1
|
Jia G, Zhou D, Tang X, Liu J, Lei P. Prognostic value of a modified pathological staging system for gastric cancer based on the number of retrieved lymph nodes and metastatic lymph node ratio. PeerJ 2024; 12:e18165. [PMID: 39372713 PMCID: PMC11451444 DOI: 10.7717/peerj.18165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
Aim The prognosis for gastric cancer (GC) remains grim, underscoring the importance of accurate staging and treatment. Given the potential benefits of using lymph node ratio (LNR) for improved prognostication and treatment planning, it is critical to incorporate examined lymph nodes (ELN) count in an integrated GC staging system. Methods Patients data from the Surveillance, Epidemiology, and End Results (SEER) database between 2010 and 2015 was utilized as training set. The Mantel-Cox survival test was used to calculate chi-square values for 40 LNR segments with a 0.025 interval, defining a novel LNR-based N (rN) classification based on the cutoff points. A revised AJCC (rAJCC) staging system was established by replacing the 8th AJCC N staging with a rN classification. The relationship between the ELN count and prognosis or positive lymph node detection was conducted by using multivariable models. The series of the odds ratios and hazard ratios were fitted with a locally weighted scatterplot smoothing (LOWESS) smoother, and the structural break points were determined by Chow test to clarify an optimal minimum ELN count. The integrated GC staging system incorporated both rAJCC system and the ideal ELN count. Discriminatory ability and prognostic homogeneity of the rAJCC and integrated staging system was compared with AJCC staging system in the SEER validation set (2016-2017), the Cancer Genome Atlas Program (TCGA) database, and the Third Affiliated Hospital of Sun Yat-sen University database. Results The current study found that LNR and ELN count are both significantly associated with the prognosis of GC patients (HR = 0.98, p < 0.001 and HR = 2.51, p < 0.001). Four peaks of the chi-square value were identified as LNR cut-off points at 0.025, 0.175, 0.45 and 0.6 to define a novel rN stage. In comparison to the 8th AJCC staging system, the rAJCC staging system demonstrated significant prognostic advantages and discriminatory ability in the training set (5-Y OS AUC: 71.7 vs. 73.0; AIC: 57,290.7 vs. 57,054.9). The superiority of the rAJCC staging system was confirmed in all validation sets. Using a LOWESS smoother and Chow test, a threshold ELN count of 30 was determined to maximum improvement in the prognosis of node-negative patients without downgrading due to potential metastasis, while also maximizing the detection efficiency of at least one involved lymph node. The integrated staging system, combining the refined rAJCC classification with an optimized ELN count threshold, has demonstrated superior discriminatory performance compared to the standalone rAJCC or the traditional AJCC system. Conclusion The development of a novel GC staging system, which integrated the LNR-based N classification and the minimum ELN count, has exhibited superior prognostic accuracy, holding promise as a valuable asset in the clinical management of GC. However, it is crucial to recognize the limitations from the retrospective database, which should be addressed in subsequent analyses.
Collapse
Affiliation(s)
- Guiru Jia
- Department of Gastrointestinal Surgery, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Dagui Zhou
- Department of Gastrointestinal Surgery, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiao Tang
- Department of Gastrointestinal Surgery, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jianpei Liu
- Department of Gastrointestinal Surgery, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Purun Lei
- Department of Gastrointestinal Surgery, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Song Y, Hirata Y, Ajani JA, Blum Murphy M, Li JJ, Das P, Minsky BD, Mansfield PF, Ikoma N, Badgwell BD. Survival Outcomes in Patients with Resectable Gastric Cancer Treated with Total Neoadjuvant Therapy. Ann Surg Oncol 2024; 31:6918-6930. [PMID: 39048909 DOI: 10.1245/s10434-024-15893-7] [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] [Received: 05/28/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Perioperative chemotherapy has become the standard of care for locally advanced gastric cancer. Total neoadjuvant therapy (TNT), including both chemotherapy and chemoradiation, is utilized in other gastrointestinal malignancies. We determined survival in a contemporary cohort of gastric cancer patients treated with TNT. METHODS Using a prospective institutional database, patients diagnosed with cT2-4 or cN+ gastric adenocarcinoma (January 2012 to June 2022) who underwent staging laparoscopy, received TNT, and underwent gastrectomy were identified. Overall survival (OS) and disease-specific survival (DSS) were determined using standard statistical methods. RESULTS The study included 203 patients. The most common TNT sequence was induction chemotherapy followed by chemoradiation (n = 186 [91.6%]). A total of 195 (96.1%) patients completed planned neoadjuvant treatments. Surgery included total gastrectomy in 108 (53.2%), extended (D1+/D2) lymphadenectomy in 193 (95.1%), and adjacent organ resection in 19 (9.4%) patients. Pathologic complete response (pCR) was achieved in 32 (15.8%) patients. The 5-year OS rate was 65.2% (95% confidence interval [CI] 57.8-73.5%), and the 5-year DSS rate was 70.8% (95% CI 63.6-78.9%) in the study cohort. Among patients with pCR, the 5-year OS rate was 89.1% (95% CI 78.1-100.0%), and the 5-year DSS rate was 96.9% (95% CI 91-100%). Posttreatment pathologic N and M stages were the strongest prognostic indicators associated with both OS and DSS. CONCLUSIONS Total neoadjuvant therapy for resectable gastric cancer is associated with a high rate of treatment completion and promising survival outcomes. Prospective comparisons with perioperative treatment are needed to identify patients most likely to benefit from TNT.
Collapse
Affiliation(s)
- Yun Song
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuki Hirata
- Department of Surgery, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Jaffer A Ajani
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mariela Blum Murphy
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jenny J Li
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prajnan Das
- Division of Radiation Oncology, Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bruce D Minsky
- Division of Radiation Oncology, Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul F Mansfield
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naruhiko Ikoma
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian D Badgwell
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
3
|
Toji Y, Takeuchi S, Ebihara Y, Kurashima Y, Harada K, Hayashi M, Abe H, Wada H, Yorinaga S, Shichinohe T, Tomaru U, Komatsu Y, Hirano S. Perioperative chemotherapy with nivolumab for HER2-negative locally advanced gastric cancer: a case series. Surg Case Rep 2024; 10:200. [PMID: 39192090 DOI: 10.1186/s40792-024-02001-w] [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: 06/25/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Nivolumab with chemotherapy has been transformative for metastatic gastric cancer (GC). The potential of this regimen for local tumor control could be utilized for perioperative chemotherapy in locally advanced GC with bulky tumors or lymph node metastasis involving other organs. CASE PRESENTATION Five patients with HER2-negative advanced GC were treated with nivolumab and oxaliplatin-based chemotherapy. All patients presented with clinical stage III or IVA GC with tumors in contact with either the pancreas or liver. Following chemotherapy, all tumors demonstrated shrinkage, allowing successful radical gastrectomies including four minimally invasive approach without postoperative complications. Four patients avoided combined resection of other organs. CONCLUSIONS Perioperative chemotherapy with nivolumab was effective for local disease control in this case series. This regimen could be a promising treatment approach for locally advanced GC; however, its survival benefits should be evaluated in clinical trials.
Collapse
Affiliation(s)
- Yuta Toji
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Shintaro Takeuchi
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Yuma Ebihara
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Yo Kurashima
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Kazuaki Harada
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Mariko Hayashi
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Hirotake Abe
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Hideyuki Wada
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Satoko Yorinaga
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
- Department of Surgical Pathology, Hokkaido University Hospital, West-5, North-14, Kita-ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Toshiaki Shichinohe
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Utano Tomaru
- Department of Surgical Pathology, Hokkaido University Hospital, West-5, North-14, Kita-ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, West-5, North-14, Kita-ku, Sapporo, Hokkaido, 060-8648, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Hokkaido University, West-7, North-15, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| |
Collapse
|
4
|
Li Y, Huang LH, Zhu HD, He P, Li BB, Wen LJ. Postoperative body weight change and its influencing factors in patients with gastric cancer. World J Gastrointest Surg 2024; 16:2242-2254. [PMID: 39087095 PMCID: PMC11287674 DOI: 10.4240/wjgs.v16.i7.2242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/16/2024] [Accepted: 06/03/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND The high incidence and mortality of gastric cancer (GC) pose a significant threat to human life and health, and it has become an important public health challenge in China. Body weight loss is a common complication after surgical treatment in patients with GC and is associated with poor prognosis and GC recurrence. However, current attention to postoperative weight change in GC patients remains insufficient, and the descriptions of postoperative weight change and its influencing factors are also different. AIM To investigate body weight changes in patients with GC within 6 mo after gastrectomy and identify factors that influence dynamic body weight changes. METHODS We conducted a prospective longitudinal study of 121 patients with GC and collected data before (T0) and 1 (T1), 3 (T2), and 6 (T3) mo after gastrectomy using a general data questionnaire, psychological distress thermometer, and body weight measurements. The general estimation equation (GEE) was used to analyze the dynamic trends of body weight changes and factors that influence body weight changes in patients with GC within 6 mo of gastrectomy. RESULTS The median weight loss at T1, T2, and T3 was 7.29% (2.84%, 9.40%), 11.11% (7.64%, 14.91%), and 14.75% (8.80%, 19.84%), respectively. The GEE results showed that preoperative body mass index (BMI), significant psychological distress, religious beliefs, and sex were risk factors for weight loss in patients with GC within 6 mo after gastrectomy (P < 0.05). Compared with preoperative low-weight patients, preoperative obese patients were more likely to have weight loss (β = 14.685, P < 0.001). Furthermore, patients with significant psychological distress were more likely to lose weight than those without (β = 2.490, P < 0.001), and religious patients were less likely to lose weight 6 mo after gastrectomy than those without religious beliefs (β = -6.844, P = 0.001). Compared to female patients, male patients were more likely to experience weight loss 6 mo after gastrectomy (β = 4.262, P = 0.038). CONCLUSION Male patients with GC with high preoperative BMI, significant psychological distress, and no religious beliefs are more likely to lose weight after gastrectomy.
Collapse
Affiliation(s)
- Yan Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Li-Hua Huang
- Department of Nursing, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Hui-Di Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ping He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Bei-Bei Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Li-Jing Wen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
5
|
Zhao Y, Li D, Zhuang J, Li Z, Xia Q, Li Z, Yu J, Wang J, Zhang Y, Li K, Xu S, Li S, Ma P, Cao Y, Liu C, Xu C, Liu Z, Wei J, Zhang C, Qiao L, Gao X, Hou Z, Liu C, Zheng R, Wang D, Liu Y. Comprehensive multi-omics analysis of resectable locally advanced gastric cancer: Assessing response to neoadjuvant camrelizumab and chemotherapy in a single-center, open-label, single-arm phase II trial. Clin Transl Med 2024; 14:e1674. [PMID: 38685486 PMCID: PMC11058238 DOI: 10.1002/ctm2.1674] [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: 11/27/2023] [Revised: 04/07/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The current standard of care for locally advanced gastric cancer (GC) involves neoadjuvant chemotherapy followed by radical surgery. Recently, neoadjuvant treatment for this condition has involved the exploration of immunotherapy plus chemotherapy as a potential approach. However, the efficacy remains uncertain. METHODS A single-arm, phase 2 study was conducted to evaluate the efficacy and tolerability of neoadjuvant camrelizumab combined with mFOLFOX6 and identify potential biomarkers of response through multi-omics analysis in patients with resectable locally advanced GC. The primary endpoint was the pathological complete response (pCR) rate. Secondary endpoints included the R0 rate, near pCR rate, progression-free survival (PFS), disease-free survival (DFS), and overall survival (OS). Multi-omics analysis was assessed by whole-exome sequencing, transcriptome sequencing, and multiplex immunofluorescence (mIF) using biopsies pre- and post-neoadjuvant therapy. RESULTS This study involved 60 patients, of which 55 underwent gastrectomy. Among these, five (9.1%) attained a pathological complete response (pCR), and 11 (20.0%) reached near pCR. No unexpected treatment-emergent adverse events or perioperative mortality were observed, and the regimen presented a manageable safety profile. Molecular changes identified through multi-omics analysis correlated with treatment response, highlighting associations between HER2-positive and CTNNB1 mutations with treatment sensitivity and a favourable prognosis. This finding was further supported by immune cell infiltration analysis and mIF. Expression data uncovered a risk model with four genes (RALYL, SCGN, CCKBR, NTS) linked to poor response. Additionally, post-treatment infiltration of CD8+ T lymphocytes positively correlates with pathological response. CONCLUSION The findings suggest the combination of PD-1-inhibitor and mFOLFOX6 showed efficacy and acceptable toxicity for locally advanced GC. Extended follow-up is required to determine the duration of the response. This study lays essential groundwork for developing precise neoadjuvant regimens.
Collapse
Affiliation(s)
- Yuzhou Zhao
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Danyang Li
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Jing Zhuang
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Zhimeng Li
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Qingxin Xia
- Department of PathologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Zhi Li
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Juan Yu
- Department of Endoscopy CenterThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Jinbang Wang
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Yong Zhang
- Department of ImmunotherapyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Ke Li
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Shuning Xu
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Sen Li
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Pengfei Ma
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Yanghui Cao
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Chenyu Liu
- Department of Surgical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Chunmiao Xu
- Department of RadiologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Zhentian Liu
- Department of translational medicineGeneplus‐Beijing InstituteBeijingChina
| | - Jinwang Wei
- GenomiCare Biotechnology LA Co., Ltd.ShanghaiChina
| | - Chengjuan Zhang
- Department of PathologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Lei Qiao
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Xuan Gao
- Department of translational medicineGeneplus‐Beijing InstituteBeijingChina
| | - Zhiguo Hou
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Chenxuan Liu
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | | | - Du Wang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Ying Liu
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| |
Collapse
|
6
|
Tian Y, Yang P, Guo H, Liu Y, Zhang Z, Ding P, Zheng T, Deng H, Ma W, Li Y, Fan L, Zhang Z, Wang D, Zhao X, Tan B, Liu Y, Zhao Q. Neoadjuvant docetaxel, oxaliplatin plus capecitabine versus oxaliplatin plus capecitabine for patients with locally advanced gastric adenocarcinoma: long-term results of a phase III randomized controlled trial. Int J Surg 2023; 109:4000-4008. [PMID: 37678277 PMCID: PMC10720837 DOI: 10.1097/js9.0000000000000692] [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: 05/16/2023] [Accepted: 08/04/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Neoadjuvant chemotherapy with docetaxel, oxaliplatin, and capecitabine (DOX regimen) is rarely used in Eastern countries and its efficacy and safety in advanced gastric cancer have not been reported. In this open-label, randomized, controlled trial, the authors aimed to assess the clinical efficacy of neoadjuvant chemotherapy using the DOX and oxaliplatin plus capecitabine (XELOX) regimens, in comparison to surgery alone. MATERIALS AND METHODS Three hundred patients younger than 60 years with potentially resectable advanced gastric cancer (cT3-4, Nany, M0) were enrolled in this randomized controlled clinical trial between November 2014 and June 2018. The primary endpoint of the study was the pathological complete response (pCR) rate. Secondary endpoints included 3-year overall survival (OS), 3-year disease-free survival. RESULTS In total, 280 patients (93 in the DOX group, 92 in the XELOX group, and 95 in the surgery group) were included in the per-protocol analysis. The DOX group demonstrated a significantly higher pCR rate compared to the XELOX group (16.1 vs. 4.3%, P =0.008). For patients with intestinal type, the DOX group exhibited significantly higher rates of both pCR and major pathological response compared to the XELOX group ( P =0.007, P <0.001). The 3-year OS rates of the DOX group, the XELOX group and the surgery group were 56.9, 44.6, and 34.7%, respectively. The 3-year disease-free survival rates were 45.2, 40.2, and 28.4%, respectively. The neoadjuvant DOX regimen demonstrated a significant improvement in the 3-year OS of patients compared to the neoadjuvant XELOX regimen ( P =0.037). CONCLUSION The neoadjuvant DOX regimen has shown the potential to increase the pCR rate and improve the prognosis of patients with advanced gastric cancer who are under 60 years old.
Collapse
Affiliation(s)
- Yuan Tian
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Peigang Yang
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Honghai Guo
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Yang Liu
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Ze Zhang
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Pingan Ding
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Tao Zheng
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Huiyan Deng
- Department of Pathology, The Fourth Hospital of Hebei Medical University
| | | | - Yong Li
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Liqiao Fan
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Zhidong Zhang
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Dong Wang
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Xuefeng Zhao
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Bibo Tan
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Yu Liu
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| | - Qun Zhao
- The Third Department of Surgery
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, People’s Republic of China
| |
Collapse
|
7
|
Tsagkalidis V, Blaszczyk MB, In H. Interpretation of Tumor Response Grade following Preoperative Therapy for Gastric Cancer: An Overview. Cancers (Basel) 2023; 15:3662. [PMID: 37509323 PMCID: PMC10377503 DOI: 10.3390/cancers15143662] [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: 06/15/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Gastric cancer is among the top five causes of cancer-related death worldwide. Preoperative chemotherapy has been established as an option in patients with locally advanced gastric cancer. However, chemotherapy yields variable results, owing to the cellular and molecular heterogeneity of this disease. Identifying patients who did or did not respond to preoperative therapy can allow clinicians to alter treatment modalities and provide important information related to prognostication. A pathologic response to preoperative therapies, called the Tumor Response Grade (TRG), has been evaluated to quantify treatment response. Multiple systems for TRG have been established. However, the literature has demonstrated inconsistent results for TRG systems and prognosis, possibly due to variability in interpretation of tumor response between systems and interobserver variability. Radiographic responses to preoperative therapies using RECIST 1.1 criteria and endoscopically assessed tumor response have demonstrated association with survival; however, their use in gastric cancer remains challenging given the inability to accurately and consistently identify and measure the tumor, especially in the setting of neoadjuvant therapy, where treatment-related changes can obscure the gastric wall layers. While the response to preoperative therapies with positron emission tomography (PET) has shown promising results in esophageal and esophagogastric junction (EGJ) malignancies, its role in gastric cancer is still under investigation. This review is focused on summarizing the available literature related to evaluating TRG in gastric cancer, as well as providing a brief overview of the use of radiographic and endoscopic methods to assess response to preoperative therapies. Lastly, we outline future directions regarding the use of a universal TRG system to guide care and assist with prognosis.
Collapse
Affiliation(s)
- Vasileios Tsagkalidis
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Maryjka B Blaszczyk
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08901, USA
| | - Haejin In
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Department of Health Behavior, Society and Policy, Rutgers School of Public Health, Piscataway, NJ 08854, USA
| |
Collapse
|
8
|
Que SJ, Zhong Q, Chen QY, Truty MJ, Yan S, Ma YB, Ding FH, Zheng CH, Li P, Wang JB, Lin JX, Lu J, Cao LL, Lin M, Tu RH, Lin JL, Zheng HL, Huang CM. A Novel ypTLM Staging System Based on LODDS for Gastric Cancer After Neoadjuvant Therapy: Multicenter and Large-Sample Retrospective Study. World J Surg 2023; 47:1762-1771. [PMID: 37069317 DOI: 10.1007/s00268-023-06994-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND The accuracy of the eighth AJCC ypTNM staging system on the prognosis of gastric cancer (GC) patients after neoadjuvant therapy (NAT) is controversial. This study aimed to develop and validate a novel staging system using the log odds of positive lymph nodes scheme (LODDS). METHODS A retrospective analysis of 606 GC patients who underwent radical gastrectomy after neoadjuvant therapy was conducted as the development cohort. (Fujian Medical University Affiliated Union Hospital (n = 183), Qinghai University Affiliated Hospital (n = 169), Mayo Clinic (n = 236), Lanzhou University First Hospital (n = 18)). The validation cohort came from the SEER database (n = 1701). A novel ypTLoddsS (ypTLM) staging system was established using the 3-year overall survival. The predictive performance of two systems was compared. RESULTS Two-step multivariate Cox regression analysis in both cohorts showed that ypTLM was an independent predictor of overall survival of GC patients after neoadjuvant therapy (HR: 1.57, 95% CI: 1.30-1.88, p < 0.001). In the development cohort, ypTLM had better discrimination ability than ypTNM (C-index: 0.663 vs 0.633, p < 0.001), better prediction homogeneity (LR: 97.7 vs. 70.9), and better prediction accuracy (BIC: 3067.01 vs 3093.82; NRI: 0.36). In the validation cohort, ypTLM had a better prognostic predictive ability (C-index: 0.614 vs 0.588, p < 0.001; LR: 11,909.05 vs. 11,975.75; BIC: 13,263.71 vs 13,328.24; NRI: 0.22). The time-dependent ROC curve shows that the predictive performance of ypTLM is better than ypTNM, and the analysis of the decision curve shows that ypTLM achieved better net benefits. CONCLUSION A LODDS-based ypTLM staging system based on multicenter data was established and validated. The predictive performance was superior to the eighth AJCC ypTNM staging system.
Collapse
Affiliation(s)
- Si-Jin Que
- 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
| | - Qing Zhong
- 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
| | - 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, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Mark J Truty
- Section of Hepatobiliary and Pancreatic Surgery, Division of Subspecialty General Surgery, Department of Surgery, Mayo Clinic, Rochester, USA
| | - Su Yan
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, China
| | - Fang-Hui Ding
- Department of Gastrointestinal Surgery, First Hospital of Lanzhou University, Lanzhou, 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
| | - 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
| | - 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
| | - 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
| | - 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, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 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, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 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, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 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, 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
| | - 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
|
9
|
Wang C, Wang Z, Zhao Y, Wang F. Neoadjuvant PD-1 Inhibitor Plus Apatinib and Chemotherapy Versus Apatinib Plus Chemotherapy in Treating Patients With Locally Advanced Gastric Cancer: A Prospective, Cohort Study. J Gastric Cancer 2023; 23:328-339. [PMID: 37129156 PMCID: PMC10154141 DOI: 10.5230/jgc.2023.23.e17] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/23/2023] [Accepted: 01/28/2023] [Indexed: 05/03/2023] Open
Abstract
PURPOSE This study aimed to evaluate the efficacy and safety of neoadjuvant programmed cell death-1 (PD-1) inhibitors plus apatinib and chemotherapy (PAC) in patients with locally advanced gastric cancer (LAGC). MATERIALS AND METHODS Seventy-three patients with resectable LAGC were enrolled and named the PAC group (n=39) or apatinib plus chemotherapy (AC) group (n=34) based on the treatment they chose. Neoadjuvant therapy was administered in a 21-day cycle for 3 consecutive cycles, after which surgery was performed. RESULTS The PAC group exhibited a higher objective response rate than the AC group (74.4% vs. 58.8%, P=0.159). Moreover, the PAC group showed a numerically better response profile than the AC group (P=0.081). Strikingly, progression-free survival (PFS) (P=0.019) and overall survival (OS) (P=0.049) were prolonged, whereas disease-free survival (DFS) tended to be longer in the PAC group than in the AC group (P=0.056). Briefly, the 3-year PFS, DFS, and OS rates were 76.1%, 76.1%, and 86.7% in the PAC group and 46.9%, 49.9%, and 70.3% in the AC group, respectively. Furthermore, PAC (vs. AC) treatment (hazard ratio=0.286, P=0.034) was independently associated with prolonged PFS in multivariate Cox regression analyses. The incidence of adverse events did not differ between the two groups (all P>0.05), where leukopenia, anemia, hypertension, and other adverse events were commonly observed in the PAC group. CONCLUSIONS Neoadjuvant PAC therapy may achieve a preferable pathological response, delayed progression, and prolonged survival compared to AC therapy with a similar safety profile in patients with LAGC; however, further validation is warranted.
Collapse
Affiliation(s)
- Chunjing Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhen Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Zhao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fujing Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
10
|
Otsuka R, Hayano K, Hayashi H, Uesato M, Murakami K, Toyozumi T, Matsumoto Y, Kurata Y, Nakano A, Matsubara H. ypTNM staging is a potentially useful prognostic stratification tool in patients with advanced gastric cancer after preoperative chemotherapy. Langenbecks Arch Surg 2023; 408:133. [PMID: 37000278 DOI: 10.1007/s00423-023-02872-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 03/27/2023] [Indexed: 04/01/2023]
Abstract
PURPOSE Although the usefulness of the ypStage in neoadjuvant chemotherapy for advanced gastric cancer (GC) has been reported, whether or not the ypStage is applicable to all GC patients who receive preoperative chemotherapy, including conversion surgery cases, is unclear. Therefore, this retrospective study evaluated the value of the ypTNM staging system in all advanced GC patients who received chemotherapy prior to gastrectomy. METHODS A total of 66 patients who underwent chemotherapy prior to gastrectomy for advanced GC at Chiba University Hospital from January 2008 to December 2020 were enrolled in the current study. The prognostic impact of the ypStage on the overall survival (OS) and relapse-free survival (RFS) were examined via univariate and multivariate analyses. RESULTS The 5-year OS rates for ypStage I, II, III, and IV were 87.5%, 64.7%, 52.9%, and 28.6%, respectively, while the 5-year RFS rates were 81.3%, 57.4%, 44.4%, and 28.6%, respectively. The univariate analysis revealed that the ypStage was significantly correlated with the OS (p = 0.037) and the ypT status and ypStage showed a significant correlation with the RFS (p = 0.043 and p = 0.021, respectively). The multivariate analysis demonstrated that only the ypStage was an independent prognostic factor for the OS and RFS (p = 0.024 and p = 0.018, respectively). CONCLUSION The ypTNM stage may be a useful tool for the risk stratification of all advanced GC patients treated with chemotherapy followed by gastrectomy, including not only neoadjuvant but also conversion surgery cases.
Collapse
Affiliation(s)
- Ryota Otsuka
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan.
| | - Koichi Hayano
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Hideki Hayashi
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Masaya Uesato
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Kentaro Murakami
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Takeshi Toyozumi
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Yasunori Matsumoto
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Yoshihiro Kurata
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Akira Nakano
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| |
Collapse
|
11
|
Tian Y, Pang Y, Yang P, Guo H, Liu Y, Zhang Z, Ding P, Zheng T, Li Y, Fan L, Zhang Z, Zhao X, Tan B, Wang D, Zhao Q. The safety and efficacy of carbon nanoparticle suspension injection versus indocyanine green tracer-guided lymph node dissection during radical gastrectomy (FUTURE-01): A single-center randomized controlled trial protocol. Front Oncol 2023; 12:1044854. [PMID: 36686792 PMCID: PMC9852878 DOI: 10.3389/fonc.2022.1044854] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Background The use of lymph node (LN) tracers can help obtain a complete dissection of the lymph nodes and increase the detection rate of LNs and metastatic LNs. Carbon nanoparticle suspension injection (CNSI) and indocyanine green (ICG) have been widely used in radical gastrectomy in recent years. Nevertheless, the comparison of their clinical effects has not been studied. Method/design The FUTURE-01 trial will be the first randomized, open-label, single-center trial to compare CNSI and ICG. The study started in 2021 and enrolled 96 patients according to a prior sample size calculation. The primary outcome is the number of LNs retrieved. The secondary outcomes are LN staining rate, LN metastasis rate, stained LN metastasis rate, perioperative recovery and survival. Conclusion By comparing the safety and efficacy of CNSI and ICG tracer-guided LN dissection in patients with gastric cancer, we can determine the most appropriate LN tracer at present. With the help of LN tracers, the operation is simplified, and the prognosis of these patients is improved. Our study is a prospective exploration of the safety, efficacy, and prognosis of CNSI and ICG. Clinical trial registration https://clinicaltrials.gov/ct2/show/NCT05229874?cond=NCT05229874&draw=2&rank=1, identifier NCT05229874.
Collapse
|
12
|
Bu Z, Jiang Y, Luo S, He X, Qin H, Tang W. Weight Loss During Neoadjuvant Therapy Is Associated With Poor Response Among the Patients With Gastrointestinal Cancer: A Propensity Score Matching Analysis. Cancer Control 2023; 30:10732748231164016. [PMID: 37071968 PMCID: PMC10126799 DOI: 10.1177/10732748231164016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 04/20/2023] Open
Abstract
PURPOSE The aim of the current study was to identify the relationship between body composition changes during neoadjuvant therapy (NAT) and the treatment efficiency of NAT in gastrointestinal cancer (GC) patients. METHODS From January 2015 to July 2020, 277 GC patients treated with NAT had included for retrospective analysis. The body mass index (BMI) and computed tomography (CT) imaging before and after NAT were recorded. The BMI change optimal cut-off value were calculated by ROC curve. Balancing essential characteristic variables using propensity score matching (PSM) method. Exploring the association between BMI changes and tumor response to NAT using logistic regression analysis. The survival outcome of matched patients between different BMI change groups was compared. RESULTS A cutoff point of BMI change >2% during NAT was defined as BMI loss. Among the 277 patients, 110 (39.7%) patients showed BMI change with a loss after NAT. In total, 71 pairs of patients were selected for further analysis. The median follow-up time was 22 months (range 3 to 63 months). Univariate and multivariate logistic regression analyses in matched cohort showed that BMI change was a prognostic factor for tumor response after NAT in GC patients (odds ratio (OR), .471; 95% confidence interval (CI), .233-.953; P = .036). In addition, patients who experienced BMI loss after NAT showed worse overall survival than those who had BMI gain or stable. CONCLUSION BMI loss during NAT probably may has negative effects on NAT efficiency and survival for gastrointestinal cancer patients. It is necessary to monitor and maintain weight for patients during treatment.
Collapse
Affiliation(s)
- Zhaoting Bu
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, The People’s Republic of China
| | - Yuting Jiang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, The People’s Republic of China
| | - Shanshan Luo
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, The People’s Republic of China
| | - Xinxin He
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, The People’s Republic of China
| | - Haiquan Qin
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, The People’s Republic of China
| | - Weizhong Tang
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People’s Republic of China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, The People’s Republic of China
| |
Collapse
|
13
|
Li Z, Li X, Zhu X, Ai S, Guan W, Liu S. Tracers in Gastric Cancer Surgery. Cancers (Basel) 2022; 14:cancers14235735. [PMID: 36497216 PMCID: PMC9741333 DOI: 10.3390/cancers14235735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The treatment of gastric cancer mainly depends on radical gastrectomy. Determination of appropriate surgical margins and adequate lymph node (LN) resection are two major surgical steps that directly correlate with prognosis in gastric cancer. Due to the expanding use of minimally invasive procedures, it is no longer possible to locate tumors and LNs through touch. As an alternative, tracers have begun to enter the field due to their capacities for intraoperative visualization. Herein, we summarize the application of contemporary tracers in gastric cancer surgery, including isosulfan blue, methylene blue, patent blue, indocyanine green, carbon particles, and radioactive tracers. Their mechanisms, administration methods, detection efficiency, and challenges, as well as perspectives on them, are also outlined.
Collapse
Affiliation(s)
| | | | | | | | - Wenxian Guan
- Correspondence: (W.G.); (S.L.); Tel.: +86-25-68182222-60931 (W.G.); +86-25-68182222-60930 (S.L.)
| | - Song Liu
- Correspondence: (W.G.); (S.L.); Tel.: +86-25-68182222-60931 (W.G.); +86-25-68182222-60930 (S.L.)
| |
Collapse
|
14
|
Kang H, Kim SS, Sung MJ, Jo JH, Lee HS, Chung MJ, Park JY, Park SW, Song SY, Park MS, Bang S. Evaluation of the 8th Edition AJCC Staging System for the Clinical Staging of Pancreatic Cancer. Cancers (Basel) 2022; 14:4672. [PMID: 36230595 PMCID: PMC9563770 DOI: 10.3390/cancers14194672] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
The 8th edition of the American Joint Committee on Cancer (AJCC) staging system for pancreatic cancer (PC) has been validated for pathological staging; however, its significance for clinical staging remains uncertain. We validated the prognostic performance and suitability of the current staging system for the clinical staging of PC. We identified 1043 patients from our PC registry who were staged by imaging according to the 8th edition staging system and conducted analysis, including overall survival (OS) comparison. Gradual prognostic stratification according to stage hierarchy yielded significant OS differences between stage groups, except between stage I and II (p = 0.193). A substage comparison revealed no survival differences between IB (T2N0) and IIA (T3N0), which were divided by the T3 criterion only (p = 0.278). A higher N stage had significantly shorter OS than a lower N stage (all pairwise p < 0.05). However, among the 150 patients who received upfront surgery, the pathological stage was more advanced than the clinical stage in 86 (57.3%), mostly due to a false-negative cN0 (70.9%). Our results suggest that the new definition of T3 and the number-based N criteria in the 8th edition AJCC staging system may be not adequate for clinical staging. Establishing separate criteria more suitable for clinical staging should be considered.
Collapse
Affiliation(s)
- Huapyong Kang
- Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon 21565, Korea
- Department of Medicine, Yonsei University Graduate School, Seoul 03722, Korea
| | - Seung-seob Kim
- Department of Radiology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Min Je Sung
- Digestive Disease Center, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Korea
| | - Jung Hyun Jo
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hee Seung Lee
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Seung Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Mi-Suk Park
- Department of Radiology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
15
|
Neoadjuvant PD-1 inhibitor and apatinib combined with S-1 plus oxaliplatin for locally advanced gastric cancer patients: a multicentered, prospective, cohort study. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04302-9. [PMID: 36042044 DOI: 10.1007/s00432-022-04302-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/15/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE Programmed cell death protein 1 (PD-1) inhibitor and apatinib have been utilized in metastatic gastric cancer patients. The current study aimed to further investigate the efficacy and safety of neoadjuvant S-1 plus oxaliplatin combined with PD-1 inhibitor and apatinib (SOXPA) in locally advanced gastric cancer (LAGC) patients. METHODS This two-centered, prospective, cohort study analyzed 30 resectable LAGC patients receiving SOXPA as neoadjuvant therapy. RESULTS Two (6.7%), 18 (60.0%), and 10 (33.3%) patients achieved complete response (CR), partial response (PR), and stable disease (SD), separately. The objective response rate (ORR) and disease control rate (DCR) were 66.7% and 100.0%, respectively. The R0 resection rate was 93.3%. Beyond that, 6 (20.0%), 18 (60.0%), and 6 (20.0%) patients achieved grade 1, 2, and 3 pathological responses. The pathological complete response (pCR) rate was 20%. The 1-year and 2-year disease-free survival (DFS) rates were 96.6% and 77.7% respectively; meanwhile, the 1-year and 2-year overall survival (OS) rates were 96.6% and 90.1%, separately. What's more, better clinical response (P = 0.046); achievement of ORR (P = 0.014), and better pathological response (P = 0.020) were correlated with longer DFS. Besides, ORR achievement was linked with longer OS (P = 0.040). Most adverse events were relatively mild and manageable. Grade 3 adverse events included leukopenia, anemia, neutropenia, fatigue, hand-foot syndrome, nausea and vomiting. No grade 4 adverse events were witnessed. CONCLUSION SOXPA as neoadjuvant therapy achieves a satisfying clinical response, pathological response, survival profile, and tolerable safety in LAGC patients.
Collapse
|
16
|
Mranda GM, Xue Y, Zhou XG, Yu W, Wei T, Xiang ZP, Liu JJ, Ding YL. Revisiting the 8th AJCC system for gastric cancer: A review on validations, nomograms, lymph nodes impact, and proposed modifications. Ann Med Surg (Lond) 2022; 75:103411. [PMID: 35386808 PMCID: PMC8977912 DOI: 10.1016/j.amsu.2022.103411] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/23/2022] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer is the fifth most frequently diagnosed cancer worldwide, behind breast, lung, colorectal, and prostate cancers. In gastric cancer, multimodality treatment shows prospective benefits and also improves survival. Surgery, however, is the mainstay of curative treatment. The staging of gastric cancer patients is critical for harmonization of care. Accurate stages assure that informed clinical decisions are timely made. The American Joint Committee on Cancer (AJCC) staging system is the most widely applied system in to determine the disease's prognosis and survival prediction. The recently adopted 8th AJCC TNM staging system has been revised to enhance its survival predictive power. Subsequent studies have established the validity of the current edition, demonstrating improved stage stratification, discriminatory power, and survival prediction. However, other studies have cast doubt on the superiority of the new edition. Innovations aimed at further improving its prognosis have resulted in developing of novel models. Advances in our understanding of the tumor microenvironment and molecular categorization of cancer have resulted in proposals for their inclusion in TNM staging as potential complementary factors that enhance survival prediction and prognostic assessment ability. The purpose of this study is to conduct a review of the published literature regarding the validity of the 8th AJCC TNM staging system, proposed modifications, and nomograms. The 8th AJCC is valid in prognostic stratification of gastric cancer, however, revisions are still required. The yPT staging requires some modifications and inclusion of stages that currently don't exist in the 8th AJCC. High lymph nodes count and anatomical localization improve the prediction ability of the current AJCC. Nomograms comprising of individual prognostic factors are crucial to the current AJCC. Molecular markers positively influence survival prediction of gastric cancer.
Collapse
Affiliation(s)
- Geofrey Mahiki Mranda
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
- Department of General Surgery, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Ying Xue
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Xing-Guo Zhou
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Wang Yu
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Tian Wei
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Zhi-Ping Xiang
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Jun-Jian Liu
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
| | - Yin-Lu Ding
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China
- Corresponding author. Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247 Beiyuan Street, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
17
|
Zhao K, Shan BQ, Gao YP, Xu JY. Role of carbon nanotracers in lymph node dissection of advanced gastric cancer and the selection of preoperative labeling time. World J Clin Cases 2022; 10:870-881. [PMID: 35127902 PMCID: PMC8790434 DOI: 10.12998/wjcc.v10.i3.870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/19/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The incidence of gastric cancer is high. The number of dissected lymph nodes was an independent factor affecting prognosis. Although preoperative labeling is helpful in lymph nodes resection, there are no guidelines for when to perform preoperative labeling.
AIM To investigate the role of nanocarbon in lymph node dissection during gastrectomy, and to discuss the relationship between the timing of preoperative injection of carbon nanoparticles and the extent of lymph node dissection.
METHODS A prospective analysis was performed on the clinical data of 307 patients with advanced gastric cancer who underwent laparoscopic surgery in the General Surgery Department of Weifang People’s Hospital between June 2018 and February 2021. The patients were randomly divided into experimental group and control group based on whether they received preoperative nanocarbon injection or not. The experimental group was divided into different groups according to the preoperative labeling time. The number of dissected lymph nodes and the number of lymph nodes with black staining were compared in each group after surgery, and the role of nanocarbon in the number of dissected lymph nodes, pathological staging, and the relationship with prognosis were discussed.
RESULTS The average number of dissected lymph nodes in the experimental group was higher than that in the control group. In the experimental group, the number of lymph node dissections and number of black-staining lymph nodes in the nanocarbon-labeling group at 2 d and 1 d before surgery were higher than in the labeling group on the day before surgery (P < 0.05).
CONCLUSION Preoperative nanocarbon labeling can safely and effectively guide lymph node dissection. To improve the detection rate of lymph nodes is conducive to subsequent comprehensive anti-tumor therapy.
Collapse
Affiliation(s)
- Kai Zhao
- Department of General Surgery, WFPH, Weifang 261400, Shandong Province, China
| | - Bao-Qiang Shan
- Department of General Surgery, WFPH, Weifang 261400, Shandong Province, China
| | - Yan-Peng Gao
- Department of General Surgery, WFPH, Weifang 261400, Shandong Province, China
| | - Jia-You Xu
- Department of General Surgery, WFPH, Weifang 261400, Shandong Province, China
| |
Collapse
|
18
|
Huang WP, Li LM, Li J, Yuan JH, Hou P, Liu CC, Ma YH, Liu XN, Han YJ, Liang P, Gao JB. Computed Tomography Features and Clinical Prognostic Characteristics of Hepatoid Adenocarcinoma of the Stomach. Front Oncol 2021; 11:772636. [PMID: 34956891 PMCID: PMC8696206 DOI: 10.3389/fonc.2021.772636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Hepatoid adenocarcinoma of the stomach (HAS) is a highly malignant and aggressive tumor. The purpose of this study was to describe the clinical, computed tomography (CT), and prognostic features of HAS to increase the awareness of this entity and determine its distinguishing features from non-HAS tumors. METHODS The CT features and clinical data of 47 patients in our hospital with pathologically documented HAS were retrospectively analyzed, and the relevant differences between pure HAS (pHAS) and mixed HAS (mHAS) were determined. In addition, 141 patients with non-HAS tumors in the same T stage in the same period were selected as the control group. The data were compared between the two groups, and factors affecting the prognosis of HAS were analyzed. In addition, we included 9 patients with HAS and 27 patients with non-HAS tumors from another center for external validation. RESULTS The patients in the HAS group were predominantly men (n = 33), and the tumor location was mostly the cardia or fundus (n = 27). Between the HAS and non-HAS groups, there were observed differences in terms of: sex, serum alpha-fetoprotein (AFP), carbohydrate antigen (CA)-125, and CA-724 levels; longest tumor diameter; degree of differentiation; vascular invasion; N stage, M stage, and tumor-node-metastasis (TNM) stage; thickest tumor diameter; plain CT attenuation; arterial-phase CT attenuation; CT attenuation between the venous and arterial phases; enhancement modes; and degrees of enhancement (all P < 0.05). In the data from another center for external validation, there were observed differences in terms of: age, degree of differentiation, vascular invasion, thickest tumor diameter, the ratio of arterial CT attenuation to CT attenuation of the abdominal aorta at the same level (RA), CT attenuation difference between the venous phase and arterial phase (HUv-a) (all P < 0.05). The results of the multivariate analysis revealed that the independent factors for differentiation were serum AFP level (P = 0.001), M stage (P = 0.038), and tumor enhancement on CT (P = 0.014). Among patients in the HAS group, 72.34% had pHAS and 27.66% had mHAS. The thickest tumor diameter and the longest short diameter of the metastatic lymph nodes of the mHAS group were on average 6.39 cm and 1.45 cm, respectively, which were larger than those in the pHAS group. The median progression-free survival time was 18.25 months in the HAS group, which was shorter than that in the non-HAS group (72.96 months; P = 0.001). The median overall survival time in the HAS group was 24.80 months, which was shorter than that in the non-HAS group (67.96 months; P = 0.001). The factors affecting the prognosis of HAS were M stage (P = 0.001), overall TNM stage (P = 0.048), presence of vascular cancer emboli (P = 0.040), and pHAS type (P = 0.046). Multifactorial analysis revealed that M stage (P = 0.027) and pHAS type (P = 0.009) were independent risk factors affecting the prognosis of HAS. CONCLUSION Although HAS is a rare clinical entity, it should be considered in the differential diagnosis of gastric tumors. Patients with HAS often have advanced-stage disease at presentation and a worse prognosis than patients with non-HAS tumors. CT findings, combined with laboratory results, can support the diagnosis of HAS. However, the final diagnosis needs to be confirmed with a histopathologic examination. If the postoperative pathologic findings reveal the mHAS type, a rapid clinical intervention and a detailed follow-up with CT are essential.
Collapse
Affiliation(s)
- Wen-peng Huang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li-ming Li
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Li
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Jun-hui Yuan
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Ping Hou
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen-chen Liu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi-hui Ma
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-nan Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi-jing Han
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pan Liang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian-bo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Lymph node ratio-based the ypTNrM staging system for gastric cancer after neoadjuvant therapy: a large population-based study. Surg Today 2021; 52:783-794. [PMID: 34724107 DOI: 10.1007/s00595-021-02386-3] [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] [Received: 04/04/2021] [Accepted: 08/21/2021] [Indexed: 12/24/2022]
Abstract
PURPOSES The lymph node ratio (LNR) has been considered a better prognostic factor than traditional N staging in patients with gastric cancer (GC), but its accuracy is unclear for those who receive neoadjuvant therapy (NAT). We aimed to compare the node ratio (Nr) staging with the ypN staging and to thereby develop a modified staging system incorporating Nr staging. METHODS A total of 1791 patients who underwent gastrectomy after NAT in the Surveillance, Epidemiology, and End Results database were retrospectively analyzed. ypTNrM staging was established based on the overall survival (OS). RESULTS The Nr staging was generated using 0.2 and 0.5 as the cutoff values of LNR and represented patients with more homogeneous OS compared with ypN staging. The 5-year OS rates for ypTNrM stages IA, IB, II, IIIA, and IIIB were 70.2%, 54.2%, 36.0%, 21.2%, and 6.6%, respectively, compared with 58.8%, 39.1%, and 21.6% for ypTNM stages I, II, and III, respectively. Compared with the ypTNM staging system, the ypTNrM staging system had a lower misclassification rate (3.0% vs. 50.9%) and better prognostic predictive power (C-index: 0.645 vs. 0.589, P < 0.001). CONCLUSIONS The ypTNrM staging system incorporating Nr staging may provide a more accurate assessment in the clinical decision-making process for GC after NAT.
Collapse
|
20
|
Bando E, Ji X, Kattan MW, Bencivenga M, de Manzoni G, Terashima M. Development and validation of pretreatment nomogram for disease-specific mortality in gastric cancer-A competing risk analysis. Cancer Med 2021; 10:7561-7571. [PMID: 34628732 PMCID: PMC8559461 DOI: 10.1002/cam4.4279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022] Open
Abstract
Background In several reports, gastric cancer nomograms for predicting overall or disease‐specific survival have been described. The American Joint Committee on Cancer (AJCC) introduced the attractiveness of disease‐specific mortality (DSM) as an endpoint of risk model. This study aimed to develop the first pretreatment gastric cancer nomogram for predicting DSM that considers competing risks (CRs). Methods The prediction model was developed using data for 5231 gastric cancer patients. Fifteen prognosticators, which were registered at diagnosis, were evaluated. The nomogram for DSM was created as visualizations of the multivariable Fine and Gray regression model. An independent cohort for external validation consisted of 389 gastric cancer patients from a different institution. The performance of the model was assessed by discrimination (Harrell's concordance (C)‐index), calibration, and decision curve analysis. DSM and CRs were evaluated, paying special attention to host‐related factors such as age and Eastern Cooperative Oncology Group performance status (ECOG PS), by using Gray's univariable method. Results Fourteen prognostic factors were selected to develop the nomogram. The new nomogram for DSM exhibited good discrimination. Its C‐index of 0.887 surpassed that of the American Joint Committee on Cancer (AJCC) clinical staging (0.794). The C‐index was 0.713 (AJCC, 0.582) for the external validation cohort. The nomogram showed good performance internally and externally, in the calibration and decision curve analysis. Host‐related factors including age and ECOG PS, were strongly correlated with competing risks. Conclusions The newly developed nomogram accurately predicts DSM, which can be used for patient counseling in clinical practice.
Collapse
Affiliation(s)
- Etsuro Bando
- Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Xinge Ji
- Department of Quantitative Health Sciences, The Cleveland Clinic, Cleveland, OH, USA
| | - Michael W Kattan
- Department of Quantitative Health Sciences, The Cleveland Clinic, Cleveland, OH, USA
| | - Maria Bencivenga
- Division of General and Upper Gastrointestinal Surgery, Department of Surgery, University of Verona, Verona, Italy
| | - Giovanni de Manzoni
- Division of General and Upper Gastrointestinal Surgery, Department of Surgery, University of Verona, Verona, Italy
| | | |
Collapse
|
21
|
Kumagai K, Sano T. Revised points and disputed matters in the eighth edition of the TNM staging system for gastric cancer. Jpn J Clin Oncol 2021; 51:1024-1027. [PMID: 33987661 DOI: 10.1093/jjco/hyab069] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/26/2021] [Indexed: 12/30/2022] Open
Abstract
The American Joint Committee on Cancer and the Union for International Cancer Control have now released the eighth edition of the cancer staging system, which incorporates some major revisions concerning gastric cancer. First, grouping of pathological stages has been revised in accordance with a proposal from the International Gastric Cancer Association. That revision was based on analysis of survival of more than 25 000 patients worldwide who had undergone curative gastrectomy without neoadjuvant therapy. Stratification of survival in Stage III subgroups has been improved by subdividing N3 into N3a and N3b. Second, a simplified grouping of clinical stages that differs completely from grouping of pathological stages has been proposed. Pre-treatment depth of tumour invasion is now categorized as T1/T2, T3/T4a or T4b, and lymph node status is simply categorized as N0 or N+. Additionally, a 'yp-stage' for specimens resected after neoadjuvant therapy has been newly proposed. These clinical and post-neoadjuvant stages were considered useful now that neoadjuvant chemotherapy is increasingly being administered to patients with potentially resectable gastric cancer. Third, staging of oesophagogastric junction tumours has been modified; Siewert type 3 tumours are now classified as gastric tumours and are staged according to the gastric, rather than the oesophageal, system. More appropriate staging of gastric cancers treated with neoadjuvant therapy, and/or new staging systems incorporating variables other than T/N/M, such as biological or genomic markers, are likely future developments.
Collapse
Affiliation(s)
- Koshi Kumagai
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Sano
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
22
|
Wang Y, Lei X, Liu Z, Shan F, Ying X, Li Z, Ji J. Short-term outcomes of laparoscopic versus open total gastrectomy after neoadjuvant chemotherapy: a cohort study using the propensity score matching method. J Gastrointest Oncol 2021; 12:237-248. [PMID: 34012622 DOI: 10.21037/jgo-20-374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Until now, little is known about the benefit of laparoscopic total gastrectomy (LTG) after neoadjuvant chemotherapy (NACT). This study was designed to compare the safety and efficacy of the LTG versus the open total gastrectomy (OTG) approach after NACT treatment in patients with advanced gastric cancer (AGC). Methods This study involved a cohort of 145 patients with AGC who underwent total gastrectomy after NACT at our centre between April 2013 and August 2018 including 24 cases of LTG and 121 OTG. The baseline characteristics were matched based on 1:2 balanced propensity score-matching method. Results Sixty-nine marched cases were finally analysed (23 LTG vs. 46 OTG). All patients underwent R0 resection. Compared to the OTG group, the LTG group had a longer surgery duration (P<0.001), but a shorter incision length (P<0.001) and less intravenous patient-controlled analgesia (IV-PCA) time after surgery (P=0.027). No statistical differences were observed in terms of blood loss, retrieved lymph nodes (LNs), resection margin, length of stay, postoperative pain intensity, and complications (P>0.05). Conclusions LTG had comparable safety and histological findings to OTG after NACT in the perioperative period; however, LTG is less invasive and patients can benefit from less IV-PCA use. Further research is needed to investigate long-term effects. Keywords Gastric cancer (GC); neoadjuvant chemotherapy (NACT); safety; laparoscopic technique.
Collapse
Affiliation(s)
- Yinkui Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaokang Lei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zining Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Fei Shan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangji Ying
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ziyu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
23
|
Lombardi PM, Mazzola M, Achilli P, Aquilano MC, De Martini P, Curaba A, Gualtierotti M, Bertoglio CL, Magistro C, Ferrari G. Prognostic value of pathological tumor regression grade in locally advanced gastric cancer: New perspectives from a single-center experience. J Surg Oncol 2021; 123:923-931. [PMID: 33497471 DOI: 10.1002/jso.26391] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Perioperative chemotherapy (PC) with radical surgery represents the gold standard of treatment for resectable advanced gastric cancer (GC). The prognostic value of pathological tumor regression grade (TRG) induced by neoadjuvant chemotherapy (NACT) is not clearly established. This study aimed to investigate the correlation between TRG and survival in GC. METHODS Patients affected by advanced GC undergoing PC and radical surgery were considered. TRG was assessed for each patient according to Becker's grading system. The correlation between TRG and survival was investigated. RESULTS One-hundred patients were selected; 25 showed a good response (GR) (TRG 1a/1b), while 75 had a poor response (PR) (TRG 2/3) to NACT. GR patients showed better disease-free survival (DFS) (52 vs. 19 months, p < .001) and disease-specific survival (DSS) (57 vs. 25 months, p < .0001) when compared to PR patients. On univariate analysis, TRG, lymph node ratio (LNR), tumor size, grading, and post-neoadjuvant therapy TNM stage were significantly correlated with survival. On multivariate analysis, TRG, LNR and tumor size were independent prognostic factors for DFS and DSS. CONCLUSIONS TRG, LNR, and tumor size are independent prognostic factors for DFS and DSS in patients with advanced GC undergoing NACT.
Collapse
Affiliation(s)
- Pietro Maria Lombardi
- Division of Minimally-invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| | - Michele Mazzola
- Division of Minimally-invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| | - Pietro Achilli
- Division of Minimally-invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| | - Maria Costanza Aquilano
- Department of Oncology and Hemat-Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| | - Paolo De Martini
- Division of Minimally-invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| | - Annabella Curaba
- Department of Pathology and Cytogenetics, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| | - Monica Gualtierotti
- Division of Minimally-invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| | - Camillo L Bertoglio
- Division of Minimally-invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| | - Carmelo Magistro
- Division of Minimally-invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| | - Giovanni Ferrari
- Division of Minimally-invasive Surgical Oncology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, Milan, Italy
| |
Collapse
|
24
|
Kim G, Friedmann P, Solsky I, Muscarella P, McAuliffe J, In H. Providing Reliable Prognosis to Patients with Gastric Cancer in the Era of Neoadjuvant Therapies: Comparison of AJCC Staging Schemata. J Gastric Cancer 2020; 20:385-394. [PMID: 33425440 PMCID: PMC7781744 DOI: 10.5230/jgc.2020.20.e41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/23/2020] [Indexed: 11/28/2022] Open
Abstract
Purpose Patients with gastric cancer who receive neoadjuvant therapy are staged before treatment (cStage) and after treatment (ypStage). We aimed to compare the prognostic reliability of cStage and ypStage, alone and in combination. Materials and Methods Data for all patients who received neoadjuvant therapy followed by surgery for gastric adenocarcinoma from 2004 to 2015 were extracted from the National Cancer Database. Kaplan-Meier (KM)curves were used to model overall survival based on cStage alone, ypStage alone, cStage stratified by ypStage, and ypStage stratified by cStage. P-values were generated to summarize the differences in KM curves. The discriminatory power of survival prediction was examined using Harrell's C-statistics. Results We included 8,977 patients in the analysis. As expected, increasing cStage and ypStage were associated with worse survival. The discriminatory prognostic power provided by cStage was poor (C-statistic 0.548), while that provided by ypStage was moderate (C-statistic 0.634). Within each cStage, the addition of ypStage information significantly altered the prognosis (P<0.0001 within cStages I–IV). However, for each ypStage, the addition of cStage information generally did not alter the prognosis (P=0.2874, 0.027, 0.061, 0.049, and 0.007 within ypStages 0–IV, respectively). The discriminatory prognostic power provided by the combination of cStage and ypStage was similar to that of ypStage alone (C-statistic 0.636 vs. 0.634). Conclusions The cStage is unreliable for prognosis, and ypStage is moderately reliable. Combining cStage and ypStage does not improve the discriminatory prognostic power provided by ypStage alone. A ypStage-based prognosis is minimally affected by the initial cStage.
Collapse
Affiliation(s)
- Gina Kim
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Patricia Friedmann
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Cardiothoracic and Vascular Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ian Solsky
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Peter Muscarella
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - John McAuliffe
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Haejin In
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
25
|
Zhong Q, Chen QY, Parisi A, Ma YB, Lin GT, Desiderio J, Yan S, Xie JW, Wang JB, Hou JF, Lin JX, Lu J, Cao LL, Lin M, Tu RH, Huang ZN, Lin JL, Liu ZY, Que SJ, Li P, Zheng CH, Huang CM. Modified ypTNM Staging Classification for Gastric Cancer after Neoadjuvant Therapy: A Multi-Institutional Study. Oncologist 2020; 26:e99-e110. [PMID: 32864840 DOI: 10.1634/theoncologist.2020-0022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 07/23/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The benefits of neoadjuvant therapy for patients with locally advanced gastric cancer (GC) are increasingly recognized. The 8th edition of the American Joint Committee on Cancer (AJCC) Staging Manual first proposed ypTNM staging, but its accuracy is controversial. This study aims to develop a modified ypTNM staging. PATIENTS AND METHODS Clinicopathological data of 1,791 patients who underwent curative-intent gastrectomy after neoadjuvant therapy in the Surveillance, Epidemiology, and End Results database, as the development cohort, were retrospectively analyzed. Modified ypTNM staging was established based on overall survival (OS). We compared the prognostic performance of the AJCC 8th edition ypTNM staging and the modified staging for patients after neoadjuvant therapy. RESULTS In the development cohort, the 5-year OS for AJCC stages I, II, and III was 58.8%, 39.1%, and 21.6%, respectively, compared with 69.9%, 54.4%, 34.4%, 24.1%, and 13.6% for modified ypTNM stages IA, IB, II, IIIA, and IIIB. The modified staging had better discriminatory ability (C-index: 0.620 vs. 0.589, p < .001), predictive homogeneity (likelihood ratio chi-square: 140.71 vs. 218.66, p < .001), predictive accuracy (mean difference in Bayesian information criterion: 64.94; net reclassification index: 35.54%; integrated discrimination improvement index: 0.032; all p < .001), and model stability (time-dependent receiver operating characteristics curves) over AJCC. Decision curve analysis showed that the modified staging achieved a better net benefit than AJCC. In external validation (n = 266), the modified ypTNM staging had superior prognostic predictive power (all p < .05). CONCLUSION We have developed and validated a modified ypTNM staging through multicenter data that is superior to the AJCC 8th edition ypTNM staging, allowing more accurate assessment of the prognosis of patients with GC after neoadjuvant therapy. IMPLICATIONS FOR PRACTICE The 8th edition of the American Joint Committee on Cancer (AJCC) Staging Manual first proposed ypTNM staging, but its accuracy is controversial. Based on multi-institutional data, this study developed a modified ypTNM staging, which is superior to the AJCC 8th edition ypTNM staging, allowing more accurate assessment of the prognosis of patients with gastric cancer after neoadjuvant therapy.
Collapse
Affiliation(s)
- Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Amilcare Parisi
- Department of Digestive Surgery, St. Mary's Hospital, University of Perugia, Terni, Italy
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, People's Republic of China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jacopo Desiderio
- Department of Digestive Surgery, St. Mary's Hospital, University of Perugia, Terni, Italy
| | - Su Yan
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, People's Republic of China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jun-Fang Hou
- Department of Gastrointestinal Surgery, Qinghai University Affiliated Hospital, Xining, People's Republic of China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ju-Li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Zhi-Yu Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Si-Jin Que
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, People's Republic of China
| |
Collapse
|
26
|
Bando E, Ji X, Kattan MW, Seo HS, Song KY, Park CH, Bencivenga M, de Manzoni G, Terashima M. Development and validation of a pretreatment nomogram to predict overall survival in gastric cancer. Cancer Med 2020; 9:5708-5718. [PMID: 32588982 PMCID: PMC7433838 DOI: 10.1002/cam4.3225] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/15/2020] [Accepted: 05/26/2020] [Indexed: 12/23/2022] Open
Abstract
Background Pretreatment clinical staging is essential to select therapy. However, there have been no published pretreatment gastric cancer nomograms constructed using pretreatment clinical prognostic factors, including in nonresection patients. We aimed to develop a new pretreatment gastric cancer nomogram for individualized prediction of overall survival (OS). Methods The nomogram was developed using data of 5231 Japanese gastric cancer patients, and it was created with a Cox regression model. Fifteen clinical variables, which were obtained at pretreatment, were collected and registered. Data of two independent cohorts of patients from Seoul St. Mary's Hospital (1001 patients), and the University of Verona (389 patients) formed the external validation cohorts. The model was validated internally and externally using measures of discrimination (Harrell's C‐index), calibration, and decision curve analysis. Results The developed nomogram showed good discrimination, with a C‐index of 0.855; that of the American Joint Committee on Cancer (AJCC) clinical stage was 0.819. In the external validation procedure, the C‐indexes were 0.856 (AJCC, 0.795) in the Seoul St. Mary's cohort and 0.714 (AJCC, 0.648) in the University of Verona cohort. The nomogram performed well in the calibration and decision curve analyses when applied to both the internal and external validation cohorts. A stage‐specific subset survival analysis of the three risk groups calculated using the nomogram also showed the superiority of nomogram‐prediction when compared to AJCC. Conclusion This new pretreatment model accurately predicts OS in gastric cancer and can be used for patient counseling in clinical practice and stratification in clinical trials.
Collapse
Affiliation(s)
- Etsuro Bando
- Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan.,Department of Quantitative Health Sciences, The Cleveland Clinic, Cleveland, OH, USA
| | - Xinge Ji
- Department of Quantitative Health Sciences, The Cleveland Clinic, Cleveland, OH, USA
| | - Michael W Kattan
- Department of Quantitative Health Sciences, The Cleveland Clinic, Cleveland, OH, USA
| | - Ho Seok Seo
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyo Young Song
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Cho-Hyun Park
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Maria Bencivenga
- Division of General and Upper Gastrointestinal Surgery, Department of Surgery, University of Verona, Verona, Italy
| | - Giovanni de Manzoni
- Division of General and Upper Gastrointestinal Surgery, Department of Surgery, University of Verona, Verona, Italy
| | | |
Collapse
|
27
|
Abstract
Gastric cancer is the fifth most common malignancies and the third leading cause of cancer-related death worldwide, with more than 40% of new cases occurring in China. With the advancement of treatment methods, the application of adjuvant therapy and targeted drugs, the prognosis of patients with gastric cancer has been significantly improved. In recent years, more and more studies have reported that magnetic resonance imaging (MRI) showed great value in the clinical application among patients with gastric cancer, including preoperative staging, treatment response evaluation, predicting prognosis and histopathological features, treatment guidance, and molecular imaging. The remarkable research progress of MRI in gastric cancer will provide new evaluation and treatment approaches for clinical diagnosis and treatment. This article aims to review the current status of the application and research progress of MRI in patients with gastric cancer.
Collapse
Affiliation(s)
- Yingjing Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jianchun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
28
|
Nostedt J, Gibson-Brokop L, Ghosh S, Seidler M, McCall M, Schiller D. Evaluating the utility of computed tomography of the chest for gastric cancer staging. Can J Surg 2020; 63:E57-E61. [PMID: 32031765 DOI: 10.1503/cjs.000319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background International guidelines recommend routine computed tomography (CT) of the chest for gastric cancer staging. In Asian countries, where the incidence of pulmonary metastases is less than 1%, some guidelines recommend chest CT only for gastroesophageal junction cancers. If the incidence of pulmonary metastases is also low in Canada, routine chest CT may not be beneficial. Methods We performed a retrospective review of patients in northern Alberta with newly diagnosed gastric cancer from January 2010 to July 2016. The primary aim of the study was to determine the incidence of pulmonary metastases at the time of diagnosis in this population. A secondary aim was to identify potential predictors of pulmonary metastases. We reviewed CT reports for pulmonary metastases. Imaging data also included liver metastases, abdominal lymphadenopathy (> 1 cm), ascites and omental or peritoneal nodules. Other data recorded were age, sex, primary tumour location, histologic type and tumour grade. Results Four hundred and sixty-two patients (311 men, 151 women) were included in the analysis. Pulmonary metastases were identified in 25 patients (5.4%) overall and in 11 of 299 patients (3.7%) whose primary cancer was not in the cardia. On univariate analysis the presence of liver metastases (odds ratio [OR] 7.72, 95% confidence interval [CI] 3.24–18.37, p < 0.001) and abdominal lymphadenopathy (OR 3.30, 95% CI 1.29–8.48, p = 0.01) was associated with an increased risk of pulmonary metastases. Liver metastases retained statistical significance on multivariate analysis (OR 6.17, 95% CI 2.53–15.03, p < 0.001). Conclusion The incidence of pulmonary metastases at the time of gastric cancer diagnosis is higher in northern Alberta than previously reported in Asian studies. Abdominal lymphadenopathy and liver metastases confer an elevated risk of pulmonary metastases.
Collapse
Affiliation(s)
- Jordan Nostedt
- From the Department of Surgery, University of Alberta, Edmonton, Alta. (Nostedt, McCall, Schiller); the Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alta. (Gibson-Brokop); the Department of Medical Oncology and the Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Alta. (Ghosh); and the Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alta. (Seidler)
| | - Lindsay Gibson-Brokop
- From the Department of Surgery, University of Alberta, Edmonton, Alta. (Nostedt, McCall, Schiller); the Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alta. (Gibson-Brokop); the Department of Medical Oncology and the Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Alta. (Ghosh); and the Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alta. (Seidler)
| | - Sunita Ghosh
- From the Department of Surgery, University of Alberta, Edmonton, Alta. (Nostedt, McCall, Schiller); the Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alta. (Gibson-Brokop); the Department of Medical Oncology and the Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Alta. (Ghosh); and the Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alta. (Seidler)
| | - Michael Seidler
- From the Department of Surgery, University of Alberta, Edmonton, Alta. (Nostedt, McCall, Schiller); the Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alta. (Gibson-Brokop); the Department of Medical Oncology and the Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Alta. (Ghosh); and the Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alta. (Seidler)
| | - Michael McCall
- From the Department of Surgery, University of Alberta, Edmonton, Alta. (Nostedt, McCall, Schiller); the Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alta. (Gibson-Brokop); the Department of Medical Oncology and the Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Alta. (Ghosh); and the Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alta. (Seidler)
| | - Daniel Schiller
- From the Department of Surgery, University of Alberta, Edmonton, Alta. (Nostedt, McCall, Schiller); the Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alta. (Gibson-Brokop); the Department of Medical Oncology and the Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Alta. (Ghosh); and the Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alta. (Seidler)
| |
Collapse
|
29
|
Zhang N, Deng J, Wang W, Sun Z, Wang Z, Xu H, Zhou Z, Liang H. Negative lymph node count as an independent prognostic factor in stage III patients after curative gastrectomy: A retrospective cohort study based on a multicenter database. Int J Surg 2019; 74:44-52. [PMID: 31874262 DOI: 10.1016/j.ijsu.2019.12.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/26/2019] [Accepted: 12/18/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To examine the prognostic value of negative lymph node (NLN) count in stage III gastric cancer (GC) patients after curative gastrectomy. METHODS The clinicopathological data of 2942 stage III patients who underwent curative gastrectomy between 2001 and 2011 were analyzed. Only patients with ≥16 examined lymph nodes (ELNs) were included. After cut-point survival analysis, the 2942 patients were divided into three subgroups with NLN counts of 0, 1-9, and ≥10. Survival differences among the subgroups were analyzed to assess the effects of NLN count on stage migration and overall survival (OS) in stage III GC patients. Spearman's correlation coefficient was used to assess the relationships between the ELN count and the positive lymph node (PLN) count, the ELN count and the NLN count, and the NLN count and the PLN count. RESULTS Survival analyses revealed that the NLN count was significantly associated with OS (P = 0.001) and was an independent predictor (P < 0.01) of prognosis in stage III GC patients. Subgroup analysis showed that the prognostic evaluation accuracy was highest when the NLN count was ≥10 for stage III patients. Stage migrations were mainly detected in the following pathological tumor-node (pTN) subgroups: pT2N3a with 1-9 NLNs and pT2N3b with ≥10 NLNs, and pT3N3a with 1-9 NLNs and pT3N3b with ≥10 NLNs. NLN count was positively correlated with the ELN and the PLN counts for pT2N3 and pT3N3 stage GC patients (r = 0.694 and r = 0.881 for pT2N3 patients; r = 0.685 and r = 0.902 for pT3N3 patients, respectively; P < 0.001). These findings indicate that the NLN count may be a useful prognostic predictor in stage III GC patients. CONCLUSIONS The NLN count may improve the prognostic prediction efficiency of the tumor-node-metastasis (TNM) classification for GC, especially for stage III patients, and should be recommended for clinical applications.
Collapse
Affiliation(s)
- Nannan Zhang
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Cancer for Cancer, Tianjin, 300060, China
| | - Jingyu Deng
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Cancer for Cancer, Tianjin, 300060, China.
| | - Wei Wang
- Department of Gastric and Pancreatic Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Zhe Sun
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Zhenning Wang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Huimian Xu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| | - Zhiwei Zhou
- Department of Gastric and Pancreatic Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| | - Han Liang
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Cancer for Cancer, Tianjin, 300060, China.
| |
Collapse
|
30
|
Inhibiting casein kinase 2 overcomes paclitaxel resistance in gastric cancer. Gastric Cancer 2019; 22:1153-1163. [PMID: 31098863 DOI: 10.1007/s10120-019-00971-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Casein kinase (CK) 2 activation has been implicated in the proliferation of various tumor types and resistance to chemotherapy. We investigated the mechanistic basis for the association between CK2 activation and paclitaxel resistance in a gastric cancer (GC). EXPERIMENTAL DESIGN CK2 expression was evaluated in 59 advanced GC patients treated with paclitaxel as the second-line therapy. The efficacy of a CK2 inhibitor, CX-4945, and paclitaxel was evaluated in GC cell lines and a xenograft model. RESULTS Patients with high CK2 expression (29/59, 39%) showed lower disease control rates (47.7% vs. 72.3%, p = 0.017) and shorter progression-free survival (2.8 vs. 4.8 months, p = 0.009) than patients with low CK2 expression. CK2 protein expression was associated with sensitivity to paclitaxel in 49 GC cell lines. Combination therapy with CX-4945 and paclitaxel exerted synergistic antiproliferative effects and inhibited the downregulation of phosphatidylinositol 3-kinase/AKT signaling in SNU-1 cells. In the SNU-1 xenograft model, the combination treatment was significantly superior to either single agent, suppressing tumor growth without notable toxicities. CONCLUSIONS These results demonstrated that CK2 activation was related to paclitaxel resistance and that CX-4945 in combination with paclitaxel could be used as a potential treatment for paclitaxel resistance in GC.
Collapse
|
31
|
Jeong O, Jung MR, Kang JH, Ryu SY. Prognostic Performance of Preoperative Staging: Assessed by Using Multidetector Computed Tomography—Between the New Clinical Classification and the Pathological Classification in the Eighth American Joint Committee on Cancer Classification for Gastric Carcinoma. Ann Surg Oncol 2019; 27:545-551. [DOI: 10.1245/s10434-019-07845-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Indexed: 02/06/2023]
|
32
|
Lin JX, Yoon C, Desiderio J, Yi BC, Li P, Zheng CH, Parisi A, Huang CM, Strong VE, Yoon SS. Development and validation of a staging system for gastric adenocarcinoma after neoadjuvant chemotherapy and gastrectomy with D2 lymphadenectomy. Br J Surg 2019; 106:1187-1196. [PMID: 31197829 DOI: 10.1002/bjs.11181] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/21/2019] [Accepted: 02/25/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Neoadjuvant chemotherapy followed by gastrectomy with D2 lymphadenectomy is commonly used for patients with locally advanced gastric adenocarcinoma. The eighth AJCC ypTNM staging system was validated based on patients undergoing more limited lymphadenectomy (less than D2). The aim of this study was to develop a system for accurate staging of patients with locally advanced gastric adenocarcinoma who receive neoadjuvant chemotherapy followed by gastrectomy with D2 lymphadenectomy. METHODS A modified system of ypTNM was developed, based on overall survival (OS) of patients receiving neoadjuvant chemotherapy followed by gastrectomy with D2 lymphadenectomy at Memorial Sloan Kettering Cancer Center, and validated using data from an international cohort of patients who had similar treatment. RESULTS Of 325 patients in the derivation cohort, 33 (10·2 per cent) had ypT0 N0/+ tumours, which are not classifiable under the AJCC system. The 5-year OS rate for modified ypTNM stages I, II, IIIA and IIIB was 89, 71, 42·3 and 10 per cent respectively, compared with 82, 65·2 and 24·1 for AJCC stages I, II and III respectively. The concordance index (0·730 versus 0·709), estimated area under the curve (0·765 versus 0·740) and time-dependent receiver operating characteristic (ROC) curve throughout the observation period were all superior for modified ypTNM staging. For the validation cohort of 186 patients, the modified system was again better at separating patients into prognostic groups for OS. CONCLUSION The modified ypTNM staging system improves the accuracy of OS prediction for patients treated with neoadjuvant chemotherapy followed by gastrectomy with D2 lymphadenectomy.
Collapse
Affiliation(s)
- J X Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.,Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - C Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J Desiderio
- Department of Digestive Surgery, St. Mary's Hospital, University of Perugia, Terni, Italy
| | - B C Yi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - P Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - C H Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - A Parisi
- Department of Digestive Surgery, St. Mary's Hospital, University of Perugia, Terni, Italy
| | - C M Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - V E Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - S S Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
33
|
Liu JY, Deng JY, Zhang NN, Liu HF, Sun WL, He WT, Wang Y, Zhang L, Liang H. Clinical significance of skip lymph-node metastasis in pN1 gastric-cancer patients after curative surgery. Gastroenterol Rep (Oxf) 2019; 7:193-198. [PMID: 31217983 PMCID: PMC6573797 DOI: 10.1093/gastro/goz008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/30/2018] [Accepted: 01/10/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In addition to the stepwise manner of lymph-node metastasis from the primary tumour, the skip lymph-node metastasis (SLNM) was identified as a low-incidence metastasis of gastric cancer (GC). So far, both the mechanism and outcome of SLNM have not been elucidated completely. The purpose of this study was to analyse the clinical significance and the potential mechanism of SLNM in GC patients who had lymph-node metastasis. METHODS Clinicopathological data and follow-up information of 505 GC patients who had lymph-node metastasis were analysed to demonstrate the significance of SLNM in evaluating the prognostic outcome. According to the pathological results, all GC patients who had lymph-node metastasis were categorized into three groups: patients with the perigastric lymph-node metastasis, patients with the perigastric and extragastric lymph-node metastasis and patients with SLNM.Results: Among the 505 GC patients who had lymph-node metastasis, 24 (4.8%) had pathologically identified SLNM. The location of lymph-node metastasis was not significantly associated with 5-year survival rate and overall survival (OS) (P = 0.194). The stratified survival analysis results showed that the status of SLNM was significantly associated with the OS in patients with pN1 GC (P = 0.001). The median OS was significantly shorter in 19 pN1 GC patients with SLNM than in 100 patients with perigastric lymph-node metastasis (P < 0.001). The case-control matched logistic regression analysis results showed that tumour size (P = 0.002) was the only clinicopathological factor that may predict SLNM in pN1 GC patients undergoing curative surgery. Among the 19 pN1 GC patients with SLNM, 17 (89.5%) had metastatic lymph nodes along the common hepatic artery, around the celiac artery or in the hepatoduodenal ligament. CONCLUSIONS SLNM may be considered a potentially practicable indicator for prognosis among various subgroups of pN1 GC patients.
Collapse
Affiliation(s)
- Jin-Yuan Liu
- Department of Gastroenterology, Tianjin Medical University Cancer Hospital, City Key Laboratory of Tianjin Cancer Center and National Clinical Research Center for Cancer, Tianjin, P. R. China
- Department of General Surgery, The Affiliated Hospital of Nankai University, Tianjin, P. R. China
| | - Jing-Yu Deng
- Department of Gastroenterology, Tianjin Medical University Cancer Hospital, City Key Laboratory of Tianjin Cancer Center and National Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Nan-Nan Zhang
- Department of Gastroenterology, Tianjin Medical University Cancer Hospital, City Key Laboratory of Tianjin Cancer Center and National Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Hui-Fang Liu
- Department of Gastroenterology, Tianjin Medical University Cancer Hospital, City Key Laboratory of Tianjin Cancer Center and National Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Wei-Lin Sun
- Department of Gastroenterology, Tianjin Medical University Cancer Hospital, City Key Laboratory of Tianjin Cancer Center and National Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Wen-Ting He
- Department of Gastroenterology, Tianjin Medical University Cancer Hospital, City Key Laboratory of Tianjin Cancer Center and National Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Yan Wang
- Department of General Surgery, The Affiliated Hospital of Nankai University, Tianjin, P. R. China
| | - Li Zhang
- Department of Gastroenterology, Tianjin Medical University Cancer Hospital, City Key Laboratory of Tianjin Cancer Center and National Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Han Liang
- Department of Gastroenterology, Tianjin Medical University Cancer Hospital, City Key Laboratory of Tianjin Cancer Center and National Clinical Research Center for Cancer, Tianjin, P. R. China
| |
Collapse
|
34
|
Zhang H, Jiang H, Zhang H, Liu J, Hu X, Chen L. Ribophorin II potentiates P-glycoprotein- and ABCG2-mediated multidrug resistance via activating ERK pathway in gastric cancer. Int J Biol Macromol 2019; 128:574-582. [PMID: 30710584 DOI: 10.1016/j.ijbiomac.2019.01.195] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/15/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022]
Abstract
Multidrug resistance (MDR) is a critical reason of cancer chemotherapy failure. Ribophorin II (RPN2) has emerged as a vital regulator of MDR in multiple cancers including gastric cancer (GC). However, the roles and molecular mechanisms of RPN2 in MDR have not been well featured till now. The present study aimed to explore the roles and molecular mechanisms of RPN2 in MDR of drug-resistant GC cells. Results showed that the expressions of RPN2, multidrug resistance 1 (MDR1), and ATP binding cassette subfamily G member 2 (ABCG2) were upregulated in SGC7901/DDP and SGC7901/VCR cells. Knockdown of RPN2 alleviated MDR through downregulating MDR1 and ABCG2 expressions in SGC7901/DDP and SGC7901/VCR cells. RPN2 depletion inhibited the activation of MEK/ERK pathway. RPN2 overexpression enhanced MDR by upregulating P-glycoprotein (P-gp) and ABCG2 protein expressions in SGC7901/DDP or SGC7901/VCR cells, while this effect of RPN2 was abrogated by ERK knockdown or treatment with ERK inhibitor PD98059. Our findings suggested that RPN2 potentiated P-gp- and ABCG2-mediated MDR via activating MEK/ERK pathway in GC, hinting the critical values of RPN2 in ameliorating MDR and providing a promising target for GC therapy.
Collapse
Affiliation(s)
- Hongzhi Zhang
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China.
| | - Huijuan Jiang
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Huixiang Zhang
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Juncai Liu
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Xigang Hu
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Lei Chen
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| |
Collapse
|
35
|
The long-term prognostic difference between gastrectomy with and without preoperative chemotherapy in patients with clinical stage IV gastric cancer. Asian J Surg 2019; 42:922-929. [PMID: 30685146 DOI: 10.1016/j.asjsur.2019.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/28/2018] [Accepted: 01/08/2019] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND/OBJECTIVE The role of gastrectomy for stage IV gastric cancer (GC) has not yet been established. Thus, we aimed to compare the prognoses of patients with clinical stage IV GC who underwent gastrectomy with and without preoperative chemotherapy after precise recategorization of patients. METHODS We retrospectively reviewed a total of 92 clinical stage IV GC patients who underwent gastrectomy with or without preoperative chemotherapy between 2010 and 2016 at a single institution. Yoshida's classification was used to categorize the patients into the following categories: 1, technically resectable metastasis; 2, marginally resectable metastasis; 3, unresectable peritoneal dissemination; and 4, incurable peritoneal dissemination with distant organ metastasis. Two-year disease-specific survival (DSS) rates were compared between patients who underwent primary surgery and preoperative chemotherapy for each category. RESULTS The two-year DSS rates of primary surgery vs. preoperative chemotherapy in Categories 1, 2, 3, and 4 (n = 35, 39, 14, and 4, respectively) were 48.6% vs. 41.7% (p = 0.829), 52.6% vs. 40.0% (p = 0.855), 50.0% vs. 75.0% (p = 0.027), and 0% vs. 66.7% (p = 0.083), respectively. Patients in Categories 1 and 2 who underwent preoperative chemotherapy tended to have lower two-year DSS rates (p = 0.911), whereas patients in Categories 3 and 4 had significantly higher two-year DSS rates than those who underwent primary surgery (p = 0.014). CONCLUSIONS Primary surgery may be performed in patients without peritoneal dissemination when GC is technically resectable. However, if peritoneal dissemination is suspected, chemotherapy should be prioritized.
Collapse
|
36
|
Yu JI, Lim DH, Lee J, Kang WK, Park SH, Park JO, Park YS, Lim HY, Kim ST, Lee SJ, Kim S, Sohn TS, Lee JH, An JY, Choi MG, Bae JM, Yoo H, Kim K. Comparison of the 7th and the 8th AJCC Staging System for Non-metastatic D2-Resected Lymph Node-Positive Gastric Cancer Treated with Different Adjuvant Protocols. Cancer Res Treat 2018; 51:876-885. [PMID: 30282450 PMCID: PMC6639239 DOI: 10.4143/crt.2018.401] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The purpose of this study was to compare prognostic differentiation performances of the 7th and the 8th edition of American Joint Commission on Cancer (AJCC) staging system for gastric cancer (GC) patients. Materials and Methods A total of 1,633 GC patients who underwent curative D2 resection followed by adjuvant chemotherapy alone (CA) or concurrent chemo-radiotherapy (CCRT) from 2004 to 2013 were included. Concordance index (c-index) was applied to compare the discriminatory ability. RESULTS In the 8th edition, migration of stage was detected in 248 patients (15.2%). Among them, 121 patients were up-staged while 127 patients were down-staged. Overall, there was no statistically significant difference in the discriminatory ability between the 7th and 8th editions. The new edition of staging system, however, showed a trend of better prognostic performance not only in recurrence-free survival (c-index=0.734; 95% confidence interval [CI], 0.706 to 0.762 in the 7th edition vs. c-index=0.740; 95% CI, 0.712 to 0.768 in the 8th edition; p=0.14), but also in overall survival (c-index=0.717; 95% CI, 0.688 to 0.745 in the 7th edition vs. c-index=0.722; 95% CI, 0.694 to 0.751 in the 8th edition; p=0.19), especially in stage III. This finding was repeated in the subgroup analysis regardless of adjuvant CA or CCRT. CONCLUSION Generally, the 8th edition of AJCC staging system had failed to show a superior discriminatory ability for curatively D2 resected GC patients than the 7th edition, although there was a trend of better prognostic performance of the new edition, regardless of adjuvant treatment method.
Collapse
Affiliation(s)
- Jeong Il Yu
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeeyun Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Ki Kang
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se Hoon Park
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Oh Park
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Suk Park
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ho Yeong Lim
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Tae Kim
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Su Jin Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Sung Sohn
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jun Ho Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Yeong An
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Min Gew Choi
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Moon Bae
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Heejin Yoo
- Statistics and Data Center, Samsung Medical Center, Seoul, Korea
| | - Kyunga Kim
- Statistics and Data Center, Samsung Medical Center, Seoul, Korea
| |
Collapse
|