1
|
Liang XW, Liu H, Wu JC. Comment on 'Effect of Hyperthermic Intraperitoneal Chemotherapy (HIPEC) on survival and recurrence rates in advanced gastric cancer - a systematic review and meta-analysis'. Int J Surg 2023; 109:4357-4358. [PMID: 37678282 PMCID: PMC10720788 DOI: 10.1097/js9.0000000000000679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023]
Affiliation(s)
- Xian-Wen Liang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)
| | - Hui Liu
- Department of Interventional Radiology, Central South University, Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan Province, People’s Republic of China
| | - Jin-Cai Wu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)
| |
Collapse
|
2
|
Patel M, Arora A, Mukherjee D, Mukherjee S. Effect of hyperthermic intraperitoneal chemotherapy on survival and recurrence rates in advanced gastric cancer: a systematic review and meta-analysis. Int J Surg 2023; 109:2435-2450. [PMID: 37158149 PMCID: PMC10442139 DOI: 10.1097/js9.0000000000000457] [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: 12/14/2022] [Accepted: 05/01/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Around 5-20% of patients who undergo surgery for advanced gastric cancer (AGC), which invades into the muscularis propria or beyond, have peritoneal carcinomatosis. The peritoneal recurrence rate is 10-54%, which is associated with a poor prognosis. The role of hyperthermic intraperitoneal chemotherapy (HIPEC) in AGC with and without peritoneal carcinomatosis is not clearly defined. METHODS The authors conducted a meta-analysis, in accordance with the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines, of the clinical trials and high-quality nonrandomized studies evaluating the role of HIPEC in AGC over the last 10 years. The studies were searched in PubMed, EMBASE, MEDLINE, and Cochrane databases between January 2011 to December 2021. Clinical data including overall survival, recurrence free survival, overall recurrence rate, peritoneal recurrence rate, and complications analyzed using RevMan 5.4. RESULTS Six randomized controlled trials and 10 nonrandomized studies, comprising a total of 1700 patients were included. HIPEC was associated with significantly improved OS at 3 [odd ratio (OR) 1.89, 95% CI: 1.17-3.05] and 5 years (OR 1.87, 95% CI: 1.29-2.71). HIPEC was associated with reduced overall recurrence (OR 0.49, 95% CI: 0.31-0.80) and peritoneal recurrence (OR 0.22, 95% CI: 0.11-0.47). HIPEC was not associated with increased complications. The occurrence of postoperative renal dysfunction was significantly higher in the HIPEC group (OR 3.94, 95% CI: 1.85-8.38). CONCLUSION The role of HIPEC in AGC has evolved over the past decade. HIPEC may improve survival rates and reduce recurrence rates in patients with AGC, without significant increase in complications and with a favorable impact on 3 and 5-year survival.
Collapse
Affiliation(s)
- Maitreyi Patel
- Department of General Surgery, Queen’s and King George’s Hospital, Barking, Havering and Redbridge University NHS Trust, Rom Valley Way, Romford, United Kingdom
| | - Amandeep Arora
- Department of Uro-Oncology, Tata Memorial Hospital, Mumbai, India
| | - Dipankar Mukherjee
- Department of General Surgery, Queen’s and King George’s Hospital, Barking, Havering and Redbridge University NHS Trust, Rom Valley Way, Romford, United Kingdom
| | - Samrat Mukherjee
- Department of General Surgery, Queen’s and King George’s Hospital, Barking, Havering and Redbridge University NHS Trust, Rom Valley Way, Romford, United Kingdom
| |
Collapse
|
3
|
Chen Z, Ali M, Kai Z, Wang Y, Wang C. HIPEC with CRS versus cytoreductive surgery (CRS) for the gastric cancer metastasis to peritoneum. Clin Transl Oncol 2023; 25:1011-1016. [PMID: 36401054 DOI: 10.1007/s12094-022-03004-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Peritoneal metastases (PM) have a poor prognosis in gastric cancer (GC). Cytoreductive surgery (CRS) gives favorable outcomes, but the influence of hyperthermic intraperitoneal chemotherapy (HIPEC) remains contentious. We designed to distinguish results between CRS versus HIPEC-CRS in patients with peritoneal metastases from gastric cancer. MATERIALS AND METHODS PubMed, Scopus, Embase and Cochrane library accessed to collect data and language is restricted to English. RevMan 5.4 was used to perform statistical analysis. The outcomes for categorical variables are mentioned in the risk ratio. RESULTS Ten trials involving 1367 patients in which 707 were CRS-HIPEC, while 660 CRS. We got significant results in 3rd year survival (P < 0.05), while 1st and 5th years are not statistically significant P > 0.05. CONCLUSION To compare with CRS, CRS-HIPEC has improved survival rate in deprived of further morbidity or mortality.
Collapse
Affiliation(s)
- Zhuming Chen
- Department of General Surgery, Anqing First People's Hospital of Anhui Medical University, Hefei, China
| | - Muhammad Ali
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- Medical College of Yangzhou University, Yangzhou, 225001, China
| | - Zhe Kai
- Department of General Surgery, Anqing First People's Hospital of Anhui Medical University, Hefei, China
| | - Yang Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- Medical College of Yangzhou University, Yangzhou, 225001, China
| | - Chaohui Wang
- Department of General Surgery, Anqing First People's Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
4
|
Huang B, Rouvelas I, Nilsson M. Gastric and gastroesophageal junction cancer: Risk factors and prophylactic treatments for prevention of peritoneal recurrence after curative intent surgery. Ann Gastroenterol Surg 2022; 6:474-485. [PMID: 35847435 PMCID: PMC9271029 DOI: 10.1002/ags3.12565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/13/2022] [Accepted: 03/01/2022] [Indexed: 11/08/2022] Open
Abstract
Aim Relapse after curative treatment for advanced gastric cancer, and especially peritoneal recurrence, is very common and has a dismal prognosis. The aim of this review is to summarize existing evidence regarding risk factors and prophylactic treatments intending to prevent peritoneal recurrence. Methods A structured search of relevant studies was conducted in MEDLINE, Embase, and the Cochrane Library. Results The main risk factors identified are advanced pathological T-stage (pT ≥ 3), regional lymph node involvement, diffuse/poorly cohesive type tumor, poorly differentiated cancer, and positive peritoneal wash cytology. Systemic chemotherapy in the perioperative or adjuvant setting improves survival for the patients but despite this peritoneal recurrence remains a common and yet an unsolved clinical problem. Different approaches of intraperitoneal chemotherapy such as hyperthermic intraperitoneal chemotherapy and early postoperative intraperitoneal chemotherapy have shown promising results as prophylactic treatments aiming to prevent peritoneal recurrence. Conclusion Future studies are warranted to find safe and effective treatments to prevent peritoneal recurrence.
Collapse
Affiliation(s)
- Biying Huang
- Department of Upper Abdominal DiseasesKarolinska University HospitalStockholmSweden
| | - Ioannis Rouvelas
- Department of Upper Abdominal DiseasesKarolinska University HospitalStockholmSweden
- Division of SurgeryDepartment of Clinical Science, Intervention and Technology (CLINTEC)Karolinska InstitutetStockholmSweden
| | - Magnus Nilsson
- Department of Upper Abdominal DiseasesKarolinska University HospitalStockholmSweden
- Division of SurgeryDepartment of Clinical Science, Intervention and Technology (CLINTEC)Karolinska InstitutetStockholmSweden
| |
Collapse
|
5
|
Kim S, Lee CM, Lee D, Kim JH, Park S, Park SH. Can We Reboot the Role of Intraperitoneal Chemotherapy in the Treatment for Gastric Cancer with Peritoneal Carcinomatosis?: A Retrospective Cohort Study Regarding Minimally Invasive Surgery Conjoined with Intraperitoneal plus Systemic Chemotherapy. Cancers (Basel) 2022; 14:cancers14092334. [PMID: 35565463 PMCID: PMC9103395 DOI: 10.3390/cancers14092334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
Background: Peritoneal carcinomatosis (PC) is the most common form of metastasis in gastric cancer (GC) and is related with a poor prognosis. Several treatment modalities including systemic chemotherapy and intraperitoneal chemotherapy have been studied and adopted in treatment of GC patients with PC. Nevertheless, few studies have reported the comparison of the oncologic outcomes between minimally invasive surgery (MIS) with intraperitoneal (IP) chemotherapy and conventional chemotherapy for GC with PC. Methods: We retrospectively reviewed the clinical records of 74 patients who had been diagnosed as GC with PC via either intra-abdominal exploration or abdominopelvic computed tomography between January 2011 and April 2021. After performing propensity score-matching for this retrospective data, we compared the outcomes of 26 patients who underwent MIS followed by IP combined systemic chemotherapy (MIS-IP group) and 26 patients who underwent systemic chemotherapy only (SC-only group). Results: The 2-year progression free survival rate of the MIS-IP group was significantly higher than the SC-only groups (36.4% and 10.5%, respectively; p = 0.010). In multivariate analysis to detect relevant factors on PFS, IP chemotherapy (HR 0.213; p < 0.001), Eastern Cooperative Oncology Group performance status (HR 3.689; p = 0.002), and the amount of ascites (p = 0.011) were significant prognostic factors. Conclusions: This study demonstrated the therapeutic potential of MIS conjoined IP plus systemic chemotherapy for GC patients with PC. MIS conjoined by IP plus systemic chemotherapy can be adopted as a treatment option to reboot the role of IP chemotherapy in GC patients with PC.
Collapse
Affiliation(s)
- Sungho Kim
- Department of Surgery, Korea University College of Medicine, Seoul 02841, Korea; (S.K.); (J.-H.K.); (S.P.); (S.-H.P.)
- Department of Surgery, Korea University Ansan Hospital, Ansan 15355, Korea;
| | - Chang-Min Lee
- Department of Surgery, Korea University College of Medicine, Seoul 02841, Korea; (S.K.); (J.-H.K.); (S.P.); (S.-H.P.)
- Department of Surgery, Korea University Ansan Hospital, Ansan 15355, Korea;
- Correspondence: ; Tel.: +82-31-412-4936; Fax: +82-31-413-4829
| | - Danbi Lee
- Department of Surgery, Korea University Ansan Hospital, Ansan 15355, Korea;
| | - Jong-Han Kim
- Department of Surgery, Korea University College of Medicine, Seoul 02841, Korea; (S.K.); (J.-H.K.); (S.P.); (S.-H.P.)
- Department of Surgery, Korea University Guro Hospital, Seoul 08308, Korea
| | - Sungsoo Park
- Department of Surgery, Korea University College of Medicine, Seoul 02841, Korea; (S.K.); (J.-H.K.); (S.P.); (S.-H.P.)
- Department of Surgery, Korea University Anam Hospital, Seoul 02841, Korea
| | - Seong-Heum Park
- Department of Surgery, Korea University College of Medicine, Seoul 02841, Korea; (S.K.); (J.-H.K.); (S.P.); (S.-H.P.)
- Department of Surgery, Korea University Anam Hospital, Seoul 02841, Korea
| |
Collapse
|
6
|
Hyperthermic Intraperitoneal Chemotherapy plus Intravenous Chemotherapy of Paclitaxel with or without Sintilimab in Gastric Cancer: A Comparative Study. JOURNAL OF ONCOLOGY 2022; 2022:3054485. [PMID: 35242186 PMCID: PMC8888085 DOI: 10.1155/2022/3054485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 11/24/2022]
Abstract
Objective To compare the clinical efficacy of hyperthermic intraperitoneal chemotherapy (HIPEC) plus intravenous chemotherapy of paclitaxel with or without sintilimab in peritoneal metastasis of gastric cancer. Methods A total of 120 patients assessed for eligibility with peritoneal metastasis of gastric cancer treated in the oncology department of our hospital from January 2019 to June 2020 were recruited. They were concurrently randomly assigned in a 1 : 1 ratio to receive HIPEC plus sintilimab-paclitaxel intravenous chemotherapy (study group) or plus paclitaxel intravenous chemotherapy only (control group). Results The objective remission rate (ORR) of ascites in the study group was significantly higher than that in the control group. Subgroup analysis showed that an age ≤60 years or well-differentiated tumors were associated with better objective remission. After treatment, significantly higher Karnofsky Performance Status (KPS) scores were observed in the study group versus those of the control group. Adverse events reported were comparable between groups. The study group obtained longer 12-month progression-free survival (PFS) and overall survival (OS) than those of the control group. Conclusion On top of HIPEC, intravenous chemotherapy with sintilimab and paclitaxel constitute an effective alternative for patients with peritoneal metastasis of gastric cancer to enhance ascites remission, ameliorate the quality of life, and prolong survival, versus with paclitaxel alone.
Collapse
|
7
|
Xiang L, Jin S, Zheng P, Maswikiti EP, Yu Y, Gao L, Zhang J, Zhang Y, Chen H. Risk Assessment and Preventive Treatment for Peritoneal Recurrence Following Radical Resection for Gastric Cancer. Front Oncol 2022; 11:778152. [PMID: 35047394 PMCID: PMC8763009 DOI: 10.3389/fonc.2021.778152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/29/2021] [Indexed: 02/03/2023] Open
Abstract
As the most common recurrence pattern after radical gastric cancer resection, peritoneal recurrence is a major cause of mortality, which affects the prognosis of patients to a very large extent. Peritoneal status and risk of peritoneal recurrence can be evaluated by peritoneal lavage cytology, photodynamic diagnosis, imaging examination, and pathologic analysis. Presently, there is no standard approach for preventing peritoneal recurrence after radical surgery; furthermore, controversies exist regarding the effects of some preventive methods. Among the preventive methods, there are high expectations about the potential of preoperative therapy, surgical skill improvement, hyperthermic intraperitoneal chemotherapy, and postoperative treatment to reduce the incidence of peritoneal recurrence after radical gastrectomy. This study aimed to analyze the results of previous studies on the risk assessment and preventive methods of peritoneal recurrence after radical gastrectomy in recent years. We hope to provide references for better approach to clinical diagnosis and treatment strategies for peritoneal recurrence after radical gastrectomy.
Collapse
Affiliation(s)
- Lin Xiang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, China
| | - Shuai Jin
- Department of Technology, Beijing Weitai’an Pharmaceutical Ltd, Beijing, China
| | - Peng Zheng
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | | | - Yang Yu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Lei Gao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jing Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Ying Zhang
- Department of Laboratory Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Hao Chen
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
8
|
Hirahara N, Matsubara T, Kaji S, Yamamoto T, Hyakudomi R, Takai K, Ishitobi K, Uchida Y, Tajima Y. Phase II feasibility study of adjuvant chemotherapy with docetaxel/cisplatin/S-1 followed by S-1 for stage III gastric cancer. BMC Cancer 2021; 21:1073. [PMID: 34598694 PMCID: PMC8485556 DOI: 10.1186/s12885-021-08795-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/16/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND This study aimed to evaluate the feasibility, safety, and efficacy of postoperative adjuvant chemotherapy with docetaxel/cisplatin/S-1 (DCS) following S-1 therapy in patients with stage III gastric cancer after curative gastrectomy. METHODS Patients with stage III gastric cancer who underwent D2 gastrectomy were enrolled. Adjuvant chemotherapy was initiated within 8 weeks of gastrectomy. The first cycle of chemotherapy consisted of S-1 monotherapy (day 1-14), followed by a 7-day rest period. Cycles 2 and 3 consisted of the following: S-1 (day 1-14) administration, followed by a 14-day rest period, and an intravenous infusion of cisplatin and docetaxel on days 1 and 15. After two cycles, S-1 was administered for up to 1 year. RESULTS Thirty patients were enrolled between 2014 and 2017. Febrile neutropenia of grade 3 or higher was the most common hematological toxicity with 4 patients (13.3%). Other hematological toxicities of grade 3 or higher were as follows: neutropenia in 3 (10.0%), leukopenia in 3 (10.0%), and anemia in 2 (6.7%) patients. Most frequent non-hematological toxicity of grade 3 was anorexia (n = 4, 13.3%) and general fatigue (n = 3, 10.0%); no grade 4 non-hematological toxicities were observed. Twenty-five patients (83.3%) completed two cycles of DCS treatment and 18 (60.0%) completed subsequent S-1 treatment for 1 year. The relative dose intensity of docetaxel and cisplatin was 0.86 and that of S-1 was 0.88. CONCLUSION The DCS regimen can be acceptable as an adjuvant chemotherapy and offers an effective postoperative treatment option for stage III gastric cancer patients. TRIAL REGISTRATION NUMBER UMIN000012785 . DATE OF REGISTRY 08/01/2014.
Collapse
Affiliation(s)
- Noriyuki Hirahara
- Department of Digestive and General Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan.
| | - Takeshi Matsubara
- Department of Digestive and General Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Shunsuke Kaji
- Department of Digestive and General Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Tetsu Yamamoto
- Department of Digestive and General Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Ryoji Hyakudomi
- Department of Digestive and General Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Kiyoe Takai
- Department of Digestive and General Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Kazunari Ishitobi
- Department of Digestive and General Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Yuki Uchida
- Department of Digestive and General Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Yoshitsugu Tajima
- Department of Digestive and General Surgery, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| |
Collapse
|
9
|
Sugawara K, Kawaguchi Y, Seto Y, Vauthey JN. Multidisciplinary treatment strategy for locally advanced gastric cancer: A systematic review. Surg Oncol 2021; 38:101599. [PMID: 33991939 DOI: 10.1016/j.suronc.2021.101599] [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: 01/20/2021] [Accepted: 04/26/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Multidisciplinary management of patients with locally advanced gastric cancer (LAGC) remains unstandardized worldwide. We performed a systemic review to summarize the advancements, regional differences, and current recommended multidisciplinary treatment strategies for LAGC. METHODS Eligible studies were identified through a comprehensive search of PubMed, Web of Science, Cochrane Library databases and Embase. Phase 3 randomized controlled trials which investigated survival of patients with LAGC who underwent gastrectomy with pre-/perioperative, postoperative chemotherapy, or chemoradiotherapy were included. RESULTS In total, we identified 11 studies of pre-/perioperative chemotherapy, 38 of postoperative chemotherapy, and 14 of chemoradiotherapy. In Europe and the USA, the current standard of care is perioperative chemotherapy for patients with LAGC using the regimen of 5-FU, folinic acid, oxaliplatin and docetaxel (FLOT). In Eastern Asia, upfront gastrectomy and postoperative chemotherapy is commonly used. The S-1 monotherapy or a regimen of capecitabine and oxaliplatin (CapOx) are used for patients with stage II disease, and the CapOx regimen or the S-1 plus docetaxel regimen are recommended for those with stage III Gastric cancer (GC). The addition of postoperative radiotherapy to peri- or postoperative chemotherapy is currently not recommended. Additionally, clinical trials testing targeted therapy and immunotherapy are increasingly performed worldwide. CONCLUSIONS Recent clinical trials showed a survival benefit of peri-over postoperative chemotherapy and chemoradiotherapy. As such, this strategy may have a potential as a global standard for patients with LAGC. Outcome of the ongoing clinical trials is expected to establish the global standard of multidisciplinary treatment strategy in patients with LAGC.
Collapse
Affiliation(s)
- Kotaro Sugawara
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshikuni Kawaguchi
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jean-Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Khachfe HH, Salhab HA, Fares MY, Chahrour MA, Jamali FR. Landscape of interventional clinical trials involving gastrectomy for gastric cancer. Ecancermedicalscience 2021; 15:1218. [PMID: 34158822 PMCID: PMC8183643 DOI: 10.3332/ecancer.2021.1218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Indexed: 01/14/2023] Open
Abstract
Background Gastric cancer (GC) is the third most common cause of malignancy associated mortality globally. The cornerstone of curative treatment involves surgical gastrectomy. In this study, we explore clinical trials involving gastrectomy for GC, highlighting inadequacies and underlining promising surgical interventions and strategies. Materials and methods On 1 May 2020, ClinicalTrials.gov was explored for interventional trials related to gastrectomy for GC, without adding limitations for location or date. All data pertaining to the trials were collected. Characteristics such as phase, duration, enrolment size, location, treatment allocation, masking and primary endpoint were analysed. Results One hundred thirty-eight clinical trials met the search criteria. Clinical trials were performed in only 14 countries; most of them occurring in China. Most trials (33%) were still in the recruiting phase. On average, the length of trials was 3.9 years. Most trials had parallel assignment, were randomised and masked. The primary endpoint which was mostly commonly studied was overall survival (33%). The most common intervention studied is laparoscopic gastrectomy in 43 (31%) trials. Conclusions Our study exposed a small number of trials, publication rate, absence of geographic variety in clinical trials involving gastrectomy for GC. Adequate management of trial design can help decrease duration and increase validity of results. More trials comparing different surgical techniques are needed to update the surgical practice of gastrectomy for GC.
Collapse
Affiliation(s)
- Hussein H Khachfe
- Faculty of Medicine, American University of Beirut Medical Center, Beirut 00000, Lebanon.,Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, USA.,Division of GI Surgical Oncology, UPMC Pancreatic Cancer Center, University of Pittsburgh Medical Center, UPMC Cancer Pavilion, Pittsburgh, PA 15260, USA
| | - Hamza A Salhab
- Faculty of Medicine, American University of Beirut Medical Center, Beirut 00000, Lebanon.,Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut 00000, Lebanon
| | - Mohamad Y Fares
- Faculty of Medicine, American University of Beirut Medical Center, Beirut 00000, Lebanon.,Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut 00000, Lebanon
| | - Mohamad A Chahrour
- Faculty of Medicine, American University of Beirut Medical Center, Beirut 00000, Lebanon
| | - Faek R Jamali
- Division of General Surgery, Department of Surgery, Sheikh Shakhbout Medical City, Abu Dhabi 11001, UAE
| |
Collapse
|
11
|
Reutovich MY, Krasko OV, Sukonko OG. Hyperthermic intraperitoneal chemotherapy in prevention of gastric cancer metachronous peritoneal metastases: a systematic review. J Gastrointest Oncol 2021; 12:S5-S17. [PMID: 33968422 DOI: 10.21037/jgo-20-129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer progression resulting in metachronous peritoneal metastasizing is almost always associated with an adverse prognosis. This review discusses various options of preventing metachronous peritoneal metastases in radically operated gastric cancer patients. Also examined are different hyperthermic intraperitoneal chemotherapy (HIPEC) regimens employed in gastric cancer treatment, postoperative morbidity and mortality rates and long-term treatment outcomes. The authors also review their own experience of using HIPEC based on the combination of cisplatin and doxorubicin in doses of 50 mg/m2 at 42 °C for 1 h to prevent gastric cancer peritoneal dissemination. As a result, progression-free survival rose from 19.6%±5.6% to 47.1%±6.3% (Plog-rank <0.001) and dissemination-free survival-from 22.7%±6.0% to 51.9%±6.3% (Plog-rank <0.001). It is noted that the combination of the described HIPEC regimen with systemic chemotherapy helped raise metastases-free 3-year survival rate to up to 91.0%±9.0% (Plog-rank =0.025) compared with 48.6%±6.4% for patients who underwent only a combined surgery/HIPEC treatment. HIPEC is a promising combined treatment strategy for radically operated gastric cancer patients that can improve patient survival and decrease peritoneal dissemination rate. However, the number of randomized studies on adjuvant HIPEC are still insufficient for a subgroup assessment of efficacy of the given chemotherapy regimens and generation of evidence-based recommendations on the individual use of chemotherapy agents and their combinations, and HIPEC procedural techniques. Further prospective randomized studies are needed to assess the practicability of complementing HIPEC with adjuvant systemic chemotherapies.
Collapse
Affiliation(s)
- Mikhail Yu Reutovich
- Gastroesophageal Pathology Department, N.N. Alexandrov National Cancer Center, Minsk, Belarus
| | - Olga V Krasko
- United Institute of Informatics Problems, National Academy of Sciences, Minsk, Belarus
| | - Oleg G Sukonko
- Gastroesophageal Pathology Department, N.N. Alexandrov National Cancer Center, Minsk, Belarus
| |
Collapse
|
12
|
Fan B, Bu Z, Zhang J, Zong X, Ji X, Fu T, Jia Z, Zhang Y, Wu X. Phase II trial of prophylactic hyperthermic intraperitoneal chemotherapy in patients with locally advanced gastric cancer after curative surgery. BMC Cancer 2021; 21:216. [PMID: 33653317 PMCID: PMC7923597 DOI: 10.1186/s12885-021-07925-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/17/2021] [Indexed: 11/10/2022] Open
Abstract
Background HIPEC is an emerging procedure to treat peritoneal metastasis of gastric cancer. Data about HIPEC in locally advanced gastric cancer is scarce. The purpose of this trial is to evaluate the safety and toxicity of prophylactic HIPEC with cisplatin for patients with locally advanced gastric cancer. Methods From March 2015 to November 2016, a prospective, randomized phase II trial was conducted. After radical gastrectomy, patients in the experimental group underwent HIPEC with cisplatin followed by adjuvant chemotherapy with SOX regime. Patients in the other group were treated with SOX regime alone. Postoperative complications and patient survival were compared. Results In total, 50 patients were eligible for analyses. No significant difference was found in the incidence of postoperative complications including anastomotic/intestinal leakage, liver dysfunction, bone marrow suppression, wound infection and ileus (P > 0.05). Mean duration of hospitalization after radical gastrectomy was 11.7 days. 12.2 days in experimental group and 10.8 days in control group respectively (P = 0.255). The percentage of patients with elevated tumor markers was 12.1% in experimental group, which was significantly lower than 41.2% in control group (P = 0.02). 3-year RFS of patients who treated with or without prophylactic HIPEC were 84.8 and 88.2% respectively (P = 0.986). In the multivariate analysis, pathological T stage was the only independent risk factor for the RFS of patients (P = 0.012, HR =15.071). Conclusion Additional intraoperative HIPEC with cisplatin did not increase postoperative complications for locally advanced gastric cancer after curative surgery. Prophylactic HIPEC with cisplatin was safe and tolerable, while it did not reduce the risk of peritoneal recurrence in this trial, supporting further studies to validate the efficacy of it. Trial registration Chinese Clinical Trial Registry, ChiCTR2000038331. Registered 18 September 2020 - Retrospectively registered, http://www.chictr.org.cn/showproj.aspx?proj=59692.
Collapse
Affiliation(s)
- Biao Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Zhaode Bu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Ji Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Xianglong Zong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Xin Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Tao Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Ziyu Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Yinan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Xiaojiang Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China.
| |
Collapse
|
13
|
Kus T, Kose F, Aktas G, Arslan UY, Sedef AM, Cinkir HY, Dirikoc M, Akkus G, Ozdemir NY. Prediction of Peritoneal Recurrence in Patients with Gastric Cancer: a Multicenter Study. J Gastrointest Cancer 2020; 52:634-642. [PMID: 32578034 DOI: 10.1007/s12029-020-00419-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE The peritoneum is the common recurrence site of gastric cancer (GC) presenting with worse survival. Although some predictive clinicopathological factors have been identified, there is no comprehensive assessment of peritoneal recurrence risk prediction for patients treated with adjuvant chemotherapy (CR) or chemoradiotherapy (CRT) after surgery. We aimed to predict peritoneal recurrence and develop a new scoring model in GC. METHODS This retrospective study included 274 GC patients who presented with recurrence after curative gastrectomy followed by adjuvant chemotherapy (CT) or chemoradiotherapy (CRT). Risk factors for peritoneal recurrence were analyzed using the following parameters: age, gender, tumor location and characteristics, and differences between treatment modalities. All parameters were assessed by binary logistic regression analysis to compare the patients with and without peritoneal recurrence. Then, a new risk scoring model was developed. RESULTS Peritoneal recurrence was observed in 115 (44.1%) patients. Peritoneal recurrence was higher in female gender (odds ratio (OR), 1.93; 1.07-3.49, P = 0.030, 1 point), T4a-b stage (OR, 2.47; 1.14-5.36, P = 0.022, 1 point), poor/undifferentiated (OR, 2.04; 1.31-4.06, P = 0.004, 1 point), and signet cell carcinoma (OR, 2.04; 1.04-4.02, P = 0.038, 1 point) after adjusted for resection and dissection types. The risk scoring model was developed using the related parameters: Peritoneal recurrence rates were 24.6%, 42.6%, and 71.4% for group 1 (0 point), group 2 (1-2 points), and group 3 (3-4 points), respectively. CONCLUSION Female gender, T4 tumor stage, undifferentiated histopathology, and signet cell type had a tendency to peritoneal recurrence after adjusted for treatment modalities. Patients with 3 or 4 risk factors had an 8.8-fold increased risk for the development of peritoneal recurrence.
Collapse
Affiliation(s)
- Tulay Kus
- School of Medicine, Department of Medical Oncology, Gaziantep University, TR-27310, Gaziantep, Turkey.
| | - Fatih Kose
- School of Medicine, Department of Medical Oncology, Baskent University, Adana, Turkey
| | - Gokmen Aktas
- School of Medicine, Department of Medical Oncology, Kahramanmaras Sütçü İmam University, Kahramanmaraş, Turkey
| | - Ulku Yalcintas Arslan
- Ankara Oncology Training and Research Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Ali Murat Sedef
- Department of Medical Oncology, Medical Park Tarsus Hospital, Tarsus, Turkey
| | - Havva Yesil Cinkir
- School of Medicine, Department of Medical Oncology, Gaziantep University, Gaziantep, Turkey
| | - Merve Dirikoc
- Department of Medical Oncology, Ankara Numune Education and Research Hospital, Ankara, Turkey
| | - Gulsum Akkus
- School of Medicine, Department of Internal Medicine, Kahramanmaras Sütçü İmam University, Kahramanmaraş, Turkey
| | - Nuriye Yildirim Ozdemir
- Faculty of Medicine, Department of Medical Oncology, Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
14
|
Terashima M, Yoshikawa T, Boku N, Ito S, Tsuburaya A, Iwasaki Y, Fukagawa T, Tokunaga M, Sano T, Sasako M. Current status of perioperative chemotherapy for locally advanced gastric cancer and JCOG perspectives. Jpn J Clin Oncol 2020; 50:528-534. [PMID: 32134452 DOI: 10.1093/jjco/hyaa005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/11/2019] [Accepted: 01/28/2020] [Indexed: 02/05/2023] Open
Abstract
Perioperative treatment for locally advanced gastric cancer has been inconsistent between Japan and the Western countries. In Japan, D2 gastrectomy followed by adjuvant chemotherapy is regarded as standard treatment, while neoadjuvant or perioperative chemotherapy is considered to be a standard in the Western countries. Stomach Cancer Study Group of Japan Clinical Oncology Group (JCOG) has conducted many perioperative chemotherapy trials. After the publishing of positive results of ACTS-GC trial, stage-specific adjuvant chemotherapy protocols are planned. JCOG1104 was conducted as to demonstrate the non-inferiority of four courses of S-1 to standard eight courses of S-1, because the efficacy of S-1 appears to be sufficient in stage II. The trial failed to demonstrate the non-inferiority of four courses of S-1. S-1 for 1 year is still recognized to be a standard for stage II gastric cancer. For stage III, studies with more intensive treatments were planned as the efficacy of S-1 monotherapy seems to be insufficient. As in the Western countries, JCOG planned the perioperative chemotherapy. However, the clinical staging is a serious issue to select optimal patients for perioperative chemotherapy. JCOG conducted a prospective cohort study to evaluate the validity of clinical staging in JCOG1302A. From the results of this study, cT3-4 and cN1-3 are selected as optimal candidate for perioperative chemotherapy. JCOG1509 was conducted to demonstrate the superiority of perioperative chemotherapy to adjuvant chemotherapy in these cohorts. Perioperative chemotherapy for marginally resectable tumours such as linitis plastica or extensive nodal disease and special type of cancer like HER2 positive are also conducted.
Collapse
Affiliation(s)
| | - Takaki Yoshikawa
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Narikazu Boku
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Seiji Ito
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Akira Tsuburaya
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center Hospital, Yokohama, Japan
| | - Yoshiaki Iwasaki
- Department of Surgery, IMS Tokyo Katsushika General Hospital, Tokyo, Japan
| | - Takeo Fukagawa
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Masanori Tokunaga
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Sano
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mitsuru Sasako
- Department of Surgery, Yodogawa Christian Hospital, Osaka, Japan
| |
Collapse
|
15
|
Ellebæk SB, Graversen M, Detlefsen S, Lundell L, Fristrup CW, Pfeiffer P, Mortensen MB. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) of peritoneal metastasis from gastric cancer: a descriptive cohort study. Clin Exp Metastasis 2020; 37:325-332. [PMID: 32002724 DOI: 10.1007/s10585-020-10023-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/23/2020] [Indexed: 12/23/2022]
Abstract
Pressurized intraperitoneal aerosol chemotherapy (PIPAC) represents a novel approach to deliver intraperitoneal chemotherapy. We report our experience with PIPAC in patients with peritoneal metastasis (PM) from gastric cancer (GC). Data from GC patients (n = 20) included in the prospective PIPAC-OPC1 and PIPAC-OPC2 studies are reported. All patients had received prior systemic chemotherapy. The mean peritoneal cancer index (PCI) was 10.5 (range 0-39) and nine patients had diffuse GC. PIPAC with cisplatin 7.5 mg/m2 and doxorubicin 1.5 mg/m2 were administered at 4-6-week intervals. Outcome criteria were objective tumour response, survival and adverse events. Twenty patients had 52 PIPAC procedures with a median follow-up of 10.4 months (3.3-26.5). Median survival from the time of PM diagnosis and after the first PIPAC procedure was 11.5 months and 4.7 months, respectively. Fourteen patients had repeated PIPAC (> 2), and the objective tumour response according to the histological peritoneal regression grading score (PRGS) was observed in 36%, whereas 36% had stable disease. Ten patients completed the three prescheduled sessions (per protocol group) and 40% of those displayed an objective tumour response, while 20% had stable disease. Only minor postoperative complications were noted, and none were considered causally related to the PIPAC treatment. PIPAC with low-dose cisplatin and doxorubicin can induce a quantifiable objective tumour response in selected patients with PM from GC. Survival data are encouraging and warrant further clinical studies.
Collapse
Affiliation(s)
- S Bremholm Ellebæk
- Odense PIPAC Center, Odense University Hospital, J.B. Winsloews Vej 4, 5000, Odense, Denmark. .,Department of Surgery, Upper GI and HPB Section, Odense University Hospital, Odense, Denmark.
| | - M Graversen
- Odense PIPAC Center, Odense University Hospital, J.B. Winsloews Vej 4, 5000, Odense, Denmark.,Department of Surgery, Upper GI and HPB Section, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,OPEN, Open Patient Data Explorative Network, Odense, Denmark
| | - S Detlefsen
- Odense PIPAC Center, Odense University Hospital, J.B. Winsloews Vej 4, 5000, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Odense University Hospital, Odense, Denmark
| | - L Lundell
- Odense PIPAC Center, Odense University Hospital, J.B. Winsloews Vej 4, 5000, Odense, Denmark.,Division of Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - C W Fristrup
- Odense PIPAC Center, Odense University Hospital, J.B. Winsloews Vej 4, 5000, Odense, Denmark.,Department of Surgery, Upper GI and HPB Section, Odense University Hospital, Odense, Denmark
| | - P Pfeiffer
- Odense PIPAC Center, Odense University Hospital, J.B. Winsloews Vej 4, 5000, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark
| | - M B Mortensen
- Odense PIPAC Center, Odense University Hospital, J.B. Winsloews Vej 4, 5000, Odense, Denmark.,Department of Surgery, Upper GI and HPB Section, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,OPEN, Open Patient Data Explorative Network, Odense, Denmark
| |
Collapse
|
16
|
Wang Z, Chen JQ, Liu JL, Tian L. Issues on peritoneal metastasis of gastric cancer: an update. World J Surg Oncol 2019; 17:215. [PMID: 31829265 PMCID: PMC6907197 DOI: 10.1186/s12957-019-1761-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022] Open
Abstract
Background Peritoneal metastasis (PM) is one of the most common forms of metastasis with a very poor prognosis in patients with gastric cancer (GC). The mechanisms, diagnosis, and management of PM remain controversial. Main body Stephen Paget’s “seed-and-soil” hypothesis gives us an illustration of the mechanisms of PM. Recently, hematogenous metastasis and exosomes from GC are identified as novel mechanisms for PM. Diagnostic accuracy of conventional imaging modalities for PM is not satisfactory, but texture analysis may be a useful adjunct for the prediction of PM. Biological markers in peritoneal washings are helpful in identifying patients at high risk of PM, but many limitations remain to be overcome. Response of PM from systemic chemotherapy alone is very limited. However, conversion therapy is confirmed to be safe and able to prolong the survival of GC patients with PM. As an important part of conversion therapy, intraperitoneal chemotherapy with taxanes has become an ideal approach with several advantages. Additionally, gastrectomy should be considered in patients who would tolerate surgery if a remarkable response to chemotherapy was observed. Conclusion Texture analysis is a reliable adjunct for the prediction of PM, and conversion therapy provides a new choice for GC patients with PM. The underlying mechanisms and new biological markers for GC patients with PM should be the direction of future studies. Furthermore, significant aspects of conversion therapy, such as timing and method of the operation, and the indications remain to be clarified.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| | - Jun-Qiang Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| | - Jin-Lu Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Lei Tian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
17
|
Misawa K, Mochizuki Y, Sakai M, Teramoto H, Morimoto D, Nakayama H, Tanaka N, Matsui T, Ito Y, Ito S, Tanaka K, Uemura K, Morita S, Kodera Y. Randomized clinical trial of extensive intraoperative peritoneal lavage versus standard treatment for resectable advanced gastric cancer (CCOG 1102 trial). Br J Surg 2019; 106:1602-1610. [PMID: 31573086 DOI: 10.1002/bjs.11303] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/27/2019] [Accepted: 06/10/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND A survival benefit of extensive intraoperative peritoneal lavage (EIPL) has been reported in patients with gastric cancer with positive peritoneal cytology. The hypothesis of this study was that EIPL may reduce peritoneal recurrence in patients with advanced gastric cancer who undergo surgery with curative intent. METHODS This was an open-label, multi-institutional, randomized, phase 3 trial to assess the effects of EIPL versus standard treatment after curative gastrectomy for resectable gastric cancer of T3 status or above. The primary endpoint was disease-free survival (DFS); secondary endpoints were overall survival, peritoneal recurrence-free survival and incidence of adverse events. RESULTS Between July 2011 and January 2014, 314 patients were enrolled from 15 institutions and 295 patients were analysed (145 and 150 in the EIPL and no-EIPL groups respectively). The 3-year DFS rate was 63·9 (95 per cent c.i. 55·5 to 71·2) per cent in the EIPL group and 59·7 (51·3 to 67·1) per cent in the control group (hazard ratio (HR) 0·81, 95 per cent c.i. 0·57 to 1·16; P = 0·249). The 3-year overall survival rate was 75·0 (67·1 to 81·3) per cent in the EIPL group and 73·7 (65·9 to 80·1) per cent in the control group (HR 0·91, 0·60 to 1·37; P = 0·634). Peritoneal recurrence-free survival was not significantly different between the two groups (HR 0·92, 0·62 to 1·36; P = 0·676). No intraoperative complications related to EIPL were observed. CONCLUSION EIPL did not improve survival or peritoneal recurrence in patients who underwent gastrectomy for advanced gastric cancer. Registration number: 000005907 (http://www.umin.ac.jp/ctr/index.htm).
Collapse
Affiliation(s)
- K Misawa
- Department of Gastroenterological Surgery, Aichi Cancer Centre Hospital, Nagoya, Japan
| | - Y Mochizuki
- Department of Surgery, Komaki Municipal Hospital, Komaki, Japan
| | - M Sakai
- Department of Surgery, Ichinomiya Municipal Hospital, Ichinomiya, Japan
| | - H Teramoto
- Department of General Surgery, Yokkaichi Municipal Hospital, Yokkaichi, Japan
| | - D Morimoto
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Surgery, JA Kainan Hospital, Yatomi, Japan
| | - H Nakayama
- Department of Surgery, National Hospital Organization Nagoya Medical Centre, Nagoya, Japan
| | - N Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Surgery, JA Aichi Konan Kosei Hospital, Konan, Japan
| | - T Matsui
- Department of Gastroenterological Surgery, Aichi Cancer Centre, Aichi Hospital, Okazaki, Japan
| | - Y Ito
- Department of Gastroenterological Surgery, Aichi Cancer Centre Hospital, Nagoya, Japan
| | - S Ito
- Department of Gastroenterological Surgery, Aichi Cancer Centre Hospital, Nagoya, Japan
| | - K Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - K Uemura
- Department of Biostatistics and Bioinformatics, Interfaculty Initiative in Information Studies, University of Tokyo, Tokyo, Japan
| | - S Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Y Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | |
Collapse
|
18
|
Intraperitoneal chemotherapy for peritoneal metastases using sustained release formula of cisplatin-incorporated gelatin hydrogel granules. Surg Today 2019; 49:785-794. [PMID: 30847629 DOI: 10.1007/s00595-019-01792-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/13/2019] [Indexed: 01/10/2023]
Abstract
PURPOSE We previously reported the effectiveness of gelatin microspheres incorporating cisplatin in a mouse model of peritoneal metastases. In this study, we report our new complete sustained-release formula of gelatin hydrogel granules incorporating cisplatin (GHG-CDDP), which exerted a good anti-tumor effect with less toxicity. METHODS GHG-CDDP was prepared without organic solvents to enable its future clinical use. The pharmaceutical characterization of GHG-CDDP was performed, and its in vivo degradability was evaluated. The anti-tumor effect was evaluated using a murine peritoneal metastasis model of the human gastric cancer MKN45-Luc cell line. RESULTS Our new manufacturing process dramatically reduced the initial burst of CDDP release to approximately 2% (wt), while the previous product had a 25-30% initial burst. In intraperitoneal degradation tests, approximately 30% of GHG-CDDP remained in the murine abdominal cavity 7 days after intraperitoneal injection and disappeared within 3 weeks. GHG-CDDP significantly suppressed the in vivo tumor growth (p = 0.02) and prolonged the survival time (p = 0.0012) compared with the control. In contrast, free CDDP did not show a significant therapeutic effect at any dose. Weight loss and hematological toxicity were also significantly ameliorated. CONCLUSIONS GHG-CDDP is a promising treatment option for peritoneal metastases through the complete sustained-release of CDDP with less systemic toxicity.
Collapse
|
19
|
Arai H, Sunakawa Y, Nakajima TE. Co-operative groups in the development of chemotherapy for gastric cancer. Jpn J Clin Oncol 2019; 49:210-227. [PMID: 30508188 DOI: 10.1093/jjco/hyy176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/14/2018] [Accepted: 11/01/2018] [Indexed: 01/05/2023] Open
Abstract
In the multimodality treatment strategy for gastric cancer, chemotherapy has an important role in conferring survival benefit. For the last three decades, great progress has been achieved in adjuvant and palliative chemotherapy. Powerful combination regimens using doublet or triplet cytotoxic agents have been developed and new molecular targeted drugs, including trastuzumab and ramucirumab, have been introduced in clinical practice. These advances have resulted from the accumulation of many clinical trials. A well-designed phase III trial can change standard treatment; however, such a trial is hard to complete due to its huge cost and need to recruit many patients. Some co-operative groups have actively made efforts at fundraising and patient recruitment, which can make implementation of high-quality and large-scale phase III trials possible. This review summarizes the development of chemotherapy for gastric cancer with focus on co-operative groups around the world, considering effective treatment developments in gastric cancer. We studied 11 active co-operative groups, including six in Europe, two in the United States, and three in Japan, that have completed one or more phase III trials cited in the major guidelines. Each co-operative group had its own characteristics and contributed to the establishment of standard treatment in each region. International collaboration in the development of gastric cancer treatment may be difficult due to regional differences in standards of care, particularly for resectable gastric cancer. Whereas, intergroup collaboration within each region is a reasonable method to effectively develop treatments for resectable and advanced gastric cancer.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki-shi, Kanagawa, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki-shi, Kanagawa, Japan
| | - Takako Eguchi Nakajima
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki-shi, Kanagawa, Japan
| |
Collapse
|
20
|
Dahdaleh FS, Turaga KK. Evolving Treatment Strategies and Outcomes in Advanced Gastric Cancer with Peritoneal Metastasis. Surg Oncol Clin N Am 2018; 27:519-537. [PMID: 29935687 DOI: 10.1016/j.soc.2018.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gastric cancer (GC) has a predilection to metastasize to the peritoneum, denoting a poor prognosis. Treatment strategies available for advanced GC have significantly evolved over time and can be categorized into systemic, regional, and surgical. Although systemic therapies have been the mainstay for the treatment of advanced GC, their ability in achieving long-term survival in patients with peritoneal involvement is modest at best. This article describes advances in combined modality treatment of peritoneal metastases, specifically with an emphasis on peritoneal-directed therapies.
Collapse
Affiliation(s)
- Fadi S Dahdaleh
- Complex General Surgical Oncology, Section of General Surgery/Surgical Oncology, The University of Chicago Medicine, 5841 South Maryland Avenue, Room S214, MC 5094, Chicago, IL 60637, USA
| | - Kiran K Turaga
- The University of Chicago Medicine, Section of General Surgery/Surgical Oncology, 5841 South Maryland Avenue, Room G207, MC 5094, Chicago, IL 60637, USA.
| |
Collapse
|
21
|
Kodera Y. Surgery with curative intent for stage IV gastric cancer: Is it a reality of illusion? Ann Gastroenterol Surg 2018; 2:339-347. [PMID: 30238074 PMCID: PMC6139716 DOI: 10.1002/ags3.12191] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 06/13/2018] [Indexed: 12/22/2022] Open
Abstract
Gastric cancer with metastases outside of the regional lymph nodes is deemed oncologically unresectable. Nevertheless, some metastatic lesions are technically resectable by applying established surgical techniques such as para-aortic lymphadenectomy and hepatectomy. At the time of compilation of the Japanese gastric cancer treatment guidelines version 4, systematic reviews were conducted to see whether it is feasible to make any recommendation to dissect both the primary and metastatic lesions with intent to cure, possibly as part of multimodality treatment. Long-term survivors were found among carefully selected groups of patients both in prospective and retrospective studies. In addition, there is a growing list of publications reporting encouraging outcomes of gastrectomy conducted after exceptionally good response to chemotherapy, usually among patients who underwent R0 resection. This type of surgery is often referred to as conversion surgery. It is sometimes difficult to define a clear borderline between curative surgery scheduled after neoadjuvant chemotherapy and the conversion surgery. This review summarizes what we knew after the literature reviews conducted at the time of compiling the Japanese guidelines and in addition reflects some new findings obtained thereafter through clinical trials and retrospective studies. Metastases were divided into three categories based on the major metastatic pathways: lymphatic, hematogenous, and peritoneal. In each of these categories, there were findings that could provide hope for patients with metastatic disease. These findings implied that the surgical technique that we already use could become more useful upon further developments in antineoplastic agents and drug delivery.
Collapse
Affiliation(s)
- Yasuhiro Kodera
- Department of Gastroenterological SurgeryNagoya University Graduate School of MedicineNagoyaAichiJapan
| |
Collapse
|
22
|
Ishigami H, Fujiwara Y, Fukushima R, Nashimoto A, Yabusaki H, Imano M, Imamoto H, Kodera Y, Uenosono Y, Amagai K, Kadowaki S, Miwa H, Yamaguchi H, Yamaguchi T, Miyaji T, Kitayama J. Phase III Trial Comparing Intraperitoneal and Intravenous Paclitaxel Plus S-1 Versus Cisplatin Plus S-1 in Patients With Gastric Cancer With Peritoneal Metastasis: PHOENIX-GC Trial. J Clin Oncol 2018; 36:1922-1929. [PMID: 29746229 DOI: 10.1200/jco.2018.77.8613] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Purpose Intraperitoneal paclitaxel plus systemic chemotherapy demonstrated promising clinical effects in patients with gastric cancer with peritoneal metastasis. We aimed to verify its superiority over standard systemic chemotherapy in overall survival. Patients and Methods This randomized phase III trial enrolled patients with gastric cancer with peritoneal metastasis who had received no or short-term (< 2 months) chemotherapy. Patients were randomly assigned at a two-to-one ratio to receive intraperitoneal and intravenous paclitaxel plus S-1 (IP; intraperitoneal paclitaxel 20 mg/m2 and intravenous paclitaxel 50 mg/m2 on days 1 and 8 plus S-1 80 mg/m2 per day on days 1 to 14 for a 3-week cycle) or S-1 plus cisplatin (SP; S-1 80 mg/m2 per day on days 1 to 21 plus cisplatin 60 mg/m2 on day 8 for a 5-week cycle), stratified by center, previous chemotherapy, and extent of peritoneal metastasis. The primary end point was overall survival. Secondary end points were response rate, 3-year overall survival rate, and safety. Results We enrolled 183 patients and performed efficacy analyses in 164 eligible patients. Baseline characteristics were balanced between the arms, except that patients in the IP arm had significantly more ascites. The median survival times for the IP and SP arms were 17.7 and 15.2 months, respectively (hazard ratio, 0.72; 95% CI, 0.49 to 1.04; stratified log-rank P = .080). In the sensitivity analysis adjusted for baseline ascites, the hazard ratio was 0.59 (95% CI, 0.39 to 0.87; P = .008). The 3-year overall survival rate was 21.9% (95% CI, 14.9% to 29.9%) in the IP arm and 6.0% (95% CI, 1.6% to 14.9%) in the SP arm. Both regimens were well tolerated. Conclusion This trial failed to show statistical superiority of intraperitoneal paclitaxel plus systemic chemotherapy. However, the exploratory analyses suggested possible clinical benefits of intraperitoneal paclitaxel for gastric cancer.
Collapse
Affiliation(s)
- Hironori Ishigami
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Yoshiyuki Fujiwara
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Ryoji Fukushima
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Atsushi Nashimoto
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Hiroshi Yabusaki
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Motohiro Imano
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Haruhiko Imamoto
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Yasuhiro Kodera
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Yoshikazu Uenosono
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Kenji Amagai
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Shigenori Kadowaki
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Hiroto Miwa
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Hironori Yamaguchi
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Takuhiro Yamaguchi
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Tempei Miyaji
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Joji Kitayama
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| |
Collapse
|
23
|
Wang PL, Huang JY, Zhu Z, Gong BC, Huang HW, Duan SJ, Xu HM, Liu FN. Development of a risk-scoring system to evaluate the serosal invasion for macroscopic serosal invasion positive gastric cancer patients. Eur J Surg Oncol 2018; 44:600-606. [DOI: 10.1016/j.ejso.2018.01.240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/06/2017] [Accepted: 01/30/2018] [Indexed: 12/22/2022] Open
|
24
|
Lauren Histologic Type Is the Most Important Factor Associated With Pattern of Recurrence Following Resection of Gastric Adenocarcinoma. Ann Surg 2017; 267:105-113. [PMID: 27759618 DOI: 10.1097/sla.0000000000002040] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To examine sites of initial recurrence in patients after resection of gastric and gastroesophageal junction Siewert II/III adenocarcinoma (GA). BACKGROUND There are few recent studies on recurrence for Western patients following potentially curative resection of GA. METHODS A review of a prospectively maintained, single institution database was performed. Clinicopathologic factors, site(s) of initial recurrence, disease-free survival, and overall survival were examined. RESULTS From January 2000 to June 2010, 957 patients underwent potentially curative resection for GA, 435 patients (46%) had recurrent disease, and complete data on recurrence site(s) could be obtained in 386 patients. Tumors were Lauren intestinal type in 206 (53%) and diffuse or mixed-type in 180 (47%). Median time to recurrence was 12 months and 75% of recurrences occurred within 2 years. There was a significant difference in pattern of initial recurrence between the intestinal and diffuse/mixed cohorts (P < 0.001). For intestinal tumors, distant metastasis was the most common site (54%), followed by locoregional (20%), peritoneal (15%), and multifocal (11%). For diffuse/mixed tumors, peritoneal recurrence was the most common (37%), followed by distant (32%), locoregional (22%), and multifocal (9%). On multivariate analysis, Lauren histologic type was the only significant factor that was associated with both peritoneal recurrence (diffuse, hazard ratio 2.22, confidence interval 1.38-3.94) and distant recurrence (intestinal, hazard ratio 1.888, confidence interval 1.202-2.966). After recurrence, median overall survival was only 8.4 months. CONCLUSIONS In GA patients who recur after resection, patterns of recurrence vary significantly based on Lauren histologic type.
Collapse
|
25
|
Irino T, Takeuchi H, Terashima M, Wakai T, Kitagawa Y. Gastric Cancer in Asia: Unique Features and Management. Am Soc Clin Oncol Educ Book 2017; 37:279-291. [PMID: 28561675 DOI: 10.1200/edbk_175228] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gastric cancer (GC) poses a burden to patients across the globe as the third leading cause of cancer deaths worldwide. Incidence of GC is particularly high in Asian countries, which is attributed to the prevalence of Helicobacter pylori (H. pylori) infection and has prompted the establishment of unique treatment strategies. D2 gastrectomy, which was established in the 1950s in Japan, has served as a gold standard for locally advanced GC for over half a century. Since the beginning of the 21st century, endoscopic resection (ER) techniques and minimally invasive laparoscopic surgery have greatly changed the treatment of patients with early GC. S-1, which showed a striking survival benefit in a large randomized trial in Japan, has been used as adjuvant therapy for the last decade. Likewise, S-1-based chemotherapy regimens are currently the standard of care for the treatment of unresectable/metastatic GC in Asia. Along with the development of standardized therapy, novel techniques and new drugs have been rapidly brought into clinical practice. State-of-the-art sentinel node (SN) navigation surgery enables clinicians to perform truly minimally invasive surgery for early GC, and appropriate chemotherapy regimens are now determined by a tumor's molecular expression. New classifications based on gene signatures are proposed and may replace conventional clinical classifications. Such highly individualized treatment has the potential to alter our clinical practice in GC in the near future. The best practice in each geographic region should be shared and integrated, resulting in the best practice without borders.
Collapse
Affiliation(s)
- Tomoyuki Irino
- From the Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan; Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroya Takeuchi
- From the Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan; Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masanori Terashima
- From the Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan; Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Toshifumi Wakai
- From the Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan; Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- From the Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan; Division of Gastric Surgery, Shizuoka Cancer Center, Shizuoka, Japan; Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan; Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
26
|
He Z, Zhao TT, Xu HM, Wang ZN, Xu YY, Song YX, Ni ZR, Xu H, Yin SC, Liu XY, Miao ZF. Efficacy and safety of intraperitoneal chemotherapy in patients with advanced gastric cancer: a cumulative meta-analysis of randomized controlled trials. Oncotarget 2017; 8:81125-81136. [PMID: 29113372 PMCID: PMC5655267 DOI: 10.18632/oncotarget.20818] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/23/2017] [Indexed: 01/04/2023] Open
Abstract
Even when a curative gastrectomy is conducted, the majority of advanced gastric cancer patients with invasion die due to peritoneal recurrence. We performed electronic searches to identify randomized controlled trials published through April 2017 evaluating the effect of intraperitoneal chemotherapy (IPC) on survival rates. We included 23 trials reporting data on 2,767 patients with advanced gastric cancer. Overall, we noted that patients who received IPC had a significantly increased 1-year survival rate, and the treatment effect of IPC on 1-year survival was most prominent in studies conducted in Japan or those with a mean age of less than 60 years. IPC was also associated with an increased incidence of 2-year survival rate, but it was not seen to have this effect in studies conducted in China or Australia or with a mean age greater than 60 years. Similarly, IPC associated with a significantly increased 3-year survival rate, but this difference was not detected in studies conducted in Austria or with a mean age greater than 60 years. IPC has no significant effect on the 5-year survival rate. Finally, IPC was associated with a lower risk of recurrence in patients with advanced gastric cancer. The findings of this study suggest that gastric cancer patients who receive IPC associate with increased 1-year, 2-year, and 3-year survival rates, but this does not extend out to a 5-year survival rate. IPC is also shown to play a protective role against the risk of recurrence in patients with advanced gastric cancer.
Collapse
Affiliation(s)
- Zheng He
- Department of Radiation Oncology, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ting-Ting Zhao
- Department of Breast Surgery, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Hui-Mian Xu
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhen-Ning Wang
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ying-Ying Xu
- Department of Breast Surgery, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yong-Xi Song
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhong-Ran Ni
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, Liaoning Province, China.,School of Life Science, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Hao Xu
- Department of Medical Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Song-Cheng Yin
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xing-Yu Liu
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhi-Feng Miao
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
27
|
Kurokawa Y, Sasako M. The Asian Perspective on the Surgical and Adjuvant Management of Esophagogastric Cancer. Surg Oncol Clin N Am 2017; 26:213-224. [PMID: 28279465 DOI: 10.1016/j.soc.2016.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In East Asia, D2 dissection has been routine surgical procedure for curable advanced gastric cancer. More extended surgery than D2 is reserved for borderline resectable disease with extended nodal metastasis. The addition of radiation therapy to adjuvant chemotherapy failed to improve the outcome after D2 dissection. Because many patients are diagnosed in East Asia with early-stage disease, postoperative adjuvant chemotherapy is preferred, and S-1 monotherapy or capecitabine-oxaliplatin is standard care. Neoadjuvant chemotherapy may be preferred for stage III tumors; for borderline resectable tumors, preoperative chemotherapy is under study given the limitations of postoperative adjuvant chemotherapy in high-risk patients.
Collapse
Affiliation(s)
- Yukinori Kurokawa
- Department of Gastrointestinal Surgery, Osaka University Graduate School of Medicine, 2-2, Yamadagaoka, Suita, Osaka Prefecture 565-0871, Japan
| | - Mitsuru Sasako
- Department of Multidisciplinary Surgical Oncology, Hyogo College of Medicine, 1-1, Mukogawacho, Nishinomiya, Hyogo Prefecture 663-8501, Japan.
| |
Collapse
|
28
|
Kobayashi D, Kodera Y. Intraperitoneal chemotherapy for gastric cancer with peritoneal metastasis. Gastric Cancer 2017; 20:111-121. [PMID: 27803990 DOI: 10.1007/s10120-016-0662-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/15/2016] [Indexed: 02/07/2023]
Abstract
Peritoneal metastasis is the most frequent pattern of gastric cancer recurrence or metastasis and is a definitive determinant of prognosis. However, an effective means of treating peritoneal disease has not yet been established. Systemic chemotherapy has only a limited effect on peritoneal metastasis, although some progress has been shown in terms of median survival time, especially among patients with a minimal or moderate disease burden. Clinical research related to intraperitoneal administration of anticancer drugs is currently underway. An advantage of intraperitoneal chemotherapy is the ability to achieve high concentrations of anticancer drugs in the peritoneal cavity and the direct exposure of peritoneal deposits and free cancer cells to those drugs. In addition, pharmacokinetic studies with taxanes have shown that these high intraperitoneal drug concentrations are sustained for a considerable length of time, allowing prolonged exposure. As taxanes are the most appropriate drugs for intraperitoneal administration, the development of repeated intraperitoneal chemotherapy using taxanes for gastric cancer peritoneal metastasis-either alone or in combination with systemic chemotherapy-has taken place over the past decade, mostly in Japan. Several phase II trials and a phase III trial have recently demonstrated the efficacy of this therapy, including median survival times of 14.4-24.6 months and one-year overall survival rates of 67-91%. These results may lead to the approval of intraperitoneal taxanes, especially paclitaxel, for official insurance coverage in the near future.
Collapse
Affiliation(s)
- Daisuke Kobayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
29
|
Kodera Y. Neoadjuvant chemotherapy for gastric adenocarcinoma in Japan. Surg Today 2017; 47:899-907. [PMID: 28247105 DOI: 10.1007/s00595-017-1473-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/21/2016] [Indexed: 12/23/2022]
Abstract
Surgery had been and remains a mainstay in the treatment of gastric cancer. The Japanese surgical oncologists employed surgery-first approach to treat gastric cancer because of the widespread use of D2 lymph node dissection and the high incidence of oncologically resectable cancer, and early attempts at the multimodality treatment strategy featured surgery followed by postoperative chemotherapy. Although evidence to treat Stage II/III gastric cancer with this strategy is now abundant in the Far East, poor compliance of the post-gastrectomy patients to intense combination chemotherapies has been a limitation associated with this strategy. Evidence in support of neoadjuvant chemotherapy in the West and in various other types of cancer prompted the Japan Clinical Oncology Group (JCOG) researchers to explore this strategy, primarily for a selected population of locally advanced cancer that could either be unresectable by the surgery-first approach or is known to suffer from a poor prognosis; cancers with bulky lymph node metastases or those with a scirrhous phenotype. Encouraged by some promising results from these neoadjuvant trials and taking into account the aforementioned limitations associated with postoperative chemotherapy, the JCOG researchers decided to embark on a phase III trial to explore neoadjuvant chemotherapy among patients with clinically Stage III cancer. This review describes the development of the neoadjuvant strategy for gastric cancer in Japan, mainly by going through a series of clinical trials conducted by the JCOG.
Collapse
Affiliation(s)
- Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
30
|
Kodera Y, Takahashi N, Yoshikawa T, Takiguchi N, Fujitani K, Ito Y, Miyamoto K, Takayama O, Imano M, Kobayashi D, Miyashita Y, Morita S, Sakamoto J. Feasibility of weekly intraperitoneal versus intravenous paclitaxel therapy delivered from the day of radical surgery for gastric cancer: a preliminary safety analysis of the INPACT study, a randomized controlled trial. Gastric Cancer 2017; 20:190-199. [PMID: 26879545 DOI: 10.1007/s10120-016-0598-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 01/25/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Peritoneal carcinomatosis is common after curative resection of gastric cancer. Intraperitoneal administration of paclitaxel (PTX) is known to control ovarian peritoneal metastases. PATIENTS AND METHODS Patients with either linitis plastica or T4 cancer with high risk of peritoneal metastasis or recurrence but whose cancer was considered resectable were preregistered. After their cancer had been confirmed intraoperatively as resectable, the patients were randomized into either group A (PTX at 60 mg/m2 intraperitoneally on the day of surgery and on days 14, 21, 28, 42, 49, and 56) or group B (PTX at 80 mg/m2 administered intravenously by the identical schedule) before being treated by evidence-based chemotherapy. The primary end point was the 2-year survival rate. Safety, the secondary end point, was also analyzed. The study has been registered as UMIN000002957. RESULTS Of 177 preregistered patients, 83 underwent treatment (39 by intraperitoneal administration and 44 by intravenous administration). There was no difference in patient demographics between the two groups. The incidences of surgical complications were similar between the groups, except for transient bowel obstruction observed exclusively in group A. The relative dose intensity of PTX was 81.4 % for group A and 76.3 % for group B. There was one death due to pulmonary thrombosis and a case of anaphylaxis that led to termination of the protocol treatment (group B). Other adverse events were mild and manageable. CONCLUSIONS Intraperitoneal administration of PTX from the day of gastrectomy did not result in a higher incidence of surgical complications and adverse reactions when compared with intravenous administration of PTX.
Collapse
Affiliation(s)
- Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan.
| | - Naoto Takahashi
- Department of Surgery, Jikei University School of Medicine, Kashiwa Hospital, Chiba, Japan
| | - Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Nobuhiro Takiguchi
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | | | - Yuichi Ito
- Department of Gastroenterological Surgery, Aichi Cancer Center, Nagoya, Japan
| | - Katsufumi Miyamoto
- Department of Surgery, Hyogo Prefectural Awaji Medical Center, Sumoto, Japan
| | - Osamu Takayama
- Department of Surgery, Itami City Hospital, Itami, Japan
| | - Motohiro Imano
- Department of Surgery, Faculty of Medicine, Kinki University, Higashiosaka, Japan
| | - Daisuke Kobayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Yumi Miyashita
- Data Center, Epidemiological and Clinical Research Information Network, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
Collapse
|
31
|
Feingold PL, Kwong MLM, Davis JL, Rudloff U. Adjuvant intraperitoneal chemotherapy for the treatment of gastric cancer at risk for peritoneal carcinomatosis: A systematic review. J Surg Oncol 2016; 115:192-201. [PMID: 27878811 DOI: 10.1002/jso.24476] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/02/2016] [Indexed: 12/29/2022]
Abstract
The peritoneal surface is a frequent site of recurrence following surgery for gastric cancer. A systematic review and random effect analysis was undertaken to analyze current literature regarding the role of adjuvant intraperitoneal chemotherapy in gastric cancer. While pooled analysis supports the use of adjuvant IP chemotherapy in resectable gastric cancer, maximal benefit occured with intra-operative delivery, and possibly the use of MMC. J. Surg. Oncol. 2017;115:192-201. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Paul L Feingold
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mei Li M Kwong
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jeremy L Davis
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Udo Rudloff
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
32
|
Coccolini F, Celotti A, Ceresoli M, Montori G, Marini M, Catena F, Ansaloni L. Hyperthermic intraperitoneal chemotherapy (HIPEC) and neoadjuvant chemotherapy as prophylaxis of peritoneal carcinosis from advanced gastric cancer-effects on overall and disease free survival. J Gastrointest Oncol 2016; 7:523-9. [PMID: 27563441 DOI: 10.21037/jgo.2016.06.05] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The possibility to enlarge criteria for intra-peritoneal chemotherapy (IPC) to all patients at high-risk to develop peritoneal carcinosis (i.e., with serosal invasion) is still discussed. METHODS Retrospective case-control study. Three-groups: advanced-gastric-cancer (AGC) (pT4) without proven carcinosis: prophylactic group (PG), those with PC: treatment group (TG), AGC (pT3-pT4) operated without hyperthermic intraperitoneal chemotherapy (HIPEC), surgery alone group (SG T3, SG T4). RESULTS Forty four patients. 26 (59.1%) were male. Sixteen (36%) patients underwent 16 HIPEC: 6 (38%) had AGC (pT4) without PC (PG), 10 (62%) had carcinosis (TG), 28 were operated without HIPEC (SG T3, SG T4). The mean disease free survival (DFS): TG: 7.7 months, SG T4: 21.6 months, SG T3: 27.7 months, PG: 34.5 months. DFS was significantly different for TG (P=0.03, P=0.021, P=0.013 respectively). The mean OS TG: 10 months, SG T4: 27.1 months, SG T3: 28.2 months, PG: 34.6 months. OS was significantly different for TG (P=0.04, P=0.04, P=0.045 respectively). Severe complication rate: TG: 60%, PG: 16.7%, SG T3: 7.7% and SG T4: 25% (P=0.035). Length-of-stay differs significantly (P=0.003); overall length-of-stay: 19.41 days [standard deviation (SD) ±15.03]; TG: 33.01 (SD ±23.08), PG: 20.17 (SD ±6.21), SG T3: 11.33 (SD ±3.22), SG T4: 15.36 (SD ±5.48). CONCLUSIONS Prophylactic intraperitoneal chemotherapy associated to neoadjuvant chemotherapy increases the DFS and OS in patients with AGC without carcinosis. More data are needed in order to confirm these results.
Collapse
Affiliation(s)
- Federico Coccolini
- General Surgery Department, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Andrea Celotti
- General Surgery Department, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Marco Ceresoli
- General Surgery Department, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Giulia Montori
- General Surgery Department, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Michele Marini
- General Surgery Department, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Fausto Catena
- General and Emergency surgery, Parma Maggiore Hospital, Parma, Italy
| | - Luca Ansaloni
- General Surgery Department, Papa Giovanni XXIII Hospital, Bergamo, Italy
| |
Collapse
|
33
|
Kilic L, Ordu C, Yildiz I, Sen F, Keskin S, Ciftci R, Pilanci KN. Current adjuvant treatment modalities for gastric cancer: From history to the future. World J Gastrointest Oncol 2016; 8:439-449. [PMID: 27190583 PMCID: PMC4865711 DOI: 10.4251/wjgo.v8.i5.439] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 01/24/2016] [Accepted: 02/24/2016] [Indexed: 02/05/2023] Open
Abstract
The discrepancy between the surgical technique and the type of adjuvant chemotherapy used in clinical trials and patient outcomes in terms of overall survival rates has led to the generation of different adjuvant treatment protocols in distinct parts of the world. The adjuvant treatment recommendation is generally chemoradiotherapy in the United States, perioperative chemotherapy in the United Kingdom and parts of Europe, and chemotherapy in Asia. These options mainly rely on the United States Intergroup-0116, United Kingdom British Medical Research Council Adjuvant Gastric Infusional Chemotherapy, and the Asian Adjuvant Chemotherapy Trial of S-1 for Gastric Cancer and Capecitabine and Oxaliplatin Adjuvant Study in Stomach Cancer trials. However, the benefits were evident for only certain patients, which were not very homogeneous regarding the type of surgery, chemotherapy regimens, and stage of disease. Whether the dissimilarities in survival are attributable to surgical technique or intrinsic biological differences is a subject of debate. Regardless of the extent of surgery, multimodal therapy may offer modest survival advantage at least for diseases with lymph node involvement. Moreover, in the era of individualized treatment for most of the other cancer types, identification of special subgroups comprising those who will derive more or no benefit from adjuvant therapy merits further investigation. The aim of this review is to reveal the historical evolution and future reflections of adjuvant treatment modalities for resected gastric cancer patients.
Collapse
|
34
|
Zhang T, Pan Q, Xiao S, Li L, Xue M. Docetaxel combined with intraperitoneal hyperthermic perfusion chemotherapy and hyperthermia in the treatment of advanced ovarian cancer. Oncol Lett 2016; 11:3287-3292. [PMID: 27123104 PMCID: PMC4840847 DOI: 10.3892/ol.2016.4414] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/04/2016] [Indexed: 01/10/2023] Open
Abstract
Ovarian cancer is a clinical type of gynecological malignant tumor with poor prognosis and a high mortality rate. At present, the primary treatment method used is surgery, with chemotherapy as an ajdunctive therapy. Thus, new short-term treatments should be identified. The aim of the present study was to investigate the short-term curative effects and safety of docetaxel combined with intraperitoneal cisplatin chemotherapy and hyperthermia treatment of advanced ovarian cancer. A total of 112 cases of advanced (stage III–IV) ovarian cancer patients confirmed by clinical diagnosis between October 2014 and December 2015 were included in the study. The patients were randomly divided into the study and control groups (n=56 cases). The control group was treated with docetaxel and intraperitoneal cisplatin hyperthermic perfusion chemotherapy, while the study group was treated with docetaxel venous chemotherapy and intraperitoneal cisplatin cyclical hyperthermic perfusion chemotherapy with BR-TRG-1 body cavity hyperthermic perfusion treatment system. Clinical treatment results for short-term curative effects and adverse reactions were compared and analyzed 8 weeks after treatment. The total effective rate of the study and control groups were 87.5 and 62.5%, respectively, and the difference was statistically significant (P<0.05). The controlled rate of ascites, remission rate of tumor and descent rate of CA125 of patients in the study group were better than patients in the control group (P<0.05). The rate of adverse reactions of patients in the study group was 39.3%, and the grade of toxicity was from I to II, while the rate of adverse reactions of patients in the control group was 55.4%, and the grade of toxicity was from II to III. The difference between the two groups was statistically significant (P<0.05). In conclusion, applying the combination of docetaxel, intraperitoneal cisplatin hyperthermic perfusion chemotherapy and hyperthermia to treat advanced ovarian cancer can improve the curative effects with little toxicity; thus, it is worthy of promotion and application.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Qiong Pan
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Songshu Xiao
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Lijie Li
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Min Xue
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
35
|
Wang ZX, Yang XL, He MM, Wang F, Zhang DS, Li YH, Zhou ZW, Zhan YQ, Xu RH. The Efficacy of Adjuvant FOLFOX6 for Patients With Gastric Cancer after D2 Lymphadenectomy: A Propensity Score-matched Analysis. Medicine (Baltimore) 2016; 95:e3214. [PMID: 27100411 PMCID: PMC4845815 DOI: 10.1097/md.0000000000003214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Adjuvant 5-fluorouracil, folinic acid, and oxaliplatin (FOLFOX6) are widely used for treating resected gastric cancer in clinics in China, but only few clinical trials have investigated its efficacy. Using propensity score matching, we evaluated the efficacy of adjuvant FOLFOX6 following D2 lymphadenectomy. Patients who received adjuvant FOLFOX6 following D2 lymphadenectomy (FOLFOX6, n = 113) or D2 lymphadenectomy only (surgery-only, n = 512) between 1998 and 2007 at our center were propensity score-matched; we identified a balanced 1:2 cohort, with 96 patients in the FOLFOX6 group and 192 patients in the surgery-only group. The overall survival (OS) was estimated using the Kaplan-Meier method; factors affecting survival were identified by Cox regression models. A nomogram incorporating independent prognosticators was constructed for predicting the 3-, 5-, and 7-year OS, and bootstrap validation was performed. The median follow-up was 9.3 years, and the 7-year OS was 52.1% in the FOLFOX6 group and 43.8% in the surgery-only group (P = 0.04), with an adjusted hazard ratio of 0.69 (95% confidence interval = 0.49-0.98). A prognostic nomogram was generated with the identified significant prognosticators (adjuvant FOLFOX6, number of total harvested nodes, the interaction effect between these two variables, tumor size, T and N stage). Internal validation of the nomogram revealed good predictive abilities, with a bootstrap-corrected concordance index of 0.70. Adjuvant FOLFOX6 following D2 lymphadenectomy is associated with survival benefit in resected gastric cancer. Receiving adjuvant FOLFOX6 can be developed into a nomogram with other independent prognosticators to refine OS prediction and estimation of benefit from adjuvant FOLFOX6 for resected gastric cancer.
Collapse
Affiliation(s)
- Zi-Xian Wang
- From the Department of Medical Oncology (Z-XW, M-MH, FW, D-SZ, Y-HL, R-HX); Faculty of Medical Sciences, (Z-XW, X-LY); and Department of Gastric and Pancreatic Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China (Z-WZ, Y-QZ)
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Feingold PL, Kwong MLM, Sabesan A, Sorber R, Rudloff U. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for gastric cancer and other less common disease histologies: is it time? J Gastrointest Oncol 2016; 7:87-98. [PMID: 26941987 DOI: 10.3978/j.issn.2078-6891.2015.098] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the fourth most commonly diagnosed cancer worldwide, and once spread to the peritoneum, has a 5-year survival of less than 5%. Recent years have demonstrated advances in the use of cytoreductive surgery (CRS) in combination with heated intraperitoneal chemotherapy (HIPEC) for the treatment of peritoneal carcinomatosis due to various malignancies. The frequent desmoplastic stroma and poor vascularization impeding drug delivery particularly in the diffuse form of gastric cancer is thought to provide a sound rationale for a regionalized treatment approach in this disease. Here, we seek to review the available data to define the role of CRS and HIPEC in gastric cancer metastatic to the peritoneal surface, and furthermore, analyze the use of CRS and HIPEC in malignancies less commonly treated with the regionalized perfusion approach.
Collapse
Affiliation(s)
- Paul L Feingold
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mei Li M Kwong
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arvind Sabesan
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca Sorber
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Udo Rudloff
- Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
|
38
|
Peng YF, Imano M, Itoh T, Satoh T, Chiba Y, Imamoto H, Tsubaki M, Nishida S, Yasuda T, Furukawa H. A phase II trial of perioperative chemotherapy involving a single intraperitoneal administration of paclitaxel followed by sequential S-1 plus intravenous paclitaxel for serosa-positive gastric cancer. J Surg Oncol 2015; 111:1041-6. [PMID: 26060133 DOI: 10.1002/jso.23928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/06/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVES We carried out a phase II trial to evaluate the feasibility, efficacy, and tolerability of perioperative chemotherapy including single intraperitoneal(IP) administration of paclitaxel(PTX) followed by intravenous(IV) administrations of PTX with S-1 in a neoadjuvant setting for serosa-positive gastric cancer. METHODS Patients with cT4a gastric cancer were enrolled. A laparoscopic survey was performed before study inclusion for the confirmation of serosal invasion, negative lavage cytology, and negative peritoneal metastasis. IP PTX (80 mg/m(2)) was administered, followed by systemic chemotherapy. Surgery was performed after the completion of chemotherapy. The primary endpoint was the treatment completion rate. RESULTS 37 patients were recruited. The treatment completion rate was 67.6% (25/37; 90% CI, 52.8-80.1%), which was significantly higher than 50%; we set this as a threshold value (P = 2.4% [one-sided]). 14 patients had target lesions; of these, 10 showed a partial response (71.4%), three had stable disease (21.4%), and one had progressive disease(7.2%). The response rate was 71.4% (10/14). All patients underwent gastrectomy with D2 lymph node dissection. The 3- and 5-year OS rates were 78.0 and 74.9%, respectively. CONCLUSIONS Perioperative chemotherapy including neoadjuvant IP PTX followed by sequential IV PTX with S-1 for serosa-positive gastric cancer is feasible, safe, and efficient.
Collapse
Affiliation(s)
- Ying-Feng Peng
- Department of Surgery, Kinki University, Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Motohiro Imano
- Department of Surgery, Kinki University, Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Tatsuki Itoh
- Department of Pathology, Kinki University, Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Takao Satoh
- Department of Pathology, Kinki University, Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Yasutaka Chiba
- Clinical Research Center, Kinki University Hospital, Osaka-sayama, Osaka, Japan
| | - Haruhiko Imamoto
- Department of Surgery, Kinki University, Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Masahiro Tsubaki
- Division of Pharmacotherapy, Kinki University, Faculty of Pharmacy, Higashi-osaka, Osaka, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kinki University, Faculty of Pharmacy, Higashi-osaka, Osaka, Japan
| | - Takushi Yasuda
- Department of Surgery, Kinki University, Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Hiroshi Furukawa
- Department of Surgery, Kinki University, Faculty of Medicine, Osaka-sayama, Osaka, Japan
| |
Collapse
|
39
|
Wang J, Sun Y, Bertagnolli MM. Comparison of gastric cancer survival between Caucasian and Asian patients treated in the United States: results from the Surveillance Epidemiology and End Results (SEER) database. Ann Surg Oncol 2015; 22:2965-71. [PMID: 25631065 DOI: 10.1245/s10434-015-4388-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prognosis for gastric cancer is better for Asian than for Caucasian patients. The primary driver of this difference is unknown. This study determined whether the survival advantage of Asian ethnicity continued to hold after control was used for other well-known prognostic factors. METHODS In this study, 12,773 patients who underwent gastrectomy for treatment of adenocarcinoma of the stomach were identified from the Surveillance, Epidemiology, and End Results cancer registry. Patients with cardia tumor were excluded from the study. The independent prognostic effect of ethnicity was evaluated by adjusting for other known factors. RESULTS The Asian patients tended to have a diagnosis at an earlier age (66.8 vs. 68.5 years), more lymph nodes examined (16 vs. 13), and more positive lymph nodes (5.1 vs. 4.8). Survival was better for the Asian patients than for the Caucasian patients, with a 12 % 5-year survival difference. Among the patients with IB, IIA, and IIB disease, the Asian patients had 37, 72, and 13 months longer median survival time than the corresponding Caucasian patients. The multivariate Cox model showed persistence of this result after adjustment for imbalances of age, gender, tumor grade, and number of examined and positive lymph nodes. The largest risk reduction was observed for the stage IA patients (31 %) and the smallest for the stage IIIC patients (9 %). CONCLUSION After excluding proximal gastric cancers, controlling for the imbalance of known prognostic factors, and decreasing in the influence of D2 lymphadenectomy, stage migration, and chemo/radiation therapy by including only patients treated in the United States, this study found that the survival advantage of Asian ethnicity continued to be present.
Collapse
Affiliation(s)
- Jiping Wang
- Division of Surgical Oncology, Brigham and Women's Hospital, Boston, MA, USA,
| | | | | |
Collapse
|
40
|
Markar SR, Wiggins T, Ni M, Steyerberg EW, Van Lanschot JJB, Sasako M, Hanna GB. Assessment of the quality of surgery within randomised controlled trials for the treatment of gastro-oesophageal cancer: a systematic review. Lancet Oncol 2015; 16:e23-31. [DOI: 10.1016/s1470-2045(14)70419-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
41
|
Nakajima T, Fujii M. What make differences in the outcome of adjuvant treatments for resected gastric cancer? World J Gastroenterol 2014; 20:11567-11573. [PMID: 25206264 PMCID: PMC4155350 DOI: 10.3748/wjg.v20.i33.11567] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/09/2014] [Accepted: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
After a long history of Dark Age of adjuvant chemotherapy for gastric cancer, definite evidences of survival benefit from adjuvant treatment have been reported since 2000s. These survival benefits are likely attributed to something new approach different from pervious studies. In 2001, South West Oncology Group INT0116 trial yielded survival benefit in curatively resected gastric cancer patients with postoperative chemoradiotherapy [5-fluorouracil (5-FU) + Leucovorin + radiotherapy], followed by positive result by MAGIC Trial, employing peri-operative(pre- and postoperative chemotherapy with Epirubicin, cisplatin (CDDP), 5-fluorouracil (ECF) regimen in patients with curative resection. A novel drug [S1: ACTS-GC (Adjuvant chemotherapy trial of TS-1 for gastric cancer) in 2007], or new drug combination chemotherapys [CDDP + 5-FU: FNCLCC/FFCD (Federation Nationale des Centres de Lutte contre le cancer/Federation Francophone de Cancerologie Digestive) in 2011, Capecitabine + Oxaliplatin: CLASSIC in 2012] also produced positive results in terms of improved prognosis. Neoadjuvant or perioperative chemotherapy, novel anti-cancer drugs, and chemoradiotherapy might be the key words to develop further improvement in the adjuvant treatment of resectable gastric cancer. Moreover, it is not new but still true to stress the importance of D2 surgery as the baseline treatment in order to minimize the amount of residual tumor after surgery.
Collapse
|
42
|
Cao J, Qi F, Liu T. Adjuvant chemotherapy after curative resection for gastric cancer: a meta-analysis. Scand J Gastroenterol 2014; 49:690-704. [PMID: 24731211 DOI: 10.3109/00365521.2014.907337] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The aim of this article is to review up-to-date clinical data published in the literature in regard to adjuvant chemotherapy in patients with gastric cancer after radical surgical resection. MATERIALS AND METHODS Medline, Embase, PubMed, the Cochrane Library and CBMDisc were searched to identify data published regarding this issue from 1966 to 2013. All the calculations and statistical tests were done using RevMan5.2 software. RESULTS A total of 29 trials with 8580 patients met all inclusion criteria. Among them, 27 studies reported survival rates at the end of follow-ups, 64.2% alive among 3981 patients in the adjuvant chemotherapy arm and 57.3% alive among 4027 patients in the observation arm. Statistical results showed that the observation arm had a shorter disease-free survival (RR: 1.11, 95% CI: 1.07-1.15), and the treatment arm had a lower recurrence rate (RR: 0.79, 95% CI: 0.74-0.84). Leucopenia, anemia, nausea and vomiting, diarrhea, alopecia and infection occurred more frequently in the treatment arm. Adjuvant chemotherapy decreased the occurrence of peritoneum relapse [RR = 0.77, 95% CI (0.66-0.90)], lymphoid nodes relapse [RR = 0.58, 95% CI (0.45-0.75)] and local relapse [RR = 0.57, 95% CI (0.41-0.80)]. CONCLUSIONS Adjuvant chemotherapy can improve the survival rate and disease-free survival rate and reduce the relapse rate after curative resection. Adjuvant chemotherapy cannot induce thrombocytopenia and mucositis or affect liver function. The tumor in situ recurrence and peritoneum, lymph nodes relapse decrease after chemotherapy, and patients benefit from adjuvant chemotherapy regardless of the numbers of positive lymph node, depth of local invasion, Asian or non-Asian, the length of follow-up, and numbers of cycles.
Collapse
Affiliation(s)
- Jisen Cao
- Department of General Surgery, Tianjin Medical University General Hospital , Tianjin , China
| | | | | |
Collapse
|
43
|
Zhang YW, Zhang YL, Pan H, Wei FX, Zhang YC, Shao Y, Han W, Liu HP, Wang ZY, Yang SH. Chemotherapy for patients with gastric cancer after complete resection: A network meta-analysis. World J Gastroenterol 2014; 20:584-592. [PMID: 24574729 PMCID: PMC3923035 DOI: 10.3748/wjg.v20.i2.584] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/15/2013] [Accepted: 11/03/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To conduct a network meta-analysis to evaluate the effectiveness of different chemotherapy regimens for patients with gastric cancer.
METHODS: PubMed (1966-2011.12), the Cochrane Library (2011 Issue 2) and EMBASE (1974-2011.12) were searched with the terms “gastric cancer” and “chemotherapy”, as well as the medical subject headings. References from relevant articles and conferences were also included. Patients who had previous gastric surgery, radiation before or after surgery or chemotherapy before surgery were excluded. In this study, only randomized controlled trials (RCTs) were considered, and the end-point was the overall mortality. Direct comparisons were performed using traditional meta-analysis whereas indirect comparisons were performed using network meta-analysis.
RESULTS: In total, 31 RCTs with 7120 patients were included. Five chemotherapy regimens, fluorouracil (FU) + BCNU, FU + methyl-CCNU (mCCNU), FU + cisplatin, FU + anthracyclines and FU + mitomycin c (MMC) + cytarabine (Ara-c), were found to be less beneficial in terms of overall mortality. In contrast, four chemotherapy regimens were effective for the patients after surgery, including FU + MMC + adriamycin (FMA), FU + MMC (FM), Tegafur and MMC, There was no significant difference in terms of overall mortality among these regimens. The evidence for the FM regimen and MMC regimen was poor. Additionally, the FMA regimen, which includes a variety of chemotherapy drugs and causes many side effects, was not better than the Tegafur regimen.
CONCLUSION: Although the four chemotherapy regimens were effective in patients with gastric cancer after surgery and the overall mortality revealed no significant difference among them in the network meta-analysis, thorough analysis of the results recommends Tegafur as the first-line adjuvant chemotherapy regimen for patients after complete resection.
Collapse
|
44
|
Coccolini F, Cotte E, Glehen O, Lotti M, Poiasina E, Catena F, Yonemura Y, Ansaloni L. Intraperitoneal chemotherapy in advanced gastric cancer. Meta-analysis of randomized trials. Eur J Surg Oncol 2013; 40:12-26. [PMID: 24290371 DOI: 10.1016/j.ejso.2013.10.019] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/20/2013] [Accepted: 10/23/2013] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION An important component of treatment failure in gastric cancer (GC) is cancer dissemination within the peritoneal cavity and nodal metastasis. Intraperitoneal chemotherapy (IPC) is considered to give a fundamental contribute in treating advanced GC. The purpose of the study is to investigate the effects of IPC in patients with advanced GC. MATERIAL AND METHODS A systematic review with meta-analysis of randomized controlled trials (RCTs) of IPC + surgery vs. control in patients with advanced GC was performed. RESULTS Twenty prospective RCTs have been included (2145 patients: 1152 into surgery + IPC arm and 993 into control arm). Surgery + IPC improves: 1, 2 and 3-year mortality (OR = 0.31, 0.27, 0.29 respectively), 2 and 3-year mortality in patients with loco-regional nodal metastasis (OR = 0.28, 0.16 respectively), 1 and 2-year mortality rate in patients with serosal infiltration (OR = 0.33, 0.27 respectively). Morbidity rate was increased by surgery + IPC (OR = 1.82). The overall recurrence and the peritoneal recurrence rates were improved by surgery + IPC (OR = 0.46 and 0.47 respectively). There was no statistically significant difference in lymph-nodal recurrence rate. The rate of haematogenous metastasis was improved by surgery + IPC (OR = 0.63). CONCLUSIONS 1, 2 and 3-year overall survival is incremented by the IPC. No differences have been found at 5-year in overall survival rate. 2 and 3-year mortality rates in patients with nodal invasion and 1 and 2-year mortality rates in patients with serosal infiltration are improved by the use of IPC. IPC has positive effect on peritoneal recurrence and distant metastasis. Morbidity rate is incremented by IPC. Loco-regional lymph-nodes invasion in patients affected by advanced gastric cancer is not a contraindication to IPC.
Collapse
Affiliation(s)
- F Coccolini
- General Surgery Dept., Papa Giovanni XXIII Hospital, Bergamo, Italy; General Surgery Dept., Centre Hospitalier Lyon Sud, Hospices Civils de Lyon and EMR 3738, Université Lyon 1, France.
| | - E Cotte
- General Surgery Dept., Centre Hospitalier Lyon Sud, Hospices Civils de Lyon and EMR 3738, Université Lyon 1, France
| | - O Glehen
- General Surgery Dept., Centre Hospitalier Lyon Sud, Hospices Civils de Lyon and EMR 3738, Université Lyon 1, France
| | - M Lotti
- General Surgery Dept., Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - E Poiasina
- General Surgery Dept., Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - F Catena
- General Surgery Dept., Ospedale Maggiore, Parma, Italy
| | - Y Yonemura
- General Surgery Dept., Kusatsu General Hospital, Yabase 1660, Japan
| | - L Ansaloni
- General Surgery Dept., Papa Giovanni XXIII Hospital, Bergamo, Italy
| |
Collapse
|
45
|
Kang YK, Yook JH, Chang HM, Ryu MH, Yoo C, Zang DY, Lee JL, Kim TW, Yang DH, Jang SJ, Park YS, Lee YJ, Jung HY, Kim JH, Kim BS. Enhanced efficacy of postoperative adjuvant chemotherapy in advanced gastric cancer: results from a phase 3 randomized trial (AMC0101). Cancer Chemother Pharmacol 2013; 73:139-49. [PMID: 24162381 DOI: 10.1007/s00280-013-2332-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 10/17/2013] [Indexed: 12/21/2022]
Abstract
PURPOSE To improve the efficacy of adjuvant chemotherapy with mitomycin-C and fluoropyrimidine (Mf) in gastric cancer, we designed a new regimen (iceMFP) and investigated in a phase III study. METHODS We randomly assigned 640 patients with resectable and macroscopically recognizable serosa-invading gastric cancer to Mf or iceMFP group during operation. The Mf consisted of intravenous mitomycin-C (20 mg/m(2)) at 3-6 weeks after surgery and oral doxifluridine (460-600 mg/m(2)/day) starting 4 weeks after the administration of mitomycin-C and continuing for 3 months. The iceMFP consisted of intraoperative intraperitoneal cisplatin (100 mg), intravenous mitomycin-C (15 mg/m(2)) on postoperative day 1, followed by oral doxifluridine for 12 months, and six monthly intravenous cisplatin (60 mg/m(2)). The primary endpoint was 3-year recurrence-free survival (RFS). RESULTS A total of 521 patients (258 in Mf, 263 in iceMFP) were eligible for analysis after excluding patients with stage I disease, distant metastasis, or R1 resection. With a median follow-up of 3.5 years, the iceMFP group had a higher RFS (hazard ratio [HR] 0.70; 95 % confidence interval [CI] 0.54-0.90; p = 0.006; 3-year RFS 60 % vs. 50 %) and overall survival (HR 0.71; 95 % CI 0.53-0.95; p = 0.02; 3-year overall survival, 71 vs. 60 %) compared with the Mf group. This was confirmed at extension analysis after a median 6.6 years of follow-up. Both regimens were well tolerated with no differences in surgical complications. CONCLUSION The efficacy of adjuvant Mf was significantly improved by the additional therapeutic strategies of iceMFP. Considering negative results of AMC0201, these suggest that early initiation of chemotherapy and/or intraperitoneal cisplatin played a distinct role in the improved efficacy.
Collapse
Affiliation(s)
- Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 138-736, Korea,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
This review presents the current status of adjuvant and neoadjuvant treatment options for primary resectable gastric cancer in the East, with updated data from recent studies. Marked disparities between the East and the West in standard surgical procedures (D2 vs. D1/0 lymphadenectomy) and their outcomes result in significant geographical variation in preferred adjuvant treatments. Currently, oral fluoropyrimidine-based postoperative adjuvant chemotherapy, 1 year of S-1 chemotherapy, or capecitabine plus oxaliplatin for 6 months are the standards of care after curative resection with D2 lymphadenectomy for stage II/III gastric cancer in the East, though there is still some room for improvement. The role of postoperative adjuvant chemoradiotherapy (CRT) following curative D2 gastrectomy has long been debated in the East. However, the first prospective randomized controlled trial comparing CRT with chemotherapy alone failed to demonstrate a survival benefit, thus further studies are required. Chemotherapy has been pursued as a neoadjuvant approach in East Asia because of a rare locoregional recurrence after curative D2 gastrectomy. Locally advanced, marginally resectable gastric cancer with poor prognosis, such as large type 3 or 4 tumors, para-aortic and/or bulky nodal disease, and serosa-positive gastric cancer, is the main target of neoadjuvant chemotherapy. Promising efficacy has been demonstrated in several phase II studies with the safe use of D2 or more extended surgery following neoadjuvant chemotherapy. Although the results of ongoing phase III trials are awaited, Asian findings could be relevant and generalizable to other regions when D2 surgery is performed by experienced surgeons.
Collapse
Affiliation(s)
- Kazumasa Fujitani
- Department of Surgery, Osaka National Hospital, Osaka 540-006, Japan.
| |
Collapse
|
47
|
Abstract
Esophagogastric junction (EGJ) adenocarcinomas are usually classified into one of the three categories of the Siewert system. The clinicopathological features of EGJ adenocarcinomas vary according to this classification scheme. The lymphatic flow in EGJ tumors of any type is mainly toward the abdomen, and nodal metastasis to the upper or middle mediastinum from Siewert type II or III tumors is relatively uncommon. Thus, the transhiatal approach is regarded as the standard in surgery for Siewert type II or III tumors, while the transthoracic approach via a right thoracotomy is recommended for Siewert type I tumors. Chemoradiotherapy followed by surgery is regarded as the standard treatment for resectable cancer of the EGJ in Western countries, but the necessity of adding radiation therapy to preoperative chemotherapy remains unknown. In the East, postoperative adjuvant chemotherapy is the current standard of care since the survival benefit was proven in pivotal randomized trials for stage II/III gastric cancer, including adenocarcinoma of the EGJ.
Collapse
Affiliation(s)
- Yukinori Kurokawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | | |
Collapse
|
48
|
Wu Y, Wei ZW, He YL, Schwarz RE, Smith DD, Xia GK, Zhang CH. Efficacy of adjuvant XELOX and FOLFOX6 chemotherapy after D2 dissection for gastric cancer. World J Gastroenterol 2013; 19:3309-3315. [PMID: 23745033 PMCID: PMC3671083 DOI: 10.3748/wjg.v19.i21.3309] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 04/16/2013] [Accepted: 05/08/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To compare the efficacy of capecitabine and oxaliplatin (XELOX) with 5-fluorouracil, folinic acid and oxaliplatin (FOLFOX6) in gastric cancer patients after D2 dissection.
METHODS: Between May 2004 and June 2010, patients in our gastric cancer database who underwent D2 dissection for gastric cancer at the First Affiliated Hospital of Sun Yat-Sen University were retrospectively analyzed. A total of 896 patients were enrolled into this study according to the established inclusion and exclusion criteria. Of these patients, 214 received the XELOX regimen, 48 received FOLFOX6 therapy and 634 patients underwent surgery only without chemotherapy. Overall survival was compared among the three groups using Cox regression and propensity score matched-pair analyses.
RESULTS: Patients in the XELOX and FOLFOX6 groups were younger at the time of treatment (median age 55.2 years; 51.2 years vs 58.9 years), had more undifferentiated tumors (70.1%; 70.8% vs 61.4%), and more lymph node metastases (80.8%; 83.3% vs 57.7%), respectively. Overall 5-year survival was 57.3% in the XELOX group which was higher than that (47.5%) in the surgery only group (P = 0.062) and that (34.5%) in the FOLFOX6 group (P = 0.022). Multivariate analysis showed that XELOX therapy was an independent prognostic factor (hazard ratio = 0.564, P < 0.001). After propensity score adjustment, XELOX significantly increased overall 5-year survival compared to surgery only (58.2% vs 44.2%, P = 0.025) but not compared to FOLFOX6 therapy (48.5% vs 42.7%, P = 0.685). The incidence of grade 3/4 adverse reactions was similar between the XELOX and FOLFOX6 groups, and more patients suffered from hand-foot syndrome in the XELOX group (P = 0.018).
CONCLUSION: Adjuvant XELOX therapy is associated with better survival in patients after D2 dissection, but does not result in a greater survival benefit compared with FOLFOX6 therapy.
Collapse
|
49
|
Neoadjuvant chemotherapy for gastric cancer in Japan: a standing position by comparing with adjuvant chemotherapy. Surg Today 2013; 44:11-21. [PMID: 23508452 DOI: 10.1007/s00595-013-0529-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 11/05/2012] [Indexed: 02/06/2023]
Abstract
Adjuvant chemotherapy aims to eradicate micrometastatic tumor cells before and after curative surgery. Many Phase III trials have been conducted to study the efficacy of postoperative adjuvant chemotherapy; however, most trials have failed to show any survival benefit because of their low statistical power and/or poor patient compliance. Since 2000, two pivotal Phase III trials, the ACTS-GC and the CLASSIC, have demonstrated the efficacy of postoperative adjuvant chemotherapy following D2 gastrectomy. Although treatment with S-1 for 1 year or combination therapy with capecitabine and oxaliplatin for 6 months is effective, more intensive chemotherapy is necessary to further improve the survival rates. In Europe, two Phase III trials, the MAGIC and the FNCLCC/FFCD, have produced results that strongly suggest that neoadjuvant chemotherapy is beneficial. The advantages of neoadjuvant chemotherapy include a high rate of R0 resection, tumor regression, high compliance and the avoidance of unnecessary surgery. The disadvantage of neoadjuvant chemotherapy is over-diagnosis. In Japan, the Japan Clinical Oncology Group has conducted several clinical trials using neoadjuvant chemotherapy to target extensive nodal disease and/or scirrhous carcinomas. The optimal courses and regimens of neoadjuvant chemotherapy should, therefore, be clarified in the future.
Collapse
|
50
|
Markar SR, Karthikesalingam A, Jackson D, Hanna GB. Long-term survival after gastrectomy for cancer in randomized, controlled oncological trials: comparison between West and East. Ann Surg Oncol 2013; 20:2328-38. [PMID: 23340695 DOI: 10.1245/s10434-012-2862-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND The presence of mixed evidence about the value of lymphadenectomy in gastric cancer surgery coupled with the difference in patients' demographics and tumor stage between the West and East have doubted the needs to standardize surgical techniques in Western clinical practice. The purpose of this study was to compare survival rates between the West and East following gastrectomy in randomized, controlled, oncological trials with appropriate adjustment for confounding variables. METHODS Systematic search revealed 25 trials that have randomization into surgery and chemotherapy versus surgery alone between 1995 and 2012 (n = 7 (East) and n = 18 (West)). End points were 5-year survival and cancer recurrence. RESULTS There was association between gastrectomy performed in the East and improved 5-year survival (pooled odds ratio (OR) 4.83; 95 % confidence interval (CI) 3.27-7.12) and reduced cancer recurrence (pooled OR 0.33; 95 % CI 0.2-0.54). Association of improved 5-year survival with surgery in the East remained when meta-regression adjusted for the effect of age, sex, chemotherapy, tumor depth and nodal status, and gastrectomy type. Association of reduced cancer recurrence also persisted with meta-regression adjusting for age, chemotherapy, nodal status, and gastrectomy type. However, when adjustment for the percentage of patients with tumor depth T1 or 2 was made statistical significance was lost. CONCLUSIONS This analysis shows association between gastrectomy performed in Eastern countries and improved survival. The known difference in surgical techniques between the East and the West is one potential unexamined variable that may be responsible in part for such discrepancy in outcomes.
Collapse
Affiliation(s)
- Sheraz R Markar
- Department of Surgery and Cancer, St. Mary's Hospital, Imperial College, London, UK
| | | | | | | |
Collapse
|