201
|
Yu J, Wang Z, Li Z, Liu Y, Fan Y, Di J, Cui M, Xing J, Zhang C, Yang H, Yao Z, Zhang N, Chen L, Liu M, Xu K, Tan F, Gao P, Su X. Health-Related Quality of Life in Patients With Locally Advanced Gastric Cancer Undergoing Perioperative or Postoperative Adjuvant S-1 Plus Oxaliplatin With D2 Gastrectomy: A Propensity Score-Matched Cohort Study. Front Oncol 2022; 12:853337. [PMID: 35444949 PMCID: PMC9013949 DOI: 10.3389/fonc.2022.853337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/09/2022] [Indexed: 11/22/2022] Open
Abstract
Background Some high-quality clinical trials have proven the efficacy and safety of perioperative and postoperative S-1 with oxaliplatin (peri-SOX and post-SOX) for patients with locally advanced gastric cancer (LAGC) undergoing D2 gastrectomy. However, little is known about how health-related quality of life (HRQOL) changes over time in patients receiving peri-SOX or post-SOX chemotherapy. Methods A prospective observational cohort (NCT04408859) identified 151 eligible patients with LAGC who underwent D2 gastrectomy with at least six cycles of peri-SOX or post-SOX chemotherapy from 2018 to 2020. HRQOL was assessed using the EROTC QLQ-C30 and its gastric module, QLQ-STO22, at indicated measurements, including the baseline, 1st, 3rd, 6th and 12th month after initiation of therapy. Baseline characteristics, therapeutic effects, and longitudinal HRQOL were compared between the peri-SOX and post-SOX groups after propensity score matching. HRQOL changes over time and the risk factors for scales with severe deterioration were further analyzed. Results No statistically significant differences in longitudinal HRQOL were observed between patients in the peri-SOX and post-SOX groups, with comparable surgical outcomes and adverse chemotherapy events. Scales of social functioning, abnormal taste, and anxiety improved earlier in the peri-SOX group than in the post-SOX group. Score changes in both groups indicated that general deterioration and slower recovery usually occurred in the scales of physical, social, and role functioning, as well as symptoms of fatigue, reflux, diarrhea, and anxiety. Conclusion Peri-SOX showed a longitudinal HRQOL comparable to post-SOX in patients with LAGC who underwent D2 gastrectomy. The peri-SOX group had better performance in social functioning, abnormal taste, and anxiety at some measurements.
Collapse
Affiliation(s)
- Jianhong Yu
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zaozao Wang
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhexuan Li
- Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ying Liu
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yingcong Fan
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiabo Di
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ming Cui
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiadi Xing
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Chenghai Zhang
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Hong Yang
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhendan Yao
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Nan Zhang
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Lei Chen
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Maoxing Liu
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kai Xu
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Fei Tan
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Pin Gao
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiangqian Su
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
202
|
Garcia-Nebreda M, Zorrilla-Vaca A, Ripollés-Melchor J, Abad-Motos A, Alvaro Cifuentes E, Abad-Gurumeta A, Mena GE, Grant MC, Paseiro-Crespo G. Early Return to Intended Oncologic Therapy after implementation of an Enhanced Recovery After Surgery pathway for gastric cancer surgery. Langenbecks Arch Surg 2022; 407:2293-2300. [PMID: 35441358 DOI: 10.1007/s00423-022-02515-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Time to initiation and completion of adjuvant therapy are critical to improve postoperative oncologic outcomes. This study aims to determine whether an Enhanced Recovery After Surgery (ERAS) pathway for gastric cancer surgery promotes early Return to Intended Oncologic Therapy (RIOT). METHODS This is a before-after intervention study including patients with gastric adenocarcinoma who underwent surgery from January 2016 to January 2021. Two periods were denoted based upon the implementation date of our institutional ERAS pathway (June 2018). Our primary outcome was time to RIOT after surgery. Hodges-Lehmann analysis was used to estimate median differences of non-parametric outcomes. RESULTS Seventy patients with gastric adenocarcinoma were included (35 in pre-ERAS period and 35 in post-ERAS period). Fourteen of the pre-ERAS and twenty-two patients of the post-ERAS period received adjuvant therapy. Time to RIOT was reduced in the post-ERAS period (median 39 days, IQR 31-49) by 12 days (95% CI 3-14 days, p = 0.01) compared to the pre-ERAS period (median 51 days, IQR 42-62). Length of hospital stay (LOS) was lower in the ERAS group (6 days, IQR 5-11 vs 10 days, IQR 8-13, p < 0.01). CONCLUSION Our institutional ERAS pathway for gastric cancer surgery was associated with earlier RIOT and shorter LOS.
Collapse
Affiliation(s)
- Maria Garcia-Nebreda
- Department of Surgery, Infanta Leonor University Hospital, Madrid, Spain. .,Grupo Español de Rehabilitación Multimodal (GERM), Zaragoza, Spain. .,Universidad Complutense de Madrid, Madrid, Spain.
| | - Andrés Zorrilla-Vaca
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Javier Ripollés-Melchor
- Grupo Español de Rehabilitación Multimodal (GERM), Zaragoza, Spain.,Universidad Complutense de Madrid, Madrid, Spain.,Department of Anesthesiology, Infanta Leonor University Hospital, Madrid, Spain.,Spanish Perioperative Audit and Research Network (REDGERM), Zaragoza, Spain
| | - Ane Abad-Motos
- Grupo Español de Rehabilitación Multimodal (GERM), Zaragoza, Spain.,Universidad Complutense de Madrid, Madrid, Spain.,Department of Anesthesiology, Infanta Leonor University Hospital, Madrid, Spain
| | - Edurne Alvaro Cifuentes
- Department of Surgery, Infanta Leonor University Hospital, Madrid, Spain.,Universidad Complutense de Madrid, Madrid, Spain
| | - Alfredo Abad-Gurumeta
- Grupo Español de Rehabilitación Multimodal (GERM), Zaragoza, Spain.,Universidad Complutense de Madrid, Madrid, Spain.,Department of Anesthesiology, Infanta Leonor University Hospital, Madrid, Spain.,Spanish Perioperative Audit and Research Network (REDGERM), Zaragoza, Spain
| | - Gabriel E Mena
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael C Grant
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Gloria Paseiro-Crespo
- Department of Surgery, Infanta Leonor University Hospital, Madrid, Spain.,Grupo Español de Rehabilitación Multimodal (GERM), Zaragoza, Spain.,Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
203
|
Wang YF, Yin X, Fang TY, Wang YM, Zhang L, Zhang XH, Zhang DX, Zhang Y, Wang XB, Wang H, Xue YW. Prognostic significance of serum inflammation indices for different tumor infiltrative pattern types of gastric cancer. World J Gastrointest Oncol 2022; 14:897-919. [PMID: 35582101 PMCID: PMC9048526 DOI: 10.4251/wjgo.v14.i4.897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/08/2021] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammatory indices are considered to be potential prognostic biomarkers for patients with gastric cancer (GC). However, there is no evidence defining the prognostic significance of inflammatory indices for GC with different tumor infiltrative pattern (INF) types.
AIM To evaluate the significance of inflammatory indices and INF types in predicting the prognosis of patients with GC.
METHODS A total of 962 patients who underwent radical gastrectomy were retrospectively selected for this study. Patients were categorized into the expansive growth type (INFa), the intermediate type (INFb), and the infiltrative growth type (INFc) groups. The cutoff values of inflammatory indices were analyzed by receiver operating characteristic curves. The Kaplan–Meier method and log-rank test were used to analyze overall survival (OS). The chi-square test was used to analyze the association between inflammatory indices and clinical characteristics. The independent risk factors for prognosis in each group were analyzed by univariate and multivariate analyses based on logistic regression. Nomogram models were constructed by R studio.
RESULTS The INFc group had the worst OS (P < 0.001). The systemic immune-inflammation index (P = 0.039) and metastatic lymph node ratio (mLNR) (P = 0.003) were independent risk factors for prognosis in the INFa group. The platelet-lymphocyte ratio (PLR) (P = 0.018), age (P = 0.026), body mass index (P = 0.003), and postsurgical tumor node metastasis (pTNM) stage (P < 0.001) were independent risk factors for prognosis in the INFb group. The PLR (P = 0.021), pTNM stage (P = 0.028), age (P = 0.021), and mLNR (P = 0.002) were independent risk factors for prognosis in the INFc group. The area under the curve of the nomogram model for predicting 5-year survival in the INFa group, INFb group, and INFc group was 0.787, 0.823, and 0.781, respectively.
CONCLUSION The outcome of different INF types GC patients could be assessed by nomograms based on different inflammatory indices and clinicopathologic features.
Collapse
Affiliation(s)
- Yu-Fei Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Xin Yin
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Tian-Yi Fang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Yi-Min Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Lei Zhang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Xing-Hai Zhang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Dao-Xu Zhang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Yao Zhang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Xi-Bo Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Hao Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Ying-Wei Xue
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| |
Collapse
|
204
|
Yang L, Sun J, Yu X, Li Y, Li M, Liu J, Wang X, Shi G. Diagnosis of Serosal Invasion in Gastric Adenocarcinoma by Dual-Energy CT Radiomics: Focusing on Localized Gastric Wall and Peritumoral Radiomics Features. Front Oncol 2022; 12:848425. [PMID: 35387116 PMCID: PMC8977467 DOI: 10.3389/fonc.2022.848425] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives To build a radiomics model and combined model based on dual-energy CT (DECT) for diagnosing serosal invasion in gastric adenocarcinoma. Materials and methods 231 gastric adenocarcinoma patients were enrolled and randomly divided into a training (n = 132), testing (n = 58), and independent validation (n = 41) cohort. Radiomics features were extracted from the rectangular ROI of the 120-kV equivalent mixed images and iodine map (IM) images in the venous phase of DECT, which was manually delineated perpendicularly to the gastric wall in the deepest location of tumor infiltration, including the peritumoral adipose tissue within 5 mm outside the serosa. The random forest algorithm was used for radiomics model construction. Traditional features were collected by two radiologists. Univariate and multivariate logistic regression was used to construct the clinical model and combined model. The diagnostic efficacy of the models was evaluated using ROC curve analysis and compared using the Delong's test. The calibration curves were used to evaluate the calibration performance of the combined model. Results Both the radiomics model and combined model showed high efficacy in diagnosing serosal invasion in the training, testing and independent validation cohort, with AUC of 0.90, 0.90, and 0.85 for radiomics model; 0.93, 0.93, and 0.89 for combined model. The combined model outperformed the clinical model (AUC: 0.76, 0.76 and 0.81). Conclusion The radiomics model and combined model constructed based on tumoral and peritumoral radiomics features derived from DECT showed high diagnostic efficacy for serosal invasion in gastric adenocarcinoma.
Collapse
Affiliation(s)
- Li Yang
- Department of Computed Tomography and Magnetic Resonance, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Junyi Sun
- Department of Computed Tomography and Magnetic Resonance, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xianbo Yu
- CT Collaboration, Siemens Healthineers Ltd., Beijing, China
| | - Yang Li
- Department of Computed Tomography and Magnetic Resonance, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Min Li
- Department of Computed Tomography and Magnetic Resonance, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Liu
- Department of Computed Tomography and Magnetic Resonance, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiangming Wang
- Department of Computed Tomography and Magnetic Resonance, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gaofeng Shi
- Department of Computed Tomography and Magnetic Resonance, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
205
|
Song Q, Lv X, Ru Y, Dong J, Chang R, Wu D, Chen L, Wang X, Guo X. Circulating exosomal gastric cancer-associated long noncoding RNA1 as a noninvasive biomarker for predicting chemotherapy response and prognosis of advanced gastric cancer: A multi-cohort, multi-phase study. EBioMedicine 2022; 78:103971. [PMID: 35349826 PMCID: PMC8965144 DOI: 10.1016/j.ebiom.2022.103971] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/23/2022] [Accepted: 03/14/2022] [Indexed: 12/24/2022] Open
Abstract
Background A previous validated study has identified the diagnostic value of circulating exosomal lncRNA-GC1 for detecting and monitoring gastric cancer. We aimed to further determine the predictive role of circulating exosomal lncRNA-GC1 for prognosis and chemotherapy response. Methods We retrospectively conducted a multi-phase analysis with four independent cohorts of 981 patients. A training cohort was used to generate the predictive model. One internal and two external cohorts were recruited as validation cohorts. Patients with stage II or III gastric cancer in the combined cohort were used to evaluate the predictive value of circulating exosomal lncRNA-GC1 for chemotherapy response. Findings In the training cohort, circulating exosomal lncRNA-GC1 was identified as an independent prognostic predictor for disease-free and overall survival. A prognostic risk stratification model based on circulating exosomal lncRNA-GC1 and AJCC stage revealed better predictive accuracy for disease-free and overall survival than the traditional AJCC stage system alone (C-index: DFS 0.701 vs 0.614; OS 0.720 vs 0.611, both P<0.05). And it has been further verified in the validation cohorts. In interaction analysis, for stage II and III GC, patients with low-level of circulating exosomal lncRNA-GC1 derived more survival benefit from adjuvant chemotherapy (P < 0.05); while those with high-level did not. Interpretation Measurement of circulating exosomal lncRNA-GC1 provides clinically important prognostic information and could complement the AJCC stage to optimize decision-making for selecting patients who could benefit more from fluorouracil-based chemotherapy after surgery. Funding The funders are listed in the Acknowledgement.
Collapse
Affiliation(s)
- Qiying Song
- Department of General Surgery, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiaohui Lv
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Ru
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Jian Dong
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Rongyan Chang
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; Department of Endoscopic Surgery, Air Force 986(th) Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Di Wu
- Department of General Surgery, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lubin Chen
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; Department of Endoscopic Surgery, Air Force 986(th) Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xinxin Wang
- Department of General Surgery, Chinese PLA General Hospital, Beijing, 100853, China..
| | - Xin Guo
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; Department of Endoscopic Surgery, Air Force 986(th) Hospital, Fourth Military Medical University, Xi'an, 710032, China; Department of General Surgery, Chinese PLA General Hospital, Beijing, 100853, China..
| |
Collapse
|
206
|
Song R, Cui Y, Ren J, Zhang J, Yang Z, Li D, Li Z, Yang X. CT-based radiomics analysis in the prediction of response to neoadjuvant chemotherapy in locally advanced gastric cancer: A dual-center study. Radiother Oncol 2022; 171:155-163. [DOI: 10.1016/j.radonc.2022.04.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/26/2022] [Accepted: 04/21/2022] [Indexed: 12/24/2022]
|
207
|
Lin JX, Tang YH, Lin GJ, Ma YB, Desiderio J, Li P, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Tu RH, Zheng CH, Parisi A, Truty MJ, Huang CM. Association of Adjuvant Chemotherapy With Overall Survival Among Patients With Locally Advanced Gastric Cancer After Neoadjuvant Chemotherapy. JAMA Netw Open 2022; 5:e225557. [PMID: 35363268 PMCID: PMC8976237 DOI: 10.1001/jamanetworkopen.2022.5557] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/15/2022] [Indexed: 11/26/2022] Open
Abstract
IMPORTANCE Neoadjuvant chemotherapy (NAC) is a standard treatment option for locally advanced gastric cancer (LAGC); however, the indications for adjuvant chemotherapy (AC) in patients with LAGC who received NAC remain controversial. OBJECTIVE To compare survival rates between patients with LAGC who received AC and those who did not after NAC followed by surgery. DESIGN, SETTING, AND PARTICIPANTS This multicenter, international cohort study included 353 patients with LAGC undergoing curative-intent gastrectomy after NAC at 2 tertiary referral teaching hospitals in China between June 1, 2008, and December 31, 2017. To externally validate the findings in the Chinese patients, 109 patients from the US and Italy between June 1, 2006, and June 30, 2013, were reviewed. The follow-up period of the Chinese patients was completed in December 2020, and the follow-up period of the Western patients was completed between February and July 2017. Data analysis was performed from December 1, 2020, to February 28, 2021. EXPOSURES Patients who received AC and those who did not were propensity score matched to evaluate the association of AC with survival. MAIN OUTCOMES AND MEASURES Overall survival (OS), disease-free survival, and disease-specific survival. RESULTS Of 353 patients from China (275 [78.1%] male; mean [SD] age, 58.0 [10.7] years), 262 (74.1%) received AC and 91 (25.9%) did not. After propensity score matching, the 3-year OS was significantly higher in patients who received AC (60.1%; 95% CI, 53.1%-68.1%) than in those who did not (49.3%; 95% CI, 39.8%-61.0%) (P = .02). Lymph node ratio (LNR) was significantly associated with AC benefit (P < .001 for interaction), and a plot of the interaction between LNR and AC demonstrated that AC was associated with improved OS in patients with higher (≥9%) LNRs (3-year OS: 46.6% vs 21.7%; P < .001), but not in patients with LNRs less than 9% (3-year OS: 73.9% vs 71.3%; P = .30). When stratified by AC cycles, only those patients who completed at least 4 AC cycles exhibited a significant survival benefit in the 6-month (hazard ratio, 0.56; 95% CI, 0.33-0.96; P = .03) and 9-month landmark analysis (hazard ratio, 0.50; 95% CI, 0.27-0.94; P = .03). In the external cohort, improved OS with AC administration was also found in patients with LNRs of 9% or greater (3-year OS: 53.0% vs 26.3%; P = .04). CONCLUSIONS AND RELEVANCE In this cohort study, the administration of AC after NAC and resection of LAGC was associated with improved prognosis in patients with LNRs of 9% or greater. These findings suggest that LNR might be valuable in AC selection in future decision-making processes.
Collapse
Affiliation(s)
- Jian-Xian Lin
- Department of Gastric 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
| | - Yi-Hui Tang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Guan-Jie Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Jacopo Desiderio
- Department of Digestive Surgery, St Mary’s Hospital, Terni, Italy
- Department of Surgical Sciences, La Sapienza University of Rome, Rome, Italy
| | - Ping Li
- Department of Gastric 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
| | - Jian-Wei Xie
- Department of Gastric 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
| | - Jia-Bin Wang
- Department of Gastric 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
| | - Jun Lu
- Department of Gastric 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
| | - Qi-Yue Chen
- Department of Gastric 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
| | - Long-Long Cao
- Department of Gastric 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
| | - Mi Lin
- Department of Gastric 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
| | - Ru-Hong Tu
- Department of Gastric 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
| | - Chao-Hui Zheng
- Department of Gastric 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
| | - Amilcare Parisi
- Department of Digestive Surgery, St Mary’s Hospital, Terni, Italy
- Department of Surgical Sciences, La Sapienza University of Rome, Rome, Italy
| | - Mark J. Truty
- Section of Hepatobiliary and Pancreatic Surgery, Division of Subspecialty General Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Chang-Ming Huang
- Department of Gastric 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
| |
Collapse
|
208
|
Sivacoumarane S, Dutta S, Dubashi B, Adithan S, Toi PC, Nelamangala Ramakrishnaiah VP. Role of Neoadjuvant Chemotherapy in Locally Advanced Carcinoma Stomach: An Analysis of the Short-Term Outcomes. Cureus 2022; 14:e23936. [PMID: 35535287 PMCID: PMC9079323 DOI: 10.7759/cureus.23936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Neoadjuvant chemotherapy (NACT) in carcinoma stomach was introduced in an effort to eliminate micro-metastasis and to improve resectablity before surgery which improves R0 resection rates. We aimed to study the short term outcomes of neoadjuvant chemotherapy on the Tumor Node Metastasis (TNM) stage and the operative outcomes including R0 resection rate in locally advanced gastric cancer. METHODS We prospectively included patients with locally advanced adenocarcinoma stomach staged by contrast-enhanced computed tomography (CECT) in our study. Patients in Group I were started on neoadjuvant chemotherapy (epirubicin, oxaliplatin, and capecitabine). Surgery was done following response assessment CECT. Patients in Group II underwent upfront surgery. We assessed R0 resection rate, number of harvested and metastatic lymph nodes, lymph node ratio, duration of surgery, blood loss, hospital stay and complications between two groups. Response to NACT was assessed in Group I. RESULTS Out of 47 patients who received NACT, two patients had complete response (4.2%), 13 had partial response (27.7%), 10 had stable disease (21.3%) and 22 patients had progressive disease (46.8%). We found no significant difference in the rate of R0 resection between the two groups (88.2% in NACT group vs 85.1% in surgery group, P=0.55). CONCLUSIONS The rate of R0 resection does not significantly improve with neoadjuvant chemotherapy. In view of high progression rates, patient selection is required when NACT is planned in carcinoma stomach which are surgically resectable at presentation. We await survival analysis to further validate the role of NACT.
Collapse
Affiliation(s)
| | - Souradeep Dutta
- Surgery, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Biswajit Dubashi
- Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Subathra Adithan
- Radiodiagnosis, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Pampa C Toi
- Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | | |
Collapse
|
209
|
You W, Cai Z, Sheng N, Yan L, Wan H, Wang Y, Ouyang J, Xie L, Wu X, Wang Z. Construction and Validation of Convenient Clinicopathologic Signatures for Predicting the Prognosis of Stage I-III Gastric Cancer. Front Oncol 2022; 12:848783. [PMID: 35402221 PMCID: PMC8987912 DOI: 10.3389/fonc.2022.848783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background Patients with stage I-III gastric cancer (GC) undergoing R0 radical resection display extremely different prognoses. How to discriminate high-risk patients with poor survival conveniently is a clinical conundrum to be solved urgently. Methods Patients with stage I-III GC from 2010 to 2016 were included in our study. The associations of clinicopathological features with disease-free survival (DFS) and overall survival (OS) were examined via Cox proportional hazard model. Nomograms were developed which systematically integrated prognosis-related features. Kaplan–Meier survival analysis was performed to compare DFS and OS among groups. The results were then externally validated by The Sixth Affiliated Hospital, Sun Yat-sen University. Results A total of 585 and 410 patients were included in the discovery cohort and the validation cohort, respectively. T stage, N stage, lymphatic/vascular/nerve infiltration, preoperative CEA, and CA19-9 were independent prognostic factors (P < 0.05). Two prognostic signatures with a concordance index (C-index) of 0.7502 for DFS and 0.7341 for OS were developed based on the nomograms. The 3-year and 5-year calibration curves showed a perfect correlation between predicted and observed outcomes. Patients were divided into three risk groups (low, intermediate, high), and distinct differences were noticed (p < 0.001). Similar results were achieved in the validation cohort. Notably, a free website was constructed based on our signatures to predict the recurrence risk and survival time of patients with stage I-III GC. Conclusions The signatures demonstrate the powerful ability to conveniently identify distinct subpopulations, which may provide significant suggestions for individual follow-up and adjuvant therapy.
Collapse
Affiliation(s)
- Weiqiang You
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Zerong Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Nengquan Sheng
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Li Yan
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Huihui Wan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yongkun Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jian Ouyang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Lu Xie
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- *Correspondence: Zhigang Wang, ; Xiaojian Wu, ; Lu Xie,
| | - Xiaojian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
- *Correspondence: Zhigang Wang, ; Xiaojian Wu, ; Lu Xie,
| | - Zhigang Wang
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Zhigang Wang, ; Xiaojian Wu, ; Lu Xie,
| |
Collapse
|
210
|
Jiang F, Hu X, Cao H, Shen X. Hsa_circ_0000081 promotes the function of gastric cancer through sponging hsa-miR-423-5p to influence 3-phosphoinositide-dependent kinase 1 expression. Bioengineered 2022; 13:8277-8290. [PMID: 35302432 PMCID: PMC9162021 DOI: 10.1080/21655979.2022.2053796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies in the world, and effective therapeutic targets need to be identified for this type of cancer. In this study, circular RNA (circRNA) microarray analysis was utilized to screen differentially expressed circRNA in GC. Using quantitative reverse transcription polymerase chain reaction (qRT-PCR), hsa_circ_0000081 (circRNA-0000081) expression was found to be up-regulated in tissues and cells and was negative correlated with patients' survival time. RNase R and Actinomycin D assays indicated that circRNA-0000081 was significantly more resistant to R enzyme and had a longer half-life than linear RNA. Moreover, the knockdown or overexpression of circRNA-000081 could influence the proliferation, migration, and invasion potential of GC. Finally, dual luciferase reporter, RNA immunoprecipitation, qRT-PCR, and western blotting assays were used to verify the targeting relationship between circRNA-000081 and miRNA-423-5p or miRNA-423-5p and 3-phosphoinositide-dependent kinase 1 (PDPK1). In conclusion, circRNA-0000081 promotes the function of GC through sponging hsa-miR-423-5p to influence PDPK1 expression, which has a promising therapeutic potential for treating patients with GC.
Collapse
Affiliation(s)
- Fei Jiang
- Key Laboratory of Environmental Medical Engineering and Education Ministry, Nanjing Public Health College, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Xueju Hu
- Key Laboratory of Environmental Medical Engineering and Education Ministry, Nanjing Public Health College, Southeast University, Nanjing, China.,Department of Occupational and Environmental Health, School of Public Health, Southeast University, Nanjing, China
| | - Hongyong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaobing Shen
- Key Laboratory of Environmental Medical Engineering and Education Ministry, Nanjing Public Health College, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China.,Department of Occupational and Environmental Health, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
211
|
A population-based study on treatment and outcomes in patients with gastric adenocarcinoma diagnosed with distant interval metastases. Eur J Surg Oncol 2022; 48:1964-1971. [DOI: 10.1016/j.ejso.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/06/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022] Open
|
212
|
Dong X, Song S, Li Y, Fan Y, Wang L, Wang R, Huo L, Scott A, Xu Y, Pizzi MP, Ma L, Wang Y, Jin J, Zhao W, Yao X, Johnson R, Wang L, Wang Z, Peng G, Ajani JA. Loss of ARID1A activates mTOR signaling and SOX9 in gastric adenocarcinoma-rationale for targeting ARID1A deficiency. Gut 2022; 71:467-478. [PMID: 33785559 PMCID: PMC9724309 DOI: 10.1136/gutjnl-2020-322660] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/20/2021] [Accepted: 03/02/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Gastric adenocarcinoma (GAC) is a lethal disease with limited therapeutic options. Genetic alterations in chromatin remodelling gene AT-rich interactive domain 1A (ARID1A) and mTOR pathway activation occur frequently in GAC. Targeting the mechanistic target of rapamycin (mTOR) pathway in unselected patients has failed to show survival benefit. A deeper understanding of GAC might identify a subset that can benefit from mTOR inhibition. METHODS Genomic alterations in ARID1A were analysed in GAC. Mouse gastric epithelial cells from CK19-Cre-Arid1Afl/fl and wild-type mice were used to determine the activation of oncogenic genes due to loss of Arid1A. Functional studies were performed to determine the significance of loss of ARID1A and the sensitivity of ARID1A-deficient cancer cells to mTOR inhibition in GAC. RESULTS More than 30% of GAC cases had alterations (mutations or deletions) of ARID1A and ARID1A expression was negatively associated with phosphorylation of S6 and SOX9 in GAC tissues and patient-derived xenografts (PDXs). Activation of mTOR signalling (increased pS6) and SOX9 nuclear expression were strongly increased in Arid1A-/- mouse gastric tissues which could be curtailed by RAD001, an mTOR inhibitor. Knockdown of ARID1A in GAC cell lines increased pS6 and nuclear SOX9 and increased sensitivity to an mTOR inhibitor which was further amplified by its combination with fluorouracil both in vitro and in vivo in PDXs. CONCLUSIONS The loss of ARID1A activates pS6 and SOX9 in GAC, which can be effectively targeted by an mTOR inhibitor. Therefore, our studies suggest a new therapeutic strategy of clinically targeting the mTOR pathway in patients with GAC with ARID1A deficiency.
Collapse
Affiliation(s)
- Xiaochuan Dong
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030;,Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuan Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030;,Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, P.R. China
| | - Yibo Fan
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Lulu Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Ruiping Wang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Longfei Huo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Ailing Scott
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Yan Xu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030;,Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, P.R. China
| | - Melissa Pool Pizzi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Lang Ma
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Ying Wang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Wei Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Xiaodan Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Randy Johnson
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Linghua Wang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, P.R. China
| | - Guang Peng
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030;,Corresponding authors: Shumei Song, MD, Ph.D, Department of Gastrointestinal Medical Oncology, Unit 426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009; phone: 713-834-6144; fax: 713-745-1163; . Jaffer A. Ajani, MD, Department of Gastrointestinal Medical Oncology, Unit 426, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009; phone: 713-792-3685; fax: 713-792-8864;
| |
Collapse
|
213
|
Harada K, Yamashita K, Iwatsuki M, Baba H, Ajani JA. Intraperitoneal therapy for gastric cancer peritoneal carcinomatosis. Expert Rev Clin Pharmacol 2022; 15:43-49. [PMID: 35184625 DOI: 10.1080/17512433.2022.2044790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
INTRODUCTION Gastric adenocarcinoma (GAC) is one of the most aggressive malignancies worldwide and has a poor prognosis. Multidisciplinary therapies are used in its treatment, but the prognosis for GAC patients with peritoneal metastases (PM) remains poor and there is no effective established approach. AREAS COVERED This review summarizes the results of recent clinical studies and recent advances in the management, including surgery, chemotherapy, targeted therapy, and immunotherapy. In this review, keywords were searched in combination with 'peritoneal carcinomatosis' and 'gastric cancer' in PubMed, and then studies that evaluated peritoneal carcinomatosis associated with gastric cancer were identified through reading them. Several studies were quoted at second hand. Despite recent advances in therapeutic approaches such as systemic chemotherapy, immunotherapy, intraperitoneal chemotherapy, debulking surgery, thermal hyperthermic intraperitoneal chemotherapy, pressurized intraperitoneal aerosol chemotherapy, immunotherapy, and best supportive therapy, further studies are necessary. This review also summarizes molecular biology of GAC patients with PM. EXPERT OPINION Each modality is advancing and some have shown therapeutic effects, but none have become standard treatments that exhibit remarkable effects. To improve the prognosis of GAC patients with PM, large-scale clinical trials and further basic research are required.
Collapse
Affiliation(s)
- Kazuto Harada
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Kohei Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.,Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
214
|
Zhang X, Zhao L, Niu P, Wang T, Wang W, Sun C, Li Z, Chen Y, Zhao D. Adjuvant Chemotherapy Might Be Recommended to Patients With Positive Margin After Gastrectomy: A 20-Year Retrospective Analysis in a Single Center. Front Oncol 2022; 11:794032. [PMID: 35186712 PMCID: PMC8847385 DOI: 10.3389/fonc.2021.794032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Background Margin positivity after gastric cancer resection is associated with poorer outcomes. However, the prognostic factors and the choice of postoperative adjuvant treatment of patients with positive margin (PM) after gastrectomy are still being debated. Methods A single-center, retrospective analysis was conducted for patients with PM after gastrectomy from the China National Cancer Center Gastric Cancer Database (NCCGCDB) from 1998 to 2018. Univariate and multivariate Cox regression analyses were performed to identify prognostic factors of overall survival (OS) and recurrence-free survival (RFS). Results A total of 449 patients were included in the study, including 192 (42.8%) in the proximal PM group (PPM), 205 (45.7%) in the distal PM group (DPM), and 52 (11.6%) in the bilateral PM group (BPM). The 3- and 5-year OS rates for the PM patients investigated were 47.5% and 39.3%, respectively, and the 3- and 5-year RFS rates were 60.0% and 53.6%, respectively. Multivariate Cox regression analysis proved total gastrectomy (hazard ratio (HR): 1.783, 95%CI: 1.133–2.805, p = 0.012), pT4 (HR: 5.264, 95%CI: 1.493–18.565, p = 0.01), pN2 (HR: 2.263, 95%CI: 1.164–4.397, p = 0.016), pN3 (HR: 2.327, 95%CI: 1.233–4.393, p = 0.009), and combined resection (HR: 1.952, 95%CI: 1.256–3.034, p = 0.003) to be independent risk factors of OS, and pT3 (HR: 9.257, 95%CI: 1.152–74.386, p = 0.036) and pT4 (HR: 11.361, 95%CI: 1.469–87.847, p = 0.020) to be independent risk factors for RFS. Adjuvant chemotherapy prolonged OS in the PPM group (p = 0.032) and prolonged RFS in the PPM group (p < 0.001) and the DPM group (p = 0.035) compared with surgery alone. Conclusions Advanced pathologic stage was associated with poor prognosis, and postoperative adjuvant chemotherapy might be recommended in PM patients after gastrectomy. Still, further prospective trials are warranted to verify and support our conclusions.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lulu Zhao
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Penghui Niu
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tongbo Wang
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Wang
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chongyuan Sun
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zefeng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingtai Chen
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongbing Zhao
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
215
|
Xiang YY, Deng CC, Liu HY, Kuo ZC, Zhang CH, He YL. The Prognostic Effect of Multidisciplinary Team Intervention in Patients with Advanced Gastric Cancer. Curr Oncol 2022; 29:1201-1212. [PMID: 35200601 PMCID: PMC8871247 DOI: 10.3390/curroncol29020102] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/31/2022] [Accepted: 02/11/2022] [Indexed: 12/24/2022] Open
Abstract
Background: The effect of multidisciplinary team intervention (MDT) on the prognosis of advanced gastric cancer (GC) is still controversial. This study aims to analyze the effect of MDTs on the overall survival time of advanced gastric cancer patients. Methods: Patients with advanced GC who underwent surgical treatment between 2007 and 2014 were included in the study. They were divided into two groups; the MDT group received MDT treatment and the non-MDT group received conventional treatment. The Kaplan-Meier method was used to compare the overall survival (OS) of the two groups. The prognostic factors of advanced GC were evaluated by multivariate Cox regression analysis. Results: 394 patients were included in our study. Kaplan-Meier survival analysis showed that the prognosis of advanced GC patients with who underwent MDT intervention was better than those without (3-year OS of 55.6% vs. 46.1%, p = 0.005), Multivariate analysis indicated that MDT intervention could reduce mortality (HR = 0.493, p < 0.001). Conclusions: MDT intervention is an effective measure that improves the survival of patients with advanced GC.
Collapse
Affiliation(s)
- Yuan-Yuan Xiang
- Digestive Disease Center, Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China; (Y.-Y.X.); (C.-C.D.); (H.-Y.L.); (Z.-C.K.)
| | - Cun-Can Deng
- Digestive Disease Center, Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China; (Y.-Y.X.); (C.-C.D.); (H.-Y.L.); (Z.-C.K.)
| | - Han-Yuan Liu
- Digestive Disease Center, Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China; (Y.-Y.X.); (C.-C.D.); (H.-Y.L.); (Z.-C.K.)
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Zi-Chong Kuo
- Digestive Disease Center, Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China; (Y.-Y.X.); (C.-C.D.); (H.-Y.L.); (Z.-C.K.)
| | - Chang-Hua Zhang
- Digestive Disease Center, Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China; (Y.-Y.X.); (C.-C.D.); (H.-Y.L.); (Z.-C.K.)
- Correspondence: (C.-H.Z.); (Y.-L.H.)
| | - Yu-Long He
- Digestive Disease Center, Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China; (Y.-Y.X.); (C.-C.D.); (H.-Y.L.); (Z.-C.K.)
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
- Correspondence: (C.-H.Z.); (Y.-L.H.)
| |
Collapse
|
216
|
Su S, Shi YT, Chu Y, Jiang MZ, Wu N, Xu B, Zhou H, Lin JC, Jin YR, Li XF, Liang J. Sec62 promotes gastric cancer metastasis through mediating UPR-induced autophagy activation. Cell Mol Life Sci 2022; 79:133. [PMID: 35165763 PMCID: PMC11073224 DOI: 10.1007/s00018-022-04143-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Sec62 is a membrane protein of the endoplasmic reticulum that facilitates protein transport. Its role in cancer is increasingly recognised, but remains largely unknown. We investigated the functional role of Sec62 in gastric cancer (GC) and its underlying mechanism. METHODS Bioinformatics, tissue microarray, immunohistochemistry (IHC), western blotting (WB), quantitative polymerase chain reaction (qPCR), and immunofluorescence were used to examine the expression of target genes. Transwell, scratch healing assays, and xenograft models were used to evaluate cell migration and invasion. Transmission electron microscopy and mRFP-GFP-LC3 double-labeled adenoviruses were used to monitor autophagy. Co-immunoprecipitation (CO-IP) was performed to evaluate the binding activity between the proteins. RESULTS Sec62 expression was upregulated in GC, and Sec62 upregulation was an independent predictor of poor prognosis. Sec62 overexpression promoted GC cell migration and invasion both in vitro and in vivo. Sec62 promoted migration and invasion by affecting TIMP-1 and MMP2/9 balance. Moreover, Sec62 could activate autophagy by upregulating PERK/ATF4 expression and binding to LC3II with concomitant FIP200/Beclin-1/Atg5 activation. Furthermore, autophagy blockage impaired the promotive effects of Sec62 on GC cell migration and invasion, whereas autophagy activation rescued the inhibitory effect of Sec62 knockdown on GC metastasis. Notably, Sec62 inhibition combined with autophagy blockage exerted a synergetic anti-metastatic effect in vitro and in vivo. CONCLUSION Sec62 promotes GC metastasis by activating autophagy and subsequently regulating TIMP-1 and MMP2/9 balance. The activation of autophagy by Sec62 may involve the unfolded protein response (UPR)-related PERK/ATF4 pathway and binding of LC3II during UPR recovery involving FIP200/Beclin-1/Atg5 upregulation. Specifically, the dual inhibition of Sec62 and autophagy may provide a promising therapeutic strategy for GC metastasis.
Collapse
Affiliation(s)
- Song Su
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University (Air Force Medical University), Changle West Road 127, Xi'an, Shaanxi, 710032, China
- The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yan-Ting Shi
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University (Air Force Medical University), Changle West Road 127, Xi'an, Shaanxi, 710032, China
| | - Yi Chu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University (Air Force Medical University), Changle West Road 127, Xi'an, Shaanxi, 710032, China
| | - Ming-Zuo Jiang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University (Air Force Medical University), Changle West Road 127, Xi'an, Shaanxi, 710032, China
| | - Nan Wu
- College of Life Sciences, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Bing Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University, 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - He Zhou
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University (Air Force Medical University), Changle West Road 127, Xi'an, Shaanxi, 710032, China
| | - Jun-Chao Lin
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University (Air Force Medical University), Changle West Road 127, Xi'an, Shaanxi, 710032, China
| | - Yi-Rong Jin
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University (Air Force Medical University), Changle West Road 127, Xi'an, Shaanxi, 710032, China
| | - Xiao-Fei Li
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University (Air Force Medical University), Changle West Road 127, Xi'an, Shaanxi, 710032, China
| | - Jie Liang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University (Air Force Medical University), Changle West Road 127, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
217
|
Chen Y, Gong W, Dai W, Jiang H, Xu X. E2F1/2/4 mRNA is associated with immune infiltration and are potential biomarkers for the prognosis of human gastric carcinoma. Transl Cancer Res 2022; 10:2801-2811. [PMID: 35116590 PMCID: PMC8797903 DOI: 10.21037/tcr-21-45] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/27/2021] [Indexed: 12/19/2022]
Abstract
Background E2Fs are genes that regulate DNA synthesis and the cell cycle by encoding a family of transcription factors. Increasing experimental evidence has revealed that E2Fs play key roles in tumor progression in various types of cancer. Methods We investigated the survival, expression and transcriptional data of E2F1/2/4 in gastric cancer (GC) patients using the immunohistochemistry assay, Kaplan-Meier Plotter, cBioPortal, String, and GEPIA databases. The plasma of GC patients was analyzed using the real-time reverse transcription polymerase chain reaction (RT-PCR) assay. The correlation between E2F1/2/4 expression and clinical features was analyzed using the quartile method. As well, the correlation between E2F1/2/4 and GC immune infiltration was also investigated using the TIMER database. Database of Immune Cell Expression (DICE) was also used to analyze correlations between SOX4 and immune responses. Results RT-PCR and tissue immunohistochemistry confirmed that E2F1/2/4 was highly expressed in serum and GC tissue samples of GC patients, the expression of which was not affected by patient age and gender. Also, the survival analysis revealed that low levels of E2F1/2/4 expression were significantly associated with a longer overall survival (OS) in GC patients. E2F1/2/4 was correlated with patient prognosis and immune cell infiltration, including B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and DCs in GC. Our findings indicated that E2F1/2/4 could be used as a prognostic biomarker and indicator of immune infiltration in GC. Conclusions This study revealed that E2F1/2/4 could be a promising indicator for tumor-associated immune infiltration and prognosis in GC patients.
Collapse
Affiliation(s)
- Yongyi Chen
- Department of Clinical Lab, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wangang Gong
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.,Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Wumin Dai
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.,Zhejiang Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Huifen Jiang
- Department of Clinical Lab, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaohong Xu
- Department of Clinical Lab, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
218
|
Gęca K, Rawicz-Pruszyński K, Mlak R, Sadok I, Polkowski WP, Staniszewska M. Kynurenine and Anthranilic Acid in the Peritoneum Correlate With the Stage of Gastric Cancer Disease. Int J Tryptophan Res 2022; 15:11786469211065620. [PMID: 35140473 PMCID: PMC8819753 DOI: 10.1177/11786469211065620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND This study aimed to assess the importance of selected kynurenines measured in peritoneal fluid, lavage washings, and blood serum in patients with advanced gastric cancer (GC) based on the clinical and pathological staging of TNM for a more precise evaluation of the stage of the disease. METHODS Data were collected from a prospectively maintained database of all patients operated on advanced GC between July 2018 and August 2020. In total, 98 patients were eligible for the analysis according to the REMARK guidelines. RESULTS Among the various kynurenines analyzed in this study, we found that the median concentration of anthranilic acid (AA) in the peritoneal lavage washings was significantly higher in patients with positive nodes (pN1-3) compared to those with negative nodes (pN0) (P = 0.0100). Based on the ROC analysis, AA showed diagnostic utility in the differentiation of the pN staging (P = 0.0047). Furthermore, there was a positive correlation between AA in peritoneal fluid with stage pN (P = 0.0116) and a positive correlation between AA in peritoneal lavage washings with stage cT (P = 0.0101). We found that the median concentration of kynurenine (Kyn) in peritoneal lavage washings was significantly higher in patients with cM1 compared to cM0 patients (P = 0.0047). Based on the ROC analysis, Kyn showed diagnostic utility in cM staging differentiation (P < 0.0001). There was a positive correlation between peritoneal Kyn and stage of cM (P = 0.0079). CONCLUSIONS AA and Kyn measured in peritoneal lavage indicate advanced GC and may be considered in the future as valuable adjunct tools in TNM staging of advanced GC.
Collapse
Affiliation(s)
- Katarzyna Gęca
- Department of Surgical Oncology,
Medical University of Lublin, Lublin, Poland
| | | | - Radosław Mlak
- Department of Human Physiology, Medical
University of Lublin, Lublin, Poland
| | - Ilona Sadok
- Laboratory of Separation and
Spectroscopic Method Applications, Centre for Interdisciplinary Research, Faculty of
Science and Health, The John Paul II Catholic University of Lublin, Lublin,
Poland
| | | | - Magdalena Staniszewska
- Laboratory of Separation and
Spectroscopic Method Applications, Centre for Interdisciplinary Research, Faculty of
Science and Health, The John Paul II Catholic University of Lublin, Lublin,
Poland
| |
Collapse
|
219
|
AKÇAKAYA A. Staging Laparoscopy in Stomach Cancer. BEZMIALEM SCIENCE 2022. [DOI: 10.14235/bas.galenos.2021.04696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
220
|
Cai Z, Song H, Huang Z, Fingerhut A, Xu X, Zhong H, Li Z, Zhang Y, Sha D, Bao D, Wang H, Cai B, Hua S, Zhang Y, Sun J, Ye K, Li J, Lu Y, Feng B. Safety and feasibility of laparoscopic surgery for colorectal and gastric cancer under the Chinese multi-site practice policy: admittance standards of competence are needed. Gastroenterol Rep (Oxf) 2022; 10:goac046. [PMID: 36196254 PMCID: PMC9522381 DOI: 10.1093/gastro/goac046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/02/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Background The multi-site practice (MSP) policy has been practiced in China over 10 years. This study aimed to investigate the safety and feasibility of performing laparoscopic surgery for colorectal cancer (LSCRC) and gastric cancer (LSGC) under the Chinese MSP policy. Methods We collected and analysed the data from 1,081 patients who underwent LSCRC or LSGC performed by one gastrointestinal surgeon in his original hospital (n = 573) and his MSP institutions (n = 508) between January 2017 and December 2020. Baseline demographics, intraoperative outcomes, post-operative recovery, and pathological results were compared between the original hospital and MSP institutions, as well as between MSP institutions with and without specific competence (surgical skill, operative instrument, perioperative multi-discipline team). Results In our study, 690 patients underwent LSCRC and 391 patients underwent LSGC. The prevalence of post-operative complications was comparable for LSCRC (11.5% vs 11.1%, P = 0.89) or LSGC (15.2% vs 12.6%, P = 0.46) between the original hospital and MSP institutions. However, patients in MSP institutions without qualified surgical assistant(s) and adequate instruments experienced longer operative time and greater intraoperative blood loss. The proportion of patients with inadequate lymph-node yield was significantly higher in MSP institutions than in the original hospital for both LSCRC (11.5% vs 21.2%, P < 0.01) and LSGC (9.8% vs 20.5%, P < 0.01). Conclusion For an experienced gastrointestinal surgeon, performing LSCRC and LSGC outside his original hospital under the MSP policy is safe and feasible, but relies on the precondition that the MSP institutions are equipped with qualified surgical skills, adequate operative instruments, and complete perioperative management.
Collapse
Affiliation(s)
| | | | | | - Abraham Fingerhut
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- Shanghai Minimally Invasive Surgery Center, Shanghai, P. R. China
- Section for Surgical Research, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Ximo Xu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- Shanghai Minimally Invasive Surgery Center, Shanghai, P. R. China
| | - Hao Zhong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
- Shanghai Minimally Invasive Surgery Center, Shanghai, P. R. China
| | - Zhigang Li
- Department of General Surgery, Wuxi Huishan District People’s Hospital, Jiangsu, P. R. China
| | - Yingjie Zhang
- Department of General Surgery, Zhoushan Hospital, Zhejiang, P. R. China
| | - Dachong Sha
- Department of General Surgery, Cixi Third People’s Hospital, Zhejiang, P. R. China
| | - Dandan Bao
- Department of General Surgery, People’s Hospital of Wenzhou, Zhejiang, P. R. China
| | - Haibo Wang
- Department of General Surgery, Chinese PLA 107 Hospital, Shandong, P. R. China
| | - Binghua Cai
- Department of General Surgery, Rudong People’s Hospital, Jiangsu, P. R. China
| | - Shangbo Hua
- Department of General Surgery, Kunshan Hospital of Chinese Medicine, Jiangsu, P. R. China
| | - Yanhui Zhang
- Department of General Surgery, Jingjiang Chinese Medicine Hospital, Jiangsu, P. R. China
| | - Jianguang Sun
- Department of General Surgery, Haiyan People’s Hospital, Zhejiang, P. R. China
| | - Ke Ye
- Department of General Surgery, Yueqing People’s Hospital, Zhejiang, P. R. China
| | - Jianwen Li
- Corresponding author. Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. No. 197 Ruijin Er Road, Shanghai 200025, P. R. China. Tel: +86-21-64370045; ; Department of Medical Affairs, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. No. 197 Ruijin Er Road, Shanghai 200025, P. R. China; Tel: +86-21-64370045; ; Shanghai Minimally Invasive Surgery Center, No. 197 Ruijin Er Road, Shanghai 200025, P. R. China. Tel: +86-21-64458887;
| | - Yong Lu
- Corresponding author. Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. No. 197 Ruijin Er Road, Shanghai 200025, P. R. China. Tel: +86-21-64370045; ; Department of Medical Affairs, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. No. 197 Ruijin Er Road, Shanghai 200025, P. R. China; Tel: +86-21-64370045; ; Shanghai Minimally Invasive Surgery Center, No. 197 Ruijin Er Road, Shanghai 200025, P. R. China. Tel: +86-21-64458887;
| | - Bo Feng
- Corresponding author. Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. No. 197 Ruijin Er Road, Shanghai 200025, P. R. China. Tel: +86-21-64370045; ; Department of Medical Affairs, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. No. 197 Ruijin Er Road, Shanghai 200025, P. R. China; Tel: +86-21-64370045; ; Shanghai Minimally Invasive Surgery Center, No. 197 Ruijin Er Road, Shanghai 200025, P. R. China. Tel: +86-21-64458887;
| |
Collapse
|
221
|
Beeraka NM, Gu H, Xue N, Liu Y, Yu H, Liu J, Chen K, Nikolenko VN, Fan R. Testing lncRNAs signature as clinical stage–related prognostic markers in gastric cancer progression using TCGA database. Exp Biol Med (Maywood) 2022; 247:658-671. [PMID: 35068210 DOI: 10.1177/15353702211067173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
LncRNA expression can be conducive to gastric cancer (GC) prognosis. The objective of this study is to ascertain five specific lncRNAs involved in tumor progression of GC and their role as prognostic markers to diagnose clinical stage-wise GC. High-throughput RNA sequencing data were obtained from The Cancer Genome Atlas (TCGA) database and performed genome-wide lncRNA expression analysis using edgeR package, Bioconductor.org , and R-statistical computing to analyze differentially expressed lncRNA analysis. Cutoff parameters were FDR < 0.05 and |Log2FC| > 2. Total 351 tumor samples with differentially expressed lncRNAs were divided into group-1 lncRNAs such as AC019117.2 and LINC00941, and group-2 lncRNAs such as LINC02410, AC012317.2, and AC141273.1 by 2:1. The Spearman correlation coefficients ( p < 0.05) and correlation test function (cor.test ()) were performed for lncRNAs as per clinical stage. Cytoscape software was used to construct lncRNA–mRNA interaction networks. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway ( p < 0.05) analysis were conducted using the clusterProfiler package. Kaplan–Meier survival analysis was performed to determine the overall survival of patients based on the expression of five lncRNAs in different clinical stages of GC. AC019117.2 and LINC00941 of group 1 inferred a positive correlation with clinical stages of stage I to stage IV, and their expressions were higher in tumor tissues than normal tissues. On the contrary, LINC02410, AC012317.2, and AC141273.1 of group 2 exhibited a negative correlation with clinical stage, and they exhibited more expression in normal tissues compared to tumor tissues. GO and KEGG pathway analysis reported that AC019117.2 may interact with LINC00941 via ITGA3 and trophoblast glycoprotein (TPBG) to foster tumor progression. Tumor-specific group-1 lncRNAs were conducive to the poor overall survival and exhibited a positive correlation with the clinical stages of stage I to stage IV in GC as per the lncRNA–mRNA networking analysis. These five lncRNAs could be considered as clinically useful lncRNA-based prognostic markers to predict clinical stage-wise GC progression.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow 119991, Russia
| | - Hao Gu
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Nannan Xue
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450052, China
| | - Huiming Yu
- Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing 450052, China
| | - Junqi Liu
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Kuo Chen
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Vladimir N Nikolenko
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow 119991, Russia
- M.V. Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Ruitai Fan
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
222
|
Ding P, Yang P, Sun C, Tian Y, Guo H, Liu Y, Li Y, Zhao Q. Predictive Effect of Systemic Immune-Inflammation Index Combined With Prognostic Nutrition Index Score on Efficacy and Prognosis of Neoadjuvant Intraperitoneal and Systemic Paclitaxel Combined With Apatinib Conversion Therapy in Gastric Cancer Patients With Positive Peritoneal Lavage Cytology: A Prospective Study. Front Oncol 2022; 11:791912. [PMID: 35127498 PMCID: PMC8807517 DOI: 10.3389/fonc.2021.791912] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Background Gastric cancer with only peritoneal lavage cytology (GC-CY1) is a special type of gastric cancer, which is defined as stage IV. The pre-treatment systemic immune-inflammation index (SII) and prognostic nutritional index (PNI) are representative blood indexes of systemic inflammatory response and nutritional status. However, the clinical significance of combined detection of these two indexes is still unclear. This study aims to evaluate the clinical value of the new score system by combining SII and PNI (SII-PNI score) as a predictor of efficacy and prognosis after neoadjuvant intraperitoneal and systemic (NIPS) paclitaxel combined with Apatinib conversion therapy for GC-CY1 patients. Methods We registered a prospective clinical study involving 36 GC-CY1 patients from April 2018 to August 2019 (NCT03718624). All patients underwent re-laparoscopic exploration after treatment. According to free cancer cells (FCCs) status, these patients were divided into FCCs group and non-FCCs group. The SII-PNI score ranged from 0 to 2 as follows: score of 2, high SII (≥512.1) and low PNI (≤52.9); score of 1, either high SII or low PNI; score of 0, no high SII nor low PNI. Results All patients underwent re-laparoscopic exploration after 3 cycles of NIPS paclitaxel and Apatinib conversion therapy. Among them, 28 cases (77.78%) were in non-FCCs group, and 8 cases (22.22%) were in FCCs group. The SII-PNI score of non-FCCs patients was significantly lower than that of FCCs patients (p=0.041). The prognosis of patients with high SII-PNI score was significantly worse than that of patients with low SII-PNI score (p<0.001). Multivariate analysis showed that SII-PNI score was an independent prognostic factor for predicting overall survival and progression-free survival (p=0.001, 0.002). Conclusion Pretreatment SII-PNI score is an important predictor for the efficacy of GC-CY1 patients after NIPS paclitaxel combined with Apatinib conversion therapy, which can help to identify high-risk groups and predict prognosis.
Collapse
Affiliation(s)
- Ping’an Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Peigang Yang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chenyu Sun
- Internal Medicine, AMITA Health Saint Joseph Hospital Chicago, Chicago, IL, United States
| | - Yuan Tian
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Honghai Guo
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yang Liu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qun Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Qun Zhao,
| |
Collapse
|
223
|
Castro JSLD, Pelosof AG, Andrade-Cabral JGGD, Seraphim AM, Taglieri E, Coimbra FJF, Zitron C. ENDOSCOPIC CHARACTERISTICS OF PATIENTS WITH COMPLETE PATHOLOGICAL RESPONSE AFTER NEOADJUVANT CHEMOTHERAPY FOR GASTRIC AND ESOPHAGOGASTRIC JUNCTION ADENOCARCINOMAS. ABCD-ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA 2022; 34:e1616. [PMID: 35019128 PMCID: PMC8735268 DOI: 10.1590/0102-672020210002e1616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/04/2021] [Indexed: 12/24/2022]
Abstract
Background:
Gastric and esophagogastric junction adenocarcinoma are responsible for approximately 13.5% of cancer-related deaths. Given the fact that these tumors are not typically detected until they are already in the advanced stages, neoadjuvancy plays a fundamental role in improving long-term survival. Identification of those with complete pathological response (pCR) after neoadjuvant chemotherapy (NAC) is a major challenge, with effects on organ preservation, extent of resection, and additional surgery. There is little or no information in the literature about which endoscopic signs should be evaluated after NAC, or even when such re-evaluation should occur.
Aim: To describe the endoscopic aspects of patients with gastric and esophagogastric junction adenocarcinomas who underwent NAC and achieved pCR, and to determine the accuracy of esophagogastroduodenoscopy (EGD) in predicting the pCR.
Methods: A survey was conducted of the medical records of patients with these tumors who were submitted to gastrectomy after NAC, with anatomopathological result of pCR.
Results: Twenty-nine patients were identified who achieved pCR after NAC within the study period. Endoscopic responses were used to classify patients into two groups: G1-endoscopic findings consistent with pCR and G2-endoscopic findings not consistent with pCR. Endoscopic evaluation in G1 was present in an equal percentage (47.4%; p=0.28) in Borrmann classification II and III. In this group, the predominance was in the gastric body (57.9%; p=0.14), intestinal subtype with 42.1% (p=0.75), undifferentiated degree, 62.5% (p=0.78), Herb+ in 73.3% (p=0.68). The most significant finding, however, was that the time interval between NAC and EGD was longer for G1 than G2 (24.4 vs. 10.2 days, p=0.008).
Conclusion: EGD after NAC seems to be a useful tool for predicting pCR, and it may be possible to use it to create a reliable response classification. In addition, the time interval between NAC and EGD appears to significantly influence the predictive power of endoscopy for pCR.
Collapse
Affiliation(s)
| | | | | | | | - Eloy Taglieri
- A.C. Camargo Cancer Center, Endoscopy Unit, São Paulo, SP, Brazil
| | | | - Claudia Zitron
- A.C. Camargo Cancer Center, Endoscopy Unit, São Paulo, SP, Brazil
| |
Collapse
|
224
|
Dhiman A, Vining CC, Witmer HDD, Sood D, Shergill A, Kindler H, Roggin KK, Posner MC, Ahmed OS, Liauw S, Pitroda S, Liao CY, Karrison T, Weichselbaum R, Polite B, Eng OS, Catenacci DVT, Turaga KK. Phase II Prospective, Open-Label Randomized Controlled Trial Comparing Standard of Care Chemotherapy With and Without Sequential Cytoreductive Interventions for Patients with Oligometastatic Foregut Adenocarcinoma and Undetectable Circulating Tumor Deoxyribose Nucleic Acid (ctDNA) Levels. Ann Surg Oncol 2022; 29:10.1245/s10434-021-11249-7. [PMID: 34988836 PMCID: PMC8730296 DOI: 10.1245/s10434-021-11249-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Metastatic adenocarcinomas of foregut origin are aggressive and have limited treatment options, poor quality of life, and a dismal prognosis. A subset of such patients with limited metastatic disease might have favorable outcomes with locoregional metastasis-directed therapies. This study investigates the role of sequential cytoreductive interventions in addition to the standard of care chemotherapy in patients with oligometastatic foregut adenocarcinoma. METHODS This is a single-center, phase II, open-label randomized clinical trial. Eligible patients include adults with synchronous or metachronous oligometastatic (metastasis limited to two sites and amenable for curative/ablative treatment) adenocarcinoma of the foregut without progression after induction chemotherapy and having undetectable ctDNA. These patients will undergo induction chemotherapy and will then be randomized (1:1) to either sequential curative intervention followed by maintenance chemotherapy versus routine continued chemotherapy. The primary endpoint is progression-free survival (PFS), and a total of 48 patients will be enrolled to detect an improvement in the median PFS in the intervention arm with a hazard ratio (HR) of 0.5 with 80% power and a one-sided alpha of 0.1. Secondary endpoints include disease-free survival (DFS) in the intervention arm, overall survival (OS), ctDNA conversion rate pre/post-induction chemotherapy, ctDNA PFS, PFS2, adverse events, quality of life, and financial toxicity. DISCUSSION This is the first randomized study that aims to prospectively evaluate the efficacy and safety of surgical/ablative interventions in patients with ctDNA-negative oligometastatic adenocarcinoma of foregut origin post-induction chemotherapy. The results from this study will likely develop pertinent, timely, and relevant knowledge in oncology.
Collapse
Affiliation(s)
- Ankit Dhiman
- Section of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago Medicine, Chicago, IL, USA
| | - Charles C Vining
- Section of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago Medicine, Chicago, IL, USA
| | - Hunter D D Witmer
- Section of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago Medicine, Chicago, IL, USA
| | - Divya Sood
- Section of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago Medicine, Chicago, IL, USA
| | - Ardaman Shergill
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Hedy Kindler
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Kevin K Roggin
- Section of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago Medicine, Chicago, IL, USA
| | - Mitchell C Posner
- Section of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago Medicine, Chicago, IL, USA
| | | | - Stanley Liauw
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Sean Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Chih-Yi Liao
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Theodore Karrison
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Ralph Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Blase Polite
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Oliver S Eng
- Section of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago Medicine, Chicago, IL, USA
| | - Daniel V T Catenacci
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Kiran K Turaga
- Section of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago Medicine, Chicago, IL, USA.
| |
Collapse
|
225
|
Lee IS, Zhu Z, Lee J, Park JO, Wu X, Ong T, Li SM, Wang X, Chao J, Goel A. A liquid biopsy signature predicts treatment response to fluoropyrimidine plus platinum therapy in patients with metastatic or unresectable gastric cancer: implications for precision oncology. Mol Cancer 2022; 21:9. [PMID: 34980153 PMCID: PMC8722275 DOI: 10.1186/s12943-021-01483-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/19/2021] [Indexed: 01/12/2023] Open
Affiliation(s)
- In-Seob Lee
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Comprehensive Cancer Center, 1218 S. Fifth Avenue, Monrovia, CA, 91016, USA
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Zhongxu Zhu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Tiffany Ong
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Sierra Min Li
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Joseph Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Comprehensive Cancer Center, 1218 S. Fifth Avenue, Monrovia, CA, 91016, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
226
|
Levy J, Gupta V, Amirazodi E, Allen-Ayodabo C, Jivraj N, Jeong Y, Davis LE, Mahar AL, De Mestral C, Saarela O, Coburn NG. Textbook Outcome and Survival in Patients With Gastric Cancer: An Analysis of the Population Registry of Esophageal and Stomach Tumours in Ontario (PRESTO). Ann Surg 2022; 275:140-148. [PMID: 32149825 DOI: 10.1097/sla.0000000000003849] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To examine the association between Textbook Outcome (TO)-a new composite quality measurement-and long-term survival in gastric cancer surgery. BACKGROUND Single-quality indicators do not sufficiently reflect the complex and multifaceted nature of perioperative care in patients with gastric adenocarcinoma. METHODS All patients undergoing gastrectomy for nonmetastatic gastric adenocarcinoma registered in the Population Registry of Esophageal and Stomach Tumours of Ontario (PRESTO) between 2004 and 2015 were included. TO was defined according to negative margins; >15 lymph nodes sampled; no severe complications; no re-interventions; no unplanned ICU admission; length of stay ≤21 days; no 30-day readmission; and no 30-day mortality. Three-year survival was estimated using the Kaplan-Meier method. A marginal multivariable Cox proportional-hazards model was used to estimate the association between achieving TO metrics and long-term survival. E-value methodology was used to assess for risk of residual confounding. RESULTS Of the 1836 patients included in this study, 402 (22%) achieved all TO metrics. TO patients had a higher 3-year survival rate compared to non-TO patients (75% vs 55%, log-rank P < 0.001). After adjustments for covariates and clustering within hospitals, TO was associated with a 41% reduction in mortality (adjusted hazards ratio 0.59, 95% confidence interval 0.48, 0.72, P < 0.001). These results were robust to potential residual confounding. CONCLUSIONS Achieving TO is strongly associated with improved long-term survival in gastric cancer patients and merits further focus in surgical quality improvement efforts.
Collapse
Affiliation(s)
- Jordan Levy
- Division of General Surgery, Department of Surgery and Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Vaibhav Gupta
- Division of General Surgery, Department of Surgery and Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Elmira Amirazodi
- Evaluative Clinical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | | - Naheed Jivraj
- Department of Anesthesia and Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Yunni Jeong
- Division of General Surgery, Department of Surgery and Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Laura E Davis
- Evaluative Clinical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Alyson L Mahar
- Manitoba Centre for Health Policy and Department of Community Health Sciences, University of Manitoba, Canada
| | - Charles De Mestral
- Division of General Surgery, Department of Surgery and Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Li Ka Shing Knowledge Institute and St. Michael's Hospital, Toronto, Ontario, Canada
| | - Olli Saarela
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Natalie G Coburn
- Division of General Surgery, Department of Surgery and Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
227
|
Huang J, Chen Y, Zhang Y, Xie J, Liang Y, Yuan W, Zhou T, Gao R, Wen R, Xia Y, Long L. Comparison of clinical-computed tomography model with 2D and 3D radiomics models to predict occult peritoneal metastases in advanced gastric cancer. Abdom Radiol (NY) 2022; 47:66-75. [PMID: 34636930 DOI: 10.1007/s00261-021-03287-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 02/05/2023]
Abstract
PURPOSE To compare the ability of a clinical-computed tomography (CT) model vs. 2D and 3D radiomics models for predicting occult peritoneal metastasis (PM) in patients with advanced gastric cancer (AGC). METHODS In this retrospective study, we included 49 patients with occult PM and 49 control patients (without PM) who underwent preoperative CT and subsequent surgery between January 2016 and December 2018. Clinical information and CT semantic features were collected, and CT radiomics features were extracted. A predictive clinical-CT model was created using multivariate logistic regression. The least absolute shrinkage and selection operator algorithm and logistic regression were used for constructing 2D and 3D radiomics models. These models were validated with an external cohort (n = 30). Receiver operating characteristics curve with area under the curve (AUC), sensitivity, and specificity were used to evaluate predictive performance. RESULTS Tumor size, mild ascites, and serum CA125 were independent factors predictive of occult PM. The clinical-CT model of these independent factors showed better diagnostic performance than 2D and 3D radiomics models. In the external validation cohort, the AUCs of different models were as follows-clinical-CT model: 0.853 (sensitivity, 66.7%; specificity, 93.3%); 2D radiomics model: 0.622 (sensitivity, 80.0%; specificity, 46.7%); and 3D radiomics model: 0.676 (sensitivity, 60.0%; specificity, 86.0%). The clinical-CT model nomogram showed good clinical predictive efficiency to assess occult PM. CONCLUSION The clinical-CT model was better than the radiomics models in predicting occult PM in AGC.
Collapse
Affiliation(s)
- Jiang Huang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yidi Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuying Zhang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Jinhuan Xie
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yiqiong Liang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Wenzhao Yuan
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Ting Zhou
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Ruizhi Gao
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Rong Wen
- Department of Medical Ultrasound, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuwei Xia
- Huiying Medical Technology Co. Ltd, Beijing, 100192, China
| | - Liling Long
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
228
|
Abstract
The intimate involvement of pathogens with the heightened risk for developing certain cancers is an area of research that has captured a great deal of attention over the last 10 years. One firmly established paradigm that highlights this aspect of disease progression is in the instance of Helicobacter pylori infection and the contribution it makes in elevating the risk for developing gastric cancer. Whilst the molecular mechanisms that pinpoint the contribution that this microorganism inflicts towards host cells during gastric cancer initiation have come into greater focus, another picture that has also emerged is one that implicates the host's immune system, and the chronic inflammation that can arise therefrom, as being a central contributory factor in disease progression. Consequently, when taken with the underlying role that the extracellular matrix plays in the development of most cancers, and how this dynamic can be modulated by proteases expressed from the tumor or inflammatory cells, a complex and detailed relationship shared between the individual cellular components and their surroundings is coming into focus. In this review article, we draw attention to the emerging role played by the cathepsin proteases in modulating the stage-specific progression of Helicobacter pylori-initiated gastric cancer and the underlying immune response, while highlighting the therapeutic significance of this dynamic and how it may be amenable for novel intervention strategies within a basic research or clinical setting.
Collapse
|
229
|
Chao J, He TF, D'Apuzzo M, Chen YJ, Frankel P, Tajon M, Chen H, Solomon S, Klempner SJ, Fakih M, Lee P. A Phase 2 Trial Combining Pembrolizumab and Palliative Radiation Therapy in Gastroesophageal Cancer to Augment Abscopal Immune Responses. Adv Radiat Oncol 2022; 7:100807. [PMID: 35071830 PMCID: PMC8767243 DOI: 10.1016/j.adro.2021.100807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/10/2021] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Single agent PD-1 inhibitors have yielded durable responses in a minority of gastroesophageal cancers. Radiation therapy has been recognized to promote antitumor immune responses and may synergize with anti-PD-1 agents. We sought to evaluate if combining palliative radiation therapy with pembrolizumab can augment antitumor immune responses in gastroesophageal cancer. METHODS AND MATERIALS Patients had metastatic gastroesophageal cancer with indication for palliative radiation therapy with ≥2 disease sites outside of the radiation field assessable for abscopal response and biopsies for laboratory correlative analyses. Palliative radiation was delivered to a dose of 30 Gy over 10 fractions. Pembrolizumab, 200 mg, was administered concurrently intravenously every 3 weeks until disease progression, unacceptable toxicity, or study withdrawal, for up to 2 years. Endpoints included PD-L1 expression in pre- and posttreatment biopsies and abscopal objective response rate per Response Evaluation Criteria in Solid Tumors. RESULTS Of 14 enrolled patients, the objective response rate was 28.6% (95% confidence interval, 8.4%-58.1%), and the median duration of response was not reached (95% confidence interval, 6.9-NR months). Overall, 2 patients had treatment-related grade 3 to 4 adverse events with no grade 5 events. One patient discontinued therapy due to grade 4 colitis. We did not observe an association between radiation and abscopal changes in PD-L1 expression via assessment of an analogous PD-L1 Combined Positive Score, Tumor Proportion Score, Mononuclear Immune Cell Density Score, or proportion of PD-L1-expressing immune cells between pre- and posttreatment tumor biopsies. CONCLUSIONS Combining palliative radiation therapy and pembrolizumab provided promising durable responses in this patient population but we were unable to definitively distinguish abscopal biologic changes. Biomarker analyses beyond PD-L1 expression are needed to better understand putative mechanisms and identify patients who will benefit from this approach.
Collapse
Affiliation(s)
- Joseph Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Ting-Fang He
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Massimo D'Apuzzo
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Yi-Jen Chen
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Paul Frankel
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Michael Tajon
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Helen Chen
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Shawn Solomon
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Samuel J. Klempner
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Marwan Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Peter Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
230
|
Yıldırım S, Yılmaz C. Trıplet or Doublet Chemotherapy Regimens in Metastatic Gastric Cancer. CLINICAL CANCER INVESTIGATION JOURNAL 2022. [DOI: 10.51847/rpfscains5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
231
|
Kang YK, Chen LT, Ryu MH, Oh DY, Oh SC, Chung HC, Lee KW, Omori T, Shitara K, Sakuramoto S, Chung IJ, Yamaguchi K, Kato K, Sym SJ, Kadowaki S, Tsuji K, Chen JS, Bai LY, Oh SY, Choda Y, Yasui H, Takeuchi K, Hirashima Y, Hagihara S, Boku N. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022; 23:234-247. [DOI: 10.1016/s1470-2045(21)00692-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023]
|
232
|
Tanaka K, Tanabe H, Sato H, Ishikawa C, Goto M, Yanagida N, Akabane H, Yokohama S, Hasegawa K, Kitano Y, Sugiyama Y, Uehara K, Kobayashi Y, Murakami Y, Kunogi T, Sasaki T, Takahashi K, Ando K, Ueno N, Kashima S, Moriichi K, Sato K, Yuzawa S, Tanino M, Taruiishi M, Sumi Y, Mizukami Y, Fujiya M, Okumura T. Prognostic factors to predict the survival in patients with advanced gastric cancer who receive later-line nivolumab monotherapy-The Asahikawa Gastric Cancer Cohort Study (AGCC). Cancer Med 2022; 11:406-416. [PMID: 34845844 PMCID: PMC8729046 DOI: 10.1002/cam4.4461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/21/2021] [Accepted: 11/08/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Chemotherapy for advanced gastric cancer is recommended in the guidelines; however, later-line treatment remains controversial. Since immune checkpoint inhibitors have been used for the treatment of various malignancies, trials have been performed for gastric cancer. A phase 3 trial indicated the survival benefit of nivolumab monotherapy for gastric cancer patients treated with prior chemotherapy regimens. PATIENTS AND METHODS A regional cohort study was undertaken to determine the real-world data of nivolumab treatment for patients with advanced or recurrent gastric cancer. The patients were enrolled for 2 years from October 2017 to October 2019 and were prospectively followed for 1 year to examine the overall survival (OS). The patient characteristics were analyzed in a multivariate analysis and a nomogram to predict the probability of survival was generated. RESULTS In total, 70 patients who received nivolumab as ≥third-line chemotherapy were included in the Asahikawa Gastric Cancer Cohort. The median OS was 7.5 (95% CI, 4.8-10.2) months and the response rate was 18.6%. Diffuse type classification, bone metastasis, high neutrophil/lymphocyte ratio, and high CRP were associated with poor OS/prognosis in the multivariate analysis. A nomogram was developed based on these clinical parameters and the concordance index was 0.80 (95% CI, 0.68-0.91). The responders were aged and were frequently diagnosed with intestinal type gastric cancer, including patients with a HER2-positive status (27.3%) or microsatellite instability-high (27.3%) status. CONCLUSIONS The regional cohort study of nivolumab monotherapy for gastric cancer patients revealed prognostic factors and a nomogram was developed that could predict the probability of survival.
Collapse
Affiliation(s)
- Kazuyuki Tanaka
- Department of GastroenterologyAsahikawa Kosei HospitalAsahikawa HokkaidoJapan
| | - Hiroki Tanabe
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Hiroki Sato
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Chisato Ishikawa
- Department of GastroenterologyJapanese Red Cross Asahikawa HospitalAsahikawa HokkaidoJapan
| | - Mitsuru Goto
- Department of GastroenterologyAsahikawa Kosei HospitalAsahikawa HokkaidoJapan
| | - Naoyuki Yanagida
- Department of SurgeryAsahikawa Kosei HospitalAsahikawa HokkaidoJapan
| | - Hiromitsu Akabane
- Department of SurgeryAsahikawa Kosei HospitalAsahikawa HokkaidoJapan
| | - Shiro Yokohama
- Department of GastroenterologyAsahikawa Medical CenterAsahikawa HokkaidoJapan
| | - Kimiharu Hasegawa
- Division of Gastrointestinal SurgeryAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Yohei Kitano
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Yuya Sugiyama
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Kyoko Uehara
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Yu Kobayashi
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Yuki Murakami
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Takehito Kunogi
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Takahiro Sasaki
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Keitaro Takahashi
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Katsuyoshi Ando
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Nobuhiro Ueno
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Shin Kashima
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Kentaro Moriichi
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Keisuke Sato
- Department of PathologyAsahikawa Kosei HospitalAsahikawa HokkaidoJapan
| | - Sayaka Yuzawa
- Department of Diagnostic PathologyAsahikawa Medical University HospitalAsahikawa Medical UniversityAsahikawaHokkaidoJapan
| | - Mishie Tanino
- Department of Diagnostic PathologyAsahikawa Medical University HospitalAsahikawa Medical UniversityAsahikawaHokkaidoJapan
| | - Masaki Taruiishi
- Department of GastroenterologyAsahikawa City HospitalAsahikawa HokkaidoJapan
| | - Yasuo Sumi
- Division of Gastrointestinal SurgeryAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Yusuke Mizukami
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Mikihiro Fujiya
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| | - Toshikatsu Okumura
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology Department of MedicineAsahikawa Medical UniversityAsahikawa HokkaidoJapan
| |
Collapse
|
233
|
Ren J, Pan G, Yang J, Xu N, Zhang Q, Li W. Circ_0000620 acts as an oncogenic factor in gastric cancer through regulating MMP2 expression via sponging miR-671-5p. JOURNAL OF BIOLOGICAL RESEARCH (THESSALONIKE, GREECE) 2021; 28:23. [PMID: 34972532 PMCID: PMC8720221 DOI: 10.1186/s40709-021-00154-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 12/10/2021] [Indexed: 04/14/2023]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common cancers in the digestive system. Circular RNAs (circRNAs) have been found to function as important regulators in the pathogenesis of GC. This study focused on the biological role and molecular mechanism of circ_0000620 in GC progression. METHODS The expression levels of circ_0000620, microRNA-671-5p (miR-671-5p) and Matrix MetalloProteinase 2 (MMP2) were measured by quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry (IHC) assay or western blot. The stability of circ_0000620 was confirmed by Ribonuclease R (RNase R) assay. The protein levels were determined by western blot assay. Cell viability, colony formation, cell migratory ability, cell invasive ability and tube formation capacity were respectively examined by CCK-8 assay, colony formation assay, wound healing assay, transwell invasion assay and tube formation assay. The interaction between miR-671-5p and circ_0000620 or MMP2 was validated by dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull-down assay. The role of circ_0000620 in GC undefined was explored by xenograft tumor assay. RESULTS Circ_0000620 was conspicuously upregulated in GC tissues and cells. Circ_0000620 knockdown reduced cell viability, colony formation, migration, invasion and tube formation capacity of GC cells in vitro. Furthermore, MMP2 was upregulated in GC and MMP2 overexpression reversed the anti-tumor response of circ_0000620 knockdown in GC progression. Moreover, circ_0000620 directly interacted with miR-671-5p and circ_0000620 downregulation regulated malignant behaviors of GC cells by upregulating miR-671-5p. In addition, silencing of circ_0000620 inhibited tumor growth in vivo. CONCLUSIONS Circ_0000620 knockdown inhibited the malignant development of GC partly through modulating the miR-671-5p/MMP2 axis.
Collapse
Affiliation(s)
- Junyu Ren
- Department of Oncology, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, 650032, Kunming, China
| | - Guoqing Pan
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Yang
- Department of Oncology, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, 650032, Kunming, China
| | - Ning Xu
- Department of Oncology, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, 650032, Kunming, China
| | - Qiong Zhang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wenliang Li
- Department of Oncology, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, 650032, Kunming, China.
| |
Collapse
|
234
|
Liu Z, Wang Y, Shan F, Ying X, Zhang Y, Li S, Jia Y, Miao R, Xue K, Li Z, Li Z, Ji J. Duration of Perioperative Chemotherapy in Locally Advanced Gastric Cancer: A "Less Is More" Question When ypN0 Is Achieved. Front Oncol 2021; 11:775166. [PMID: 34926284 PMCID: PMC8671134 DOI: 10.3389/fonc.2021.775166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Backgrounds Perioperative chemotherapy (PEC) and neoadjuvant chemotherapy (NAC) have become a vital part of locally advanced gastric cancer (LAGC) treatment, but the optimal duration of PEC has not been studied. The aim of this study was to demonstrate the possibility of duration reduction in PEC in the adjuvant chemotherapy (AC) phase for ypN0 patients. Methods We included LAGC patients who achieved ypN0 after NAC in our institution from 2005 to 2018. The risk/benefit of AC and other covariates were majorly measured by overall survival (OS) and progression-free survival (PFS). We developed a survival-tree-based model to determine the optimal PEC duration for ypN0 patients in different classes. Results A total of 267 R0 resection patients were included. There were 55 patients who did not receive AC. The 5-year OS was 74.34% in the non-AC group and 83.64% in the AC group with a significant difference (p = 0.012). Multivariate Cox regression revealed that both AC (AC vs. non-AC: HR, 0.49; 95%CI, 0.27–0.88; p = 0.018) and ypT stages (ypT3-4 vs. ypT0-2: HR, 2.00; 95%CI, 1.11–3.59; p = 0.021) were significant protective/risk factors on patients OS and PFS. A decision tree model for OS indicated an optimal four to six cycles of PEC, which was recommended for ypT0-2N0 patients, while a minimum of five PEC cycles was recommended for ypT3-4N0 patients. Conclusion AC treatment is still necessary for ypN0. The duration reduction could be applied for the ypT0-2N0 stage patients but may not be suitable for higher ypT stages and beyond. A multicenter-based study is required.
Collapse
Affiliation(s)
- Zining Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yinkui Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Fei Shan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangji Ying
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shuangxi Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yongning Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Rulin Miao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Kan Xue
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhemin Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ziyu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
235
|
Shi C, Badgwell BD, Grabsch HI, Gibson MK, Hong SM, Kumarasinghe P, Lam AK, Lauwers G, O'Donovan M, van der Post RS, Tang L, Ushiku T, Vieth M, Selinger CI, Webster F, Nagtegaal ID. Data Set for Reporting Carcinoma of the Stomach in Gastrectomy. Arch Pathol Lab Med 2021; 146:1072-1083. [DOI: 10.5858/arpa.2021-0225-oa] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 11/06/2022]
Abstract
Context.—
A standardized detailed surgical pathology report is the cornerstone of gastric cancer management.
Objective.—
To guide management and prognostication for patients with gastric carcinomas globally, the International Collaboration on Cancer Reporting aimed to produce an evidence-based international pathology reporting data set with a panel of globally recognized expert pathologists and clinicians.
Design.—
Based on published guidelines/data sets for gastric carcinomas, a working draft was developed by the chair of the expert panel of pathologists and clinicians. The draft was then circulated to the panel and discussed in a series of teleconferences and email communications until consensus was achieved. The draft data set was uploaded on the International Collaboration on Cancer Reporting Web site for public comment. The data set was reviewed in consideration of the feedback, and a final version was approved by the panel.
Results.—
This data set was developed for gastrectomy specimens for primary gastric carcinomas, including neuroendocrine carcinomas and mixed neuroendocrine-nonneuroendocrine neoplasms. Well-differentiated neuroendocrine tumors, nonepithelial malignancies, and secondary tumors were excluded from this data set. The final data set contains 15 core (required) elements and 8 noncore (recommended) elements. A commentary is provided for each element.
Conclusions.—
The International Collaboration on Cancer Reporting has published freely available, evidence-based data sets for gastric cancer reporting. Standardized reporting has been shown to improve patient care and facilitates data exchange and analysis for quality assurance, cancer epidemiology, and clinical and basic research.
Collapse
Affiliation(s)
- Chanjuan Shi
- From the Department of Pathology, Duke University School of Medicine, Durham, North Carolina (Shi)
| | - Brian D. Badgwell
- The Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston (Badgwell)
| | - Heike I. Grabsch
- The Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands (Grabsch)
- The Division of Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom (Grabsch)
| | - Michael K. Gibson
- The Division of Hematology and Oncology, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee (Gibson)
| | - Seung-Mo Hong
- The Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea (Hong)
| | - Priyanthi Kumarasinghe
- PathWest Laboratory Medicine, PathWest QEII Medical Center, Perth, Australia (Kumarasinghe)
| | - Alfred K. Lam
- Pathology, School of Medicine, Gold Coast Campus, Griffith University, Gold Coast, Australia (Lam)
- Pathology Queensland, Gold Coast University Hospital, Southport, Australia (Lam)
- Faculty of Medicine, The University of Queensland, Herston, Australia (Lam)
| | - Gregory Lauwers
- The Department of Pathology, Moffitt Cancer Center, Tampa, Florida (Lauwers)
| | - Maria O'Donovan
- The Histopathology Department, Cambridge University Hospitals NHS Foundation Trust Addenbrookes Hospital, Cambridge, United Kingdom (O'Donovan)
| | - Rachel S. van der Post
- The Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands (van der Post and Nagtegaal)
| | - Laura Tang
- The Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, New York, (Tang)
| | - Tetsuo Ushiku
- The Department of Pathology and Diagnostic Pathology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (Ushiku)
| | - Michael Vieth
- The Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, Klinikum Bayreuth, Germany (Vieth)
| | | | - Fleur Webster
- The International Collaboration on Cancer Reporting, Sydney, Australia (Webster)
| | - Iris D. Nagtegaal
- The Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands (van der Post and Nagtegaal)
| |
Collapse
|
236
|
Chen Y, He J, Liu D, Xiao J, Chen X, Tang H, Luo D, Shang C, Lian L, Peng J. Triplet versus doublet neoadjuvant chemotherapy regimens for locally advanced gastric cancer: a propensity score matching analysis. BMC Cancer 2021; 21:1328. [PMID: 34903191 PMCID: PMC8667459 DOI: 10.1186/s12885-021-09093-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/06/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND To investigate the differences between doublet and triplet neoadjuvant chemotherapy (NAC) regimens in efficacy and safety profile. METHODS A total of 227 locally advanced gastric cancer (LAGC) patients who received NAC and sequential radical gastrectomy were reviewed. After propensity score matching (PSM), 140 patients with similar baseline characteristics were selected. Among them, 70 received doublet NAC regimens consisted of platinum and fluorouracil; the other 70 received triplet NAC regimens consisted of docetaxel, platinum, and fluorouracil. RESULTS The efficacy of doublet and triplet regimens was comparable after propensity score matching in terms of tumor regression (pathological complete response, Doublet 11.4% vs. Triplet 15.7%, p = 0.642), achieving of R0 resection (Doublet 88.6% vs. Triplet 88.6%, p = 1), 1-year disease-free survival (DFS) (Doublet 77.1% vs. Triplet 68.6%, p = 0.178), 3-years overall survival (OS) (Doublet 54.3% vs. Triplet 60.9%, p = 0.941). Post-surgery complications were more common in the triplet cohort (Doublet 5.7% vs. Triplet 27.1%, p = 0.001), especially abdominal infection (Doublet 0% vs. Triplet 11.1%, p = 0.001). CONCLUSIONS A more intense preoperative triplet NAC regimen does not bring extra downstage effect and survival benefit compared to a doublet regimen. It may even result in a higher risk of post-surgery complications.
Collapse
Affiliation(s)
- Yonghe Chen
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, 510655, China
| | - Jiasheng He
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, 510655, China
| | - Dan Liu
- Department of Laboratory Science, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510105, China
| | - Jian Xiao
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Xijie Chen
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, 510655, China
| | - Haijie Tang
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, 510655, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Dandong Luo
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, 510655, China
| | - Chenyu Shang
- Department of Laboratory Science, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510105, China
| | - Lei Lian
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, China.
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, 510655, China.
| | - Junsheng Peng
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, China.
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, 510655, China.
| |
Collapse
|
237
|
Sun Z, Jiang Y, Chen C, Zheng H, Huang W, Xu B, Tang W, Yuan Q, Zhou K, Liang X, Chen H, Han Z, Feng H, Yu S, Hu Y, Yu J, Zhou Z, Wang W, Xu Y, Li G. Radiomics signature based on computed tomography images for the preoperative prediction of lymph node metastasis at individual stations in gastric cancer: A multicenter study. Radiother Oncol 2021; 165:179-190. [PMID: 34774652 DOI: 10.1016/j.radonc.2021.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 10/19/2021] [Accepted: 11/03/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Specific diagnosis and treatment of gastric cancer (GC) require accurate preoperative predictions of lymph node metastasis (LNM) at individual stations, such as estimating the extent of lymph node dissection. This study aimed to develop a radiomics signature based on preoperative computed tomography (CT) images, for predicting the LNM status at each individual station. METHODS We enrolled 1506 GC patients retrospectively from two centers as training (531) and external (975) validation cohorts, and recruited 112 patients prospectively from a single center as prospective validation cohort. Radiomics features were extracted from preoperative CT images and integrated with clinical characteristics to construct nomograms for LNM prediction at individual lymph node stations. Performance of the nomograms was assessed through calibration, discrimination and clinical usefulness. RESULTS In training, external and prospective validation cohorts, radiomics signature was significantly associated with LNM status. Moreover, radiomics signature was an independent predictor of LNM status in the multivariable logistic regression analysis. The radiomics nomograms revealed good prediction performances, with AUCs of 0.716-0.871 in the training cohort, 0.678-0.768 in the external validation cohort and 0.700-0.841 in the prospective validation cohort for 12 nodal stations. The nomograms demonstrated a significant agreement between the actual probability and predictive probability in calibration curves. Decision curve analysis showed that nomograms had better net benefit than clinicopathologic characteristics. CONCLUSION Radiomics nomograms for individual lymph node stations presented good prediction accuracy, which could provide important information for individual diagnosis and treatment of gastric cancer.
Collapse
Affiliation(s)
- Zepang Sun
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yuming Jiang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chuanli Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huan Zheng
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weicai Huang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | | | - Weijing Tang
- Department of Pathology, Stanford University School of Medicine, USA
| | - Qingyu Yuan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kangneng Zhou
- School of Computer and Communication Engineering, University of Science and Technology Beijing, China
| | - Xiaokun Liang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen Colleges of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Hao Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhen Han
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hao Feng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shitong Yu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanfeng Hu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
238
|
Zhang Y, Zhang PS, Rong ZY, Huang C. One stomach, two subtypes of carcinoma-the differences between distal and proximal gastric cancer. Gastroenterol Rep (Oxf) 2021; 9:489-504. [PMID: 34925847 PMCID: PMC8677565 DOI: 10.1093/gastro/goab050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/13/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors of the digestive tract, posing a significant risk to human health. Over the past 10 years, the pathological characteristics and the prognosis of GC have been determined based on the locations of the tumors that were then classified into two types-proximal and distal GC. This review focuses on the differences in epidemiology, etiology, cell source, pathological characteristics, gene expression, molecular markers, manifestations, treatment, prognosis, and prevention between proximal and distal GC to provide guidance and a basis for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Peng-Shan Zhang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Ze-Yin Rong
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Chen Huang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
239
|
Song S, Chen Q, Li Y, Lei G, Scott AW, Huo L, Li CY, Estrella JS, Correa AM, Pizzi M, Ma L, Jin J, Liu B, Wang Y, Xiao L, Hofstetter WL, Lee JH, Weston B, Bhutani MS, Shanbhag ND, Johnson RL, Gan B, Wei S, Ajani JA. Targeting cancer stem cells with a pan-BCL-2 inhibitor in preclinical and clinical settings in patients with gastroesophageal carcinoma. Gut 2021; 70:2238-2248. [PMID: 33487592 PMCID: PMC9720890 DOI: 10.1136/gutjnl-2020-321175] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Gastro-oesophageal cancers (GEC) are resistant to therapy and lead to poor prognosis. The cancer stem cells (CSCs) and antiapoptotic pathways often confer therapy resistance. We sought to elucidate the antitumour action of a BCL-2 inhibitor, AT101 in GEC in vitro, in vivo and in a clinical trial. METHODS Extensive preclinical studies in vitro and in vivo were carried out to establish the mechanism action of AT101 on targeting CSCs and antiapoptotic proteins. A pilot clinical trial in patients with GEC was completed with AT-101 added to standard chemoradiation. RESULTS Overexpression of BCL-2 and MCL-1 was noted in gastric cancer tissues (GC). AT-101 induced apoptosis, reduced proliferation and tumour sphere formation in MCL-1/BCL-2 high GC cells. Interestingly, AT101 dramatically downregulated genes (YAP-1/Sox9) that control CSCs in GEC cell lines regardless of BCL-2/MCL-1 expression. Addition of docetaxel to AT-101 amplified its antiproliferation and induced apoptosis effects. In vivo studies confirmed the combination of AT101 and docetaxel demonstrated stronger antitumour activity accompanied with significant decrease of CSCs biomarkers (YAP1/SOX9). In a pilot clinical trial, 13 patients with oesophageal cancer (EC) received AT101 orally concurrently with chemoradiation. We observed dramatic clinical complete responses and encouraging overall survival in these patients. Clinical specimen analyses revealed that AT-101 dramatically reduced the expression of CSCs genes in treated EC specimens indicating antitumour activity of AT101 relies more on its anti-CSCs activity. CONCLUSIONS Our preclinical and clinical data suggest that AT-101 overcomes resistance by targeting CSCs pathways suggesting a novel mechanism of action of AT101 in patients with GEC.
Collapse
Affiliation(s)
- Shumei Song
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Qiongrong Chen
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030;,Department of Pathology & Surgical Oncology, Hubei Cancer Hospital, Wuhan, Hubei, CN, 430079
| | - Yuan Li
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Guang Lei
- Department of Experimental Radiation Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Ailing W Scott
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Longfei Huo
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Cordelia Y. Li
- Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Jeannelyn S. Estrella
- Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Arlene M. Correa
- Department of Thoracic and Cardiovascular Surgery, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Melissa Pizzi
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Lang Ma
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Bin Liu
- Department of Epigenetic&Molecular Carcinogenesis, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Ying Wang
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Lianchun Xiao
- Department of Biostatistics, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Wayne L. Hofstetter
- Department of Thoracic and Cardiovascular Surgery, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Jeffrey H. Lee
- Department of Gastroenterology Hepat&Nutr, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Brian Weston
- Department of Gastroenterology Hepat&Nutr, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Manoop S. Bhutani
- Department of Gastroenterology Hepat&Nutr, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Namita D. Shanbhag
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Randy L. Johnson
- Department of Cancer Biology at the University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Boyi Gan
- Department of Experimental Radiation Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Shaozhong Wei
- Department of Pathology & Surgical Oncology, Hubei Cancer Hospital, Wuhan, Hubei, CN, 430079
| | - Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030;,Corresponding Authors: Shumei Song, Tel: 713-834-6144; or Jaffer A. Ajani, Tel: 713-792-3685; ; Department of Gastrointestinal Medical Oncology, Unit 426, The University of Texas, M.D. Anderson Cancer Center; 1515 Holcombe Blvd., Houston, TX, 77030
| |
Collapse
|
240
|
Lin HW, Loh EW, Shen SC, Tam KW. Gastrectomy with or without omentectomy for gastric cancer: A systematic review and meta-analysis. Surgery 2021; 171:1281-1289. [PMID: 34857385 DOI: 10.1016/j.surg.2021.10.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/05/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Omentectomy is conventionally performed in the procedure of gastrectomy for gastric cancer. However, the clinical value and importance of omentectomy remain unclear. This meta-analysis investigated the benefits and safety of gastrectomy with or without omentectomy for patients with gastric cancer. METHODS A literature search was conducted in PubMed, Embase, and the Cochrane Library for studies comparing complete omentectomy with omentum preservation or partial omentectomy. Primary outcomes were overall survival, relapse-free survival, and incidences of recurrence and complications, whereas secondary outcomes were the total length of operation and the amount of blood loss. RESULTS Nine studies involving 3,561 patients were included. Our meta-analysis revealed no significant differences between omentectomy and omentum preservation in terms of the 5-year overall survival (risk ratio [RR] = 0.95, 95% confidence interval [CI] = 0.89-1.01), 5-year relapse free survival (RR: 0.96, 95% CI: 0.89-1.03), incidence of recurrence in the peritoneum or other visceral organs (RR: 1.13, 95% CI: 0.80-1.60 and RR: 1.06, 95% CI: 0.78-1.45, respectively), and incidence of complications (RR: 1.15, 95% CI: 0.89-1.50). Moreover, omentum preservation significantly reduced the total length of operation (mean difference [MD] 25.70, 95% CI: 3.23-48.17) and the amount of blood loss (MD: 56.29, 95% CI: 14.02-98.56). CONCLUSION Omentectomy may not be necessary and can be omitted during gastrectomy for gastric cancer.
Collapse
Affiliation(s)
- Ho-Wei Lin
- School of Medicine, College of Medicine, Taipei Medical University, Taiwan. https://twitter.com/linhewei1
| | - El-Wui Loh
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taiwan; Center for Evidence-Based Health Care, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Cochrane Taiwan, Taipei Medical University, Taiwan
| | - Shih-Chiang Shen
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taiwan
| | - Ka-Wai Tam
- Center for Evidence-Based Health Care, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Cochrane Taiwan, Taipei Medical University, Taiwan; Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taiwan.
| |
Collapse
|
241
|
Dong YP, Cai FL, Wu ZZ, Wang PL, Yang Y, Guo SW, Zhao ZZ, Zhao FC, Liang H, Deng JY. Risk of station 12a lymph node metastasis in patients with lower-third gastric cancer. World J Gastrointest Surg 2021; 13:1390-1404. [PMID: 34950428 PMCID: PMC8649572 DOI: 10.4240/wjgs.v13.i11.1390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/06/2021] [Accepted: 10/24/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Controversy over the issue that No. 12a lymph node involvement is distant or regional metastasis remains, and the possible inclusion of 12a lymph nodes in D2 lymphadenectomy is unclear. As reported, gastric cancer (GC) located in the lower third is highly related to the metastasis of station 12a lymph nodes. AIM To investigate whether the clinicopathological factors and metastasis status of other perigastric nodes can predict station 12a lymph node metastasis and evaluate the prognostic significance of station 12a lymph node dissection in patients with lower-third GC. METHODS A total of 147 patients with lower-third GC who underwent D2 or D2+ lymphadenectomy, including station 12a lymph node dissection, were included in this retrospective study from June 2003 to March 2011. Survival prognoses were compared between patients with or without station 12a lymph node metastasis. Logistic regression analyses were used to clarify the association between station 12a lymph node metastasis and clinicopathological factors or metastasis status of other perigastric nodes. The metastasis status of each regional lymph node was evaluated to identify the possible predictors of station 12a lymph node metastasis. RESULTS Metastasis to station 12a lymph nodes was observed in 18 patients with lower-third GC, but not in 129 patients. The incidence of station 12a lymph node involvement was reported as 12.2% in patients with lower-third GC. The overall survival of patients without station 12a lymph node metastasis was significantly better than that of patients with station 12a metastasis (P < 0.001), which could also be seen in patients with or without extranodal soft tissue invasion. Station 12a lymph node metastasis and extranodal soft tissue invasion were identified as independent predictors of poor prognosis in patients with lower-third GC. Advanced pN stage was defined as independent risk factor significantly correlated with station 12a lymph node positivity. Station 3 lymph node staus was also proven to be significantly correlated with station 12a lymph node involvement. CONCLUSION Metastasis of station 12a lymph nodes could be considered an independent prognosis factor for patients with lower-third GC. The dissection of station 12a lymph nodes may not be ignored in D2 or D2+ lymphadenectomy due to difficulties in predicting station 12a lymph node metastasis.
Collapse
Affiliation(s)
- Yin-Ping Dong
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Feng-Lin Cai
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zi-Zhen Wu
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Peng-Liang Wang
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yang Yang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Shi-Wei Guo
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhen-Zhen Zhao
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Fu-Cheng Zhao
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Han Liang
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jing-Yu Deng
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
242
|
Li X, Zheng NR, Wang LH, Li ZW, Liu ZC, Fan H, Wang Y, Dai J, Ni XT, Wei X, Liu MW, Li K, Li ZX, Zhou T, Zhang Y, Zhang JY, Kadeerhan G, Huang S, Wu WH, Liu WD, Wu XZ, Zhang LF, Xu JM, Gerhard M, You WC, Pan KF, Li WQ, Qin J. Proteomic profiling identifies signatures associated with progression of precancerous gastric lesions and risk of early gastric cancer. EBioMedicine 2021; 74:103714. [PMID: 34818622 PMCID: PMC8617343 DOI: 10.1016/j.ebiom.2021.103714] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 12/21/2022] Open
Abstract
Background Molecular features underlining the multistage progression of gastric lesions and development of early gastric cancer (GC) are poorly understood, restricting the ability to GC prevention and management. Methods We portrayed proteomic landscape and explored proteomic signatures associated with progression of gastric lesions and risk of early GC. Tissue proteomic profiling was conducted for a total of 324 subjects. A case-control study was performed in the discovery stage (n=169) based on populations from Linqu, a known high-risk area for GC in China. We then conducted two-stage validation, including a cohort study from Linqu (n = 56), with prospective follow-up for progression of gastric lesions (280–473 days), and an independent case-control study from Beijing (n = 99). Findings There was a clear distinction in proteomic features for precancerous gastric lesions and GC. We derived four molecular subtypes of gastric lesions and identified subtype-S4 with the highest progression risk. We found 104 positively-associated and 113 inversely-associated proteins for early GC, with APOA1BP, PGC, HPX and DDT associated with the risk of gastric lesion progression. Integrating these proteomic signatures, the ability to predict progression of gastric lesions was significantly strengthened (areas-under-the-curve=0.88 (95%CI: 0.78–0.99) vs. 0.56 (0.36–0.76), Delong's P = 0.002). Immunohistochemistry assays and examination at mRNA level validated the findings for four proteins. Interpretation We defined proteomic signatures for progression of gastric lesions and risk of early GC, which may have translational significance for identifying particularly high-risk population and detecting GC at an early stage, improving potential for targeted GC prevention. Funding The funders are listed in the Acknowledgement.
Collapse
Affiliation(s)
- Xue Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Nai-Ren Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Lin-Heng Wang
- Department of Gastroenterology, Second Clinical Medical College of Beijing University of Chinese Medicine (Dongfang Hospital), Beijing 100078, China
| | - Zhong-Wu Li
- Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zong-Chao Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jin Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiao-Tian Ni
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xin Wei
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Ming-Wei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Kai Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Zhe-Xuan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Tong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yang Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jing-Ying Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Gaohaer Kadeerhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Sha Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wen-Hui Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wei-Dong Liu
- Linqu County Public Health Bureau, Shandong 262600, China
| | - Xiu-Zhen Wu
- Linqu County People's Hospital, Shandong 262600, China
| | - Lan-Fu Zhang
- Linqu County People's Hospital, Shandong 262600, China
| | - Jian-Ming Xu
- Department of Gastrointestinal Oncology, The Fifth Medical Center, General Hospital of PLA, Beijing 100071, China
| | - Markus Gerhard
- PYLOTUM Key joint laboratory for upper GI cancer, Technische Universität München/Peking University Cancer Hospital & Institute, Munich/Beijing, Germany/ China
| | - Wei-Cheng You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China; PYLOTUM Key joint laboratory for upper GI cancer, Technische Universität München/Peking University Cancer Hospital & Institute, Munich/Beijing, Germany/ China
| | - Kai-Feng Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China; PYLOTUM Key joint laboratory for upper GI cancer, Technische Universität München/Peking University Cancer Hospital & Institute, Munich/Beijing, Germany/ China.
| | - Wen-Qing Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing 100142, China; PYLOTUM Key joint laboratory for upper GI cancer, Technische Universität München/Peking University Cancer Hospital & Institute, Munich/Beijing, Germany/ China.
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.
| |
Collapse
|
243
|
Jing C, Bai Z, Zhang J, Jiang H, Yang X, Yan S, Yin J, Cai J, Zhang Z, Deng W. Apatinib plus S-1 for previously treated, advanced gastric or gastro-oesophageal junction adenocarcinoma: a phase 2, single-arm, prospective study. J Gastrointest Oncol 2021; 12:2035-2044. [PMID: 34790371 DOI: 10.21037/jgo-21-186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022] Open
Abstract
Background The current management of advanced gastric or gastro-oesophageal junction adenocarcinoma remains unsatisfactory. We investigated the efficacy and safety of the combination therapy of apatinib and S-1, considering the potential advantage of home-based treatment without hospital admission, in patients with platinum-refractory gastric or gastro-oesophageal junction adenocarcinoma. Methods In this open-label, single-arm, phase 2 trial, in each 21-day cycle, eligible patients received apatinib at an initial dose of 500 mg once daily continuously and S-1 at a dose of 40-60 mg twice daily on days 1-14 until the trail was discontinued disease progression, development of intolerable toxicity, or withdrawal of consent. The primary endpoints were progression-free survival. The secondary endpoints were objective response rates, disease control rates, and safety, and overall survival. This study was registered at ClinicalTrials.gov, NCT04338438. Results Between April 2015 and May 2019, we included 37 patients with advanced gastric or gastro-oesophageal junction adenocarcinoma refractory to first-line platinum-containing therapy. At the data cutoff, the 6-month progression-free survival was 31.5%, the median progression-free survival and overall survival were 4.2 (95% CI: 3.50-4.90) months and 8.2 (95% CI: 4.69-11.71) months, respectively. Of 37 eligible patients, 8 (21.6%) patients reached objective responses, 31 (83.8%) patients reached disease control. Grade 3 or 4 adverse events occurred in 8 (21.6%) patients, including hand-foot syndrome, hypertension, and diarrhea, etc. Conclusions The combination of Apatinib and S-1 showed promising efficacy and manageable toxicity as a home-based, second-line therapy in patients with advanced gastric or gastro-oesophageal junction adenocarcinoma, especially for the elder patients with poor performance status. Trial Registration NCT04338438.
Collapse
Affiliation(s)
- Chao Jing
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jun Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hongpeng Jiang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaobao Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shu Yan
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jie Yin
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jun Cai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Deng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
244
|
Yang P, Tian Y, Tan B, Ding P, Guo H, Liu Y, Zhang Z, Li Y, Zhao Q. Clinical application of nano-carbon to improve the accuracy of lymph node staging in patients with advanced gastric cancer receiving neoadjuvant chemotherapy: a prospective randomized controlled trial. J Gastrointest Oncol 2021; 12:2052-2060. [PMID: 34790373 DOI: 10.21037/jgo-21-457] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/27/2021] [Indexed: 11/06/2022] Open
Abstract
Background The significance of nano-carbon for lymph node staging in radical gastrectomy for gastric cancer (GC) has been confirmed, but studies on its application for GC patients treated with neoadjuvant chemotherapy (NCT) are rare. The purpose of this study was to explore the clinical value of using carbon nanoparticles suspension injections (CNS) to improve the accuracy of lymph node staging (N staging) of NCT for advanced GC. Methods 160 advanced GC patients receiving preoperative NCT were enrolled, according to the random number generated by computer, the enrolled patients were randomly divided into two groups: experimental group (n=80) and control group (n=80). The experimental group received endoscopic injection of CNS within 24 hours prior to NCT, while the control group received this within 24 hours post NCT and before D2 radical resection. SOX [oxaliplatin: 130 mg/(body surface area, BSA): m2, first day + S-1: (BSA: <1.25 m2, 40 mg each time; ≥1.25 to <1.5 m2, 50 mg each time; ≥1.5 m2, 60 mg each time), 2 times a day, for 2 weeks] was chosen as the NCT regimen, repeat every 3 weeks, 4 cycles were performed preoperative. Surgery was performed 3 weeks after the end of the 4 cycles of chemotherapy. The staining rate, metastasis rate, metastasis rate of stained lymph nodes, postoperative complication rate, and N staging of the two groups were analyzed and compared. Results A total of 3,197 lymph nodes were harvested in the experimental group, including 384 metastatic lymph nodes, 1,424 stained lymph nodes, and 210 metastatic stained lymph nodes. The total number of lymph nodes harvested in the control group was 2,565, including 244 metastatic lymph nodes, 796 stained lymph nodes, and 94 metastatic stained lymph nodes. Compared with the control group, a higher rate of stained lymph nodes, a higher total number of lymph nodes, and an increased number of metastatic lymph nodes were detected in the experimental group. Conclusions The application of CNS before NCT in patients with advanced GC can minimize lymph node staging bias after NCT and improve its accuracy. Trial Registration Chinese Clinical Trial Registry ChiCTR2100047407.
Collapse
Affiliation(s)
- Peigang Yang
- Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuan Tian
- Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bibo Tan
- Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Pingan Ding
- Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Honghai Guo
- Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yang Liu
- Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhidong Zhang
- Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yong Li
- Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qun Zhao
- Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
245
|
Necroptosis-Related lncRNAs: Predicting Prognosis and the Distinction between the Cold and Hot Tumors in Gastric Cancer. JOURNAL OF ONCOLOGY 2021; 2021:6718443. [PMID: 34790235 PMCID: PMC8592775 DOI: 10.1155/2021/6718443] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/21/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022]
Abstract
Background In the face of poor prognosis and immunotherapy failure of gastric cancer (GC), this project tried to find new potential biomarkers for predicting prognosis and precision medication to ameliorate the situation. Methods To form synthetic matrices, we retrieved stomach adenocarcinoma transcriptome data from Genotype-Tissue Expression Project (GTEx) and The Cancer Genome Atlas (TCGA). Necroptosis-related prognostic lncRNA was identified by coexpression analysis and univariate Cox regression. Then we performed the least absolute shrinkage and selection operator (LASSO) to construct the necroptosis-related lncRNA model. Next, the Kaplan–Meier analysis, time-dependent receiver operating characteristics (ROC), univariate Cox (uni-Cox) regression, multivariate Cox (multi-Cox) regression, nomogram, and calibration curves were made to verify and evaluate the model. Gene set enrichment analyses (GSEA), principal component analysis (PCA), immune analysis, and prediction of the half-maximal inhibitory concentration (IC50) in risk groups were also analyzed. For further discussing immunotherapy between the cold and hot tumors, we divided the entire set into two clusters based on necroptosis-related lncRNAs. Results We constructed a model with 16 necroptosis-related lncRNAs. In the model, we found the calibration plots showed a good concordance with the prognosis prediction. The area's 1-, 2-, and 3-year OS under the ROC curve (AUC) were 0.726, 0.763, and 0.770, respectively. Risk groups could be a guide of systemic treatment because of significantly different IC50 between risk groups. Above all, clusters could help distinguish between the cold and hot tumors effectively and contribute to precise mediation. Cluster 2 was identified as the hot tumor and more susceptible to immunotherapeutic drugs. Conclusion The results of this project supported that necroptosis-related lncRNAs could predict prognosis and help make a distinction between the cold and hot tumors for improving individual therapy in GC.
Collapse
|
246
|
Aigner K, Vashist YK, Selak E, Gailhofer S, Aigner KR. Efficacy of Regional Chemotherapy Approach in Peritoneal Metastatic Gastric Cancer. J Clin Med 2021; 10:jcm10225322. [PMID: 34830604 PMCID: PMC8624731 DOI: 10.3390/jcm10225322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/04/2021] [Accepted: 11/13/2021] [Indexed: 12/22/2022] Open
Abstract
Peritoneal spread is frequent in gastric cancer (GC) and a palliative condition. After failure to systemic chemotherapy (sCTx) remaining therapeutic options are very limited. We evaluated the feasibility and efficacy of locoregional chemotherapy (RegCTx) in peritoneal metastatic GC. In total, 38 (23 male and 15 female) patients with peritoneal metastatic GC after failure of previous sCTx and unresectable disease were enrolled in this study. Using the hypoxic abdominal stop-flow perfusion, upper abdominal perfusion and intraarterial infusion technique in total 114 cycles with Cisplatin, Adriamycin and Mitomycin C were applied. No significant procedure related toxicity was noticed- especially no Grade 3 or 4 toxicity occurred. With the RegCTx approach a median overall survival of 17.4 months was achieved. Patients who had undergone previously resection of the GC the median overall survival was even better with 23.5 months. RegCTx is a promising, safe and efficient approach in diffuse metastatic GC. The evaluation of RegCTx in the setting of multimodal treatment approach at less advanced stages is also warranted.
Collapse
|
247
|
Construction, Validation, and Visualization of Two Web-Based Nomograms to Predict Overall and Cancer-Specific Survival in Patients with Gastric Cancer and Lung Metastases. JOURNAL OF ONCOLOGY 2021; 2021:5495267. [PMID: 34759968 PMCID: PMC8575630 DOI: 10.1155/2021/5495267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022]
Abstract
Background The lung is one of the most common sites of metastasis in gastric cancer. Our study developed two nomograms to achieve individualized prediction of overall survival (OS) and cancer-specific survival (CSS) in patients with gastric cancer and lung metastasis (GCLM) to better guide follow-up and planning of subsequent treatment. Methods We reviewed data of patients diagnosed with GCLM in the Surveillance, Epidemiology, and End Results (SEER) database from 2010 to 2015. The endpoints of the study were the OS and CSS. We used the “caret” package to randomly divide patients into training and validation cohorts in a 7 : 3 ratio. Multivariate Cox regression analysis was performed using univariate Cox regression analysis to confirm the independent prognostic factors. Afterward, we built the OS and CSS nomograms with the “rms” package. Subsequently, we evaluated the two nomograms through calibration curves, receiver operating characteristic (ROC) curves, and decision curve analysis (DCA). Finally, two web-based nomograms were built on the basis of effective nomograms. Results The OS analysis included 640 patients, and the results of the multivariate Cox regression analysis showed that grade, chemotherapy, and liver metastasis were independent prognostic factors for patients with GCLM. The CSS analysis included 524 patients, and the results of the multivariate Cox regression analysis showed that the independent prognostic factors for patients with GCLM were chemotherapy, liver metastasis, marital status, and tumor site. The ROC curves, calibration curves, and DCA revealed favorable predictive power in the OS and CSS nomograms. We created web-based nomograms for OS (https://zhenghh.shinyapps.io/aclmos/) and CSS (https://zhenghh.shinyapps.io/aslmcss/). Conclusions We created two web-based nomograms to predict OS and CSS in patients with GCLM. Both web-based nomograms had satisfactory accuracy and clinical usefulness and may help clinicians make individualized treatment decisions for patients.
Collapse
|
248
|
Vos EL, Carr RA, Hsu M, Nakauchi M, Nobel T, Russo A, Barbetta A, Tan KS, Tang L, Ilson D, Ku GY, Wu AJ, Janjigian YY, Yoon SS, Bains MS, Jones DR, Coit D, Molena D, Strong VE. Prognosis after neoadjuvant chemoradiation or chemotherapy for locally advanced gastro-oesophageal junctional adenocarcinoma. Br J Surg 2021; 108:1332-1340. [PMID: 34476473 PMCID: PMC8599637 DOI: 10.1093/bjs/znab228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Trials typically group cancers of the gastro-oesophageal junction (GOJ) with oesophageal or gastric cancer when studying neoadjuvant chemoradiation and perioperative chemotherapy, so the results may not be fully applicable to GOJ cancer. Because optimal neoadjuvant treatment for GOJ cancer remains controversial, outcomes with neoadjuvant chemoradiation versus chemotherapy for locally advanced GOJ adenocarcinoma were compared retrospectively. METHODS Data were collected from all patients who underwent neoadjuvant treatment followed by surgery for adenocarcinoma located at the GOJ at a single high-volume institution between 2002 and 2017. Postoperative major complications and mortality were compared between groups using Fisher's exact test. Overall survival (OS) and disease-free survival (DFS) were assessed by log rank test and multivariable Cox regression analyses. Cumulative incidence functions were used to estimate recurrence, and groups were compared using Gray's test. RESULTS Of 775 patients, 650 had neoadjuvant chemoradiation and 125 had chemotherapy. These groups were comparable in terms of clinical tumour and lymph node categories, although the chemoradiation group had greater proportions of white men, complete pathological response to chemotherapy, and smaller proportions of diffuse cancer, poor differentiation, and neurovascular invasion. Postoperative major complications (20.0 versus 17.6 per cent) and 30-day mortality (1.7 versus 1.6 per cent) were not significantly different between the chemoradiation and chemotherapy groups. After adjustment, type of therapy (chemoradiation versus chemotherapy) was not significantly associated with OS (hazard ratio (HR) 1.26, 95 per cent c.i. 0.96 to 1.67) or DFS (HR 1.27, 0.98 to 1.64). Type of recurrence (local, regional, or distant) did not differ after neoadjuvant chemoradiation versus chemotherapy. CONCLUSION In patients undergoing surgical resection for locally advanced adenocarcinoma of the GOJ, OS and DFS did not differ significantly between patients who had neoadjuvant chemoradiation compared with chemotherapy.
Collapse
Affiliation(s)
- E L Vos
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - R A Carr
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - M Hsu
- Department of Bioinformatics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - M Nakauchi
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - T Nobel
- Department of Surgery, Mount Sinai Health System, New York, New York, USA
| | - A Russo
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - A Barbetta
- Department of Surgery, University of Southern California, Los Angeles, California, USA
| | - K S Tan
- Department of Bioinformatics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - L Tang
- Department of Pathology, Experimental and Gastrointestinal Pathology Services, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - D Ilson
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - G Y Ku
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - A J Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Y Y Janjigian
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - S S Yoon
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - M S Bains
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - D R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - D Coit
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - D Molena
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - V E Strong
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
249
|
Li Y, Zhong Y, Xu Q, Zhu Z, Tian Y. Prognostic Significance of Signet Ring Cells in Gastric Cancer: The Higher Proportion, The Better Survival. Front Oncol 2021; 11:713587. [PMID: 34858807 PMCID: PMC8630623 DOI: 10.3389/fonc.2021.713587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 10/18/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Due to the fact that the definition of gastric signet ring cell cancer (GSRC) was still controversial in the past decades, the prognosis affected by the proportion of signet ring cells within gastric cancer is uncertain. This study compared the clinicopathological features and prognosis of GSRC with the various proportions of signet ring cells. METHODS We collected GSRC cases without metastasis who underwent curative (R0) resection between 2011 and 2018. Individuals who were in the low-proportion signet ring cell group (LSRC, <50%) were matched to those who were in the high-proportion signet ring cell group (HSRC, >50%) through propensity score matching (1:1). We used Cox proportional hazard regression to calculate the adjusted hazard ratios (HR) and 95% confidence intervals (CI) and explored interactions with gender and stage. RESULTS We had 1:1 matched individuals including 231 cases from the LSRC group and 231 cases from the HSRC group. Patients with HSRC had a significantly higher overall survival rate in the multivariable model (aHR = 0.56, 95%CI = 0.38, 0.84) compared with those with LSRC. The association of HSRC appeared to be more substantial among individuals at early stage and N0 stage (p-interaction < 0.01). CONCLUSIONS This study confirms that GSRC with different proportions of signet ring cells could affect the survival of the patient. Further clinical studies should be developed in the future to provide an appropriate treatment strategy for GSRC.
Collapse
Affiliation(s)
- Yang Li
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Quan Xu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhikai Zhu
- Center for Big Data, National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yantao Tian
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
250
|
Yu Z, Huang R, Zhao L, Wang X, Shangguan X, Li W, Li M, Yin X, Zhang C, Liu D. Safety Profile of Oxaliplatin in 3,687 Patients With Cancer in China: A Post-Marketing Surveillance Study. Front Oncol 2021; 11:757196. [PMID: 34745993 PMCID: PMC8567037 DOI: 10.3389/fonc.2021.757196] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/06/2021] [Indexed: 12/19/2022] Open
Abstract
Background Oxaliplatin (OXA), a third-generation platinum derivative, has become one of the main chemotherapeutic drugs for colorectal cancer and other cancers, but reports of adverse reactions are also increasing with the extensive application of OXA. In this study, post-marketing surveillance was carried out to investigate the safety profile of OXA in a real-world setting in Chinese cancer patients to provide a reference for the rational application of OXA. Methods All patients with cancer who received OXA-based chemotherapy in 10 tertiary hospitals in Hubei Province, China, between May 2016 and November 2016 were enrolled. A central registration method was used to document patients' demographics, clinical use, and any incidence of adverse reactions to OXA. All adverse drug reactions (ADRs) were collected and analyzed to assess causality, severity, treatment, and outcome. Results In total, 3687 patients were enrolled in this study. Approximately 64.6% of the patients were male, and 68.8% were aged 50-70 years, with a mean age of 55.3 years. The proportions of patients diagnosed with colorectal and gastric cancers were 59.3% and 31.6%, respectively. In this study, the overall incidence of ADRs and serious ADRs was 42.7% and 1.3%, respectively. The most common ADRs were gastrointestinal disorders (25.7%), blood disorders (21.1%), and peripheral nervous system disorders (8.0%). The serious ADRs identified were hypersensitivity reactions, thrombocytopenia, abnormal hepatic function, and leukopenia/neutropenia. The median onset of gastrointestinal toxicity, myelosuppression, peripheral neurotoxicity, and abnormal hepatic function was 1 d, 5 d, 1 d, and 14 d, respectively. The majority (84.7%) of hypersensitivity reactions were mild to moderate, and the median time to onset of these reactions was within the first 20 min of OXA infusion. Almost 88.0% of patients who experienced ADRs recovered or improved with treatment. Conclusion Our data suggest that OXA-induced ADRs are very common in Chinese patients with cancer; however, more attention should be paid to hypersensitivity reactions caused by OXA. This study provides a valuable reference regarding the safe application of OXA in a real-world setting.
Collapse
Affiliation(s)
- Zaoqin Yu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Huang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zhao
- Hubei Center for Adverse Drug Reaction Monitoring, Wuhan, China
| | - Ximin Wang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofang Shangguan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianguo Yin
- Hubei Center for Adverse Drug Reaction Monitoring, Wuhan, China
| | - Chengliang Zhang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|