% vs 8.8%), neutropenia (3.2% vs 2.9%), anemia (9.8% vs 8.8%), thrombocytopenia (6.4% vs 2.9%), proteinuria (3.2% vs 2.9%), aminotransferase (0% vs 11.8%), hyperbilirubinemia (9.8% vs 5.9%), hypertension (9.8% vs5.9%), hand–foot syndrome (3.2% vs 8.8%), nausea and vomiting (0% vs 11.8%), diarrhea (0% vs 5.9%), and fatigue (6.5% vs 2.9%). Conclusion Patients with advanced GAC benefit more from concurrent apatinib and docetaxel therapy than apatinib monotherapy.
Collapse
Affiliation(s)
- Haimin Lin
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong 250117, China, .,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong 250200, China
| | - Dali Han
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong 250117, China,
| | - Guobin Fu
- Department of Medical Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Chengxin Liu
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong 250117, China,
| | - Lili Wang
- Department of Bone and Soft Tumor, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong 250117, China
| | - Shumei Han
- Department of Medical Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong 250117, China,
| | - Bo Liu
- Department of Medical Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong 250117, China,
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong 250117, China,
| |
Collapse
|
545
|
Skill N, Maluccio M. Contemporary paradigm for the evaluation and treatment of hereditary gastric cancer. Transl Gastroenterol Hepatol 2019; 4:14. [PMID: 30976717 DOI: 10.21037/tgh.2019.01.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 01/27/2019] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the third leading cause of cancer mortality worldwide. Survival is linked to stage at diagnosis and tolerance to surgery and adjuvant therapy. The emergence of sophisticated methods to identify patients at high risk for the development of gastric cancer has given us an opportunity to eliminate a lethal disease in an identifiable patient population. Guidelines and recommendations have been established and prophylactic total gastrectomy is considered the most effective treatment. However, this requires substantial physical and emotional investment. It is imperative that patients and families are supported by genetic counseling, ongoing surveillance, and survivorship studies.
Collapse
Affiliation(s)
- Nicholas Skill
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mary Maluccio
- Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
546
|
Guo J, Chen Y, Liu Z, Guan Y, Wang W, Sun X, Li W, Xu D. The Diagnostic and Prognostic Value of Digital Rectal Examination in Gastric Cancer Patients with Peritoneal Metastasis. J Cancer 2019; 10:1489-1495. [PMID: 31031858 PMCID: PMC6485225 DOI: 10.7150/jca.29814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/09/2019] [Indexed: 12/27/2022] Open
Abstract
Background: Peritoneal metastasis (PM) is the most common cause of death in gastric cancer (GC) patients. However, diagnosis of PM is still difficult in clinical practice. This study aimed to explore the diagnostic and prognostic value of digital rectal examination (DRE) in GC. Methods: 247 GC patients with PM confirmed by operation were included. The diagnostic yield of DRE compared with computed tomography (CT) was calculated. In another group of 1330 cases receiving radical gastrectomy, 38 cases with DRE (+) postoperatively were analyzed to identify risk factors. A nomogram was constructed to predict postoperative DRE (+). Results: The specificity, positive predictive value and positive likelihood ratio of DRE in diagnosis of PM was 99.8%, 91.2% and 58.4, higher than CT (97.6%, 64.9% and 10.4). Though the sensitivity of DRE (12.6%) was lower than CT (24.7%), 17 of 31 patients with DRE (+) could not be found by CT. Moreover, the overall survival of confirmed PM patients with DRE (+) (PM-DRE (+)) was much lower than PM-DRE (-) patients (P<0.001). In addition, the nomogram to predict postoperative DRE (+) had a bootstrap-corrected concordance index of 0.73 and was well calibrated. Conclusions: GC patients with DRE (+) could be regarded as a special subtype of stage IV ones with poorer prognosis. Supply of palliative care and chemotherapy rather than unnecessary operation might be a better alternative for these patients. DRE was an effective supplement for CT and should be generally recommended for GC patients.
Collapse
Affiliation(s)
- Jing Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yongming Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhimin Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanxiang Guan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaowei Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dazhi Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
547
|
He W, Liang B, Wang C, Li S, Zhao Y, Huang Q, Liu Z, Yao Z, Wu Q, Liao W, Zhang S, Liu Y, Xiang Y, Liu J, Shi M. MSC-regulated lncRNA MACC1-AS1 promotes stemness and chemoresistance through fatty acid oxidation in gastric cancer. Oncogene 2019; 38:4637-4654. [PMID: 30742067 PMCID: PMC6756048 DOI: 10.1038/s41388-019-0747-0] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 01/10/2019] [Accepted: 01/29/2019] [Indexed: 02/08/2023]
Abstract
Chemotherapy is the preferred treatment for advanced stage gastric cancer (GC) patients and chemotherapy resistance is the major obstacle to effective cancer therapy. Increasing evidence suggests that mesenchymal stem cells (MSCs) make important contributions to development of drug resistance. However, the underlying mechanism remains elusive. In this study, we discovered that abundant MSCs in tumor tissues predicted a poor prognosis in GC patients. MSCs promoted stemness and chemoresistance in GC cells through fatty acid oxidation (FAO) in vitro and in vivo. Mechanically, transforming growth factor β1 (TGF-β1) secretion by MSCs activated SMAD2/3 through TGF-β receptors and induced long non-coding RNA (lncRNA) MACC1-AS1 expression in GC cells, which promoted FAO-dependent stemness and chemoresistance through antagonizing miR-145-5p. Moreover, pharmacologic inhibition of FAO with etomoxir (ETX) attenuated MSC-induced FOLFOX regiment resistance in vivo. These results suggest that FAO plays an important role in MSC-mediated stemness and chemotherapy resistance in GC and FAO inhibitors in combination with chemotherapeutic drugs present as a promising strategy to overcome chemoresistance.
Collapse
Affiliation(s)
- Wanming He
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bishan Liang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunlin Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaowei Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Zhao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiong Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zexian Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Zhiqi Yao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qijing Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuyi Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yajing Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Xiang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
548
|
van den Boorn HG, Abu-Hanna A, Ter Veer E, van Kleef JJ, Lordick F, Stahl M, Ajani JA, Guimbaud R, Park SH, Dutton SJ, Bang YJ, Boku N, Mohammad NH, Sprangers MAG, Verhoeven RHA, Zwinderman AH, van Oijen MGH, van Laarhoven HWM. SOURCE: A Registry-Based Prediction Model for Overall Survival in Patients with Metastatic Oesophageal or Gastric Cancer. Cancers (Basel) 2019; 11:187. [PMID: 30764578 PMCID: PMC6406639 DOI: 10.3390/cancers11020187] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/19/2018] [Accepted: 01/10/2019] [Indexed: 02/08/2023] Open
Abstract
Prediction models are only sparsely available for metastatic oesophagogastric cancer. Because treatment in this setting is often preference-based, decision-making with the aid of a prediction model is wanted. The aim of this study is to construct a prediction model, called SOURCE, for the overall survival in patients with metastatic oesophagogastric cancer. Data from patients with metastatic oesophageal (n = 8010) or gastric (n = 4763) cancer diagnosed during 2005⁻2015 were retrieved from the nationwide Netherlands cancer registry. A multivariate Cox regression model was created to predict overall survival for various treatments. Predictor selection was performed via the Akaike Information Criterion and a Delphi consensus among experts in palliative oesophagogastric cancer. Validation was performed according to a temporal internal-external scheme. The predictive quality was assessed with the concordance-index (c-index) and calibration. The model c-indices showed consistent discriminative ability during validation: 0.71 for oesophageal cancer and 0.68 for gastric cancer. The calibration showed an average slope of 1.0 and intercept of 0.0 for both tumour locations, indicating a close agreement between predicted and observed survival. With a fair c-index and good calibration, SOURCE provides a solid foundation for further investigation in clinical practice to determine its added value in shared decision making.
Collapse
Affiliation(s)
- Héctor G van den Boorn
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Ameen Abu-Hanna
- Department of Medical Informatics, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Emil Ter Veer
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Jessy Joy van Kleef
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Florian Lordick
- 1st Medical Department, University Cancer Center Leipzig (UCCL), University Hospital Leipzig, 04103 Leipzig, Germany.
| | - Michael Stahl
- Department of Medical Oncology and Hematology, Kliniken Essen-Mitte, 45136 Essen, Germany.
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, TX 77030, USA.
| | - Rosine Guimbaud
- Department of Medical Oncology, Centre Hospitalo-Univeristaire de Toulouse, 31400 Toulouse, France.
| | - Se Hoon Park
- University School of Medicine, Samsung Medical Center, Sungkyunkwan, 06351 Seoul, Korea.
| | - Susan J Dutton
- Oxford Clinical Trials Research Unit and Centre for Statistics in Medicine, University of Oxford, OX1 2JD Oxford, UK.
| | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul National University Hospital, 03080 Seoul, Korea.
| | - Narikazu Boku
- Department of Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, 104-0045 Tokyo, Japan.
| | - Nadia Haj Mohammad
- Department of Medical Oncology, UMC Utrecht, 3584 CX Utrecht, Utrecht University, The Netherlands.
| | - Mirjam A G Sprangers
- Department of Medical Psychology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Rob H A Verhoeven
- Netherlands Comprehensive Cancer Organization (IKNL), 5612 HZ Eindhoven, The Netherlands.
- Department of Surgery, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Martijn G H van Oijen
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
549
|
Boku N, Ryu MH, Kato K, Chung HC, Minashi K, Lee KW, Cho H, Kang WK, Komatsu Y, Tsuda M, Yamaguchi K, Hara H, Fumita S, Azuma M, Chen LT, Kang YK. Safety and efficacy of nivolumab in combination with S-1/capecitabine plus oxaliplatin in patients with previously untreated, unresectable, advanced, or recurrent gastric/gastroesophageal junction cancer: interim results of a randomized, phase II trial (ATTRACTION-4). Ann Oncol 2019; 30:250-258. [PMID: 30566590 PMCID: PMC6386029 DOI: 10.1093/annonc/mdy540] [Citation(s) in RCA: 294] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Nivolumab is approved as an option for third- or later-line treatment of advanced gastric/gastroesophageal junction (G/GEJ) cancer in several countries after ATTRACTION-2. To further improve the therapeutic efficacy of first-line therapy, exploration of a nivolumab-chemotherapy combination is warranted. In part 1 (phase II) of ATTRACTION-4, the safety and efficacy of nivolumab combined with S-1 plus oxaliplatin (SOX) or capecitabine plus oxaliplatin (CapeOX) as first-line therapy for unresectable advanced or recurrent human epidermal growth factor receptor 2 (HER2)-negative G/GEJ cancer were evaluated. PATIENTS AND METHODS Patients were randomized (1 : 1) to receive nivolumab (360 mg intravenously every 3 weeks) plus SOX (S-1, 40 mg/m2 orally twice daily for 14 days followed by 7 days off; oxaliplatin, 130 mg/m2 intravenously on day 1 every 3 weeks) or CapeOX (capecitabine, 1000 mg/m2 orally twice daily for 14 days followed by 7 days off; oxaliplatin, 130 mg/m2 intravenously on day 1 every 3 weeks) until disease progression, unacceptable toxicity, or consent withdrawal. RESULTS Of 40 randomized patients, 39 (nivolumab plus SOX, 21; nivolumab plus CapeOX, 18) and 38 (21 and 17, respectively) comprised the safety and efficacy populations, respectively. Most frequent (>10%) grade 3/4 treatment-related adverse events were neutropenia (14.3%) in the nivolumab plus SOX group, and neutropenia (16.7%), anemia, peripheral sensory neuropathy, decreased appetite, type 1 diabetes mellitus, and nausea (11.1% each) in the nivolumab plus CapeOX group. No treatment-related death occurred. Objective response rate was 57.1% (95% confidence interval 34.0-78.2) with nivolumab plus SOX and 76.5% (50.1-93.2) with nivolumab plus CapeOX. Median overall survival was not reached (NR) in both groups. Median progression-free survival was 9.7 months (5.8-NR) and 10.6 months (5.6-12.5), respectively. CONCLUSION Nivolumab combined with SOX/CapeOX was well tolerated and demonstrated encouraging efficacy for unresectable advanced or recurrent HER2-negative G/GEJ cancer. ATTRACTION-4 has proceeded to part 2 (phase III) to compare nivolumab plus SOX/CapeOX versus placebo plus SOX/CapeOX. CLINICALTRIALS.GOV ID NCT02746796.
Collapse
Affiliation(s)
- N Boku
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - M-H Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - K Kato
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - H C Chung
- Department of Medical Oncology, Yonsei Cancer Center, Song Dang Institute for Cancer Research, Yonsei University College of Medicine, Yonsei University Health System, Seoul, South Korea
| | - K Minashi
- Clinical Trial Promotion Department, Chiba Cancer Center, Chiba, Japan
| | - K-W Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - H Cho
- Department of Surgery, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Tokyo, Japan (previously Kanagawa Cancer Center, Yokohama, Japan
| | - W K Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Y Komatsu
- Division of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Sapporo, Japan
| | - M Tsuda
- Department of Gastroenterological Oncology, Hyogo Cancer Center, Akashi, Japan
| | - K Yamaguchi
- Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - H Hara
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - S Fumita
- Department of Medical Oncology, Nara Hospital Kindai University, Ikoma, Japan
| | - M Azuma
- Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa, Japan
| | - L-T Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Y-K Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
550
|
A Simplified Two-Step Technique for Extended Lymphadenectomy During Resection of Gastroesophageal Malignancy: Early Results Compared to En Bloc Dissection. J Gastrointest Surg 2019; 23:393-401. [PMID: 30603860 DOI: 10.1007/s11605-018-4056-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/13/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Extended lymph node dissection (ELND) remains an important component of curative intent resection of mid-stage gastric cancer (GC). Benefits include enhanced staging accuracy, extending regional disease control, and optimizing potential curability. ELND during gastrectomy remains underutilized in US centers due to a low prevalence of GC operations. METHODS The traditional en bloc ELND was modified into a two-step technique to facilitate greater ease of dissection with better exposure. After completion of the gastrectomy component, retrogastric nodes are dissected in a separate, contiguous specimen. Resulting data were compared to outcomes after en bloc resection. RESULTS Of 179 consecutive patients undergoing gastrectomy, 129 underwent an ELND (73%). There were 97 men and 32 women, with a median age of 64 years (range 24-98). The median total LN count was 25 (3-86). The two-step dissection yielded an average of 18.3 (± 8.5 S.D.) perigastric and 12.1 (± 5.8) retrogastric nodes. Two-step LND was associated with lower estimated blood loss (265 vs. 448 ml, p = 0.0005), lower transfusion requirements (6 vs. 28%, p = 0.007), greater mean total LN counts (30 vs. 26, p = 0.03), and a greater rate of obtaining at least 15 or 20 LNs (91 vs. 77% and 83 vs. 65%, p = 0.05). Major morbidity (overall 16%), length of stay, and survival outcomes were not different. CONCLUSIONS The two-step LND technique as described was found to be associated with favorable operative and postoperative outcome parameters and an excellent LN yield. It can be recommended for standard ELND indications in the absence of macroscopically abnormal LNs.
Collapse
|