1
|
Chen W, Cai Z, Li J, Li Z, Xu Z, Wu J, Liu F, Zhao H, Xu Y. Five-year survival outcome of laparoscopic D2 lymphadenectomy plus complete mesogastrium excision for patients with stage II/III gastric cancer without invasion of the squamocolumnar junction. Surgery 2025; 181:109290. [PMID: 40068362 DOI: 10.1016/j.surg.2025.109290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/30/2024] [Accepted: 02/01/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Gastric cancer patients without invasion of the squamocolumnar junction have a low probability of extragastric mesenteric metastasis, and the survival benefit of conducting D2 lymphadenectomy plus complete mesogastrium excision is unclear in this group of patients. METHODS This was a retrospective analysis of patients with stage II/III gastric cancer without invasion of the squamocolumnar junction who underwent laparoscopic radical surgery for gastric cancer at the First Hospital of Putian City, Fujian Province, from January 2014 to May 2019. Five-year disease-free survival was the primary outcome, and 5-year overall survival and the recurrence pattern were the secondary outcomes. Morbidity, mortality, and surgery-related outcomes within 30 days of surgery were also analyzed. RESULTS This study included 366 patients, 133 in the D2 lymphadenectomy group and 233 in the D2 lymphadenectomy plus complete mesogastrium excision group. There was no statistically significant difference in 5-year disease-free survival and overall survival in the D2 lymphadenectomy plus complete mesogastrium excision group compared with the D2 lymphadenectomy group (P > .05). Regarding recurrence pattern, the D2 lymphadenectomy group had a higher rate of local recurrence (12.3% and 38.3%, P = .001). In subgroup analysis, patients with stage III and T3-4aN1-3 gastric cancer in the D2 lymphadenectomy plus complete mesogastrium excision group had significantly better 5-year disease-free survival compared with the D2 lymphadenectomy group (P < .05). CONCLUSION Laparoscopic D2 lymphadenectomy plus complete mesogastrium excision not only decreased the local recurrence rate of stage II/III gastric cancer without invasion of the squamocolumnar junction, but it also improved the 5-year disease-free survival of patients with stage III and T3-4aN1-3 gastric cancer without invasion of the squamocolumnar junction.
Collapse
Affiliation(s)
- Weixiang Chen
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Zhiming Cai
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Junpeng Li
- Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Zhixiong Li
- Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Zhengnan Xu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Jihuang Wu
- Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Feng Liu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Hongrui Zhao
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China
| | - Yanchang Xu
- Gastrointestinal Surgery Unit 1, The First Hospital of Putian City, Putian, Fujian, China.
| |
Collapse
|
2
|
Zhan Z, Chen B, Xu S, Lin R, Chen H, Ma X, Lin X, Huang W, Zhuo C, Chen Y, Guo Z. Neoadjuvant chemotherapy combined with antiangiogenic therapy and immune checkpoint inhibitors for the treatment of locally advanced gastric cancer: a real - world retrospective cohort study. Front Immunol 2025; 16:1518217. [PMID: 39967656 PMCID: PMC11832677 DOI: 10.3389/fimmu.2025.1518217] [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: 10/28/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Background Although immune checkpoint inhibitors (ICIs) and anti-angiogenic drugs have demonstrated effectiveness in treating advanced gastric cancer (GC), their role in neoadjuvant or conversion therapy remains uncertain. This study aimed to evaluate the efficacy and safety of combining neoadjuvant chemotherapy with anti-angiogenesis and ICIs in patients with locally advanced GC (LAGC). Methods In this cohort study, we reviewed our prospectively maintained GC database and included individuals diagnosed with clinical stage II-III GC who received neoadjuvant therapy followed by surgery between January 2022 and August 2023. The treatment protocol combined ICIs, anti-angiogenic therapy (specifically apatinib), and chemotherapy (S-1 with oxaliplatin). A systematic approach was used to document patients' clinical and pathological characteristics, pathological findings, and survival outcomes, which were subsequently analyzed in detail. Results A total of 38 individuals met the study's inclusion criteria, with the majority (32 patients, 84.2%) having clinical stage III GC. All participants underwent surgery, resulting in a notable R0 resection rate of 97.4%. The rates of major pathological response (MPR) and pathological complete response (pCR) were 47.4% and 23.7%, respectively. Post-surgery, 36 patients (92.1%) received adjuvant chemotherapy. With a median follow-up of 22 months, ten patients experienced disease recurrence, including three who died from tumor relapse. The 1-year overall survival (OS) rate stood at 100%, and the disease-free survival (DFS) rate was 94.7%, with median OS and DFS yet to be reached. The neoadjuvant therapy regimen was generally well-tolerated, with no grade 5 treatment-related adverse events (TRAEs) reported. Only one patient experienced a grade 4 TRAE (immune-related hepatitis), while the most common grade 3 TRAEs included thrombocytopenia, elevated aminotransferase levels, and neutropenia. Conclusions The combination of neoadjuvant chemotherapy, anti-angiogenic therapy, and ICIs has proven effective in treating LAGC patients, achieving high pCR rates and favorable survival outcomes while maintaining an acceptable safety profile.
Collapse
Affiliation(s)
- Zhouwei Zhan
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Bijuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Shaohua Xu
- Department of Hepatobiliary and Pancreatic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Ruyu Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Haiting Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Xiaohuan Ma
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Xuanping Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Wanting Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Changhua Zhuo
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Zengqing Guo
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| |
Collapse
|
3
|
Li C, Tian Y, Zheng Y, Yuan F, Shi Z, Yang L, Chen H, Jiang L, Wang X, Zhao P, Zhang B, Wang Z, Zhao Q, Dong J, Lian C, Xu S, Zhang A, Zheng Z, Wang K, Dang C, Wu D, Chen J, Xue Y, Liang B, Cheng X, Wang Q, Chen L, Xia T, Liu H, Xu D, Zhuang J, Wu T, Zhao X, Wu W, Wang H, Peng J, Hou Z, Zheng R, Chen Y, Yin K, Zhu Z. Pathologic Response of Phase III Study: Perioperative Camrelizumab Plus Rivoceranib and Chemotherapy Versus Chemotherapy for Locally Advanced Gastric Cancer (DRAGON IV/CAP 05). J Clin Oncol 2025; 43:464-474. [PMID: 39383487 PMCID: PMC11776878 DOI: 10.1200/jco.24.00795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/01/2024] [Accepted: 09/05/2024] [Indexed: 10/11/2024] Open
Abstract
PURPOSE This multicenter, randomized phase III trial evaluated the efficacy and safety of perioperative camrelizumab (an anti-PD-1 antibody) plus low-dose rivoceranib (a VEGFR-2 inhibitor) and S-1 and oxaliplatin (SOX) (SOXRC), high-dose rivoceranib plus SOX (SOXR), and SOX alone (SOX) for locally advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. METHODS Patients with T3-4aN + M0 G/GEJ adenocarcinoma were randomly assigned (1:1:1) to receive perioperative treatment with SOXRC, SOXR, or SOX. The primary end points were pathologic complete response (pCR) and event-free survival. The Independent Data Monitoring Committee recommended stopping enrollment in the SOXR group on the basis of the safety data of the first 103 randomly assigned patients in the three groups. The patients were then randomly assigned 1:1 to the SOXRC or SOX groups. This report presents the pCR results obtained per protocol for the first 360 randomly assigned patients who had the opportunity for surgery in the SOXRC and SOX groups. RESULTS In the SOXRC and SOX groups, of the 180 patients in each group, 99% and 98% of patients received neoadjuvant therapy, 91% and 94% completed planned neoadjuvant therapy, and 86% and 87% underwent surgery, respectively. The pCR was significantly higher in the SOXRC group at 18.3% (95% CI, 13.0 to 24.8) compared with 5.0% (95% CI, 2.3 to 9.3) in the SOX group (difference of 13.7%; 95% CI, 7.2 to 20.1; odds ratio of 4.5 [95% CI, 2.1 to 9.9]). The one-sided P value was <.0001, crossing the prespecified statistical significance threshold of P = .005. Surgical complications and grade ≥3 neoadjuvant treatment-related adverse events were 27% versus 33% and 34% versus 17% for SOXRC and SOX, respectively. CONCLUSION The SOXRC regimen significantly improved pCR compared with SOX alone in patients with G/GEJ adenocarcinoma with a tolerable safety profile.
Collapse
Affiliation(s)
- Chen Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yantao Tian
- Pancreatogastric Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Yanan Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zheng Shi
- Gastrointestinal Surgery, Shanghai Changhai Hospital, Shanghai, China
| | - Lin Yang
- Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Hao Chen
- Oncological Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Lixin Jiang
- Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Yantai, China
| | - Xixun Wang
- Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Yantai, China
| | - Ping Zhao
- Gastrointestinal Surgery, Sichuan Provincial Cancer Hospital, Chengdu, China
| | - Benyan Zhang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zhenqiang Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Qun Zhao
- Gastrointestinal Oncology Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianhong Dong
- Minimally Invasive Surgery of Digestive Endoscopy, Shanxi Provincial Cancer Hospital, Taiyuan, China
| | - Changhong Lian
- Gastrointestinal Surgery, HePing Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Sanrong Xu
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Aimin Zhang
- Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Zhichao Zheng
- Gastric Cancer Surgery, Liaoning Cancer Hospital & Institution, Shenyang, China
| | - Kang Wang
- Gastrointestinal Surgery, Sichuan Provincial People's Hospital, Chengdu, China
| | - Chengxue Dang
- Surgical Oncology, The First Affiliated Hospital of XI'AN Jiaotong University, Xi'an, China
| | - Dan Wu
- Gastrointestinal Surgery, The Second Affiliated Hospital Zhejiang University, School of Medicine, Hangzhou, China
| | - Jian Chen
- Gastrointestinal Surgery, The Second Affiliated Hospital Zhejiang University, School of Medicine, Hangzhou, China
| | - Yingwei Xue
- Gastrointestinal Surgery, Harbin Medical University Affiliated Cancer Hospital, Harbin, China
| | - Bo Liang
- General Surgery, Jilin Guowen Hospital, Changchun, China
| | | | - Qian Wang
- Gastrointestinal Surgery, The Affiliated Hospital of GuiZhou Medical University, Guiyang, China
| | - Luchuan Chen
- Gastrointestinal Cancer Surgery Department, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Tao Xia
- Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Heli Liu
- Gastrointestinal Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Dazhi Xu
- Gastrosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing Zhuang
- General Surgery, Henan Cancer Hospital, Zhengzhou, China
| | - Tao Wu
- General Surgery, Tangdu Hospital of the Fourth Military Medical, Xi'an, China
| | - Xi Zhao
- Colorectal Surgery, Hainan Cancer Hospital, Haikou, China
| | - Wei Wu
- Geriatric Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Hongzhi Wang
- Center of Gastrointestinal Tumor, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Junsheng Peng
- Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiguo Hou
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | | | - Yuting Chen
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Kai Yin
- Gastrointestinal Surgery, Shanghai Changhai Hospital, Shanghai, China
| | - Zhenggang Zhu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Panahizadeh R, Panahi P, Asghariazar V, Makaremi S, Noorkhajavi G, Safarzadeh E. A literature review of recent advances in gastric cancer treatment: exploring the cross-talk between targeted therapies. Cancer Cell Int 2025; 25:23. [PMID: 39856676 PMCID: PMC11762578 DOI: 10.1186/s12935-025-03655-8] [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: 06/08/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) ranks fourth in global mortality rates and fifth in prevalence, making it one of the most common cancers worldwide. Recent clinical studies have highlighted the potential of immunotherapies as a promising approach to treating GC. This study aims to shed light on the most impactful therapeutic strategies in the context of GC immunotherapy, highlighting both established and emerging approaches. MAIN BODY This review examines over 160 clinical studies conducted globally, focusing on the effectiveness of various immunotherapy modalities, including cancer vaccines, adoptive cell therapy, immune checkpoint inhibitors (ICIs), and monoclonal antibodies (mAbs). A comprehensive search of peer-reviewed literature was performed using databases such as Web of Science, PubMed, and Scopus. The selection criteria included peer-reviewed articles published primarily within the last 10 years, with a focus on studies that provided insights into targeted therapies and their mechanisms of action, clinical efficacy, and safety profiles. The findings indicate that these immunotherapy strategies can enhance treatment outcomes for GC, aligning with current treatment guidelines. ICIs like pembrolizumab and nivolumab have shown significant survival benefits in specific GC subgroups. Cancer vaccines and CAR-T cell therapies demonstrate potential, while mAbs targeting HER2 and VEGFR pathways enhance outcomes in combination regimens. We discuss the latest advancements and challenges in targeted therapy and immunotherapy for GC. Given the evolving nature of this field, this research emphasizes significant evidence-based therapies and those currently under evaluation rather than providing an exhaustive overview. Challenges include resistance mechanisms, immunosuppressive tumor environments, and inconsistent results from combination therapies. Biomarker-driven approaches and further research into emerging modalities like CAR-T cells and cancer vaccines are critical for optimizing treatments. CONCLUSIONS Immunotherapy is reshaping GC management by improving survival and quality of life. Ongoing research and clinical evaluations are crucial for refining personalized and effective therapies.
Collapse
Affiliation(s)
- Reza Panahizadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Padideh Panahi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shima Makaremi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ghasem Noorkhajavi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 85991-56189, Iran.
| |
Collapse
|
5
|
Yu Z, Liang C, Xu Q, Yuan Z, Chen M, Li R, Zhou S, Li P, Wei B, Zhao X. The safety and efficacy of neoadjuvant PD-1 inhibitor plus chemotherapy for patients with locally advanced gastric cancer: a systematic review and meta-analysis. Int J Surg 2025; 111:1415-1426. [PMID: 39172720 PMCID: PMC11745722 DOI: 10.1097/js9.0000000000002056] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/11/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND The extensive utilization of immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1) has achieved significant advancements in the treatment of diverse solid tumors. The present meta-analysis aims to evaluate the safety and efficacy of neoadjuvant chemotherapy (NCT) plus PD-1 inhibitors for patients with locally advanced gastric cancer (LAGC). METHODS An electronic search of PubMed, EmBase, and the Cochrane Library was performed to identify the clinical trials of NCT + PD-1 inhibitor vs. NCT in patients with LAGC. The retrieval period extended from the establishment of the corresponding database until April 2024, and meta-analysis was conducted using Stata (version 15) software. Subsequently, direct comparative analysis was used to compare pooled results of neoadjuvant immunochemotherapy (NICT) with NCT. RESULTS After screening, six phase II/III randomized controlled trials (RCTs) and nine retrospective studies with 2953 patients were included. In meta-analysis, the NICT group demonstrated a significantly higher rate of pathological complete response (pCR) ( P <0.001) and R0 resection ( P =0.001), and a lower 2-year recurrence rate ( P =0.001) compared to the NCT group. The NICT group, however, exhibited a higher incidence of severe treatment-related adverse events (TRAEs) ( P =0.044). Additionally, the NICT and NCT groups exhibited no statistical differences in terms of the number of harvested lymph nodes, the occurrence of total TRAEs and postoperative complications, as well as the duration of postoperative hospitalization. CONCLUSIONS The combination of PD-1 inhibitor + NCT in LAGC patients enhances the likelihood of achieving radical surgery and improves prognosis, albeit to some extent increasing the risk of severe TRAEs. NICT is anticipated to emerge as the preferred neoadjuvant therapy option for patients diagnosed with LAGC.
Collapse
Affiliation(s)
- Zhiyuan Yu
- Medical School of Chinese PLA
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing
- School of Medicine, Nankai University, Tianjin
| | - Chen Liang
- Department of Gastroenterology, Beijing Jishuitan Hospital, Capital Medical University, Beijing
| | - Qixuan Xu
- Medical School of Chinese PLA
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing
| | - Zhen Yuan
- Medical School of Chinese PLA
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing
- School of Medicine, Nankai University, Tianjin
| | - Miao Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Shandong, People’s Republic of China
| | - Rui Li
- Medical School of Chinese PLA
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing
- School of Medicine, Nankai University, Tianjin
| | - Sixin Zhou
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing
| | - Peiyu Li
- Medical School of Chinese PLA
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing
- School of Medicine, Nankai University, Tianjin
| | - Bo Wei
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing
| | - Xudong Zhao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing
| |
Collapse
|
6
|
Chang C, Cai Z, Cheng K, Shen C, Zhang B, Chen Z, Yin Y, Cao D. Efficacy and safety of S-1 plus oxaliplatin combined with apatinib and camrelizumab as neoadjuvant therapy for patients with locally advanced gastric or gastroesophageal junction adenocarcinoma: a protocol for a single-arm phase II trial. Updates Surg 2025; 77:165-174. [PMID: 39738886 DOI: 10.1007/s13304-024-02052-6] [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: 08/12/2023] [Accepted: 11/27/2024] [Indexed: 01/02/2025]
Abstract
Gastric cancer, as the fifth most diagnosed malignancy and the fourth leading cause of cancer-related death globally, remains a significant health concern. The potential effect of the programmed death-1 (PD-1) inhibitor, when used alongside chemotherapy and antiangiogenic agents in neoadjuvant therapy for gastric cancer, has yet to be explored in the published literature. This study aims to evaluate the efficacy and safety of the S-1 plus oxaliplatin (SOX) regimen when combined with apatinib and camrelizumab (SOXAC) as neoadjuvant therapy for patients with locally advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma. A single-arm, open-label, single-center phase II clinical trial has been designed to evaluate the safety and efficacy of the SOXAC regimen as neoadjuvant therapy for patients diagnosed with locally advanced gastric or GEJ adenocarcinoma (cT2-3N + M0 or T4NxM0). Eligible patients are to receive 2 cycles of SOXAC and 1 cycle of SOX regimen with camrelizumab (SOXC) as neoadjuvant therapy prior to radical surgery, and 3 cycles of SOXC as postoperative adjuvant therapy. The primary endpoint is major pathological remission (MPR), while secondary endpoints include pathological complete response (pCR) rate, R0 resection rate, objective response rate (ORR), operation-related outcomes, and safety. The SOX regimen remains a leading choice for neoadjuvant chemotherapy in Eastern countries. Recent studies suggest that combining chemotherapy, targeted agents, and immune checkpoint inhibitors can enhance the antitumor immune response. This phase II clinical trial seeks to assess the safety and efficacy of the SOXAC regimen as neoadjuvant therapy for patients with locally advanced resectable gastric or GEJ adenocarcinoma, while also exploring the correlation between biomarkers and efficacy.Trial Registration Chinese Clinical Trial Registry (ChiCTR): ChiCTR2200062285 ( https://www.chictr.org.cn/ ).
Collapse
Affiliation(s)
- Chen Chang
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Zhaolun Cai
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Ke Cheng
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Chaoyong Shen
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Bo Zhang
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Zhixin Chen
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yuan Yin
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Dan Cao
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Tang Y, Dai L, Wang Z, Zhang M, Xie H, Yang Y, Zhou Y, Yan Z, Wang H, Yang H, Zhang L, He T, Chen J, Wang G, Jin X, Wang Q. Short term efficacy and safety of PD-1 inhibitor and apatinib plus S-1 and oxaliplatin as neoadjuvant chemotherapy for patients with locally advanced gastric cancer. Medicine (Baltimore) 2024; 103:e40572. [PMID: 39560533 PMCID: PMC11576002 DOI: 10.1097/md.0000000000040572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 10/30/2024] [Indexed: 11/20/2024] Open
Abstract
Surgical resection is the cornerstone of treatment for locally advanced gastric cancer (LAGC). Hence, downstaging of the tumor with neoadjuvant therapy is critical for R0 resection and prolongs the overall survival. Data from related studies are lacking, and the literature is scarce. Therefore, a single arm-study was performed on PD-1 inhibitor and apatinib plus S-1 and oxaliplatin as neoadjuvant chemotherapy for patients with LAGC. The findings are expected to serve as a reference for neoadjuvant therapy for LAGC. We assessed 130 LAGC patients using PD-1 inhibitor, apatinib plus S-1, and oxaliplatin as neoadjuvant chemotherapy from January 2021 to October 2022. A total of 104 patients received gastric transcatheter chemoembolization (GTC). The primary endpoint was the rate of clinical complete response, pathological complete response, and safety, while the secondary endpoints were the R0 resection rate and objective response rate of the disease and the disease control rate. A total of 130 patients completed the clinical assessment, of which 6 patients (4.6%) achieved clinical complete response, 87 patients (66.9%) achieved partial response, 30 patients (23.0%) achieved stable disease, and 7 patients (5.5%) experienced progressive disease. The overall response rate was 71.5% (93/130), and the disease control rate was 94.5% (123/130). A remarkable downstaging effect was observed in this study. Downstaging of the T stage and N stage was achieved in 71.5% and 80% of the patients, respectively, which translated into a high R0 resection rate. The findings revealed that 125 patients underwent R0 resection, and the R0 resection rate was 96.1%. According to the observed results, 21.6% of the patients achieved pathological complete response after neoadjuvant chemotherapy. Gastric transcatheter chemoembolization in the first cycle of neoadjuvant therapy was beneficial for tumor regression (P < .001). All adverse events were relieved and disappeared after symptomatic treatment, and no grade 4 adverse events were noted. PD-1 inhibitor and apatinib plus S-1 and oxaliplatin are safe and effective as neoadjuvant treatment of LAGC. Gastric transcatheter chemoembolization is useful for tumor regression during neoadjuvant therapy.
Collapse
Affiliation(s)
- Yunchuan Tang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Li Dai
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhiqin Wang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Meifeng Zhang
- Department of Outpatient Clinic, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Haitao Xie
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yunshan Yang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yongjin Zhou
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Zhiqiang Yan
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Haibin Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hongxin Yang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lei Zhang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tong He
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jiaju Chen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Guanghai Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiangren Jin
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qian Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Guizhou Medical University, Guiyang, Guizhou Province, China
| |
Collapse
|
8
|
Yu Z, Liu H, Li R, Hu L, Xiao C, Gao Y, Li P, Liang W, Zhou S, Zhao X. Clinicopathological Factors and Nomogram Construction for Lymph Node Metastasis in Locally Advanced Gastric Cancer. Cancer Manag Res 2024; 16:1475-1489. [PMID: 39439918 PMCID: PMC11495200 DOI: 10.2147/cmar.s487247] [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: 08/17/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Background The research on lymph node metastasis (LNM) in locally advanced gastric cancer (LAGC) infiltrating the subserous tissue and serous membrane (T3-4a) is significantly inadequate. This study aims to explore the clinicopathological factors related to LNM in stages T3 and T4a LAGC, while also developing predictive nomograms. Methods After systematic searching and rigorous screening, 1995 T3 and 1244 T4a LAGC cases who underwent surgery without neoadjuvant or perioperative chemotherapy were selected. The risk factors associated with LNM were identified using both univariate and multivariate logistic regression analyses. Subsequently, the independent variables identified through the multivariate analyses were utilized to construct a nomogram. Results The incidence of LNM in T3 and T4a LAGC was 77.1% (1539/1995) and 83.8% (1043/1244), respectively. The following factors were found to be independently associated with LNM in T3 LAGC: preoperative serum albumin <41g/L (P=0.007), gastrointestinal obstruction (P<0.001), tumor location (P=0.040), tumor size >4cm (P=0.002), mixed (P=0.001) and undifferentiated histological types (P=0.002), presence of lymphovascular invasion (LVI) (P<0.001) and nerve invasion (P<0.001). Additionally, in T4a LAGC cases, serum albumin < 39g/L (P=0.004), tumor size >6cm (P=0.020), mixed (P<0.001) and undifferentiated histological types (P<0.001), presence of gastrointestinal hemorrhage (P=0.016), neuroendocrine differentiation (P=0.024), and LVI (P<0.001) independently influenced the occurrence of LNM. Conclusion This study identified the risk factors associated with LNM in T3-4a LAGC cases and constructed nomograms, thereby providing valuable guidance for formulating and implementing a multidisciplinary perioperative treatment program.
Collapse
Affiliation(s)
- Zhiyuan Yu
- Medical School of Chinese PLA, Beijing, People’s Republic of China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Haopeng Liu
- Department of Hepatobiliary Surgery, Zhangqiu District People’s Hospital, Jinan, Shandong Province, People’s Republic of China
| | - Rui Li
- Medical School of Chinese PLA, Beijing, People’s Republic of China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Liai Hu
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Chun Xiao
- Department of General Surgery, PLA Rocket Force Characteristic Medical Center, Beijing, People’s Republic of China
| | - Yunhe Gao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Peiyu Li
- Medical School of Chinese PLA, Beijing, People’s Republic of China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Wenquan Liang
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Sixin Zhou
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Xudong Zhao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
9
|
Lin JX, Xu BB, Zheng HL, Li P, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Tu RH, Huang ZN, Lin JL, Yao ZH, Zheng CH, Huang CM. Laparoscopic Spleen-Preserving Hilar Lymphadenectomy for Advanced Proximal Gastric Cancer Without Greater Curvature Invasion: Five-Year Outcomes From the Fuges-02 Randomized Clinical Trial. JAMA Surg 2024; 159:747-755. [PMID: 38691353 PMCID: PMC11238028 DOI: 10.1001/jamasurg.2024.1023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/13/2024] [Indexed: 05/03/2024]
Abstract
Importance Splenic hilar lymphadenectomy has been recommended for locally advanced proximal gastric cancer (APGC) involving the greater curvature. However, it is unclear whether laparoscopic spleen-preserving splenic hilar lymphadenectomy (LSPSHL) is associated with a long-term survival benefit for APGC without greater curvature invasion. Objective To present the 5-year follow-up data from a randomized clinical trial that compared laparoscopic total gastrectomy (D2 group) with D2 plus LSPSHL (D2 + No. 10 group) among patients with resectable APGC. Design, Setting, and Participants This is a post hoc secondary analysis of a randomized clinical trial that enrolled 536 patients with potentially resectable APGC (cT2-4a, N0 or N+, and M0) without greater curvature invasion from January 5, 2015, to October 10, 2018. All patients were tracked for at least 5 years. The final follow-up was on October 30, 2023. Interventions Patients were randomly assigned in a 1:1 ratio to the D2 + No. 10 or D2 groups. Main Outcomes and Measures The 5-year disease-free survival (DFS) and overall survival (OS) rates were measured. Recurrence patterns and causes of death were compared. Results A total of 526 patients (392 men [74.5%]; mean [SD] age, 60.6 [9.6] years) were included in the modified intent-to-treat analysis, with 263 patients in each group. The 5-year DFS rate was 63.9% (95% CI, 58.1%-69.7%) for the D2 + No. 10 group and 55.1% (95% CI, 49.1%-61.1%) for the D2 group (log-rank P = .04). A statistically significant difference was observed in the 5-year OS between the D2 + No. 10 group and the D2 group (66.2% [95% CI, 60.4%-71.9%] vs 57.4% [95% CI, 51.4%-63.4%]; log-rank P = .03). The No. 10 lymph node exhibited a therapeutic value index (TVI) of 6.5, surpassing that of Nos. 8a (TVI, 3.0), 11 (TVI, 5.8), and 12a (TVI, 0.8). A total of 86 patients in the D2 + No. 10 group (cumulative incidence, 32.7%) and 111 patients in the D2 group (cumulative incidence, 42.2%) experienced recurrence (hazard ratio, 0.72; 95% CI, 0.54-0.95; P = .02). The multivariable competing risk regression model demonstrated that D2 + No. 10 remained an independent protective factor for a lower 5-year cumulative recurrence rate after surgery (hazard ratio, 0.75; 95% CI, 0.56-1.00; P = .05). There was a significant difference in the 5-year cumulative recurrence rate at the No. 10 lymph node area between the 2 groups (D2 + No. 10 group vs D2 group: 0% vs 2.3% [n = 6]; P = .01). Conclusions This post hoc secondary analysis of a randomized clinical trial found that laparoscopic total gastrectomy with LSPSHL can improve the prognosis and reduce recurrence for APGC without greater curvature invasion. Future multicenter studies are warranted to validate these findings. Trial Registration ClinicalTrials.gov Identifier: NCT02333721.
Collapse
Affiliation(s)
- Jian-xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Bin-bin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Jian-wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Jia-bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Qi-yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Long-long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Ru-hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Ze-ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Ju-li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Zi-hao Yao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
- Fujian Province Minimally Invasive Medical Center, Fuzhou, China
| |
Collapse
|
10
|
Wei C, Du X, Hu J, Dong Y, Chen Y, Cao B. Perioperative chemotherapy versus adjuvant chemotherapy in patients with resectable gastric cancer: A systematic review with meta-analysis. Crit Rev Oncol Hematol 2024; 198:104082. [PMID: 37532103 DOI: 10.1016/j.critrevonc.2023.104082] [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: 04/05/2023] [Revised: 06/01/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023] Open
Abstract
PURPOSE The study aimed to investigate the prognosis and safety of perioperative chemotherapy (PC) compared with adjuvant chemotherapy (AC). METHODS We systematically searched and assessed studies in PubMed, Embase, and the Cochrane Library from inception to 1st September 2022. RESULTS Eighteen studies were eligible for the analysis, including 4686 patients in total. Our study found that patients with resectable gastric cancer undergoing PC had favorable prognosis on OS (HR 0.77; 95% CI 0.69-0.87) and DFS (HR 0.76; 95% CI 0.69-0.84) than those who undergoing AC. Addition of neoadjuvant chemotherapy (NAC) to AC provided higher R0 resection rate but did not increase the risk of postoperative complication rate and most of the adverse event rates. CONCLUSION Our study demonstrated that PC shows better OS and DFS in Asians with resectable gastric cancer compared with AC. PC should be preferred because of its favorable prognosis and similar safety.
Collapse
Affiliation(s)
- Chenyu Wei
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Xuelin Du
- Clinical Trial Institution, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jiexuan Hu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yin Dong
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yan Chen
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bangwei Cao
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
11
|
Wu L, Yan H, Qin Y, Huang M, Wang T, Jin Q, Wei W. Fruquintinib plus oxaliplatin combined with S-1 (SOX) as neoadjuvant therapy for locally advanced gastric cancer (GC) or gastro-oesophageal junction adenocarcinoma (GEJ): a multicentre, phase II, single-arm, open-label clinical trial (FRUTINEOGA) protocol. BMJ Open 2024; 14:e075696. [PMID: 38341203 PMCID: PMC10862274 DOI: 10.1136/bmjopen-2023-075696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
INTRODUCTION Curing locally advanced gastric cancer (GC) or gastro-oesophageal junction adenocarcinoma (GEJ) with surgery alone is challenging. Neoadjuvant chemotherapy (NCT) has become the standard treatment for patients with locally advanced GC/GEJ, and SOX is the most common neoadjuvant regimen in China. The generally good tolerability in patients and fruquintinib's low potential for drug-drug interaction suggest that it may be highly suitable for combinations with other antineoplastic therapies. A combination of fruquintinib, S-1 and oxaliplatin can be a promising neoadjuvant treatment for locally advanced GC/GEJ. In this phase II study, we aim to investigate the efficacy and toxicity of fruquintinib plus SOX as neoadjuvant treatment for locally advanced GC/GEJ. METHODS AND ANALYSIS The FRUTINEOGA trial is a prospective, multicentre, phase II, single-arm, open-label clinical trial that will enrol 54 patients. Eligible patients will be registered, enrolled and receive 2-4 cycles of fruquintinib plus SOX, after which surgery will be performed and tumour regression will be evaluated. The primary endpoint is the pathological remission rate, and the secondary endpoints are disease-free survival, overall survival, objective response rate, major pathological response rate and R0 resection rate. ETHICS AND DISSEMINATION Written informed consent will be required from all patients enrolled, and it will be provided by them. The study protocol received approval from the independent ethical review committee of Guangxi Medical University Cancer Hospital, Wuming Hospital of Guangxi Medical University and Wuzhou Red Cross Hospital, Wuzhou Gongren Hospital (approval number: CS2021(96)). We will submit the finalised paper for publication on completing the analyses. This study will provide valuable insights to clinicians regarding the safety and efficacy of incorporating fruquintinib into SOX as neoadjuvant treatment for locally advanced GC/GEJ. The findings have the potential to inform future research proposals and may guide the use of fruquintinib in the neoadjuvant setting for locally advanced GC/GEJ. TRIAL REGISTRATION NUMBER NCT05122091.
Collapse
Affiliation(s)
- Liucheng Wu
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Haiqing Yan
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yuzhou Qin
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Mingwei Huang
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Tingan Wang
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Qinwen Jin
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Weiyuan Wei
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| |
Collapse
|
12
|
Lin JX, Tang YH, Zheng HL, Ye K, Cai JC, Cai LS, Lin W, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Zheng CH, Li P, Huang CM. Neoadjuvant camrelizumab and apatinib combined with chemotherapy versus chemotherapy alone for locally advanced gastric cancer: a multicenter randomized phase 2 trial. Nat Commun 2024; 15:41. [PMID: 38167806 PMCID: PMC10762218 DOI: 10.1038/s41467-023-44309-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Prospective evidence regarding the combination of programmed cell death (PD)-1 and angiogenesis inhibitors in treating locally advanced gastric cancer (LAGC) is limited. In this multicenter, randomized, phase 2 trial (NCT04195828), patients with gastric adenocarcinoma (clinical T2-4N + M0) were randomly assigned (1:1) to receive neoadjuvant camrelizumab and apatinib combined with nab-paclitaxel plus S-1 (CA-SAP) or chemotherapy SAP alone (SAP) for 3 cycles. The primary endpoint was the major pathological response (MPR), defined as <10% residual tumor cells in resection specimens. Secondary endpoints included R0 resection rate, radiologic response, safety, overall survival, and progression-free survival. The modified intention-to-treat population was analyzed (CA-SAP [n = 51] versus SAP [n = 53]). The trial has met pre-specified endpoints. CA-SAP was associated with a significantly higher MPR rate (33.3%) than SAP (17.0%, P = 0.044). The CA-SAP group had a significantly higher objective response rate (66.0% versus 43.4%, P = 0.017) and R0 resection rate (94.1% versus 81.1%, P = 0.042) than the SAP group. Nonsurgical grade 3-4 adverse events were observed in 17 patients (33.3%) in the CA-SAP group and 14 (26.4%) in the SAP group. Survival results were not reported due to immature data. Camrelizumab and apatinib combined with chemotherapy as a neoadjuvant regimen was tolerable and associated with favorable responses for LAGC.
Collapse
Affiliation(s)
- Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Yi-Hui Tang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Kai Ye
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jian-Chun Cai
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Li-Sheng Cai
- Department of General Surgery, Zhangzhou Municipal Hospital of Fujian Province, Zhangzhou, China
| | - Wei Lin
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Putian University, Putian, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
13
|
Ling Q, Huang ST, Yu TH, Liu HL, Zhao LY, Chen XL, Liu K, Chen XZ, Yang K, Hu JK, Zhang WH. Optimal timing of surgery for gastric cancer after neoadjuvant chemotherapy: a systematic review and meta-analysis. World J Surg Oncol 2023; 21:377. [PMID: 38037067 PMCID: PMC10690980 DOI: 10.1186/s12957-023-03251-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Following neoadjuvant chemotherapy, surgical resection is one of the most preferred treatment options for locally advanced gastric cancer patients. However, the optimal time interval between chemotherapy and surgery is unclear. This review aimed to identify the optimal time interval between neoadjuvant chemotherapy and surgery for advanced gastric cancer. METHODS Beginning on November 12, 2022, we searched the PubMed, Cochrane Library, Web of Science databases, and Embase.com databases for relevant English-language research. Two authors independently screened the studies, assessed their quality, extracted the data, and analyzed the results. The primary goal was to investigate the relationship between the time interval to surgery (TTS) and long-term survival outcomes for patients. This study has been registered with PROSPERO (CRD42022365196). RESULTS After an initial search of 4880 articles, the meta-analysis review ultimately included only five retrospective studies. Ultimately, this meta-analysis included 1171 patients, of which 411 patients had TTS of < 4 weeks, 507 patients had TTS of 4-6 weeks, and 253 patients had TTS of > 6 weeks. In survival analysis, patients with TTS of > 6 weeks had poorer overall survival outcomes than patients with TTS of 4-6 weeks (HR = 1.34, 95% CI: 1.03-1.75, P = 0.03). No significant differences were found in terms of disease-free survival the groups. CONCLUSION Based on the current clinical evidence, patients with locally advanced gastric cancer may benefit better with a TTS of 4-6 weeks; however, this option still needs additional study.
Collapse
Grants
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 21PJ043 1. Medical Science and Technique Project of Health Commission of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 23NSFSC1611 2. Natural Science Foundation of Sichuan Province
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
- No. 2020HXBH162 3. Post-Doctor Research Project, West China Hospital, Sichuan University
Collapse
Affiliation(s)
- Qi Ling
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shi-Ting Huang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tian-Hang Yu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Han-Lin Liu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Lin-Yong Zhao
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiao-Long Chen
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Liu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xin-Zu Chen
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Yang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jian-Kun Hu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China.
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Wei-Han Zhang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China.
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Liu J, Li W, Zhang K, Huang J, Zhang X, Lei Y, Liu J, Sun J, Yang G, Zhang H. Low-dose apatinib in subjects with renal impairment: A pharmacokinetics study. Eur J Pharm Sci 2023; 190:106536. [PMID: 37490973 DOI: 10.1016/j.ejps.2023.106536] [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: 04/22/2023] [Revised: 07/12/2023] [Accepted: 07/21/2023] [Indexed: 07/27/2023]
Abstract
OBJECTIVE In patients with renal impairment, we studied apatinib and its major metabolites (M1-1, M1-2, M1-6, and M9-2) for pharmacokinetics. METHODS Subjects with different renal functions were given a single oral dose of apatinib mesylate tablets of 250 mg. Pharmacokinetic samples were collected at 1 hour before dosing,0.25, 0.5, 1, 2, 3, 4, 6, 8, 24, 48, 72, and 96 h after dosing. The pharmacokinetic parameters of apatinib and its major metabolites were calculated by noncompartmental analysis. RESULTS Comparing PK parameters of the mild or moderate renal impairment group with the healthy group: the geometric mean ratios of maximum observed drug concentration (Cmax), the area under the plasma drug concentration-time curve from time 0 to the final quantifiable time (AUC0-t), and the area under the plasma concentration-time curve from time 0 extrapolated to infinity (AUC0-inf) were all about one. No significant effect of mild and moderate renal impairment on apatinib pharmacokinetics was observed. Mild and moderate renal impairment was also not observed to have a significant effect on the pharmacokinetics of metabolites M1-1, M1-2, and M1-6. However, mild and moderate renal impairment had a certain increase in exposure to the metabolite M9-2. Considering that M9-2 has no inhibitory effect on protein tyrosine kinase, it has no clinical significance. In addition, the proportion of cumulative excretion of apatinib and its major metabolites was small and almost negligible in all three groups of subjects. CONCLUSION Patients with mild and moderate renal impairment do not need to adjust the dose of apatinib when using low dose (250 mg) apatinib.
Collapse
Affiliation(s)
- Jishi Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University No. 138, Tongzipo Road, Changsha, 410013, Hunan Province, China
| | - Wei Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University No. 138, Tongzipo Road, Changsha, 410013, Hunan Province, China
| | - Kaiqian Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University No. 138, Tongzipo Road, Changsha, 410013, Hunan Province, China
| | - Jie Huang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, PR China
| | - Xingfei Zhang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, PR China
| | - Yumeng Lei
- Department of Nephrology, The Third Xiangya Hospital, Central South University No. 138, Tongzipo Road, Changsha, 410013, Hunan Province, China
| | - Jun Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University No. 138, Tongzipo Road, Changsha, 410013, Hunan Province, China
| | - Jian Sun
- Department of Nephrology, The Third Xiangya Hospital, Central South University No. 138, Tongzipo Road, Changsha, 410013, Hunan Province, China
| | - Guoping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, PR China; Research Center of Drug Clinical Evaluation of Central South University, Changsha, 410013, Hunan, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China.
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University No. 138, Tongzipo Road, Changsha, 410013, Hunan Province, China.
| |
Collapse
|
15
|
Xiong H, Li Y. Neoadjuvant PD-1 inhibitor plus apatinib and chemotherapy versus apatinib plus chemotherapy versus chemotherapy alone in patients with locally advanced gastric cancer. Am J Cancer Res 2023; 13:3559-3570. [PMID: 37693166 PMCID: PMC10492097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/28/2023] [Indexed: 09/12/2023] Open
Abstract
Programed cell death protein-1 (PD-1) inhibitor, apatinib, and chemotherapy show synergistic antitumor effect in gastric cancer. This study aimed to evaluate this combination as a neoadjuvant therapy in locally advanced gastric cancer (LAGC). In this retrospective study, data from 179 LAGC patients who underwent neoadjuvant therapy with a PD-1 inhibitor plus apatinib and chemotherapy (PAC group, n=56), apatinib and chemotherapy (AC group, n=50), or chemotherapy alone (C group, n=73) were analyzed. The PAC group displayed a numerically higher radiologic objective response rate than the AC group (73.2% vs. 60.0%, P=0.149) and significantly higher than the C group (73.2% vs. 35.6%, P<0.001). Tumor resection rates between the PAC and AC groups were not significantly different (100.0% vs. 94.0%, P=0.102) but were higher in the PAC group compared to the C group (100.0% vs. 89.0%, P=0.010). Pathological evaluations revealed comparable R0 resection rates across all groups (P=0.873) and a non-significantly higher pathological complete response rate in the PAC group compared to the AC group (26.8% vs. 17.0%, P=0.236), while significantly higher than the C group (26.8% vs. 7.7%, P=0.005). Moreover, the PAC group exhibited a longer progression-free survival compared to the AC (P=0.036) and C (P<0.001) groups, an extended disease-free survival compared to the C group (P=0.002), and improved overall survival compared to the AC (P=0.028) and C (P=0.002) groups. Adverse events were generally comparable, with the highest incidence of peripheral neuropathy observed in the PAC group (26.8%, P=0.020). PD-1 inhibitor plus apatinib and chemotherapy may represent an effective neoadjuvant regimen for LAGC management, necessitating further validation.
Collapse
|
16
|
Xu D, Luo Y, Wang P, Li J, Ma L, Huang J, Zhang H, Yang X, Li L, Zheng Y, Fang G, Yan P. Clinical progress of anti-angiogenic targeted therapy and combination therapy for gastric cancer. Front Oncol 2023; 13:1148131. [PMID: 37384288 PMCID: PMC10295723 DOI: 10.3389/fonc.2023.1148131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/12/2023] [Indexed: 06/30/2023] Open
Abstract
The incidence of gastric cancer is increasing year by year. Most gastric cancers are already in the advanced stage with poor prognosis when diagnosed, which means the current treatment is not satisfactory. Angiogenesis is an important link in the occurrence and development of tumors, and there are multiple anti-angiogenesis targeted therapies. To comprehensively evaluate the efficacy and safety of anti-angiogenic targeted drugs alone and in combination against gastric cancer, we systematically searched and sorted out relevant literature. In this review, we summarized the efficacy and safety of Ramucirumab, Bevacizumab, Apatinib, Fruquintinib, Sorafenib, Sunitinib, Pazopanib on gastric cancer when used alone or in combination based on prospective clinical trials reported in the literature, and sorted response biomarkers. We also summarized the challenges faced by anti-angiogenesis therapy for gastric cancer and available solutions. Finally, the characteristics of the current clinical research are summarized and suggestions and prospects are raised. This review will serve as a good reference for the clinical research of anti-angiogenic targeted drugs in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Donghan Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Yehao Luo
- School of Second Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jiaxin Li
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Linrui Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jie Huang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Hao Zhang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Xiaoman Yang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Liqi Li
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Yuhong Zheng
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Gang Fang
- Guangxi Key Laboratory of Applied Fundamental Research of Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Peiyu Yan
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, Macau University of Science and Technology, Macao, Macao SAR, China
| |
Collapse
|
17
|
Deng YY, Jiang DY, Zhu PF, Lu H, Liu Q, Zhang X, Pan SY, Chen ZL, Yang L. Apatinib combined with SOX regimen for conversion therapy in advanced gastric cancer patients: a retrospective cohort study. World J Surg Oncol 2023; 21:129. [PMID: 37041581 PMCID: PMC10088230 DOI: 10.1186/s12957-023-02973-3] [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: 10/20/2022] [Accepted: 02/27/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Recently, many studies have shown that the progress of conversion therapy can provide surgical opportunities for patients with advanced gastric cancer (GC) and bring survival benefits. However, the results of the current study show that the regimen used in conversion therapy is still controversial. Apatinib, as the standard third-line treatment for GC, has an inconclusive status in conversion therapy. METHODS This study retrospectively analyzed GC patients admitted to Zhejiang Provincial People's Hospital from June 2016 to November 2019. All patients were pathologically diagnosed, had unresectable factors, and received SOX regimen with or without apatinib as conversion therapy. RESULTS A total of 50 patients were enrolled in the study. Altogether 33 patients (66%) received conversion surgery and 17 patients (34%) received conversion therapy without surgery. The median progression-free survival (PFS) between surgery group and non-surgery group were 21.0 versus 4.0 months (p < 0.0001), and the median overall survival (OS) were 29.0 versus 14.0 months (p < 0.0001). In conversion surgery group, 16 patients (16/33) were treated with SOX plus apatinib, and the R0 resection rate was 81.3%; 17 patients (17/33) were treated with SOX regimen along, and the R0 resection rate was 41.2% (p = 0.032). The PFS in the SOX combined with apatinib group was significantly longer than that of SOX group (25.5 versus 16 months, p = 0.045), and the median OS were 34.0 versus 23.0 months (p = 0.048). The addition of apatinib did not increase the incidence of serious adverse reactions throughout the preoperative therapy period. CONCLUSIONS Patients with advanced inoperable gastric cancer could benefit probably from conversion chemotherapy and subsequence conversion surgery. Apatinib-targeted therapy combined with SOX chemotherapy may be a safe and feasible option for conversion therapy.
Collapse
Affiliation(s)
- Ya-Ya Deng
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
- The Qingdao University Medical College, Qingdao, Shandong Province, 260075, China
| | - Ding-Yi Jiang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
- The Qingdao University Medical College, Qingdao, Shandong Province, 260075, China
| | - Peng-Fei Zhu
- The Qingdao University Medical College, Qingdao, Shandong Province, 260075, China
- Graduate School of Clinical Medicine, Bengbu Medical College, Bengbu, 233000, Anhui Province, China
| | - Hongrui Lu
- The Qingdao University Medical College, Qingdao, Shandong Province, 260075, China
- Graduate School of Clinical Medicine, Bengbu Medical College, Bengbu, 233000, Anhui Province, China
| | - Qian Liu
- The Qingdao University Medical College, Qingdao, Shandong Province, 260075, China
| | - Xinyue Zhang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
- The Qingdao University Medical College, Qingdao, Shandong Province, 260075, China
| | - Shuang-Yue Pan
- The Qingdao University Medical College, Qingdao, Shandong Province, 260075, China
- Graduate School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310014, Zhejiang Province, China
| | - Zhe-Ling Chen
- The Qingdao University Medical College, Qingdao, Shandong Province, 260075, China.
| | - Liu Yang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China.
- The Qingdao University Medical College, Qingdao, Shandong Province, 260075, China.
| |
Collapse
|
18
|
Lin JX, Lin JP, Wang ZK, Li P, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Tu RH, Lin GT, Huang ZN, Lin JL, Zheng HL, Lin GS, Huang CM, Zheng CH. Assessment of Laparoscopic Spleen-Preserving Hilar Lymphadenectomy for Advanced Proximal Gastric Cancer Without Invasion Into the Greater Curvature: A Randomized Clinical Trial. JAMA Surg 2023; 158:10-18. [PMID: 36383362 PMCID: PMC9857675 DOI: 10.1001/jamasurg.2022.5307] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022]
Abstract
IMPORTANCE The survival benefit of laparoscopic total gastrectomy combined with spleen-preserving splenic hilar lymphadenectomy (LSTG) for locally advanced proximal gastric cancer (APGC) without invasion into the greater curvature remains uncertain. OBJECTIVE To compare the long-term and short-term efficacy of LSTG (D2 + No. 10 group) and conventional laparoscopic total gastrectomy (D2 group) for patients with APGC that has not invaded the greater curvature. DESIGN, SETTING, AND PARTICIPANTS In this open-label, prospective randomized clinical trial, a total of 536 patients with clinical stage cT2 to 4a/N0 to 3/M0 APGC without invasion into the greater curvature were enrolled from January 2015 to October 2018. The final follow-up was on October 31, 2021. Data were analyzed from December 2021 to February 2022. INTERVENTIONS Eligible patients were randomized to the D2 + No. 10 group or the D2 group. MAIN OUTCOMES AND MEASURES The primary outcome was 3-year disease-free survival (DFS). The secondary outcomes were 3-year overall survival (OS) and morbidity and mortality within 30 days after surgery. RESULTS Of 526 included patients, 392 (74.5%) were men, and the mean (SD) age was 60.6 (9.6) years. A total of 263 patients were included in the D2 + No. 10 group, and 263 were included in the D2 group. The 3-year DFS was 70.3% (95% CI, 64.8-75.8) for the D2 + No. 10 group and 64.3% (95% CI, 58.4-70.2; P = .11) for the D2 group, and the 3-year OS in the D2 + No. 10 group was better than that in the D2 group (75.7% [95% CI, 70.6-80.8] vs 66.5% [95% CI, 60.8-72.2]; P = .02). Multivariate analysis revealed that splenic hilar lymphadenectomy was not an independent protective factor for DFS (hazard ratio [HR], 0.86; 95% CI, 0.63-1.16) or OS (HR, 0.81; 95% CI, 0.59-1.12). Stratification analysis showed that patients with advanced posterior gastric cancer in the D2 + No. 10 group had better 3-year DFS (92.9% vs 39.3%; P < .001) and OS (92.9% vs 42.9%; P < .001) than those in the D2 group. Multivariate analysis confirmed that patients with advanced posterior gastric cancer could have the survival benefit from No. 10 lymph node dissection (DFS: HR, 0.10; 95% CI, 0.02-0.46; OS: HR, 0.12; 95% CI, 0.03-0.52). CONCLUSIONS AND RELEVANCE Although LSTG could not significantly improve the 3-year DFS of patients with APGC without invasion into the greater curvature, patients with APGC located posterior gastric wall may benefit from LSTG. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02333721.
Collapse
Affiliation(s)
- Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jun-Peng Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Zu-Kai Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ju-Li Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Guo-Sheng Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, 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
|
19
|
Shannon AH, Ruff SM, Pawlik TM. Expert Insights on Current Treatments for Hepatocellular Carcinoma: Clinical and Molecular Approaches and Bottlenecks to Progress. J Hepatocell Carcinoma 2022; 9:1247-1261. [PMID: 36514693 PMCID: PMC9741819 DOI: 10.2147/jhc.s383922] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary liver tumor that typically occurs in the setting of chronic liver disease/cirrhosis. Treatment modalities for HCC have evolved and given the variety of treatment options, a multi-disciplinary approach requiring input from surgical, medical, and radiation oncology, hepatology, and interventional radiology is necessary. Multiple advances have been made over the last decade regarding treatment of HCC, especially advanced disease. Resection and transplantation remain as cornerstone curative-intent treatment options. For patients who are not candidates for curative-intent therapy, exciting progress has been made in molecular and cellular approaches to systemic therapy for HCC including immunotherapies and tyrosine kinase inhibitors. Although the prognosis for advanced HCC remains poor, the armamentarium of therapies has increased, and valuable years of life can be gained with these therapies. While the main therapeutic modality for early-stage disease remains resection, multimodal immunotherapy has emerged as first-line treatment for advanced disease. We herein review different clinical and molecular treatment modalities related to the treatment of HCC, as well as provide insights into future directions for HCC treatment. We highlight how research and progress are needed to move into a new era of molecular and cellular treatments.
Collapse
Affiliation(s)
- Alexander H Shannon
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Samantha M Ruff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA,Correspondence: Timothy M Pawlik, Department of Surgery, The Urban Meyer III and Shelley Meyer Chair for Cancer Research, Professor of Surgery, Oncology, Health Services Management and Policy, The Ohio State University, Wexner Medical Center, 395 W. 12th Ave., Suite 670, Columbus, OH, USA, Tel +1 614 293 8701, Fax +1 614 293 4063, Email
| |
Collapse
|
20
|
Pan L, Tian Y, Wang K, Tang J, Liu J, Zhang J, Wang M, Liu J, Xu H, Chen X. Low-dose apatinib combined with camrelizumab and the SOX regimen in the neoadjuvant treatment of locally advanced gastric/gastroesophageal junction adenocarcinoma (SPACE-neo): a protocol for an open-label, single-arm, clinical trial. J Gastrointest Oncol 2022; 13:3300-3313. [PMID: 36636043 PMCID: PMC9830353 DOI: 10.21037/jgo-22-1158] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022] Open
Abstract
Background Neoadjuvant chemotherapy with S-1 plus oxaliplatin (SOX regimen) has shown promising results in pathological response rate and survival rate in patients with locally advanced resectable gastric cancer (LAGC). We previously carried out the SPACE study to assess efficacy and safety of low-dose apatinib combined with camrelizumab and the SOX regimen as a first-line treatment of advanced gastric/gastroesophageal junction adenocarcinoma (AGC/GEJC). The preliminary results demonstrated a high objective response rate. However, the SPACE study was conducted in patients with AGC, but the efficacy of LAGC patients is not yet known. The SPACE-neo study is designed to investigate whether this combination could improve outcomes in patients with locally advanced gastric/gastroesophageal junction cancer (LAGC/GEJC) as neoadjuvant therapy. Methods SPACE-neo is a prospective, open-label, single-arm study conducted in China at the First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital). Thirty-two patients with human epidermal growth factor receptor 2 (HER2)-negative or HER2-unknown LAGC/GEJC confirmed by histopathology or cytology will be recruited. Included patients shall be clinically staged as M0 and either T3 to T4 or N+ assessed by ultrasound endoscopy and thoracoabdominal-enhanced computed tomography or magnetic resonance imaging. The patients will receive three cycles of this combined regimen as a neoadjuvant treatment. Each patient will receive screening visits within 2 weeks before the first cycle and planned visits before every cycle of treatment. Key monitoring data include imaging data, pathological findings, and adverse events associated with neoadjuvant and surgical treatment. The primary endpoints are major pathological response (MPR) and safety. MPR is the proportion of patients whose residual tumor cells make up less than 10% of the primary tumor from among the total cohort. Clopper-Pearson method will be used to estimate the 95% confidence interval of MPR and safety data will be reported as descriptive statistical analysis. Discussion The SPACE-neo trial aims to evaluate the safety and preliminary efficacy of this regimen in the neoadjuvant treatment of LAGC/GEJC. It is hoped that the study can achieve a higher pathological response rate and longer survival rate. Trial Registration ChiCTR.gov.cn: ChiCTR2100049305.
Collapse
Affiliation(s)
- Lanlan Pan
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Yitong Tian
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Kangxin Wang
- Department of Oncology, Pukou Branch Hospital of Jiangsu Province Hospital (Nanjing Pukou Central Hospital), Nanjing, China
| | - Jie Tang
- Department of Oncology, Liyang People’s Hospital, Liyang, China
| | - Jin Liu
- Clinical Medicine Research Institution, the First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Jiaguang Zhang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Min Wang
- Digestive Endoscopy Department and General Surgery Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, China
| | - Jie Liu
- Pharmacy Department, Pukou Branch Hospital of Jiangsu Province Hospital (Nanjing Pukou Central Hospital), Nanjing, China
| | - Hao Xu
- Division of Gastric Surgery, Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Xiaofeng Chen
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China;,Department of Oncology, Pukou Branch Hospital of Jiangsu Province Hospital (Nanjing Pukou Central Hospital), Nanjing, China;,Department of Oncology, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
21
|
Wang T, Li C, Li X, Zhai J, Wang S, Shen L. The optimal neoadjuvant chemotherapy regimen for locally advanced gastric and gastroesophageal junction adenocarcinoma: a systematic review and Bayesian network meta-analysis. Eur J Med Res 2022; 27:239. [DOI: 10.1186/s40001-022-00878-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022] Open
Abstract
Abstract
Background
Neoadjuvant chemotherapy (NAC) for locally advanced gastric and gastroesophageal junction adenocarcinoma (LAGC) has been recommended in several guidelines. However, there is no global consensus about the optimum of NAC regimens. We aimed to determine the optimal NAC regimen for LAGC.
Methods
A systematic review and Bayesian network meta-analysis was performed. The literature search was conducted from inception to June 2022. The odds ratio (OR) value and 95% confidence interval (95% CI) were used for assessment of R0 resection rate and pathological complete response rate (pCR) as primary outcomes. The hazard ratio (HR) value and 95% CI were interpreted for the assessment of overall survival (OS) and disease-free survival (DFS) as second outcomes. The risk ratio (RR) value and 95% CI were used for safety assessment.
Results
Twelve randomized controlled trials were identified with 3846 eligible participants. The network plots for R0 resectability, OS, and DFS constituted closed loops. The regimens of TPF (taxane and platinum plus fluoropyrimidine), ECF (epirubicin and cisplatin plus fluorouracil), and PF (platinum plus fluoropyrimidine) showed a meaningful improvement of R0 resectability, as well as OS and/or DFS, compared with surgery (including surgery-alone and surgery plus postoperative adjuvant chemotherapy). Importantly, among these regimens, TPF regimen showed significant superiority in R0 resection rate (versus ECF regimen), OS (versus ECF regimen), DFS (versus PF and ECF regimens), and pCR (versus PF regimen).
Conclusions
The taxane-based triplet regimen of TPF is likely the optimal neoadjuvant chemotherapy regimen for LAGC patients.
Collapse
|
22
|
Ma Y, Wang B, Maswikiti EP, Wang X, Wang N, Chen H. Pathological complete remission of a locally advanced gastric cancer by neoadjuvant therapy "sandwich" regimen as SOXAP+ fluorescence laparoscopic surgery +SOXAP: Case report. Front Pharmacol 2022; 13:1008755. [PMID: 36408251 PMCID: PMC9666721 DOI: 10.3389/fphar.2022.1008755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/18/2022] [Indexed: 12/23/2022] Open
Abstract
Gastric cancer is an extremely burdensome and challenging malignant tumor with a high incidence and a high mortality rate, which seriously results in a thorny prognosis for oncology patients. Surgical treatment combined with postoperative adjuvant therapy are currently the most regular methods for the treatment of locally advanced gastric cancer (LAGC), but long-term efficacy is not an ideal outcome. Therefore, herein we report a case of a pathologically confirmed complete remission of LAGC treated by the administration of neoadjuvant therapy combined with fluorescence laparoscopic surgery with more significant long-term survival. With that being mentioned, a 60-year-old man was diagnosed as moderately differentiated gastric antrum adenocarcinoma (T3N1M0). Moreover, after three cycles of SOXAP regimen (Oxaliplatin + S-1+Apatinib + Camrelizumab), and it was found out that the gastric lesion was smaller in size than before, total laparoscopic radical resection of the distal gastric cancer was performed at the time. Furthermore, no tumor cells were seen in gross specimen post operatively, achieving complete remission of the case. In addition, he also underwent three cycles of SOXAP regimen postoperatively. Interestingly and assuredly, he was in good health after an almost 2-year follow up period. These results suggest that this therapeutic regimen is a promising treatment modality for the management of locally advanced gastric cancers.
Collapse
Affiliation(s)
- Yanling Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Bofang Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | | | - Xueyan Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Na Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Hao Chen
- Department of Cancer Center, Lanzhou University Second Hospital, Lanzhou, China,*Correspondence: Hao Chen,
| |
Collapse
|
23
|
Zeng Q, Zhu Y, Li L, Feng Z, Shu X, Wu A, Luo L, Cao Y, Tu Y, Xiong J, Zhou F, Li Z. CT-based radiomic nomogram for preoperative prediction of DNA mismatch repair deficiency in gastric cancer. Front Oncol 2022; 12:883109. [PMID: 36185292 PMCID: PMC9523515 DOI: 10.3389/fonc.2022.883109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundDNA mismatch repair (MMR) deficiency has attracted considerable attention as a predictor of the immunotherapy efficacy of solid tumors, including gastric cancer. We aimed to develop and validate a computed tomography (CT)-based radiomic nomogram for the preoperative prediction of MMR deficiency in gastric cancer (GC).MethodsIn this retrospective analysis, 225 and 91 GC patients from two distinct hospital cohorts were included. Cohort 1 was randomly divided into a training cohort (n = 176) and an internal validation cohort (n = 76), whereas cohort 2 was considered an external validation cohort. Based on repeatable radiomic features, a radiomic signature was constructed using the least absolute shrinkage and selection operator (LASSO) regression analysis. We employed multivariable logistic regression analysis to build a radiomics-based model based on radiomic features and preoperative clinical characteristics. Furthermore, this prediction model was presented as a radiomic nomogram, which was evaluated in the training, internal validation, and external validation cohorts.ResultsThe radiomic signature composed of 15 robust features showed a significant association with MMR protein status in the training, internal validation, and external validation cohorts (both P-values <0.001). A radiomic nomogram incorporating a radiomic signature and two clinical characteristics (age and CT-reported N stage) represented good discrimination in the training cohort with an AUC of 0.902 (95% CI: 0.853–0.951), in the internal validation cohort with an AUC of 0.972 (95% CI: 0.945–1.000) and in the external validation cohort with an AUC of 0.891 (95% CI: 0.825–0.958).ConclusionThe CT-based radiomic nomogram showed good performance for preoperative prediction of MMR protein status in GC. Furthermore, this model was a noninvasive tool to predict MMR protein status and guide neoadjuvant therapy.
Collapse
Affiliation(s)
- Qingwen Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanyan Zhu
- Department of Radiology, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Leyan Li
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zongfeng Feng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xufeng Shu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Ahao Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Lianghua Luo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Yi Cao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
- *Correspondence: Zhengrong Li, ; Yi Cao,
| | - Yi Tu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianbo Xiong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Fuqing Zhou
- Department of Radiology, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Zhengrong Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
- Institute of Digestive Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Zhengrong Li, ; Yi Cao,
| |
Collapse
|
24
|
Zhou X, Lin J, Wang F, Chen X, Zhang Y, Hu Z, Jin X. Circular RNA-regulated autophagy is involved in cancer progression. Front Cell Dev Biol 2022; 10:961983. [PMID: 36187468 PMCID: PMC9515439 DOI: 10.3389/fcell.2022.961983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/03/2022] [Indexed: 12/05/2022] Open
Abstract
Circular RNAs (circRNAs) are a sort of long, non-coding RNA molecules with a covalently closed continuous ring structure without 5'-3' polarity and poly-A tail. The modulative role of circRNAs in malignant diseases has been elucidated by many studies in recent years via bioinformatics and high-throughput sequencing technologies. Generally, circRNA affects the proliferative, invasive, and migrative capacity of malignant cells via various mechanisms, exhibiting great potential as novel biomarkers in the diagnoses or treatments of malignancies. Meanwhile, autophagy preserves cellular homeostasis, serving as a vital molecular process in tumor progression. Mounting studies have demonstrated that autophagy can not only contribute to cancer cell survival but can also induce autophagic cell death in specific conditions. A growing number of research studies have indicated that there existed abundant associations between circRNAs and autophagy. Herein, we systemically reviewed and discussed recent studies on this topic in different malignancies and concluded that the circRNA–autophagy axis played crucial roles in the proliferation, metastasis, invasion, and drug or radiation resistance of different tumor cells.
Collapse
|
25
|
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: 9] [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
|
26
|
Wang Z, He T, Yu D, Qin X, Geng A, Yang H. Neoadjuvant apatinib plus tegafur/gimeracil/oteracil potassium (S‑1)/oxaliplatin chemotherapy vs. chemotherapy alone in patients with locally advanced gastric carcinoma. Exp Ther Med 2022; 24:625. [PMID: 36160880 PMCID: PMC9468841 DOI: 10.3892/etm.2022.11562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/20/2022] [Indexed: 02/03/2023] Open
Abstract
The current study aimed to evaluate the efficacy and safety of neoadjuvant apatinib plus tegafur/gimeracil/oteracil potassium (S-1) plus oxaliplatin (SOX) chemotherapy in patients with locally advanced gastric carcinoma (LAGC). Therefore, patients with LAGC treated with neoadjuvant apatinib plus SOX chemotherapy (apatinib + SOX group; n=25) or SOX chemotherapy (SOX group; n=35) were enrolled in the present study. Subsequently, the objective response (ORR) and disease control rates (DCR), pathological response, disease-free survival (DFS), overall survival (OS) and adverse events were recorded. The results showed that patients in the apatinib + SOX group exhibited a higher ORR (64.0 vs. 37.1%; P=0.040), but a similar DCR (96.0 vs. 88.6%; P=0.580), compared with those in the SOX group. The pathological response rates in patients with grade 0, 1, 2 and 3 LAGC were 0.0, 20.8, 62.5 and 16.7%, respectively, in the apatinib + SOX group, while in those treated with SOX alone they were 9.1, 39.4, 42.4 and 9.1%, respectively. By contrast, the pathological response was elevated in the apatinib + SOX group compared with the SOX group (P=0.030). During a median follow-up period of 21.0 months (range, 6.4-38.1 months), median DFS and OS were not reached. More specifically, the 1-, 2- and 3-year DFS rates were 91.7, 75.2 and 75.2% in the apatinib + SOX group and 71.8, 59.6 and 44.7% in the SOX group, respectively. In addition, the 1-, 2- and 3-year OS rates were 100.0, 89.6 and 78.4% in the apatinib + SOX group, while those in the SOX group were 90.3, 69.2 and 55.4%, respectively. However, no differences in DFS (P=0.094) or OS (P=0.155) were observed between the two groups. Additionally, the most common adverse events in the SOX group were mild leukopenia (42.9%) and fatigue (34.3%), while those in the apatinib + SOX group were tolerable leukopenia (44.0%) and hypertension (44.0%). In conclusion, the present study suggested that neoadjuvant apatinib plus SOX chemotherapy could be more effective and tolerable compared with SOX chemotherapy alone in patients with LAGC.
Collapse
Affiliation(s)
- Zhenfeng Wang
- Department of General Surgery, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| | - Tingbang He
- Department of General Surgery, The People's Hospital of Xiajin Affiliated to Shandong First Medical University, Xiajin, Dezhou, Shandong 253299, P.R. China
| | - Deguo Yu
- Department of Emergency Surgery, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| | - Xiantao Qin
- Department of General Surgery, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| | - Aizhi Geng
- Department of Gynecology, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| | - Hailei Yang
- Department of Gynecology, The Second People's Hospital of Liaocheng, Linqing, Liaocheng, Shandong 252699, P.R. China
| |
Collapse
|
27
|
Apatinib plus chemotherapy versus chemotherapy alone as neoadjuvant therapy in locally advanced gastric carcinoma patients: a prospective, cohort study. Ir J Med Sci 2022:10.1007/s11845-022-03075-x. [PMID: 35819743 DOI: 10.1007/s11845-022-03075-x] [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: 05/24/2022] [Accepted: 06/15/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Apatinib, a small molecule targeting VEGFR2, is commonly used for advanced gastric cancer treatment. This prospective cohort study further investigated the efficacy and safety of neoadjuvant apatinib plus chemotherapy in locally advanced gastric carcinoma patients. METHODS Ninety-six locally advanced gastric carcinoma patients were divided into the apatinib plus chemotherapy group (N = 45) and chemotherapy group (N = 51) according to their chosen treatment. Apatinib was administered (375 mg/day), and S-1 plus oxaliplatin (SOX) or oxaliplatin plus capecitabine (CapOx) was given as chemotherapy, for 3 cycles with 3 weeks a cycle before surgery. RESULTS The objective response rate (62.2% vs. 37.3%, P = 0.015) and pathological response grade (P = 0.011) were better; meanwhile, the tumor-resection rate (95.6% vs. 84.3%, P = 0.143) and pathological complete response rate (23.3% vs. 9.3%, P = 0.080) exhibited increasing trends (without statistical significance) in the apatinib plus chemotherapy group compared with the chemotherapy group. Additionally, the apatinib plus chemotherapy group achieved prolonged disease-free survival (DFS) (P = 0.019) and overall survival (OS) (P = 0.047) compared with the chemotherapy group. After adjusted by multivariate Cox's regression analysis, neoadjuvant apatinib plus chemotherapy was still superior to chemotherapy regarding DFS (hazard ratio (HR): 0.277, P = 0.014) and OS (HR: 0.316, P = 0.038). Notably, the incidences of adverse events between the two groups were not different (P > 0.050). Moreover, the most common adverse events of neoadjuvant apatinib plus chemotherapy were leukopenia (42.2%), fatigue (37.8%), hypertension (37.8%), and anemia (31.1%). CONCLUSION Neoadjuvant apatinib plus chemotherapy realizes better clinical response, pathological response, survival profile, and non-inferior safety profile compared to chemotherapy in locally advanced gastric carcinoma.
Collapse
|
28
|
Li H, Huang H, Zhang T, Feng H, Wang S, Zhang Y, Ji X, Cheng X, Zhao R. Apatinib: A Novel Antiangiogenic Drug in Monotherapy or Combination Immunotherapy for Digestive System Malignancies. Front Immunol 2022; 13:937307. [PMID: 35844616 PMCID: PMC9276937 DOI: 10.3389/fimmu.2022.937307] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/30/2022] [Indexed: 02/03/2023] Open
Abstract
Digestive system malignancies are one of the primary causes of cancer-related death. Meanwhile, angiogenesis has been proved to play an important role in the process of cancer neovascularization. Apatinib, a novel targeted antiangiogenic molecule, could generate highly selective competition in the vascular endothelial growth factor receptor-2, involved in tumor progression and metastasis. It has been implied as a promising cancer treatment agent that can prevent tumor cell proliferation meanwhile inhibit tumor angiogenesis. Furthermore, completed clinical trials demonstrated that apatinib could prolong the progression-free survival and overall survival in advanced gastric cancer and primary liver cancer. Recent studies revealed that apatinib had a synergistic effect with immunotherapy as a second-line and third-line treatment regimen for some other cancers. In this review, we summarize the pharmacological properties of apatinib and the latest clinical application in chemotherapy-refractory patients with advanced digestive system cancer. Based on the comparable survival results, the molecular mechanisms of apatinib are prospective to include the antiangiogenic, apoptosis-inducing, and autophagy-inducing properties in the corresponding signaling pathway. Treatment of apatinib monotherapy or combination immunotherapy remains the optimal option for patients with digestive system malignancies in the future.
Collapse
Affiliation(s)
- Haosheng Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoran Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaodong Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaqi Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaopin Ji
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiaopin Ji, ; Xi Cheng, ; Ren Zhao,
| | - Xi Cheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiaopin Ji, ; Xi Cheng, ; Ren Zhao,
| | - Ren Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiaopin Ji, ; Xi Cheng, ; Ren Zhao,
| |
Collapse
|
29
|
Zhao L, Yu Q, Gao C, Xiang J, Zheng B, Feng Y, Li R, Zhang W, Hong X, Zhan YY, Xiao L, Hu T. Studies of the Efficacy of Low-Dose Apatinib Monotherapy as Third-Line Treatment in Patients with Metastatic Colorectal Cancer and Apatinib’s Novel Anticancer Effect by Inhibiting Tumor-Derived Exosome Secretion. Cancers (Basel) 2022; 14:cancers14102492. [PMID: 35626097 PMCID: PMC9139438 DOI: 10.3390/cancers14102492] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 12/23/2022] Open
Abstract
Simple Summary We assessed the efficacy and safety of low-dose apatinib monotherapy as a third-line treatment in patients with metastatic colorectal cancer. The ORR and DCR were 4.0% (2/50) and 70% (35/50), and the median PFS and OS were 4.7 months and 10.1 months, which demonstrated comparable survival outcomes, significant improvements to the patient’s quality of life, and tolerable adverse reactions. We also disclosed a novel role of apatinib’s anticancer effect, i.e., inhibiting tumor-derived exosome release. Our results indicated that apatinib treatment inhibited exosome secretion through the regulation of MVB biogenesis, transport, and fusion by regulating LAMP2, RAB11, Snap23, and VAMP2. This novel regulatory mechanism provides a new perspective for the antitumor effect of apatinib in CRC treatment. Abstract Antiangiogenic therapy is an important treatment strategy for metastatic colorectal cancer (mCRC). We carried out a clinical study of low-dose apatinib (250 mg) monotherapy as a third-line treatment in patients with mCRC and assessed its efficacy and safety. It demonstrated that low-dose apatinib had comparable survival outcomes, significantly improved the patient quality of life, and caused tolerable adverse reactions. To further investigate the underlying mechanism of the effects of apatinib in CRC besides angiogenesis, we performed RNA-seq, and our results suggested that apatinib may have other potential antitumor mechanisms in CRC through multiple pathways, including exosomes secretion. In RKO and HCT116 cells, apatinib significantly reduced exosomes secretion by targeting multivesicular body (MVB) transport. Further studies have indicated that apatinib not only promoted the degradation of MVBs via the regulation of LAMP2 but also interfered with MVB transport by inhibiting Rab11 expression. Moreover, apatinib inhibited MVB membrane fusion by reducing SNAP23 and VAMP2 expression. In vivo, apatinib inhibited orthotopic murine colon cancer growth and metastasis and reduced the serum exosomes amount. This novel regulatory mechanism provides a new perspective for the antitumor effect of apatinib beyond angiogenesis inhibition.
Collapse
Affiliation(s)
- Lingying Zhao
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Qiang Yu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Chunyi Gao
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Jingzhou Xiang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Bowen Zheng
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Yujie Feng
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Runyang Li
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Wenqing Zhang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Xiaoting Hong
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Yan-yan Zhan
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
| | - Li Xiao
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
- Department of Oncology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
- Correspondence: (L.X.); (T.H.); Tel.: +86-592-2292012 (L.X.); +86-592-2188223 (T.H.)
| | - Tianhui Hu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, Xiamen University School of Medicine, Xiamen 361102, China; (L.Z.); (Q.Y.); (C.G.); (J.X.); (B.Z.); (Y.F.); (R.L.); (W.Z.); (X.H.); (Y.-y.Z.)
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- Correspondence: (L.X.); (T.H.); Tel.: +86-592-2292012 (L.X.); +86-592-2188223 (T.H.)
| |
Collapse
|
30
|
Zhang C, Xie Y, Lai R, Wu J, Guo Z. Nonsynonymous C1653T Mutation of Hepatitis B Virus X Gene Enhances Malignancy of Hepatocellular Carcinoma Cells. J Hepatocell Carcinoma 2022; 9:367-377. [PMID: 35535232 PMCID: PMC9078866 DOI: 10.2147/jhc.s348690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Methods Results Conclusion
Collapse
Affiliation(s)
- Cuifang Zhang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Oncology, The Pingshan County People’s Hospital, Shijiazhuang, People’s Republic of China
| | - Ying Xie
- Hebei Key Laboratory of Laboratory Animal Science, Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Ruixue Lai
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Jianhua Wu
- Animal Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Zhanjun Guo
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Correspondence: Zhanjun Guo, Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, People’s Republic of China, Tel + 86 311 8609 5734, Fax + 86 311 8609 5237, Email
| |
Collapse
|
31
|
Tang Z, Wang Y, Yu Y, Cui Y, Liang L, Xu C, Shen Z, Shen K, Wang X, Liu T, Sun Y. Neoadjuvant apatinib combined with oxaliplatin and capecitabine in patients with locally advanced adenocarcinoma of stomach or gastroesophageal junction: a single-arm, open-label, phase 2 trial. BMC Med 2022; 20:107. [PMID: 35382819 PMCID: PMC8985371 DOI: 10.1186/s12916-022-02309-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/21/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Adding anti-angiogenics to neoadjuvant chemotherapy for localized gastric cancer is recognized as a promising strategy, but its clinical value remains to be defined. METHODS This single-center, single-arm, phase 2 trial included patients with locally advanced (cT3/4aN+M0) adenocarcinoma of the stomach or gastroesophageal junction (GEJ) who received three cycles of intravenous oxaliplatin (135 mg/m2 on day 1), oral capecitabine (1000 mg/m2 twice daily on days 1 to 14), and oral apatinib for 21 days (250 mg once daily in the first two cycles, and further increased to 500 mg daily in the third cycle based on whether any adverse event of grade 3 or worse occurred), and an additional cycle of oxaliplatin plus capecitabine, followed by gastrectomy with D2 lymphadenectomy. The primary endpoint was the proportion of patients who achieved an objective response according to RECIST version 1.1. RESULTS Between April 28, 2017, and October 23, 2019, 37 patients were screened and 35 participants were included. Of the 32 patients assessable for efficacy and safety, objective responses were achieved in 25 (78.1%; 95% confidence interval [CI], 60.0% to 90.7%) patients. Thirty-one (96.9%) patients received R0 resection, two (6.3%) patients achieved pathological complete response, and 11 (34.4%) patients achieved pathological response. At the data cutoff date (September 30, 2021), the median event-free survival was 42.6 (95% CI, 16.2 to not reached) months, and the median overall survival was not reached. The most common grade 3 or 4 treatment-emergent adverse events were hypertension (9/32, 28.1%), thrombocytopenia (7/32, 21.9%), and neutropenia (5/32, 15.6%). Seven (21.9%) patients developed surgical complications, and the most common one was intra-abdominal abscess (4/32, 12.5%). CONCLUSIONS The concomitant use of apatinib, oxaliplatin, and capecitabine as neoadjuvant therapy showed promising efficacy and manageable safety profile in patients with locally advanced adenocarcinoma of the stomach or GEJ, and further phase 3 study is warranted. TRIAL REGISTRATION This study was registered with ClinicalTrial.gov ( NCT03229096 ).
Collapse
Affiliation(s)
- Zhaoqing Tang
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Liang Liang
- Department of Radiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhenbin Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Kuntang Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Xuefei Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
32
|
Rahman MA, Ahmed KR, Rahman MDH, Park MN, Kim B. Potential Therapeutic Action of Autophagy in Gastric Cancer Managements: Novel Treatment Strategies and Pharmacological Interventions. Front Pharmacol 2022; 12:813703. [PMID: 35153766 PMCID: PMC8834883 DOI: 10.3389/fphar.2021.813703] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC), second most leading cause of cancer-associated mortality globally, is the cancer of gastrointestinal tract in which malignant cells form in lining of the stomach, resulting in indigestion, pain, and stomach discomfort. Autophagy is an intracellular system in which misfolded, aggregated, and damaged proteins, as well as organelles, are degraded by the lysosomal pathway, and avoiding abnormal accumulation of huge quantities of harmful cellular constituents. However, the exact molecular mechanism of autophagy-mediated GC management has not been clearly elucidated. Here, we emphasized the role of autophagy in the modulation and development of GC transformation in addition to underlying the molecular mechanisms of autophagy-mediated regulation of GC. Accumulating evidences have revealed that targeting autophagy by small molecule activators or inhibitors has become one of the greatest auspicious approaches for GC managements. Particularly, it has been verified that phytochemicals play an important role in treatment as well as prevention of GC. However, use of combination therapies of autophagy modulators in order to overcome the drug resistance through GC treatment will provide novel opportunities to develop promising GC therapeutic approaches. In addition, investigations of the pathophysiological mechanism of GC with potential challenges are urgently needed, as well as limitations of the modulation of autophagy-mediated therapeutic strategies. Therefore, in this review, we would like to deliver an existing standard molecular treatment strategy focusing on the relationship between chemotherapeutic drugs and autophagy, which will help to improve the current treatments of GC patients.
Collapse
Affiliation(s)
- Md. Ataur Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- Department of Biotechnology and Genetic Engineering, Global Biotechnology and Biomedical Research Network (GBBRN), Faculty of Biological Sciences, Islamic University, Kushtia, Bangladesh
| | - Kazi Rejvee Ahmed
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, Bangladesh
| | - MD. Hasanur Rahman
- Department of Biotechnology and Genetic Engineering, Global Biotechnology and Biomedical Research Network (GBBRN), Faculty of Biological Sciences, Islamic University, Kushtia, Bangladesh
- ABEx Bio-Research Center, East Azampur, Bangladesh
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|