1
|
Jiao X, Wang Y, Fu H, Liu Y, Qu J, Fu W. Prognostic significance of lymphovascular invasion in pN0 stage gastric cancer: a propensity score matching analysis. BMC Surg 2024; 24:402. [PMID: 39709382 DOI: 10.1186/s12893-024-02705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND To explore the potential impact of lymphovascular invasion (LVI) on overall survival (OS) of pN0 stage gastric cancer (GC) after curative resection. METHODS A total of 497 GC patients who underwent curative gastrectomy and postoperative pathology proved negative lymph node metastasis between January 2015 and December 2018 in our center were enrolled in this study. All patients were divided into two groups according to the status of LVI. Their clinical and pathological features were compared and potential prognostic factors were analyzed using the propensity score matching analysis (PSM). RESULTS Ninety-nine (19.9%) patients had LVI. The presence of LVI was associated with significantly worse survival outcomes in both the overall and PSM cohorts (χ2 = 19.635, p < 0.001; χ2 = 9.367, p = 0.002). After PSM, data of 99 pairs of patients were extracted. Multivariate analysis revealed that number of examined lymph nodes (LNs), and LVI were independent predictors of OS (all p < 0.05). Following stratified analysis, patients with LNs 11-25 and those without LVI tended to have better OS than those with LVI (LNs 11-15: χ2 = 5.019, p = 0.025; LNs 16-25: χ2 = 11.876, p = 0.001). CONCLUSIONS pN0 stage GC patients with LVI have poor prognosis. More than 15 lymph nodes need to be dissected to reduce the influence of LVI on the prognosis of pN0 stage GC patients.
Collapse
Affiliation(s)
- Xuguang Jiao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, P.R. China
- Department of Gastrointestinal Surgery, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, P.R. China
| | - Yu Wang
- Department of Information Network Management, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, P.R. China
| | - Hao Fu
- Department of Gastrointestinal Surgery, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, P.R. China
| | - Yongning Liu
- Department of Gastrointestinal Surgery, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, P.R. China
| | - Jianjun Qu
- Department of Gastrointestinal Surgery, Weifang People's Hospital, Shandong Second Medical University, Weifang, Shandong, P.R. China
| | - Weihua Fu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, P.R. China.
| |
Collapse
|
2
|
Cao L, Zhu G, Wang X, Kuang Z, Song X, Ma X, Zhu X, Gao R, Li J. Yiqi Wenyang Jiedu prescription for preventing and treating postoperative recurrence and metastasis of gastric cancer: a randomized controlled trial protocol. Front Oncol 2024; 14:1326970. [PMID: 39035732 PMCID: PMC11257841 DOI: 10.3389/fonc.2024.1326970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 06/17/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction Postoperative recurrence and metastasis of gastric cancer (GC) are primary factors that contribute to poor prognosis. GC recurs at a rate of approximately 70%-80% within 2 years after local treatment and approximately 90% within 5 years. "Yang-deficient toxic node" is the core pathogenesis of GC recurrence and metastasis. The Yiqi Wenyang Jiedu prescription (YWJP), a form of complementary and alternative medicine in China, is an empirical remedy to prevent postoperative recurrence and metastasis of GC. Taking the main therapeutic principles of "nourishing Qi and warming Yang, strengthening Zhengqi, and detoxifying" can aid in preventing the recurrence and metastasis of GC in patients during the watchful waiting period after surgery and adjuvant chemotherapy. This approach aims to enhance the quality of life of patients. However, high-quality evidence to support this hypothesis is lacking. This study will aim to investigate the efficacy and safety of YWJP to prevent and treat postoperative metastasis and GC recurrence. Methods The study will be a multicenter, randomized, double-blind, placebo-parallel-controlled clinical trial. A total of 212 patients who completed adjuvant chemotherapy within 8 months of radical gastrectomy will be enrolled. Patients in the intervention group will receive the YWJP, whereas those in the control group will receive a placebo. The main outcome was the disease-free survival (DFS) rate 2 years after surgery. The secondary outcomes included DFS time, overall survival, annual cumulative recurrence and rate of metastasis after 1-3 years, cumulative annual survival after 1-3 years, fat distribution-related indicators, tumor markers, peripheral blood inflammatory indicators, prognostic nutritional index, symptoms and quality of life evaluation, medication compliance, and adverse reaction rate. Discussion There is a lack of effective therapy after the completion of adjuvant therapy during the postoperative period of watchful waiting. This study will be the first randomized clinical trial to evaluate whether complementary and alternative medical interventions can effectively prevent recurrence and metastasis during the watchful waiting period after GC surgery and to provide evidence for surveillance treatment management after GC surgery. Clinical trial registration ClinicalTrials.gov, identifier NCT05229809.
Collapse
Affiliation(s)
- Luchang Cao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guanghui Zhu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xinmiao Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ziyu Kuang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotong Song
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyi Ma
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoyu Zhu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruike Gao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Cobani E, Al Hallak MN, Shields AF, Maier J, Kelly TE, Naidoo N, Tobon M, Kim S, Beal EW. Gastric Cancer Survivorship: Multidisciplinary Management, Best Practices and Opportunities. J Gastrointest Cancer 2024; 55:519-533. [PMID: 38180678 DOI: 10.1007/s12029-023-01001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2023] [Indexed: 01/06/2024]
Abstract
PURPOSE Gastric cancer is the 5th most common malignancy worldwide. As early detection increases and treatments for gastric cancer improve, the number of gastric cancer survivors grows. METHODS Here, we review the diagnosis and management of gastric cancer and discuss important considerations for gastric cancer survivorship including cancer surveillance, weight loss, malnutrition, fatigue, specific complications related to surgery and radiation, quality of life in gastric cancer survivorship, health behavior, and models of survivorship. RESULTS Multimodality therapy with chemotherapy and surgery can result in chronic toxicities in multiple organ systems. This emphasizes the need for a multidisciplinary survivorship care model including cancer surveillance, management of chronic toxicities, and optimization of modifiable risk factors with long-term involvement of appropriate providers. CONCLUSION Adequately caring for gastric cancer survivors requires a coordinated, multidisciplinary approach.
Collapse
Affiliation(s)
- Era Cobani
- Wayne State University School of Medicine, Detroit, MI, USA
| | - Mohammed Najeeb Al Hallak
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R., Mailcode: HW04HO, 48201, Detroit, MI, USA
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R., Mailcode: HW04HO, 48201, Detroit, MI, USA
| | - Jordan Maier
- Department of Radiation Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Thomas E Kelly
- Department of Gastroenterology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Niren Naidoo
- Department of Supportive Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Miguel Tobon
- Department of Surgery, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Steve Kim
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R., Mailcode: HW04HO, 48201, Detroit, MI, USA
- Department of Surgery, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eliza W Beal
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R., Mailcode: HW04HO, 48201, Detroit, MI, USA.
- Department of Surgery, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
4
|
Costa PMD, Antunes C, Lages P, Rodrigues J, Peyroteo M, Onofre S, Lara Santos L. Challenging the Dogma: Stage migration or negative lymph nodes, which of them is the main player on gastric cancer prognosis? EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108318. [PMID: 38626587 DOI: 10.1016/j.ejso.2024.108318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/23/2024] [Accepted: 03/30/2024] [Indexed: 04/18/2024]
Abstract
Expanding loco-regional nodes harvesting is expected to increase survival. This improvement may be associated to stage migration (SM). However, the great bulk of harvested lymph nodes observed in large dissections is negative. M&M: 830 patients who received R0 gastrectomy for adenocarcinoma were included. pN+ patients with <26 nodes (n = 209) were included for a simulation to "offer 26 nodes" - SM (proportional and exponential based) was simulated and analysed through machine learning algorithms. Overall Survival (OS), in native and simulated stages, were compared. OS of extended lymphadenectomies (pN+, D ≥ 26, n = 273) was compared with the simulated curves. OS of patients in the following dissection intervals of negative nodes were compared: <16 (n = 233), 16-25 (n = 258), ≥26 (n = 339). RESULTS: After simulation to 26 nodes (pN+, D < 26 patients, n = 209), staging was recomputed. OS of native vs simulated early-stages (I-II) and advanced stages (III) were not different (p > 0.05). OS of patients with lymphadenectomy (≥26) was better than simulated for early and advanced stages (p = 0.008; p = 0.005). OS of patients included in distinct intervals of negative lymph nodes were different (p < 0.001). These intervals were an independent prognostic factor (multivariate analysis). CONCLUSIONS: The influence of Stage Migration was null in this set of simulations and Will Rogers phenomenon was not observed. Extended dissection performed better in OS. But the influence of the number of negative nodes, even in large dissections, was highlighted. By emphasizing the role of negative nodes, we aim to facilitate more informed decision-making in management of gastric cancer patients, ultimately leading to improved treatment outcomes and patient care.
Collapse
Affiliation(s)
- P Matos da Costa
- Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Hospitais de Santa Maria and Garcia de Orta, Affiliated to Universidade de Lisboa, Lisboa, Portugal.
| | - Cláudia Antunes
- Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| | - Patrícia Lages
- Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Hospitais de Santa Maria and Garcia de Orta, Affiliated to Universidade de Lisboa, Lisboa, Portugal.
| | - Jéssica Rodrigues
- Epidemiology Service, Instituto Português de Oncologia, Porto, Porto Portugal.
| | - Mariana Peyroteo
- Surgical Oncology, Instituto Português de Oncologia, Porto, Porto, Portugal.
| | - Susana Onofre
- Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Hospitais de Santa Maria and Garcia de Orta, Affiliated to Universidade de Lisboa, Lisboa, Portugal.
| | - Lúcio Lara Santos
- Surgical Oncology, Instituto Português de Oncologia, Porto, Porto, Portugal.
| |
Collapse
|
5
|
Škapars R, Gašenko E, Broza YY, Sīviņš A, Poļaka I, Bogdanova I, Pčolkins A, Veliks V, Folkmanis V, Lesčinska A, Liepniece-Karele I, Haick H, Rumba-Rozenfelde I, Leja M. Breath Volatile Organic Compounds in Surveillance of Gastric Cancer Patients following Radical Surgical Management. Diagnostics (Basel) 2023; 13:diagnostics13101670. [PMID: 37238155 DOI: 10.3390/diagnostics13101670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/20/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
As of today, there is a lack of a perfect non-invasive test for the surveillance of patients for potential relapse following curative treatment. Breath volatile organic compounds (VOCs) have been demonstrated to be an accurate diagnostic tool for gastric cancer (GC) detection; here, we aimed to prove the yield of the markers in surveillance, i.e., following curative surgical management. Patients were sampled in regular intervals before and within 3 years following curative surgery for GC; gas chromatography-mass spectrometry (GC-MS) and nanosensor technologies were used for the VOC assessment. GC-MS measurements revealed a single VOC (14b-Pregnane) that significantly decreased at 12 months, and three VOCs (Isochiapin B, Dotriacontane, Threitol, 2-O-octyl-) that decreased at 18 months following surgery. The nanomaterial-based sensors S9 and S14 revealed changes in the breath VOC content 9 months after surgery. Our study results confirm the cancer origin of the particular VOCs, as well as suggest the value of breath VOC testing for cancer patient surveillance, either during the treatment phase or thereafter, for potential relapse.
Collapse
Affiliation(s)
- Roberts Škapars
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Evita Gašenko
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Yoav Y Broza
- Department of Chemical Engineering and Russel Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Armands Sīviņš
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Inese Poļaka
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Inga Bogdanova
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Andrejs Pčolkins
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Viktors Veliks
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Valdis Folkmanis
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Anna Lesčinska
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Inta Liepniece-Karele
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| | - Hossam Haick
- Department of Chemical Engineering and Russel Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Ingrīda Rumba-Rozenfelde
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
| | - Mārcis Leja
- Institute of Clinical and Preventive Medicine, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia
- Department of Abdominal and Soft Tissue Surgery, Oncology Center of Latvia, Riga East University Hospital, LV-1038 Riga, Latvia
| |
Collapse
|
6
|
Xu Q, Kang WZ, Xiong JP, Shao XX, Li WK, Hu HT, Tian YT. A new scoring system to evaluate adjuvant chemotherapy for patients with T2N0M0 gastric cancer after D2 gastrectomy. World J Gastroenterol 2022; 28:5626-5635. [PMID: 36304088 PMCID: PMC9594010 DOI: 10.3748/wjg.v28.i38.5626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/19/2022] [Accepted: 09/21/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND At present, there is insufficient medical evidence to determine whether adjuvant chemotherapy is necessary for T2N0M0 gastric cancer. AIM To obtain a risk score to assess the need for adjuvant chemotherapy in patients with T2N0M0 gastric cancer. METHODS We identified 325 patients with pathological T2N0M0 stage primary gastric cancer at the National Cancer Center between 2011 and 2018. Univariate and multivariate Cox regression analyses were performed to predict factors affecting prognosis. Vascular invasion, tumor site, and body mass index were assessed, and a scoring system was established. We compared the survival outcomes and benefits of adjuvant chemotherapy between the different subgroups. RESULTS Five-year survival rates of the score 0, 1, 2, and 3 groups were 92%, 95%, 80%, and 50%, respectively (P < 0.001). In the score 2-3 group, five-year survival rates for patients in the adjuvant chemotherapy group and postoperative observation group were 95% and 61%, respectively (P = 0.021). CONCLUSION For patients with T2N0M0 stage gastric cancer and two or more risk factors, adjuvant chemotherapy after D2 gastrectomy may have a survival benefit.
Collapse
Affiliation(s)
- 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 100021, China
| | - Wen-Zhe Kang
- 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 100021, China
| | - Jian-Ping Xiong
- 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 100021, China
| | - Xin-Xin Shao
- 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 100021, China
| | - Wei-Kun 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 100021, China
| | - Hai-Tao Hu
- 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 100021, China
| | - Yan-Tao 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 100021, China
| |
Collapse
|
7
|
Yu C, Zhou Z, Liu B, Yao D, Huang Y, Wang P, Li Y. Pathological Nodal Staging Score for Gastric Signet Ring Cell Carcinoma: A Clinical Tool of Adequate Nodal Staging. Diagnostics (Basel) 2022; 12:diagnostics12102289. [PMID: 36291978 PMCID: PMC9600920 DOI: 10.3390/diagnostics12102289] [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: 08/06/2022] [Revised: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Gastric signet ring cell carcinoma (GSRCC) is a subset of gastric cancer with distinct histological and inconsistent prognosis outcome. Currently, the association between the adequate regional lymph node and proper nodal staging in GSRCC is rarely noticed. Materials and methods: Clinical data of GSRCC were retrieved from the Surveillance, Epidemiology, and End Results database. Beta-binomial distribution model was employed for the estimation of the probability of missing nodal disease, followed by the development of a nodal staging score (NSS). Results: A total of 561 GSRCC patients were included in this study, with 193 in lymph node-negative and 368 in lymph node-positive diagnoses. As the number of examined lymph nodes increased, the probability of missing nodal disease decreased rapidly, with T stage-specific curves. The probability of missing nodal disease in T4 was lower than that in T1. NSS calculation indicated that T1 stage patients commonly had NSS > 0.8. However, with the NSS of T2−T4 to reach 0.8, the number of examined lymph node was required to be larger than 12 in T2, 17 in T3 and 27 in T4. NSS ≥ 0.75 (quantile 75%) subgroup in T2−T4 subgroups tended to have better outcome; however, without significant prognostic value. Conclusions: NSS is served as a reliable and feasible tool in adequate nodal staging of GSRCC with statistical basis and provides further evidence for clinical decision making.
Collapse
|
8
|
Spolverato G, Capelli G, Mari V, Lorenzoni G, Gregori D, Poultsides G, Fields RC, Weber SM, Votanopoulos K, Cho CS, He J, Maithel SK, Pucciarelli S, Pawlik TM. Very Early Recurrence After Curative-Intent Surgery for Gastric Adenocarcinoma. Ann Surg Oncol 2022; 29:8653-8661. [PMID: 36018525 DOI: 10.1245/s10434-022-12434-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/31/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Recurrence after curative-intent surgery can occur in more than 50% of gastric cancer (GC) patients. We sought to identify predictors of very early recurrence (VER) among GC patients who underwent curative-intent surgery. METHODS A multi-institutional database of GC patients undergoing curative-intent surgery between 2000 and 2020 at 8 major institutions was queried. VER was defined as local or distant tumor recurrence within 6 months from surgery. Univariable Cox proportional hazard models were used to evaluate the predictive value of clinical-pathological features on VER. A regularized Cox regression model was employed to build a predictive model of VER and recurrence within 12 months. The discriminant ability of the Cox regularized models was evaluated by reporting a ROC curve together with the calibration plot, considering 200 runs. RESULTS Among 1133 patients, 65 (16.0%) patients experienced a VER. Preoperative symptoms (HR 1.198), comorbidities (HR 1.289), tumor grade (HR 1.043), LNR (HR 4.339) and T stage (HR 1.639) were associated with an increased likelihood of VER. Model performance was very good at predicting VER at 6 months (AUC of 0.722) and 12 months (AUC 0.733). Two nomograms to predict 6-month and 12-month VER were built based on the predictive model. A higher nomogram score was associated with worse prognosis. There was good prediction between observed and estimated VER with minimal evidence of overfitting and good performance on internal bootstrapping validation. CONCLUSION One in 6 patients experienced VER following curative-intent surgery for GC. Nomograms to predict risk of VER performed well on internal validation, and stratified patients into distinct prognostic groups relative to 6- and 12-months recurrence.
Collapse
Affiliation(s)
- Gaya Spolverato
- Department of Surgical Oncological and Gastrointestinal Science, University of Padova, Padova, Italy
| | - Giulia Capelli
- Department of Surgical Oncological and Gastrointestinal Science, University of Padova, Padova, Italy
| | - Valentina Mari
- Department of Surgical Oncological and Gastrointestinal Science, University of Padova, Padova, Italy
| | - Giulia Lorenzoni
- Department of Surgical Oncological and Gastrointestinal Science, University of Padova, Padova, Italy
| | - Dario Gregori
- Department of Surgical Oncological and Gastrointestinal Science, University of Padova, Padova, Italy
| | | | - Ryan C Fields
- Department of Surgery, Washington University, St. Louis, MO, USA
| | - Sharon M Weber
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin, Madison, WI, USA
| | | | - Clifford S Cho
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jin He
- Department of Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Shishir K Maithel
- Division of Surgical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Salvatore Pucciarelli
- Department of Surgical Oncological and Gastrointestinal Science, University of Padova, Padova, Italy
| | - Timothy M Pawlik
- Department of Surgery, The Urban Meyer III and Shelley Meyer Chair for Cancer Research, The Ohio State Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
9
|
Mei Y, Feng X, Feng T, Yan M, Zhu Z, Li T, Zhu Z. Adjuvant Chemotherapy in pT2N0M0 Gastric Cancer: Findings From a Retrospective Study. Front Pharmacol 2022; 13:845261. [PMID: 35250596 PMCID: PMC8891981 DOI: 10.3389/fphar.2022.845261] [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: 12/29/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background: There is no global consensus on adjuvant chemotherapy (ACT) for pT2N0M0 gastric cancer. We conducted a retrospective study to reveal the role of ACT in such patients. Methods: Patients with pT2N0M0 gastric cancer who underwent radical resection with D2 lymphadenectomy for primary gastric cancer between January 2012 and May 2016 were included. Kaplan-Meier and Cox regression were used to evaluate overall survival (OS), disease-specific survival (DSS) and predictors of prognosis. Stratified analysis based on high-risk factors was conducted. Results: Of enrolled 307 patients, 111 patients underwent surgery alone and 196 patients received ACT. Surgery alone (HR = 2.913, 95% CI: 1.494-5.682, p = 0.002) and total gastrectomy (HR = 2.445, 95% CI: 1.279-4.675, p = 0.007) were independently associated with decreased OS. With the median follow-up of 73.1 months, the 5-year OS rate was 87.9% and 5-year DSS rate was 91.8%. Patients receiving ACT showed a better 5-year OS rate (92.9 vs. 79.3%, p < 0.001) and DSS rate (96.8 vs. 83.0%, p < 0.001) than patients underwent surgery alone. Patients receiving monotherapy (n = 130) had a relatively poor prognosis compared to patients receiving dual-drug (n = 66) without a significant difference (92.3 vs. 93.9%, p = 0.637). In patients without high-risk factors based on the Chinese Society of Clinical Oncology (CSCO) Guidelines, ACT also provided survival benefit (96.0 vs 82.9%, p = 0.038). Conclusions: ACT was accompanied with higher 5-year OS and DSS rates of patients with pT2N0M0 gastric cancer. Patients with pT2N0M0 gastric cancer, regardless of high-risk factors based on the CSCO guidelines, might be considered candidates for ACT. In regard to the therapy regimen, monotherapy might be the optimal choice, considering the adverse events.
Collapse
Affiliation(s)
- Yu Mei
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xijia Feng
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tienan Feng
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Yan
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenggang Zhu
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Zhenglun Zhu
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Lu J, Wu D, Xu BB, Xue Z, Zheng HL, Xie JW, Wang JB, Lin JX, Li P, Zheng CH, Huang CM. A matched cohort study of the failure pattern after laparoscopic and open gastrectomy for locally advanced gastric cancer: does the operative approach matter? Surg Endosc 2022; 36:689-700. [PMID: 33591445 DOI: 10.1007/s00464-021-08337-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/19/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Due to lacking evidence for confirming the efficacy of performing laparoscopic surgery for locally advanced gastric cancer (LAGC). Therefore, this study aimed to compare the static and dynamic failure patterns after laparoscopic gastrectomy (LG) and open gastrectomy (OG) in LAGC. METHODS A total of 1792 LAGC patients who underwent radical resection between January 2010 and January 2017 were divided into the LG group (n = 1557) and the OG group (n = 235). Propensity score matching was performed to balance the two groups. Dynamic hazard rates of failure were calculated using the hazard function. Early and late failure were defined as failure occurring before and after 2 years since surgery, respectively. RESULTS A total of 1175 patients with LAGC were included after matching (LG group, n = 940; OG, n = 235). The failure rate of the whole cohort was 43.2% (508/1175), accounting for 41.4% (389/940) and 50.6% (119/235) in the LG and OG groups, respectively. Although the two groups showed no significant differences in failure rate for any failure type, landmark analysis showed a lower early distant recurrence rate in the stage IIa-IIIb subgroup of the LG group (OG versus LG: 30.3% versus 21.1%, P = 0.004). The dynamic hazard rate peaked at 9.4 months (peak rate = 0.0186) before gradually declining. In stage IIa-IIIb patients, the hazard rate of the OG group remained significantly higher than that of the LG group within the first 2 years in terms of distant recurrence (peak rate: OG versus LG, 0.0091 versus 0.0055). CONCLUSION Given the differences in early failure between LG and OG, more intensive surveillance for distant recurrence within the first 2 years should be considered for patients with stage IIa-IIIb after OG.
Collapse
Affiliation(s)
- Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Dong Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Bin-Bin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Zhen Xue
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
11
|
Sun JW, Liu DL, Chen JX, Lin LZ, Zhuang LP, Hou XH. Early and late recurrences in lymph node-negative gastric cancer: a retrospective cohort study. Ann Saudi Med 2021; 41:336-349. [PMID: 34873937 PMCID: PMC8650598 DOI: 10.5144/0256-4947.2021.336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Predictors of recurrence in patients with lymph node-negative gastric cancer (GC) who have undergone curative resection have been widely investigated, but not the effects of predictors on timing of recurrence. OBJECTIVE Determine the factors associated with early and late recurrence in patients with node-negative GC. DESIGN Retrospective cohort. SETTING Academic tertiary care center. PATIENTS AND METHODS The study included patients with node-negative GC after curative resection between 2008 and 2018 at two institutions. Early and late recurrences were determined using a minimum P value approach to evaluate the optimal cutoff for recurrence-free survival (RFS). A competing risk model and landmark analysis were used to analyze factors associated with early and late recurrences. MAIN OUTCOME MEASURES Recurrence-free survival and factors associated with survival. SAMPLE SIZE 606. RESULTS After a median follow-up of 70 months, 50 (8.3%) patients experienced recurrent disease. The optimal length of RFS for distinguishing between early (n=26) and late recurrence (n=24) was 24 months (P=.0013). The median RFS in the early and late recurrence groups was 11 and 32 months, respectively. Diffuse tumors (hazard ratio 3.358, P=.014), advanced T stage (HR 8.804, P=.003), perineural invasion (HR 10.955, P<.001), and anemia (HR 2.351, P=.018) were independent predictors of early recurrence. Mixed tumor location (HR 5.586, P=.002), advanced T stage (HR 5.066, P<.001), lymphovascular invasion (HR 5.902, P<.001), and elevated CA19-9 levels (HR 5.227, P<.001) were independent predictors of late recurrence. Similar results were obtained in the landmark analysis. CONCLUSIONS Individualized therapeutic and follow-up strategies should be considered in future studies because of distinct patterns in predictors of early and late recurrence. LIMITATIONS Retrospective design, small sample size. CONFLICT OF INTEREST None.
Collapse
Affiliation(s)
- Jian-Wei Sun
- From the Department of Surgery, Qingyang's People's Hospital, Qingyan, China
| | - Dao-Li Liu
- From the Department of Gastrointestinal Surgery, Anqing First People's Hospital of Anhui Medical University, Anqing, China
| | - Jia-Xian Chen
- From the Department of Surgery, Qingyang's People's Hospital, Qingyan, China
| | - Li-Zhen Lin
- From the Graduate School, Fujian Medical University, Fuzhou, China
| | - Lv-Ping Zhuang
- From the Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xian-Hai Hou
- From the Department of Surgery, Qingyang's People's Hospital, Qingyan, China
| |
Collapse
|
12
|
Jin P, Ji X, Ma S, Kang W, Liu H, Li Y, Ma F, Hu H, Xiong J, Tian Y. Adjuvant chemotherapy indications for stage I gastric cancer patients with negative lymph node. Clin Res Hepatol Gastroenterol 2021; 45:101634. [PMID: 33662780 DOI: 10.1016/j.clinre.2021.101634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/06/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Whether adjuvant chemotherapy (AC) has a survival benefit for pathological stage Ⅰ (T1N0 and T2N0) gastric cancer (GC) patients with negative lymph node (N0) remains controversial. METHODS Patients with surgically resected, histologically confirmed pT1N0 and pT2N0 GC between January 2011 and December 2017 at the National Cancer Center, China, were retrospectively reviewed. RESULTS A total of 1601 patients who met the inclusion criteria were identified. Independent risk factors for reduced overall survival (OS) identified in the Cox regression analysis were male sex (hazard ratio [HR] 1.454, 95% confidence interval [CI] 1.127-1.876), age ≥ 65 years (HR 1.367; 95% CI 1.071-1.744 ), T2 stage (HR 1.283; 95% CI 1.005-1.638), tumor size > 3 cm (HR 1.704; 95% CI 1.346-2.158), examined lymph nodes (EN) ≤ 15 (HR 1.327; 95% CI 1.058-1.664), and non-signet ring cell carcinoma (Non-SRCC) (HR 1.639; 95% CI 1.123-2.392). While only T2 stage (HR 1.410; 95% CI 1.026-1.936), tumor size > 3 cm (HR 1.755; 95% CI 1.285-2.397), examined lymph nodes (EN) ≤ 15 (HR 1.489; 95% CI 1.101-2.015) were independent risk factors for cause-specific survival (CSS). We divided patients with pT2N0 into four sub-categories according to two significant prognostic factors (size and EN) and found that only patient in group 3 (EN ≤ 15, size >3 cm) with improved CSS benefit from AC (p = 0.049). More significant CSS benefit from AC was identified in Non-SRCC patients within group 3 (p = 0.034). CONCLUSION An additional survival benefit related to AC is expected for selected pT2N0 patients. Non-SRCC patients with EN ≤ 15 and tumor size >3 cm may be particularly appropriate candidates for AC.
Collapse
Affiliation(s)
- Peng Jin
- 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, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, PR China
| | - Xiaoyan Ji
- Department of Emergency Ward, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, PR China
| | - Shuai Ma
- 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, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, PR China
| | - Wenzhe Kang
- 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, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, PR China
| | - Hao Liu
- 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, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, PR China
| | - 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, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, PR China
| | - Fuhai Ma
- 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, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, PR China
| | - Haitao Hu
- 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, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, PR China
| | - Jianping Xiong
- 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, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, PR 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, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing 100021, PR China.
| |
Collapse
|
13
|
Lelisho ME, Seid AA, Pandey D. A Case Study on Modeling the Time to Recurrence of Gastric Cancer Patients. J Gastrointest Cancer 2021; 53:218-228. [PMID: 34379265 DOI: 10.1007/s12029-021-00684-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Gastric cancer is a malignant tumor of the stomach and it is one of the leading causes of death worldwide. The study aimed to model the time to first recurrence of gastric cancer patients at the Tikur Anbesa specialized hospital. METHODS The data for this study were gastric cancer patients followed up from January 1, 2013 to February 29, 2020 at Tikur Anbesa Specialized Hospital, Oncology Center, Addis Ababa. We used Weibull, log-logistic and lognormal as baseline hazard functions with the gamma and the inverse Gaussian frailty distributions. Data analyzed with the statistical software R. RESULTS The median recurrence time of the patients was about 23.96 months with a maximum recurrence time of 60.81 months, of which about 61.2% had first recurrences of gastric cancer. The clustering effect is significant in modeling the time to recurrence of gastric cancer. According to the result of the log-logistic inverse Gaussian frailty model, the sex of the patient, the tumor size, smoking habit, the treatment carried out, the vascular invasion, the stage of the disease, the helicobacter pylori infection and the histological type were the significant prognostic factors at 5% level of significance. CONCLUSION Inverse Gaussian frailty model is the model that best describes the time to recurrence of the gastric cancer data set. Gender of the patients, tumor size, treatment taken, vascular invasion, disease stage, helicobacter pylori infection and histological type were the determining prognostic factors. This requires measures to improve patient health and prevent relapse based on significant risk factors, and particular attention should be paid to patients with such factors.
Collapse
Affiliation(s)
- Mesfin Esayas Lelisho
- Department of Statistics, College of Natural Science and Computational, Mizan Tepi University, Tepi, Ethiopia
| | - Adem Aregaw Seid
- Department of Statistics, College of Natural Science and Computational, Mizan Tepi University, Tepi, Ethiopia
| | - Digvijay Pandey
- Department of Technical Education, IET, Dr. A.P.J.Abdul Kalam Technical University Uttar Pradesh, Lucknow, 226021, India.
| |
Collapse
|
14
|
Lu J, Xu BB, Zheng ZF, Xie JW, Wang JB, Lin JX, Chen QY, Cao LL, Lin M, Tu RH, Huang ZN, Zheng CH, Huang CM, Li P. Does three-dimensional surgery affect recurrence patterns in patients with gastric cancer after laparoscopic R0 gastrectomy? Results from a 3-year follow-up phase III trial. Surg Endosc 2021; 35:113-123. [PMID: 31953725 DOI: 10.1007/s00464-020-07367-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 01/04/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Numerous studies have shown that the short-term efficacy of three-dimensional (3D) laparoscopic radical gastrectomy (LG) is comparable to that of two-dimensional (2D)-LG. Whether 3D-LG affects the recurrence patterns of gastric cancer (GC) patients has not been investigated. METHODS From January 2015 to April 2016, a total of 419 patients were recruited for a phase III clinical trial (NCT02327481), which compared the short-term outcomes between the 2D and 3D groups. The long-term efficacy including recurrence patterns was compared between the 2D and 3D groups in this retrospective study. Multivariate analyses were performed to determine whether 3D-LG affects the recurrence patterns. RESULTS Ultimately, 401 patients were analyzed (197 in the 2D-LG group and 204 in the 3D-LG group), and no differences were observed in the clinicopathological data between the two groups. There were no significant differences between the two groups in the recurrence types, first recurrence time or recurrence-free survival (RFS) (all p > 0.05). According to the 7th American Joint Committee on Cancer tumor-node-metastasis (TNM) staging system, both groups were stratified into pathological stages I, II, and III. The stratified analysis showed no significant differences in RFS or overall survival (OS) among patients in each subgroup (all p > 0.05). The multivariate analysis of RFS showed that tumor diameter, pTNM stage, lymphovascular invasion, and adjuvant chemotherapy were independent factors (all p < 0.05). The multivariate analysis of post-recurrence survival (PRS) showed that adjuvant chemotherapy was an independent protective factor (p = 0.043). CONCLUSIONS 3D-LG for GC did not differ significantly from 2D-LG in the effects on 3-year recurrence patterns, RFS and OS, which provides more tumor-related evidence for 3D technology. And due to the technological similarity, it may have certain reference value for robotic-assisted gastrectomy. Further multicenter, large-scale clinical trials are warranted.
Collapse
Affiliation(s)
- Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Bin-Bin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Zhi-Fang Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- 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, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- 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, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- 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, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
15
|
Deep sequencing of circulating tumor DNA detects molecular residual disease and predicts recurrence in gastric cancer. Cell Death Dis 2020; 11:346. [PMID: 32393783 PMCID: PMC7214415 DOI: 10.1038/s41419-020-2531-z] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/29/2022]
Abstract
Identifying locoregional gastric cancer patients who are at high risk for relapse after resection could facilitate early intervention. By detecting molecular residual disease (MRD), circulating tumor DNA (ctDNA) has been shown to predict post-operative relapse in several cancers. Here, we aim to evaluate MRD detection by ctDNA and its association with clinical outcome in resected gastric cancer. This prospective cohort study enrolled 46 patients with stage I–III gastric cancer that underwent resection with curative intent. Sixty resected tumor samples and 296 plasma samples were obtained for targeted deep sequencing and longitudinal ctDNA profiling. ctDNA detection was correlated with clinicopathologic features and post-operative disease-free (DFS) and overall survival (OS). ctDNA was detected in 45% of treatment-naïve plasma samples. Primary tumor extent (T stage) was independently associated with pre-operative ctDNA positivity (p = 0.006). All patients with detectable ctDNA in the immediate post-operative period eventually experienced recurrence. ctDNA positivity at any time during longitudinal post-operative follow-up was associated with worse DFS and OS (HR = 14.78, 95%CI, 7.991–61.29, p < 0.0001 and HR = 7.664, 95% CI, 2.916–21.06, p = 0.002, respectively), and preceded radiographic recurrence by a median of 6 months. In locoregional gastric cancer patients treated with curative intent, these results indicate that ctDNA-detected MRD identifies patients at high risk for recurrence and can facilitate novel treatment intensification studies in the adjuvant setting to improve survival.
Collapse
|
16
|
Wang J, He A, Feng Q, Hou P, Wu J, Huang Z, Xiao Z, Sun C, Liao W, Wu L. Gastrointestinal mixed adenoneuroendocrine carcinoma: a population level analysis of epidemiological trends. J Transl Med 2020; 18:128. [PMID: 32169074 PMCID: PMC7071749 DOI: 10.1186/s12967-020-02293-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/07/2020] [Indexed: 02/06/2023] Open
Abstract
Background The rise in incidence and mortality of gastrointestinal mixed adenoneuroendocrine carcinoma (MANEC) has not been well focused. The aim of our study was to examine epidemiological trends in incidence and incidence-based (IB) mortality of gastrointestinal MANEC at a population level. Methods The incidence and IB mortality of gastrointestinal MANEC as well as data on affected patients from 2000 to 2016 were obtained from the Surveillance, Epidemiology, and End Results database. Trends in incidence and IB mortality were assessed using Joinpoint regression. The Kaplan–Meier method and log-rank test were used for survival analysis. Cox proportional hazards regression was used to identify independent predictors of mortality. Results 581 patients diagnosed with gastrointestinal MANEC were enrolled. Gastrointestinal MANEC incidence was 0.23 cases per 1,000,000 individuals in 2000 and 1.16 cases per 1,000,000 individuals in 2016, with an annual percent change (APC) of 8.0% (95% CI 5.7–10.3%, P < 0.05). IB mortality also showed a sustained increase (APC 12.9%, 95% CI 9.0–16.8%, P < 0.05). In Cox regression analysis, age at diagnosis, tumor grade and stage, lymph node metastasis, surgery, and tumor size were independently associated with mortality. Median survival was 75 months (95% CI 60–128 months). Median survival of appendiceal MANEC was significantly longer than that of cecal MANEC (115 vs. 31 months; P < 0.001). Conclusions We found a sustained and rapid increase both in incidence and IB mortality of gastrointestinal MANEC, manifesting that there has been no significant improvement in patient outcomes, nor progress in prevention and treatment. Additional resources should be devoted to gastrointestinal MANEC research.
Collapse
Affiliation(s)
- Jiakun Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Aoxiao He
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Qian Feng
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Ping Hou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Junjun Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Zhihao Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Zhouqing Xiao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Chi Sun
- Department of Nursing, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China
| | - Wenjun Liao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China.
| | - Linquan Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Nanchang, 330006, China.
| |
Collapse
|
17
|
Zhao B, Huang X, Zhang J, Luo R, Lu H, Xu H, Huang B. Clinicopathologic factors associated with recurrence and long-term survival in node-negative advanced gastric cancer patients. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2020; 111:111-120. [PMID: 30404528 DOI: 10.17235/reed.2018.5829/2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND despite a better prognosis in node-negative advanced gastric cancer (GC), a proportion of patients have a tumor recurrence within five years and eventually die due to cancer-related causes. The present study aimed to evaluate the predictive factors of tumor recurrence and long-term survival in node-negative advanced GC. METHODS a total of 646 node-negative advanced GC patients who underwent a curative gastrectomy in our institution were included in the study. The impact of different clinicopathologic factors on tumor recurrence and overall survival were analyzed. RESULTS tumor recurrences were observed in 181 patients and the cumulative recurrence rate at two-years and five-years were 50.8% and 86.2%, respectively. Lymphovascular invasion, advanced T stage (T3-T4) and an inadequate number of retrieved lymph nodes (LNs) were independent predictive factors of tumor recurrence in node-negative advanced GC. Older age, an upper 1/3 tumor, lymphovascular invasion, infiltration growth pattern (INFγ) and the depth of tumor invasion (T4 stage) were independently associated with long-term survival. With regard to node-negative patients with ≥ 15 retrieved LNs, infiltration growth pattern (INFγ) and advanced T stage (T3-T4) were independent risk factors for both tumor recurrence and long-term survival. CONCLUSION in addition to lymphovascular invasion, inadequate RLNs and advanced T stage, the prognostic significance of infiltration growth pattern in node-negative advanced GC was especially emphasized. These risk factors should be considered when selecting candidates for adjuvant chemotherapy and postoperative surveillance.
Collapse
Affiliation(s)
- Bochao Zhao
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, China
| | - Xinyu Huang
- Department of Clinical Medicine of year 2013, Liaoning University of Traditional Chinese Medicine
| | - Jiale Zhang
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, China
| | - Rui Luo
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, China
| | - Huiwen Lu
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, China
| | - Huimian Xu
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, China
| | - Baojun Huang
- Department of Surgical Oncology, Affiliated Hospital of China Medical University, China
| |
Collapse
|
18
|
Ethun CG, Lopez-Aguiar AG, Switchenko JM, Gillespie TW, Delman KA, Staley CA, Maithel SK, Cardona K. The Prognostic Value of Lymphovascular Invasion in Truncal and Extremity Soft Tissue Sarcomas: An Analysis from the National Cancer Database. Ann Surg Oncol 2019; 26:4723-4729. [PMID: 31502023 DOI: 10.1245/s10434-019-07805-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The aim of this study was to determine the association between lymphovascular invasion (LVI) and overall survival (OS) in truncal/extremity soft tissue sarcomas (STS). METHODS The National Cancer Database (NCDB) was queried for all patients, ages 18-85 years, who underwent resection of primary, truncal/extremity STS between 2010 and 2012, and had LVI data. The primary endpoint was OS. RESULTS Among 6169 patients identified, the most common histology groups were (1) liposarcoma (LPS, 24%), (2) undifferentiated pleiomorphic sarcoma (UPS, 19%), and (3) leiomyosarcoma (LMS, 15%); 449 patients (7%) were LVI-positive. There were no differences in demographics or comorbidities between the LVI groups. Compared with LVI-negative patients, LVI-positive patients were more likely to have larger (> 5 cm: 80% vs. 66%), deep (80% vs. 68%), and high-grade tumors (82% vs. 57%). They were also more likely to have positive margins (27% vs. 17%), nodal (16% vs. 2%) and metastatic disease (21% vs. 4%), and receive chemotherapy (37% vs. 18%; all p < 0.001). LVI was associated with worse median OS (39 months vs. MNR; p < 0.001), which persisted on stratum-specific analyses for all tumor grades, size categories, and stages I-III, but not stage IV. On multivariable Cox regression, LVI was associated with worse OS (hazard ratio [HR] 1.84, 95% confidence interval [CI] 1.39-2.44), while accounting for other significant prognostic factors. Among non-metastatic, curative-intent resections (n = 5696), LVI was still associated with worse OS (HR 1.79, 95% CI 1.28-2.49). CONCLUSIONS LVI appears to be an important adverse pathologic factor in truncal and extremity STS. Even when taking into account other established prognostic factors, LVI was predictive of worse OS. Knowledge of LVI status may help to better risk-stratify patients and guide management strategies, and should be considered in future prognostic classification schemes and nomograms.
Collapse
Affiliation(s)
- Cecilia G Ethun
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Alexandra G Lopez-Aguiar
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jeffery M Switchenko
- Department of Biostatistics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Theresa W Gillespie
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Keith A Delman
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Charles A Staley
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Shishir K Maithel
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Kenneth Cardona
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
19
|
Xu BB, Lu J, Zheng ZF, Xie JW, Wang JB, Lin JX, Chen QY, Cao LL, Lin M, Tu RH, Huang ZN, Lin JL, Zheng CH, Huang CM, Li P. The predictive value of the preoperative C-reactive protein-albumin ratio for early recurrence and chemotherapy benefit in patients with gastric cancer after radical gastrectomy: using randomized phase III trial data. Gastric Cancer 2019; 22:1016-1028. [PMID: 30739259 DOI: 10.1007/s10120-019-00936-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/02/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The definition and predictors of early recurrence (ER) for gastric cancer (GC) patients after radical gastrectomy are unclear. METHODS A minimum-p value approach was used to evaluate the optimal cutoff value of recurrence-free survival to determine ER and late recurrence (LR). Receiver operating characteristic curves were generated for inflammatory indices. Potential risk factors for ER were assessed with a Cox regression model. A decision curve analysis was performed to evaluate the clinical utility. RESULTS A total of 401 patients recruited in a clinical trial (NCT02327481) from January 2015 to April 2016 were included in this study. The optimal length of recurrence-free survival to distinguish between ER (n = 44) and LR (n = 52) was 12 months. Factors associated with ER included a preoperative C-reactive protein-albumin ratio (CAR) ≥ 0.131, stage III and postoperative adjuvant chemotherapy (PAC) > 3 cycles. The risk model consisting of both the CAR and TNM stage had a higher predictive ability and better clinical utility than TNM stage alone. Further stratification analysis of the stage III patients found that for the patients with a CAR < 0.131, both PAC with 1-3 cycles (p = 0.029) and > 3 cycles (p < 0.001) could reduce the risk of ER. However, for patients with a CAR ≥ 0.131, a benefit was observed only if they received PAC > 3 cycles (54.2% vs 16.0%, p = 0.004), rather than 1-3 cycles (58.3% vs 54.2%, p = 0.824). CONCLUSIONS A recurrence-free interval of 12 months was found to be the optimal threshold for differentiating between ER and LR. Preoperative CAR was a promising predictor of ER and PAC response. PAC with 1-3 cycles may not exert a protective effect against ER for stage III GC patients with CAR ≥ 0.131.
Collapse
Affiliation(s)
- Bin-Bin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhi-Fang Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Ju-Li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China.
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
20
|
Liang Y, Liu L, Xie X, Xia L, Meng J, Xu R, He D. Tumor Size Improves the Accuracy of the Prognostic Prediction of Lymph Node-Negative Gastric Cancer. J Surg Res 2019; 240:89-96. [PMID: 30913463 DOI: 10.1016/j.jss.2019.02.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 02/07/2019] [Accepted: 02/22/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Tumor size has been regarded as the "T" stage of many solid tumors because of its effect on prognosis. However, the prognostic impact of tumor size in gastric cancer (GC) is still controversial. MATERIALS AND METHODS A total of 436 patients with curatively resected GC and those without lymph node metastasis in our center were retrospectively enrolled. The appropriate cutoff points for tumor size were determined. Potential prognostic factors were analyzed. In addition, a pathological tumor-size (pTS) classification system was proposed to evaluate the superiority of its prognostic prediction of node-negative GC patients compared with that of the pT staging system. RESULTS The ideal cutoff points for tumor size were 4 and 8 cm. In the multivariate analysis, tumor size was identified as an independent prognostic factor for node-negative GC patients after surgery, as was pT stage. The pTS classification was found to be more appropriate for predicting the overall survival of node-negative GC patients after curative surgery than pT stage, and the -2 log-likelihood of the pTS classification (1680.782) was smaller than the value of pT (1695.239). CONCLUSIONS As an independent prognostic factor, tumor size should be incorporated into the pT staging system to enhance the accuracy of the prognostic prediction of node-negative GC patients.
Collapse
Affiliation(s)
- Yuexiang Liang
- Department of Gastrointestinal Oncology, The First Affiliated Hospital of Hainan Medical University, Haikou City, China
| | - Lijie Liu
- Department of Gastrointestinal Oncology, The First Affiliated Hospital of Hainan Medical University, Haikou City, China
| | - Xiaoming Xie
- Department of Gastrointestinal Oncology, The First Affiliated Hospital of Hainan Medical University, Haikou City, China
| | - Liping Xia
- Department of Gastrointestinal Oncology, The First Affiliated Hospital of Hainan Medical University, Haikou City, China
| | - Jin Meng
- Department of Gastrointestinal Oncology, The First Affiliated Hospital of Hainan Medical University, Haikou City, China
| | - Ronghua Xu
- Department of Gastrointestinal Oncology, The First Affiliated Hospital of Hainan Medical University, Haikou City, China
| | - Donglei He
- Department of Gastrointestinal Oncology, The First Affiliated Hospital of Hainan Medical University, Haikou City, China.
| |
Collapse
|
21
|
Lin JX, Huang YQ, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Tu RH, Huang ZN, Lin JL, Li P, Huang CM, Zheng CH. Age-adjusted Charlson Comorbidity Index (ACCI) is a significant factor for predicting survival after radical gastrectomy in patients with gastric cancer. BMC Surg 2019; 19:53. [PMID: 31133008 PMCID: PMC6537159 DOI: 10.1186/s12893-019-0513-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 05/09/2019] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION To assess the ability of the Age-Adjusted Charlson Comorbidity Index (ACCI) to predict survival after radical gastrectomy in patients with gastric cancer (GC). METHOD Data from patients with GC who underwent radical gastrectomy from January 2008 to December 2012 in Fujian Medical University Union Hospital were retrospectively analyzed. Patients were categorized into either high ACCI group or low ACCI group based on the effect of ACCI on long-term GC prognosis. 1:1 propensity score matching (PSM) was used to reduce confounding bias. To further analyze the impact of ACCI on the long-term prognosis of patients after radical gastrectomy, a nomogram was built based on the Cox proportional hazards regression model. RESULTS A total of 1476 patients were included in the analysis. After PSM, there was no statistically significant differences in tumor location, tumor size and tumor stage between low ACCI group (429 cases) and high ACCI group (429 cases) (all P > 0.05). Before and after PSM, the incidence of postoperative complications in high ACCI group was significantly higher than that in low ACCI group (P < 0.05). The 5-year overall survival rate (OS) in low ACCI group was significantly higher than that in high ACCI group. Multivariate analysis showed that ACCI was an independent risk factor for OS (P < 0.05). The Harrell's C-statistics (C-index) of TNMA, a prognostic evaluation system combining ACCI and TNM staging system, was significantly higher than that of TNM staging system in both the modeling and validation groups (all P < 0.05). CONCLUSIONS ACCI was an independent risk factor for the long-term prognosis of GC patients after radical gastrectomy that could effectively improve the predictive efficacy of the TNM staging system for GC.
Collapse
Affiliation(s)
- Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ying-Qi Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- 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
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Ju-Li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
22
|
Lu J, Xu BB, Zheng ZF, Xie JW, Wang JB, Lin JX, Chen QY, Cao LL, Lin M, Tu RH, Huang ZN, Zheng CH, Huang CM, Li P. CRP/prealbumin, a novel inflammatory index for predicting recurrence after radical resection in gastric cancer patients: post hoc analysis of a randomized phase III trial. Gastric Cancer 2019; 22:536-545. [PMID: 30377862 DOI: 10.1007/s10120-018-0892-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/25/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Serum prealbumin (PALB) can predict the prognosis of patients with gastric cancer (GC). However, the prognostic value of combination of C-reactive protein and PALB (CRP/PALB) remains unclear. METHODS A total of 419 gastric cancer patients included in a clinical trial (NCT02327481) were analyzed. The present study is a substudy of the trial. Receiver operating characteristic (ROC) curves were generated, and by calculating the areas under the curve (AUC) and the C-index, the discriminative ability of each inflammatory index was compared, including CRP/PALB, C-reactive protein/albumin, Glasgow prognostic score (GPS), modified GPS, systemic immune-inflammation index, neutrophil-lymphocyte ratio, and platelet-lymphocyte ratio. RESULTS Ultimately, 401 patients were included in this study. The optimal cutoff value of CRP/PALB was 17.7. According to this cutoff point, the entire sample was divided into a CRP/PALB < 17.7 (LCP) group and a CRP/PALB ≥ 17.7 (HCP) group, comprising 245 and 156 patients, respectively. There were 54 and 22 patients experienced recurrence in the HCP and LCP group, respectively, p < 0.001. Compared with traditional inflammatory indices, CRP/PALB had the highest AUC (0.707) and C-index (0.716), all p < 0.05. The post-recurrence survival (PRS) of patients in the HCP group was significantly shorter than that in the LCP group (p = 0.010), especially for pathological stage III patients (p = 0.015) or patients with distant (p = 0.018) or local (p = 0.023) recurrences. CONCLUSIONS The predictive value of preoperative CRP/PALB for the recurrence of GC is significantly better than traditional inflammatory indices. HCP significantly reduces the PRS, especially for pathological stage III patients or patients with distant or local recurrences.
Collapse
Affiliation(s)
- Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Bin-Bin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhi-Fang Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China.
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
23
|
Borggreve AS, Goense L, Brenkman HJF, Mook S, Meijer GJ, Wessels FJ, Verheij M, Jansen EPM, van Hillegersberg R, van Rossum PSN, Ruurda JP. Imaging strategies in the management of gastric cancer: current role and future potential of MRI. Br J Radiol 2019; 92:20181044. [PMID: 30789792 DOI: 10.1259/bjr.20181044] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Accurate preoperative staging of gastric cancer and the assessment of tumor response to neoadjuvant treatment is of importance for treatment and prognosis. Current imaging techniques, mainly endoscopic ultrasonography (EUS), computed tomography (CT) and 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET), have their limitations. Historically, the role of magnetic resonance imaging (MRI) in gastric cancer has been limited, but with the continuous technical improvements, MRI has become a more potent imaging technique for gastrointestinal malignancies. The accuracy of MRI for T- and N-staging of gastric cancer is similar to EUS and CT, making MRI a suitable alternative to other imaging strategies. There is limited evidence on the performance of MRI for M-staging of gastric cancer specifically, but MRI is widely used for diagnosing liver metastases and shows potential for diagnosing peritoneal seeding. Recent pilot studies showed that treatment response assessment as well as detection of lymph node metastases and systemic disease might benefit from functional MRI (e.g. diffusion weighted imaging and dynamic contrast enhancement). Regarding treatment guidance, additional value of MRI might be expected from its role in better defining clinical target volumes and setup verification with MR-guided radiation treatment.
Collapse
Affiliation(s)
- Alicia S Borggreve
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands.,2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Lucas Goense
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands.,2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Hylke J F Brenkman
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Stella Mook
- 2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Gert J Meijer
- 2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Frank J Wessels
- 3 Department of Radiology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Marcel Verheij
- 4 Department of Radiation Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek (NKI-AVL) , Amsterdam , Netherlands
| | - Edwin P M Jansen
- 4 Department of Radiation Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek (NKI-AVL) , Amsterdam , Netherlands
| | - Richard van Hillegersberg
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Peter S N van Rossum
- 2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Jelle P Ruurda
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| |
Collapse
|
24
|
A Simplified Two-Step Technique for Extended Lymphadenectomy During Resection of Gastroesophageal Malignancy: Early Results Compared to En Bloc Dissection. J Gastrointest Surg 2019; 23:393-401. [PMID: 30603860 DOI: 10.1007/s11605-018-4056-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/13/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Extended lymph node dissection (ELND) remains an important component of curative intent resection of mid-stage gastric cancer (GC). Benefits include enhanced staging accuracy, extending regional disease control, and optimizing potential curability. ELND during gastrectomy remains underutilized in US centers due to a low prevalence of GC operations. METHODS The traditional en bloc ELND was modified into a two-step technique to facilitate greater ease of dissection with better exposure. After completion of the gastrectomy component, retrogastric nodes are dissected in a separate, contiguous specimen. Resulting data were compared to outcomes after en bloc resection. RESULTS Of 179 consecutive patients undergoing gastrectomy, 129 underwent an ELND (73%). There were 97 men and 32 women, with a median age of 64 years (range 24-98). The median total LN count was 25 (3-86). The two-step dissection yielded an average of 18.3 (± 8.5 S.D.) perigastric and 12.1 (± 5.8) retrogastric nodes. Two-step LND was associated with lower estimated blood loss (265 vs. 448 ml, p = 0.0005), lower transfusion requirements (6 vs. 28%, p = 0.007), greater mean total LN counts (30 vs. 26, p = 0.03), and a greater rate of obtaining at least 15 or 20 LNs (91 vs. 77% and 83 vs. 65%, p = 0.05). Major morbidity (overall 16%), length of stay, and survival outcomes were not different. CONCLUSIONS The two-step LND technique as described was found to be associated with favorable operative and postoperative outcome parameters and an excellent LN yield. It can be recommended for standard ELND indications in the absence of macroscopically abnormal LNs.
Collapse
|
25
|
Saluja H, Karapetis CS, Pedersen SK, Young GP, Symonds EL. The Use of Circulating Tumor DNA for Prognosis of Gastrointestinal Cancers. Front Oncol 2018; 8:275. [PMID: 30087854 PMCID: PMC6066577 DOI: 10.3389/fonc.2018.00275] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 07/02/2018] [Indexed: 01/10/2023] Open
Abstract
Gastrointestinal cancers, including oesophageal, gastric and colorectal cancers (CRC) have high rates of disease recurrence despite curative resection. There are a number of recent studies that have investigated the use of circulating tumor DNA (ctDNA) for prognostic value in these cancers. We reviewed studies that had been published prior to March 2018 that assessed the prognostic values of ctDNA in patients with oesophageal and gastric cancers, gastrointestinal stromal tumors (GIST) and CRC. We identified 63 eligible clinical studies that focussed on recurrence and survival. Studies assessed investigated various ctDNA biomarkers in patients with different stages of cancer undergoing surgical resection, chemotherapy and no treatment. For oesophageal squamous cell carcinoma and oesophageal adenocarcinoma, methylation of certain genes such as APC and DAPK have been highlighted as promising biomarkers for prognostication, but these studies are limited and more comprehensive research is needed. Studies focusing on gastric cancer patients showed that methylation of ctDNA in SOX17 and APC were independently associated with poor survival. Two studies demonstrated an association between ctDNA and recurrence and survival in GIST patients, but more studies are needed for this type of gastrointestinal cancer. A large proportion of the literature was on CRC which identified both somatic mutations and DNA methylation biomarkers to determine prognosis. ctDNA biomarkers that identified somatic mutations were more effective if they were personalized based on mutations found in the primary tumor tissue, but ctDNA methylation studies identified various biomarkers that predicted increased risk of recurrence, poor disease free survival and overall survival. While the use of non-invasive ctDNA biomarkers for prognosis is promising, larger studies are needed to validate the clinical utility for optimizing treatment and surveillance strategies to reduce mortality from gastrointestinal cancers.
Collapse
Affiliation(s)
- Hariti Saluja
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Department of Medicine, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Christos S Karapetis
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Department of Oncology, Flinders Medical Centre, Bedford Park, SA, Australia
| | | | - Graeme P Young
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Erin L Symonds
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Bowel Health Service, Flinders Medical Centre, Bedford Park, SA, Australia
| |
Collapse
|
26
|
Schwarz RE. Clinical trends and effects on quality metrics for surgical gastroesophageal cancer care. Transl Gastroenterol Hepatol 2018; 3:43. [PMID: 30148228 DOI: 10.21037/tgh.2018.06.07] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022] Open
Abstract
Background Surgical therapy of mid-stage gastric cancer (GC) and other neoplastic conditions requiring gastric resection remains at the center of curative outcomes, while epidemiologic changes and multimodality treatment options have evolved rapidly. Putative quality metrics for gastrectomy such as R0 rate, total lymph node (LN) count or postoperative morbidity may depend partly on changing disease and treatment patterns, and deserve evaluation under various practice conditions. Methods Data within a U.S.-based single surgical oncologist's practice over 15 years were prospectively recorded and retrospectively analyzed for clinicopathologic factors, operative treatment aspects and outcomes. Trends and spectrum changes over three time intervals were analyzed with contingency analysis and continuous data comparative statistics. Results Of 179 patients undergoing gastric resection, 119 were male and 60 female, with a median age of 63 years (range, 24-98 years). Resections included 56 total, 56 subtotal/distal, 30 proximal and 37 segmental gastrectomies. Diagnoses included 96 GCs, 31 gastroesophageal (GE) junction (GEJ) cancers, 21 GI stromal tumors (GISTs), and 31 other conditions. Significant trends from first towards last time interval were observed for resection type (16% to 32% proximal, 9% to 30% segmental, P=0.0003), curative intent (76% to 98%, P=0.002), diagnosis (5% to 42% GEJ cancer, P<0.0001) and preoperative therapy use (0% to 58%, P<0.0001), among others. Intraoperative aspects showed significantly reduced blood loss (median: 500 to 150 mL) and transfusion requirements (39% to 4%), and an increased use of minimally invasive techniques over time (all at P<0.001). Among patients undergoing curative intent GC resection with LN dissection, total LN counts remained steady (mean: 26), while the number of involved LNs decreased (9.0 to 3.7, P=0.0003) and the R0 resection rate increased from 74% to 85% (P=0.05). The number of specimens with >15 LNs examined increased from 69.0% to 92.5% (P=0.022). At the same time, spleen preservation rate (91% overall) and major morbidity (16%) remained unchanged throughout. Postoperative length of stay decreased from a median of 12 to 8 days (P<0.0001). Conclusions This experience represents some variable practice patterns within a clinicopathologic spectrum of GE diseases. Postoperative or oncologic quality metrics have been sustained or did improve, which would support their utility for various practice settings; they compare favorably to other published U.S. experiences during the same time period.
Collapse
Affiliation(s)
- Roderich E Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA.,Goshen Center for Cancer Care, Goshen, IN, USA
| |
Collapse
|
27
|
Baiocchi GL, Molfino S, Baronchelli C, Giacopuzzi S, Marrelli D, Morgagni P, Bencivenga M, Saragoni L, Vindigni C, Portolani N, Botticini M, De Manzoni G. Recurrence in node-negative advanced gastric cancer: Novel findings from an in-depth pathological analysis of prognostic factors from a multicentric series. World J Gastroenterol 2017; 23:8000-8007. [PMID: 29259375 PMCID: PMC5725294 DOI: 10.3748/wjg.v23.i45.8000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 09/20/2017] [Accepted: 10/17/2017] [Indexed: 02/07/2023] Open
Abstract
AIM To analyze the clinicopathological characteristics of patients with both node-negative gastric carcinoma and diagnosis of recurrence during follow-up. METHODS We enrolled 41 patients treated with curative gastrectomy for pT2-4aN0 gastric carcinoma between 1992 and 2010, who developed recurrence (Group 1). We retrospectively selected this group from the prospectively collected database of 4 centers belonging to the Italian Research Group for Gastric Cancer, and compared them with 437 pT2-4aN0 patients without recurrence (Group 2). We analyzed lymphatic embolization, microvascular infiltration, perineural infiltration, and immunohistochemical determination of p53, Ki67, and HER2 in Group 1 and in a subgroup of Group 2 (Group 2bis) of 41 cases matched with Group 1 according to demographic and pathological characteristics. RESULTS T4a stage and diffuse histotype were associated with recurrence in the group of pN0 patients. In-depth pathological analysis of two homogenous groups of pN0 patients, with and without recurrence during long-term follow-up (groups 1 and 2bis), revealed two striking patterns: lymphatic embolization and perineural infiltration (two parameters that pathologists can easily report), and p53 and Ki67, represent significant factors for recurrence. CONCLUSION The reported pathological features should be considered predictive factors for recurrence and could be useful to stratify node-negative gastric cancer patients for adjuvant treatment and tailored follow-up.
Collapse
Affiliation(s)
- Gian Luca Baiocchi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25121, Italy
| | - Sarah Molfino
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25121, Italy
| | - Carla Baronchelli
- Department of Pathology, Spedali Civili di Brescia, Brescia 25121, Italy
| | - Simone Giacopuzzi
- 1st Department of General Surgery, Borgo Trento Hospital, University of Verona, Verona 37129, Italy
| | - Daniele Marrelli
- Department of Human Pathology and Oncology, Surgical Oncology, University of Siena, Siena 53100, Italy
| | - Paolo Morgagni
- Department of General Surgery, Morgagni-Pierantoni Hospital, Forlì 47121, Italy
| | - Maria Bencivenga
- 1st Department of General Surgery, Borgo Trento Hospital, University of Verona, Verona 37129, Italy
| | - Luca Saragoni
- Department of Pathology, Morgagni-Pierantoni Hospital, Forlì 47121, Italy
| | - Carla Vindigni
- Department of Pathology, Azienda Ospedaliera Universitaria Senese, Siena 53100, Italy
| | - Nazario Portolani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia 25121, Italy
| | | | - Giovanni De Manzoni
- 1st Department of General Surgery, Borgo Trento Hospital, University of Verona, Verona 37129, Italy
| |
Collapse
|
28
|
Hsu JT, Le PH, Kuo CJ, Yeh TS, Jan YY. Survival impact of the number of lymph node retrieved on patients with node-negative gastric cancer: more is better? Transl Gastroenterol Hepatol 2017; 2:103. [PMID: 29354760 PMCID: PMC5763007 DOI: 10.21037/tgh.2017.12.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Jun-Te Hsu
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Puo-Hsien Le
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chia-Jung Kuo
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ta-Sen Yeh
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Yin Jan
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
29
|
Mirkin KA, Hollenbeak CS, Wong J. Greater Lymph Node Retrieval Improves Survival in Node-Negative Resected Gastric Cancer in the United States. J Gastric Cancer 2017; 17:306-318. [PMID: 29302371 PMCID: PMC5746652 DOI: 10.5230/jgc.2017.17.e35] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/25/2017] [Accepted: 11/11/2017] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Guidelines in western countries recommend retrieving ≥15 lymph nodes (LNs) during gastric cancer resection. This study sought to determine whether the number of examined lymph nodes (eLNs), a proxy for lymphadenectomy, effects survival in node-negative disease. MATERIALS AND METHODS The US National Cancer Database (2003-2011) was reviewed for node-negative gastric adenocarcinoma. Treatment was categorized by neoadjuvant therapy (NAT) vs. initial resection, and further stratified by eLN. Kaplan-Meier and Weibull models were used to analyze overall survival. RESULTS Of the 1,036 patients who received NAT, 40.5% had ≤10 eLN, and most underwent proximal gastrectomy (67.8%). In multivariate analysis, greater eLN was associated with improved survival (eLN 16-20: HR, 0.71; P=0.039, eLN 21-30: HR, 0.55; P=0.001). Of the 2,795 patients who underwent initial surgery, 42.5% had ≤10 eLN, and the majority underwent proximal gastrectomy (57.2%). In multivariate analysis, greater eLN was associated with improved survival (eLN 11-15: HR, 0.81; P=0.021, eLN 16-20: HR, 0.73; P=0.004, eLN 21-30: HR, 0.62; P<0.001, and eLN >30: HR, 0.58; P<0.001). CONCLUSIONS In the United States, the majority of node-negative gastrectomies include suboptimal eLN. In node-negative gastric cancer, greater LN retrieval appears to have therapeutic and prognostic value, irrespective of initial treatment, suggesting a survival benefit to meticulous lymphadenectomy.
Collapse
Affiliation(s)
- Katelin A. Mirkin
- Department of Surgery, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Christopher S. Hollenbeak
- Department of Surgery, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
- Department of Public Health Sciences, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Joyce Wong
- Department of Surgery, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| |
Collapse
|
30
|
Poorman CE, Ethun CG, Postlewait LM, Tran TB, Prescott JD, Pawlik TM, Wang TS, Glenn J, Hatzaras I, Shenoy R, Phay JE, Keplinger K, Fields RC, Jin LX, Weber SM, Salem A, Sicklick JK, Gad S, Yopp AC, Mansour JC, Duh QY, Seiser N, Solórzano CC, Kiernan CM, Votanopoulos KI, Levine EA, Staley CA, Poultsides GA, Maithel SK. A Novel T-Stage Classification System for Adrenocortical Carcinoma: Proposal from the US Adrenocortical Carcinoma Study Group. Ann Surg Oncol 2017; 25:520-527. [PMID: 29164414 DOI: 10.1245/s10434-017-6236-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Indexed: 01/29/2023]
Abstract
BACKGROUND The 7th AJCC T-stage system for adrenocortical carcinoma (ACC), based on size and extra-adrenal invasion, does not adequately stratify patients by survival. Lymphovascular invasion (LVI) is a known poor prognostic factor. We propose a novel T-stage system that incorporates LVI to better risk-stratify patients undergoing resection for ACC. METHOD Patients undergoing curative-intent resections for ACC from 1993 to 2014 at 13 institutions comprising the US ACC Group were included. Primary outcome was disease-specific survival (DSS). RESULTS Of the 265 patients with ACC, 149 were included for analysis. The current T-stage system failed to differentiate patients with T2 versus T3 disease (p = 0.10). Presence of LVI was associated with worse DSS versus no LVI (36 mo vs. 168 mo; p = 0.001). After accounting for the individual components of the current T-stage system (size, extra-adrenal invasion), LVI remained a poor prognostic factor on multivariable analysis (hazard ratio 2.14, 95% confidence interval 1.05-4.38, p = 0.04). LVI positivity further stratified patients with T2 and T3 disease (T2: 37 mo vs. median not reached; T3: 36 mo vs. 96 mo; p = 0.03) but did not influence survival in patients with T1 or T4 disease. By incorporating LVI, a new T-stage classification system was created: [T1: ≤ 5 cm, (-)local invasion, (+/-)LVI; T2: > 5 cm, (-)local invasion, (-)LVI OR any size, (+)local invasion, (-)LVI; T3: > 5 cm, (-)local invasion, (+)LVI OR any size, (+)local invasion, (+)LVI; T4: any size, (+)adjacent organ invasion, (+/-)LVI]. Each progressive new T-stage group was associated with worse median DSS (T1: 167 mo; T2: 96 mo; T3: 37 mo; T4: 15 mo; p < 0.001). CONCLUSIONS Compared with the current T-stage system, the proposed T-stage system, which incorporates LVI, better differentiates T2 and T3 disease and accurately stratifies patients by disease-specific survival. If externally validated, this T-stage classification should be considered for future AJCC staging systems.
Collapse
Affiliation(s)
- Caroline E Poorman
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Cecilia G Ethun
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Lauren M Postlewait
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Thuy B Tran
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jason D Prescott
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Timothy M Pawlik
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tracy S Wang
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jason Glenn
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ioannis Hatzaras
- Department of Surgery, New York University School of Medicine, New York, NY, USA
| | - Rivfka Shenoy
- Department of Surgery, New York University School of Medicine, New York, NY, USA
| | - John E Phay
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Kara Keplinger
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Linda X Jin
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Sharon M Weber
- Department of General Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ahmed Salem
- Department of General Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jason K Sicklick
- Department of Surgery, University of California San Diego, San Diego, CA, USA
| | - Shady Gad
- Department of Surgery, University of California San Diego, San Diego, CA, USA
| | - Adam C Yopp
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John C Mansour
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Quan-Yang Duh
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Natalie Seiser
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - Edward A Levine
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Charles A Staley
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - George A Poultsides
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Shishir K Maithel
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, GA, USA. .,Division of Surgical Oncology, Winship Cancer Institute, 1365C Clifton Road NE, 2nd Floor, Atlanta, GA, 30322, USA.
| |
Collapse
|
31
|
Olsen S, Jin L, Fields RC, Yan Y, Nalbantoglu ILK. Tumor budding in intestinal-type gastric adenocarcinoma is associated with nodal metastasis and recurrence. Hum Pathol 2017; 68:26-33. [PMID: 28428104 DOI: 10.1016/j.humpath.2017.03.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/14/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
Gastric adenocarcinoma (GAC) is a common cause of cancer-related death worldwide. GAC can be classified as intestinal or diffuse. Intestinal-type cancers are common and reported to have a better prognosis compared to diffuse cancers. Studies have shown the presence and amount of tumor budding in intestinal carcinomas of the colon and esophagus to predict nodal metastasis and recurrence. Our aim is to determine if tumor budding in intestinal-type GAC correlates with prognostic features. One hundred four patients treated with primary surgical excision between 1999 and 2013 were identified. Histologic type (intestinal, diffuse, or mixed), tumor grade, T-stage, and lymph node status were evaluated. Tumor bud scores were assigned to all intestinal-type cancers using methods previously described for colorectal adenocarcinoma. Scores of <1 were designated as low and ≥1 as high. Tumor characteristics were as follows: 52 intestinal (50%), 36 diffuse (35%), and 16 mixed (15%). Of the 52 cases with intestinal histology, 4 were well (8%), 28 were moderately (54%), and 20 were poorly differentiated (38%). Thirty-three (63%) of the intestinal tumors had high tumor bud scores. Cases with high scores were associated with higher T-stage, N-stage, and grade (P<.001, P<.001, and P=.002). These also had a higher likelihood of recurrence (P=.007). In our cohort, high tumor bud scores in intestinal-type GAC have higher T-stage, N-stage, grade, and likelihood of recurrence. Assessment of tumor budding may guide clinical management in a subset of patients.
Collapse
Affiliation(s)
- Stephen Olsen
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, 63110, USA.
| | - Linda Jin
- Department of Surgery, Washington University in St Louis, St Louis, MO, 63110, USA.
| | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, St Louis, MO 63110.
| | - Yan Yan
- Departments of Surgery and Biostatistics, Washington University in St Louis, St Louis, MO, 63110, USA.
| | - ILKe Nalbantoglu
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, 63110, USA.
| |
Collapse
|
32
|
Zhao LY, Chen XL, Wang YG, Xin Y, Zhang WH, Wang YS, Chen XZ, Yang K, Liu K, Xu L, Zhang B, Chen ZX, Chen JP, Zhou ZG, Hu JK. A new predictive model combined of tumor size, lymph nodes count and lymphovascular invasion for survival prognosis in patients with lymph node-negative gastric cancer. Oncotarget 2016; 7:72300-72310. [PMID: 27509175 PMCID: PMC5342163 DOI: 10.18632/oncotarget.11035] [Citation(s) in RCA: 16] [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: 02/15/2016] [Accepted: 07/18/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Various factors may affect the clinical prognosis of lymph node-negative gastric cancer (GC) patients. This study aimed to provide evaluable prognostic information of combination of tumor size (Ts), lymph nodes count (LNs) and lymphovascular invasion (LVI) in lymph node-negative GC patients. METHODS A total of 1,019 node-negative GC patients were enrolled in this retrospective study from 2000 to 2010. The cutoff points of Ts and LNs were determined using X-tile and patients were randomly categorized into training and validation sets by the sample size ratio 1:1. The clinicopathologic characteristics were analyzed and survival prognostic factors were identified, whereas the survival prediction accuracy was also compared by C-index during the different independent prognostic factors. RESULTS The cutoff points for Ts were 3cm and 5cm, while 14 was the cutoff point for LNs. Age, T stage, Ts, LNs and LVI were identified as independent prognostic factors in node-negative GC patients, and a new prognostic predictive model, TsNL staging system which was composed of Ts, LNs and LVI, was proposed in this study. Compared with T staging system, significant improvement of predictive accuracy for TsNL system was found. Furthermore, nomogram based on TsNL was more accurate in prognostic prediction than that based on Ts, LNs and LVI, separately. CONCLUSIONS Age, T stage, Ts, LNs and LVI were independent prognostic factors in lymph node-negative GC patients. The TsNL staging system, composed of Ts, LNs and LVI, which was closely associated with clinicopathologic features, may improve the prognostic prediction accuracy in node-negative GC patients.
Collapse
Affiliation(s)
- Lin-Yong Zhao
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xiao-Long Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yi-Gao Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yue Xin
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Wei-Han Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yin-Su Wang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xin-Zu Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Liu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lian Xu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhi-Xin Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jia-Ping Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zong-Guang Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jian-Kun Hu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Kong P, Wu R, Yang C, Geng Q, Liu J, Chen S, Liu X, Ye M, He W, Yang Q, Xia L, Xu D. Prognostic Impact of the Signet Ring Cell Type in Node-Negative Gastric Cancer. Sci Rep 2016; 6:26313. [PMID: 27381549 PMCID: PMC4933873 DOI: 10.1038/srep26313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 04/12/2016] [Indexed: 01/18/2023] Open
Abstract
Little is known regarding the prognostic impact of the signet ring cell (SRC) histotype on negative lymph nodes (LNs) in gastric cancer (GC). In this study, we aimed to investigate the differences between SRC and non-SRC GC patients without LN metastasis. The medical records of patients with GC who underwent gastrectomy at Sun Yat-Sen University Cancer Centre from 1996 to 2012 were reviewed to analyse the clinicopathologic characteristics associated with survival. A total of 480 cases of GC patients without LN metastasis were identified, which included 90 SRC GC patients and 390 non-SRC GC patients. Between the two groups, there were a host of significant differences in the American Joint Committee on Cancer, 7th edition (AJCC) stage. We found that SRC histology was correlated with a poor prognosis in terms of recurrence in node-negative GC patients and that SRC histologic analysis combined with AJCC staging maybe an effectual method for prediction of the recurrence rate. Additionally, we found that SRC GC presents a more dismal overall prognosis in patients with perineural or vascular invasion.
Collapse
Affiliation(s)
- Pengfei Kong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of the VIP region, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Ruiyan Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | - Chenlu Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of the Gynaecologic Oncology Surgery, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Qirong Geng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of Hematology Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianjun Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of Gastric and Pancreatic Surgery, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Shangxiang Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of Gastric and Pancreatic Surgery, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Xuechao Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of Gastric and Pancreatic Surgery, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Minting Ye
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of the VIP region, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Wenzhuo He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of the VIP region, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Qiong Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of the VIP region, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Liangping Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of the VIP region, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Dazhi Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China.,Department of Gastric and Pancreatic Surgery, Sun Yat-sen University Cancer Centre, Guangzhou, China
| |
Collapse
|
34
|
Randle RW, Swords DS, Levine EA, Fino NF, Squires MH, Poultsides G, Fields RC, Bloomston M, Weber SM, Pawlik TM, Jin LX, Spolverato G, Schmidt C, Worhunsky D, Cho CS, Maithel SK, Votanopoulos KI. Optimal extent of lymphadenectomy for gastric adenocarcinoma: A 7-institution study of the U.S. gastric cancer collaborative. J Surg Oncol 2016; 113:750-5. [PMID: 26996496 DOI: 10.1002/jso.24227] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/09/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND OBJECTIVES The optimal extent of lymphadenectomy in the treatment of gastric adenocarcinoma is debated. We compared gastrectomy outcomes following limited (D1) or extended (D2) lymphadenectomy. METHODS Using the multi-institutional US Gastric Cancer Collaborative database, we reviewed the morbidity, mortality, recurrence, and overall survival (OS) of patients receiving D1 or D2 lymphadenectomies. RESULTS Between 2000 and 2012, 266 and 461 patients received a D1 and D2 lymphadenectomy, respectively. ASA class, mean number of comorbidities, grade, and stage were similar between groups. While major morbidity was similar (P = 0.85), mortality was worse for those receiving a D1 lymphadenectomy (4.9% vs. 1.3%, P = 0.004). D2 lymphadenectomy was associated with improved median OS in stage I (4.7 years for D1 vs. not reached for D2, P = 0.003), stage II (3.6 years for D1 vs. 6.3 for D2, P = 0.42), and stage III patients (1.3 years for D1 vs. 2.1 for D2, P = 0.01). After adjusting for predictors of OS, D2 lymphadenectomy remained a significant predictor of improved survival (HR 1.5, 95%CI 1.1-2.0, P = 0.008). CONCLUSIONS D2 lymphadenectomy can be performed without increased risk of morbidity and mortality. Additionally, D2 lymphadenectomy is associated with improved survival especially in early stages, and should be considered for gastric adenocarcinoma patients. J. Surg. Oncol. 2016;113:750-755. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Reese W Randle
- Surgical Oncology Service, Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Douglas S Swords
- Surgical Oncology Service, Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Edward A Levine
- Surgical Oncology Service, Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Nora F Fino
- Department of Biostatistics, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | - George Poultsides
- Department of Surgery, Stanford University Medical Center, Stanford, California
| | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Mark Bloomston
- Department of Surgery, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Sharon M Weber
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Timothy M Pawlik
- Division of Surgical Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Linda X Jin
- Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Gaya Spolverato
- Division of Surgical Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Carl Schmidt
- Department of Surgery, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - David Worhunsky
- Department of Surgery, Stanford University Medical Center, Stanford, California
| | - Clifford S Cho
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | | | - Konstantinos I Votanopoulos
- Surgical Oncology Service, Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
35
|
Hsu JT, Yeh TS, Jan YY. Survival impact of the number of lymph node dissection on stage I-III node-negative gastric cancer. Transl Gastroenterol Hepatol 2016; 1:9. [PMID: 28138576 PMCID: PMC5244715 DOI: 10.21037/tgh.2016.03.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 01/06/2016] [Indexed: 12/11/2022] Open
Affiliation(s)
- Jun-Te Hsu
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ta-Sen Yeh
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Yin Jan
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
36
|
Qian J, Qian Y, Wang J, Gu B, Pei D, He S, Zhu F, Røe OD, Xu J, Liu L, Gu Y, Guo R, Yin Y, Shu Y, Chen X. A clinical prognostic scoring system for resectable gastric cancer to predict survival and benefit from paclitaxel- or oxaliplatin-based adjuvant chemotherapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:241-58. [PMID: 26966350 PMCID: PMC4771399 DOI: 10.2147/dddt.s88743] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Gastrectomy with D2 lymphadenectomy is a standard procedure of curative resection for gastric cancer (GC). The aim of this study was to develop a simple and reliable prognostic scoring system for GC treated with D2 gastrectomy combined with adjuvant chemotherapy. Methods A prognostic scoring system was established based on clinical and laboratory data from 579 patients with localized GC without distant metastasis treated with D2 gastrectomy and adjuvant chemotherapy. Results From the multivariate model for overall survival (OS), five factors were selected for the scoring system: ≥50% metastatic lymph node rate, positive lymphovascular invasion, pathologic TNM Stage II or III, ≥5 ng/mL preoperative carcinoembryonic antigen level, and <110 g/L preoperative hemoglobin. Two models were derived using different methods. Model A identified low- and high-risk patients for OS (P<0.001), while Model B differentiated low-, intermediate-, and high-risk patients for OS (P<0.001). Stage III patients in the low-risk group had higher survival probabilities than Stage II patients. Both Model A (area under the curve [AUC]: 0.74, 95% confidence interval [CI]: 0.69–0.78) and Model B (AUC: 0.79, 95% CI: 0.72–0.83) were better predictors compared with the pathologic TNM classification (AUC: 0.62, 95% CI: 0.59–0.71, P<0.001). Adjuvant paclitaxel- or oxaliplatin-based or triple chemotherapy showed significantly better outcomes in patients classified as high risk, but not in those with low and intermediate risk. Conclusion A clinical three-tier prognostic risk scoring system was established to predict OS of GC treated with D2 gastrectomy and adjuvant chemotherapy. The potential advantage of this scoring system is that it can identify high-risk patients in Stage II or III who may benefit from paclitaxel- or oxaliplatin-based regimens. Prospective studies are needed to confirm these results before they are applied clinically.
Collapse
Affiliation(s)
- Jing Qian
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Yingying Qian
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Jian Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Bing Gu
- Department of Laboratory Medicine, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, People's Republic of China; Medical Technology Institute, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Dong Pei
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Shaohua He
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Fang Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Oluf Dimitri Røe
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Oncology, Clinical Cancer Research Center, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark; Department of Surgery, Cancer Clinic, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Jin Xu
- Department of Molecular Cell Biology and Toxicology, Jiangsu Key Lab of Cancer Biomarkers, Prevention & Treatment, Cancer Center, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lianke Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Renhua Guo
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Xiaofeng Chen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| |
Collapse
|
37
|
Aurello P, Petrucciani N, Giulitti D, Campanella L, D'Angelo F, Ramacciato G. Pulmonary metastases from gastric cancer: Is there any indication for lung metastasectomy? A systematic review. Med Oncol 2016; 33:9. [PMID: 26708132 DOI: 10.1007/s12032-015-0718-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/10/2015] [Indexed: 02/07/2023]
Abstract
It is still not clear whether pulmonary resection may have a role in the multidisciplinary management of gastric cancer lung metastases. A systematic literature search was performed to identify all studies published between January 1998 and December 2014 about pulmonary resection of gastric cancer metastases. Ten studies published between 1998 and 2013 were retrieved, including a total of 44 patients. After gastrectomy, median disease-free interval was 35 months. Thirty-eight patients had single lung metastases, whereas six presented with more than one lesion. Median overall survival after lung resection was 45 months, and median disease-free survival was 9 months. Our analysis of the recent literature shows that lung metastasectomy for gastric cancer pulmonary metastases has been reported only in the setting of anecdotic cases or small series of highly selected patients. Lung metastasectomy has no role in the standard management of metastatic gastric patients and may actually be proposed only in individual highly selected cases.
Collapse
Affiliation(s)
- Paolo Aurello
- Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, St Andrea Hospital, Sapienza University, UOC Chirurgia 3, via di Grottarossa, 1035-1039, Rome, Italy
| | - Niccolo' Petrucciani
- Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, St Andrea Hospital, Sapienza University, UOC Chirurgia 3, via di Grottarossa, 1035-1039, Rome, Italy.
| | - Diego Giulitti
- Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, St Andrea Hospital, Sapienza University, UOC Chirurgia 3, via di Grottarossa, 1035-1039, Rome, Italy
| | - Laura Campanella
- Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, St Andrea Hospital, Sapienza University, UOC Chirurgia 3, via di Grottarossa, 1035-1039, Rome, Italy
| | - Francesco D'Angelo
- Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, St Andrea Hospital, Sapienza University, UOC Chirurgia 3, via di Grottarossa, 1035-1039, Rome, Italy
| | - Giovanni Ramacciato
- Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, St Andrea Hospital, Sapienza University, UOC Chirurgia 3, via di Grottarossa, 1035-1039, Rome, Italy
| |
Collapse
|
38
|
Zhou Y, Yu F, Wu L, Ye F, Zhang L, Li Y. Survival after Gastrectomy in Node-Negative Gastric Cancer: A Review and Meta-Analysis of Prognostic Factors. Med Sci Monit 2015; 21:1911-9. [PMID: 26134762 PMCID: PMC4500595 DOI: 10.12659/msm.893856] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Lymph node metastasis is one of the most important prognostic factors for survival of patients with gastric cancer (GC) after surgical resection. Nevertheless, a considerable number of patients have node-negative disease. We performed the present systematic review to evaluate survival and identify prognostic factors in node-negative GC patients undergoing curative intent resection. Material/Methods Relevant studies published between January 2000 and January 2015 were identified by searching the PubMed database and reviewed systematically. Summary relative risks (RR) and 95% confidence intervals (95% CI) were estimated using random-effects models. Results Thirty observational studies involving 12 504 patients were included in the review. Median 5-year overall survival was 84.3% (range, 53–96.3%). Pooled analysis showed that old age (RR, 1.26; 95%CI, 1.13–1.42), <D2 lymph node dissection (1.28; 1.05–1.55), larger tumor (1.18; 1.10–1.26), serosal invasion (2.03; 1.68–2.44), lymphatic invasion (1.25; 1.00–1.57), vascular invasion (1.67; 1.19–2.34), and lymphovascular invasion (1.93; 1.20–3.10) were significant association with decreased survival. Conclusions Surgical resection offers good overall survival for patients with node-negative GC. Tumor-related factors seem to have most prognostic significance.
Collapse
Affiliation(s)
- Yanming Zhou
- Department of Hepato-Biliary-Pancreato-Vascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China (mainland)
| | - Feng Yu
- Department of Hepatobiliary Surgery, The 101th Hospital of Chinese PLA, Wuxi, Jiangsu, China (mainland)
| | - Lupeng Wu
- Department of Hepato-Biliary-Pancreato-Vascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China (mainland)
| | - Feng Ye
- Department of Hepato-Biliary-Pancreato-Vascular Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China (mainland)
| | - Leilei Zhang
- Department of Anaesthesiology, Second Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China (mainland)
| |
Collapse
|